Download as pdf or txt
Download as pdf or txt
You are on page 1of 49

Critical Reviews in Food Science and Nutrition

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/bfsn20

A review of the role of inositols in conditions of


insulin dysregulation and in uncomplicated and
pathological pregnancy

Oliver C. Watkins, Hannah E. J. Yong, Neha Sharma & Shiao-Yng Chan

To cite this article: Oliver C. Watkins, Hannah E. J. Yong, Neha Sharma & Shiao-Yng Chan (2022)
A review of the role of inositols in conditions of insulin dysregulation and in uncomplicated and
pathological pregnancy, Critical Reviews in Food Science and Nutrition, 62:6, 1626-1673, DOI:
10.1080/10408398.2020.1845604

To link to this article: https://doi.org/10.1080/10408398.2020.1845604

© 2020 The Author(s). Published with View supplementary material


license by Taylor & Francis Group, LLC.

Published online: 07 Dec 2020. Submit your article to this journal

Article views: 5205 View related articles

View Crossmark data Citing articles: 13 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=bfsn20
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION
2022, VOL. 62, NO. 6, 1626–1673
https://doi.org/10.1080/10408398.2020.1845604

REVIEW

A review of the role of inositols in conditions of insulin dysregulation and in


uncomplicated and pathological pregnancy
Oliver C. Watkinsa , Hannah E. J. Yongb , Neha Sharmaa , and Shiao-Yng Chana,b
a
Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore;
b
Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore

ABSTRACT KEYWORDS
Inositols, a group of 6-carbon polyols, are highly bioactive molecules derived from diet and Diabetes; PCOS; myo-
endogenous synthesis. Inositols and their derivatives are involved in glucose and lipid metabolism inositol; lipid-metabolism;
and participate in insulin-signaling, with perturbations in inositol processing being associated with placenta; fetal development
conditions involving insulin resistance, dysglycemia and dyslipidemia such as polycystic ovary syn-
drome and diabetes. Pregnancy is similarly characterized by substantial and complex changes in
glycemic and lipidomic regulation as part of maternal adaptation and is also associated with
physiological alterations in inositol processing. Disruptions in maternal adaptation are postulated
to have a critical pathophysiological role in pregnancy complications such as gestational diabetes
and pre-eclampsia. Inositol supplementation has shown promise as an intervention for the allevi-
ation of symptoms in conditions of insulin resistance and for gestational diabetes prevention.
However, the mechanisms behind these affects are not fully understood. In this review, we explore
the role of inositols in conditions of insulin dysregulation and in pregnancy, and identify priority
areas for research. We particularly examine the role and function of inositols within the maternal-
placental-fetal axis in both uncomplicated and pathological pregnancies. We also discuss how ino-
sitols may mediate maternal-placental-fetal cross-talk, and regulate fetal growth and development,
and suggest that inositols play a vital role in promoting healthy pregnancy.

Introduction Brautigan, and Thorner 2010; Croze and Soulage 2013;


Heimark, McAllister, and Larner 2014; Ceriello et al. 1988;
Inositols are 6-carbon polyols present in all living cells
Croze, Gelo€en, and Soulage 2015; Sun et al. 2002), as well as
(Noventa et al. 2016). Humans synthesize approximately 4 g
perinatal disorders including gestational diabetes (GDM)
of inositol a day in the kidney (Clements 1979), but inositols
(Crawford et al. 2015; D’Anna and Santamaria 2018), pre-
are also abundant in fruits, grains and nuts, with a typical
North American diet providing about 1 g of inositol a day eclampsia (D’Oria et al. 2017) and intrauterine growth
(Holub 1986). Inositol and inositol derivatives often act as restriction (IUGR) (Dessı and Fanos 2013). Alterations are
both insulin mimics and second messengers (Asplin, most prominent in nervous and reproductive tissue, but the
Galasko, and Larner 1993; Greene et al. 1987; Huang et al. extent and foci of the inositol-related pathophysiology in
1993; Suzuki et al. 1994; Larner, Brautigan, and Thorner insulin resistant conditions and pregnancy complications
2010; Cheang et al. 2008; Unfer et al. 2012; Facchinetti et al. remains unclear (Asplin, Galasko, and Larner 1993;
2015; Croze and Soulage 2013) and their complex interac- Daughaday and Larner 1954; Kennington et al. 1990).
tions with both glucose and lipid metabolism are well dem- Several small clinical trials suggest that inositol supple-
onstrated across tissue types and in many species (Larner, mentation could be a useful intervention or preventive
Brautigan, and Thorner 2010; M€ uller et al. 1998; Romero measure for the insulin-related and metabolic abnormalities
and Larner 1993; Saltiel 1990; Hansen 2015; Yamazaki, in PCOS, GDM and type 2 diabetes (Noventa et al. 2016;
Zawalich, and Zawalich 2010; Nielson and Rutter 2018; Croze and Soulage 2013; Crawford et al. 2015; Papaleo et al.
Kunjara et al. 1999; Tabrizi et al. 2018). In humans, pertur- 2007; D’anna et al. 2012; Gateva, Unfer, and Kamenov 2018;
bations in inositol synthesis, metabolism and excretion are Colazingari et al. 2013; Bizzarri and Carlomagno 2014;
associated with metabolic conditions such as polycystic Unfer and Porcaro 2014; Muscogiuri et al. 2016; Lubin et al.
ovary syndrome (PCOS), diabetes mellitus and metabolic 2016; Brown, Crawford Tineke, and Alsweiler 2015; Farren
syndrome (Asplin, Galasko, and Larner 1993; Larner, et al. 2017; Malvasi et al. 2014; Pintaudi, Di Vieste, and

CONTACT Shiao-Yng Chan obgchan@nus.edu.sg Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of
Singapore, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore 119228, Singapore.
Supplemental data for this article is available online at https://doi.org/10.1080/10408398.2020.1845604.
ß 2020 The Author(s). Published with license by Taylor & Francis Group, LLC.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1627

Bonomo 2016; D’Anna et al., 2013; Matarrelli et al. 2013; with a focus on their role within the maternal-placental-fetal
Werner et al. 2016; DʼAnna et al. 2015; D’Anna et al., 2013; axis. This review was conducted in the Department of
Cogram et al. 2002; Cavalli et al. 2011). Inositol supplemen- Obstetrics and Gynecology at the National University of
tation appears to decrease glycemia and improve insulin Singapore and includes studies published in English up until
sensitivity, but effects vary depending on the inositol isomer March 2020. Articles were identified manually using Pubmed,
used, the dose and the population studied (D’anna et al. Science Direct and Google Scholar using the key words
2012; Dʼ Anna et al. 2015; D’Anna et al., 2013; Kim et al. described in Supplementary Table S1 and by following relevant
2005; Santamaria et al. 2012; Giordano et al. 2011; Artini references in articles of interest. Human and animal studies,
et al. 2013; Genazzani et al. 2012; Santamaria, Di Benedetto, clinical, randomized controlled trials (RCTs), cross-sectional
et al. 2016; Corrado et al. 2011; Fraticelli et al. 2018). Other and prospective studies, in-vitro studies, cell studies, genetic
small clinical trials have also suggested that inositol supple- studies, reviews and meta-analyses were all included.
mentation might reduce plasma triglycerides and total and
LDL-cholesterol levels among patients with diabetes mellitus,
Inositol biology
hyperinsulinemia, metabolic syndrome and PCOS (Tabrizi
et al. 2018), thereby highlighting a natural compound that Inositol stereoisomers and their derivatives
could address the increased risk of atherosclerotic and car-
diovascular diseases associated with these conditions. Inositol distribution and content in tissues and fluids
Current evidence suggests that each inositol isomer and Myo-inositol is the most abundant and well-studied inositol,
derivative affects metabolism in distinct ways (Thomas, Mills, but there are seven other natural inositol stereo-isomers: D-
and Potter 2016). Furthermore, these compounds appear to be chiro-inositol, L-chiro-inositol, epi-inositol, allo-inositol,
regulated differently in different tissues and in different popula- muco-inositol, neo-inositol and scyllo-inositol, and one syn-
tions even within apparently similar clinical phenotypes (Tables thetic stereo-isomer: cis-inositol (Thomas, Mills, and Potter
3–7). In this review we will examine how inositol isomers and 2016). Only myo-inositol, D-chiro-inositol, L-chiro-inositol,
derivatives regulate glucose and lipid metabolism, and discuss neo-inositol and scyllo-inositol have so far been found in
how disruptions in inositol synthesis, metabolism or excretion mammals (Thomas, Mills, and Potter 2016; Stewart,
could be involved in conditions of insulin dysregulation. Sherman, and Harris 1970). Reported inositol concentrations
Pregnancy is characterized by substantial and complex in human and mammalian adult and fetal tissues, and in the
changes in glycemic and lipidomic regulation, brought about circulation are summarized in Table 1.
by signals released by the fetal-placental unit (Di Cianni et al. Many older methods were not able to quantify different
2003). The placenta is situated at the maternal-fetal interface, inositol isomers separately (Tables 1A and 1C), whilst newer
where it ensures appropriate fetal nutrition throughout gesta- techniques such as gas chromatography (GC), high perform-
tion, by regulating maternal-fetal cross talk and nutritional ance liquid chromatography (HPLC) and nuclear magnetic
transfer (Gallo, Barrett, and Nitert 2017). Dysregulation of resonance (NMR) have enabled the quantification of some
these processes in pregnancy complications such as GDM and individual isomers (Tables 1B, 1D).
pre-eclampsia cause disordered fetal growth and threaten both Inositol is ubiquitous in the body, but content is particu-
maternal and fetal health (Gallo, Barrett, and Nitert 2017; Uhl larly high in the central nervous system (CNS), kidney, and
et al. 2015; Herrera and Ortega-Senovilla 2018; Larque et al. male and female reproductive organs, suggesting an import-
2014; Philipps et al. 2011). Impact on offspring is often long ant role for inositol in these tissues (Table 1). Several studies
term because the in-utero environment shapes life-long health suggest that inositols are critical for nervous system function
(Fisher, Novak, and Agranoff 2002; Thurston et al. 1989) and
(Developmental Origin of Health and Disease (Barker 2004)).
The fetal-placental unit is rich in inositol, and inositol inositol is known to play a key role in dopamine, serotonin,
concentrations in fetal and neonatal circulations are much noradrenaline and acetylcholine neurotransmission
higher than that in adults (Brusati et al. 2005; Toh et al. (Shaldubina et al. 2007). Indeed, brain inositol dysregulation
has been associated with Down syndrome (Berry et al. 1999),
1987; Santamaria, Di Benedetto, et al. 2016; Campling and
bipolar disorder (Calker and Belmaker, 2000), schizophrenia
Nixon 1954; Pereira et al. 1990; Islam, Selvam, et al. 2019;
(Shimon et al. 1998), aging (Stokes, Gillon, and Hawthorne
Pillai et al. 2020; Battaglia et al. 1961). Inositols are thus
1983), epilepsy (Pascente et al. 2016) and Alzheimer’s disease
postulated to be important in maintaining normal pregnancy
(Stokes and Hawthorne 1987). Inositols were also found to be
physiology and inositol dysregulation could be a key patho-
crucial for testicular and ovarian function, with dysregulation
logical element in many pregnancy pathologies. In this
associated with male and female infertility (Steegers-
review we will therefore investigate how inositols could act
Theunissen, Groenen, and Beemster 2002; Condorelli et al.
as critical signals in maternal-placental-fetal communications
2017; Unfer et al. 2017; Papaleo et al. 2009).
and how they might regulate fetal nutritional supply,
growth, adiposity and tissue programming.
Isomeric composition of inositols in circulation, urine
and tissues
Review criteria
The amount and isomeric composition of inositol in any
Overall, this review aimed to examine the current literature on cell, and the consequent biological effects, are influenced by
the biology of inositols in conditions of insulin dysregulation, systemic and local factors. Systemic inositol concentrations
1628
O. C. WATKINS ET AL.

Table 1A. Unspecified inositols (mmol/ kg wet tissue) analyzed by methods that do not differentiate between inositol isomers.
Ref Method Species Central nervous system Muscle Liver Kidney Heart Intrauterine tissues Other tissues
(Kalra, Kalra, and Sharma 2016) Inositols isolated and weighed Adult Rat 9.1-9.3 Testis: 2.2-3.7,
Epididymis:
11.4-12.3,
Seminal vesicle:
33.9-37.0
(Battaglia et al. 1961) Thin layer chroma-tography Adult Sheep Cortex: 0.8-2.7 2.1-3.9 3.6-10.9 4.6-15.2 Uterine mucosa:
11-11.6, 3.9  15.7
Cerebellum: 11.2-15.5
Fetal sheep Cortex: 12.3, Cerebellum: 15.5 0.3-4.3 0.4-3.3 0-12.7 0-3.22 Chorion: 4.2,
Placenta: 1.0
Adult Goat Cortex: 0-1.7 1.3 3.7-8.5 8.4-11.0 Uterine mucosa:
5.9-9.3, 0.5-3.4,
Cerebellum: 6.9-15.7 Chorion:
1.6-4.1,
Placenta:
0.4-1.0
Fetal goat Cortex: 2.0-4.2 12.7-15.0 2.7-3.9
10.6-14.5,
Cerebellum: 12.9-14.9
Adult Rabbit Brain: 10.3 1.9 1.7 Placenta: 1.7
Adult Rat Brain: 0.2-2.1 2.4 6.9-7.6 0.8-1.7 Uterine mucosa: 3.1,
5.0-6.7 Placenta:
0.7-1.7
Adult Pig Uterine mucosa: Ovaries:
0.4-5.8 3.3-5.3
(Campbell et al. 2004) Microbiological assay method Adult Mice 11.7 (1) 0.32 (0.04) 1.2 (0.2) 8.2 (0.6) 1.4 (0.2) Pancreas:
2.3 (0.2)
(Islam, Selvam, et al. 2019) Enzymatic assay Fetal Human Placenta (Freea):
2.7 (0.3),
Placenta (totalb):
3.5 (0.4)
Ref: Study reference. aFree inositol was extracted with water, bTotal inositol extracted by high temperature acid hydrolysis. Mean with standard deviation (SD) or range stated if available.
Table 1B. Specified inositols in tissues.
Ref Method Species Central nervous system Peripheral nervous system Liver Kidney In utero tissues Other tissue
Myo-inositol in tissues quantified in mmol/ kg wet tissue

(Stokes and Hawthorne 1987) GC Adult Human Cortex: 2.28


(Shimon et al. 1998) GC Adult Human Frontal cortex:
1.20 (0.6),
Occipital cortex:
0.81 (0.5),
Cerebellum:
0.67 (0.5)
(Toh et al. 1987) GC Fetal Human Placenta: 0.53,
Umbilical cord: 0.59
(Stull et al. 2008) GC Adult Rat Freea: 5.31-6.67 Freea: 0-0.32 Freea:
Lipid boundb: Lipid boundb: 2.06-2.55 5.72-6.53,
1.11-1.38 Lipid boundb:
1.02-1.04
(Taguchi et al. 1997) GC Adult Rat Brain: 6.62 (0.96) Sciatic nerve: 0.57 (0.12) 4.51 (0.36)
3.10 (0.34)
(Kennington 1992) GC Adult Rat 3.52 (0.04)
(Larner 2002) GC Adult Female rats Brain: 4.5 (0.29) 1.27 (0.2)
(Sherman, Goodwin, GC Adult Rabbit Cerebral cortex: Sciatic nerve: 2.0 Testis: 5.7,
and Gunnell 1971) 13.0 (0.9), 4.25 (0.19), Spleen: 0.8,
Cerebellar cortex: 13.6 (0.5), Vagus nerve: Lens: 0.3
Optic nerve: 4.26 (0.55)
14.2 (0.3),
White matter:
11.6 (0.7),
Midbrain nuclei:
13.2 (0.7)
Adult Rat Brain: 5.82 (0.29) Sciatic nerve: 6.12 (0.54)
Spinal cord: 7.4 4.06 (0.26)
Neo-inositol in tissues quantified in mmol/ kg wet tissue

(Groenen, Merkus, GC Adult Rat Brain: 1.9-6.5 <0.8 6.2-9.6 Heart: <0.8-3.5,
et al. 2003) Testis: 6.5-4,
Spleen: 1.2-4.4
Scyllo-inositol in tissues quantified in mmol/ kg wet tissue

(Michaelis et al., 1993) GC Adult Rabbit Cerebral cortex: Sciatic nerve: 0.17 Testis: 0.2,
0.53 (0.05), 0.34 (0.04), Spleen: 0.12,
Cerebellar cortex: Vagus nerve: Pancreas: 0.09,
0.07 (0.06), 0.65 (0.17) Lens: 0.59
Optic nerve:
0.63 (0.03),
White matter:
0.46 (0.03)
Midbrain nuclei: 0.57 (0.06)
GC Adult Rat Brain: 0.16 (0.01), Sciatic nerve: 0.45 (0.02)
Spinal cord: 0.18 0.29 (0.02)
(Seaquist and Gruetter 1998 ) NMR Adult Human Brain: 0.43 (0.11)
(Sherman, Stewart, NMR Adult Human Occipital cortex:
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION

Kurien, et al. 1968) 1.4 (0.1)


Ref: Study reference, GC: Gas Chromatography, NMR: Nuclear Magnetic resonance. aFree inositol was extracted with water, bLipid bound inositol was extracted with chloroform methanol (2:1). Mean with standard devi-
ation (SD) or range if available.
1629
1630 O. C. WATKINS ET AL.

Table 1C. Unspecified inositols in fluids (mmol/L) by methods that do not differentiate between inositol isomers.(Garcia-Perez and Burg 1990)Inositols isolated
and weighed Adult Boar Seminal: 110,000-167,000
Ref Method Species (status) Blood Other fluid
(Kalra, Kalra, and Sharma 2016) Inositols isolated Adult Rat Seminal: 52,400-58,900
and weighed
(Battaglia et al. 1961) Thin layer chromatography Adult Sheep Plasma: 0-89
Fetal sheep Plasma: 800-1333 Spinal: 7780, Urine: 17-83,
Allantoic: 555-167,
Amniotic: 300-528
Adult Goat Plasma: 39-89 Spinal: 460
Fetal goat Plasma: 933-1230 Spinal: 3060, Amniotic: 560
(H€aussinger 1998) Microbiological assay Adult Bull Coccygeal vein plasma: 22 (3) Rete testis: 3877 (287),
Cauda epididymidis: 2470
(240),
Seminal: 761 (121)
(Zheng et al. 2015) Microbiological assay Adult Human Semen: 3600 (1600)
(Campling and Nixon 1954) Microbiological assay Human (pregnant, term) Whole blood: 28-44
Human fetus (term) Whole blood: 61-155 Amniotic: 33-89
Human fetus (under 24 weeks) Whole blood: 188.7-888.1 Amniotic: 38.9-521.8,
Plasma: 188.7-621.7 Fetal urine: 144.3-299.7
Sheep (pregnant) Whole blood: 33-150
Sheep fetus Whole blood: 555-1721 Amniotic: 33-1117, Allantoic:
Plasma: 610.6-1465.4 278-1171
Adult monkey (pregnant) Plasma: 44.4
Monkey fetus Plasma: 199.8 Amniotic: 99.9
Adult goat (pregnant) Whole blood: 27.8-194.3
Goat fetus Whole blood: 172.1-1887.2 Amniotic: 138.8-405.2,
Allantoic: 444.0-1182.3
Adult rabbit (pregnant) Whole blood: 44.4-172.1
Rabbit fetus Whole blood: 177.6-910.3 Amniotic: 105.5-1681.8,
Allantoic: 3718.9-10629.4
Adult cat (pregnant) Whole blood: 138.8-188.7
Cat fetus Whole blood: 910.3-1254.4 Amniotic: 455.2,
Allantoic: 1509.8-2919.6
þ
(Celentano et al. 2020) NAD myo-inositol Adult Human Serum: 39 (2) Follicular fluid: 36 (3)
dehydrogenase assay
(Pereira et al. 1990) Enzymatic fluorometric assay Adult human (post partum) Serum (at birth): 52 (6) Breast milk
(preterm colostrum): 2240
(280),
Breast milk
(term colostrum): 1840
(451),
Breast milk (mature):
1456 (276)
Neonate Preterm neonate serum: 108
(10),
Term neonate serum:
86 (13)
Ref: Study reference. NAD: Nicotinamide adenine dinucleotide. Mean with standard deviation (SD) or range if available.

are determined by inositol intake, bioavailability and excre- Only a handful of studies have described the relative
tion. Locally, cellular levels are regulated by cellular uptake abundance of other inositol isomers. In Sprague Dawley
and efflux, isomeric conversion, synthesis, catabolism and rats, L-chiro-inositol was found in similar concentrations to
metabolism into inositol-containing derivatives (Holub D-chiro-inositol in muscle phospholipids (skeletal and
1986). Myo-inositol comprises 98% of adult human plasma smooth) and heart phospholipids, but D-chiro-inositol was
inositol and 71% of urinary inositol (Kalra, Kalra, and higher than L-chiro-inositol in fat, liver, brain and kidney
Sharma 2016). D-chiro-inositol is commonly quoted as the phospholipids (Kennington 1992; Larner 2002). Neo-inositol
second most abundant inositol with plasma D-chiro-inositol meanwhile was found in the brain, heart, kidney, testis, and
being 3% relative to plasma myo-inositol, and urinary D- spleen of the rat, but was not detected in rat liver (Sherman,
chiro-inositol being 21- 29% relative to urinary myo-inositol Goodwin, and Gunnell 1971). Scyllo-inositol and epi-inositol
(Campbell et al. 2004; Stull et al. 2008). However, this may quantified in human plasma were about 100 times less abun-
not be the case, since most methods used cannot distinguish dant than myo-inositol (Groenen, Merkus, et al. 2003), while
between D-chiro-inositol and L-chiro-inositol, and do not scyllo-inositol in the human brain was about five times less
quantify scyllo-inositol or neo-inositol. There is also concern abundant than myo-inositol (Michaelis et al., 1993; Seaquist
that the use of acid in some analytical methods may cause and Gruetter 1998). In rabbit tissue, the myo-inositol to
isomerization ex-vivo, leading to an artificial change in the scyllo-inositol ratio was very high in the spleen, pancreas,
ratio of different inositol isomers (Taguchi et al. 1997). testis (45:1, 30:1, 28:1, respectively) and in the brain (19:1 –
Therefore, in many studies inositols may have been missed, 25:1 depending on region), but much lower in the sciatic
mis-identified or mis-analyzed. nerve, vagus nerve and lens (12:1, 6.6:1, 10:1, respectively)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1631

Table 1D. Specified isomers in fluids (mmol/L).


Ref Method Species Blood Other Fluids
Myo-inositol

(Formuso, Stracquadanio, and GC Adult human Urine: 454 (738)


Ciotta 2015)
(Ma et al., 2012) GC Adult human Seminal: 1472 (255)
(Ma et al., 2012) GC Adult human Serum: 38 Cerebrospinal: 22-905
(Nozadze et al. 2011) GC Adult human Cerebrospinal: 167-611
(Taguchi et al. 1997) GC Adult rat Serum: 80 (10)
(Kennington 1992) GC Adult rat Serum: 48.6 (4.9)
(Michaelis et al., 1993) GC Adult rat Serum: 71.0 (0.6)
(Bersudsky et al. 1999) GC Adult human Plasma: 19.9 (2.5)
Adult human human 90 min Plasma: 96.5 (16.6)
after myo-inositol oral
ingestion
(100 mg/kg body weight)
(Santamaria, Di Benedetto, GC Adult human Amniotic: 79.0
et al. 2016)
(Einat and Belmaker 2001) GC Human (Non-pregnant) Serum: 24.5
Human (pregnant) Serum: 21.4
Human fetus Cord serum (mid-gestation):
125,
Umbilical artery serum
(term): 60.2,
Umbilical vein serum
(term): 45.4
(Strieleman et al., 1992) GC Adult human Plasma: 34 (6) Urine: 167 (99)
(Brusati et al. 2005) HPLC Human (pregnant) Plasma: 26
Human Fetus Umbilical artery (term): 75
Umbilical vein (term): 64
Other inositol isomers

(Strieleman et al., 1992) GC Adult human Plasma D-chiro-inositol, urine: 50 (35)


D-chiro-inositol: 0.90 (0.24)
(Formuso, Stracquadanio, and GC Adult human D-chiro-inositol, urine: 21 (34)
Ciotta 2015)
(Bersudsky et al. 1999) GC Adult human Plasma
Scyllo-inositol: 0.46 (0.13),
Epi-inositol: 0.19 (0.19)
Adult human 90 min after Plasma
myo-inositol oral ingestion Scyllo-inositol: 1.09 (0.17),
(100 mg/kg body weight) Epi-inositol: 0.38 (0.23)
Ref: Study reference, GC: Gas chromatography, HPLC: high performance liquid chromatography. Mean with standard deviation (SD) or range if available.

Table 2. Inositol transport proteins facilitating cellular influx of inositol.


Name/ alternative
Transporter type name Metabolites transferred (ordered by relative affinity) Organs with highest gene expression
Proton (Hþ)/ myo-inositol symporters HMIT/ GLUT13/ Myo-inositol > scyllo-inositol > D-chiro- Brain, adipose tissue and kidneys
SLC2A13 inositol > muco-inositol
Sodium/ myo-inositol transporters SMIT-1/ Myo-inositol ¼ scyllo-inositol > glucose Kidney, brain, placenta, pancreas,
SLC5A3 heart and skeletal muscle
SMIT-2/ Myo-inositol and D-chiro-inositol > L-Chiro-inositol, Heart, skeletal muscle, kidney, liver
SLC5A11 D-glucose, D-manose and D-pinitol and placenta
Note: Data from Guo et al. 1997; Davanzo et al. 2003; Davanzo et al. 2001, Willmroth et al. 2007; Davanzo et al. 2003; Harwood 2005; Davanzo et al. 2003;
Lubrich and van Calker 1999; Calker and Belmaker, 2000.

with similar results seen in rats (Sherman, Stewart, Kurien, inositol isomers have the same effects when shape does not
et al. 1968). This may suggest a more important role for affect function, such as in osmotic regulation (Thurston
scyllo-inositol in the peripheral nervous system compared et al. 1989; Garcia-Perez and Burg 1990; H€aussinger 1998).
with other tissues. Overall, further studies are required to Inositol isomers are also differently disrupted in various
clarify the inositol composition in biological systems. pathologies (Tables 3–7). The pharmacological effects of
inositol likely depend on the inositol isomer, the dose, and
the population studied. For example, daily inositol supple-
Different actions of inositol isomers mentation resulted in decreased insulin resistance as
The shape of inositol isomers affects their ability to interact assessed by the Homeostatic Model Assessment of Insulin
with enzymes giving them distinct bioactivities (Thomas, Resistance (HOMA-IR) in postmenopausal women with
Mills, and Potter 2016) and altering how these molecules are metabolic syndrome [4000 mg myo-inositol (Santamaria
transported across membranes (Table 2). However, all et al. 2012; Giordano et al. 2011)], in non-pregnant women
1632 O. C. WATKINS ET AL.

Table 3. Case-control studies of inositol concentrations in non-pregnant populations with diabetes compared with controls.
Population Comparison Country/
Ref (as specified in study) (Control) Ethnicity Sample L-chiro-inositol D-chiro-inositol Myo-inositol Method
Type 1 or Insulin-dependent population
(Pugliese et al. 1990) Overweight, Insulin Overweight/ USA Urine (24 h No change Increased Increased GCMS
dependent diabetes obese, output,
Non-diabetic creatinine
normalized)
Plasma No change No change No change GCMS
(Coppey et al. 2002) Normal weight Type Normal weight, Korea Urine – Decreased Increased HPLC
1 diabetes Non-diabetic (24 h
output)
Type 2 or Insulin-independent population
(Daughaday and Poorly Non- diabetic USA Urine Inositol Increased Yeast assay
Larner 1954) controlled diabetes Plasma No change in inositol
(Pugliese et al. 1990) Obese, non-insulin Overweight/ USA Urine No change Increased Increased GCMS
dependent diabetes obese, (24 h
(poorly controlled) Non-diabetic output)
Plasma No change No change No change
(Formuso, Stracquadanio, Overweight, Type Normal weight, Korea Urine – Increased Increased HPLC
and Ciotta 2015) 2 diabetes Non-diabetic
(Kennington et al. 1990) Non-insulin Non-diabetic USA Urine – Decreased Increased GCMS
dependent diabetes (White, (24 h output)
Black)
Non-insulin Non-diabetic USA, – Decreased Increased GCMS
dependent diabetes Pima Indian
(Coppey et al. 2002) Normal weight, Type Normal weight, Korea Urine – Decreased Increased HPLC
2 diabetes Non-diabetic (24 h
output)
(Suzuki et al. 1994) Normal weight, Type Age- matched Japan Urine – Decreased – GCMS
2 diabetes Normal
weight,
Non-diabetic
Ref: Study reference. Inositol levels refer to the non-lipid-bound portion unless otherwise stated.

Table 4. Animal studies of inositol levels in non-pregnant diabetes models.


Ref Species Model of diabetes Control Location D-Chiro-inositol Myo-inositol
(Taguchi et al. 1997) Rat Streptozocin-induced, Untreated, non- Brain, kidney, urine – Increased
long-term mild diabetes diabetic rat Sciatic nerve – Decreased
Brain – Decreased lipid-bound
Streptozocin-induced, Serum, Liver – Increased
acute diabetes Sciatic nerve – Decreased lipid-bound
(Yue et al. 1989) Rat Streptozocin- induced Untreated, non- Peripheral nerve – Decreased
diabetic rat Peripheral nerve – Decreased incorporation
of [3H]-myo-inositol
into
phosphatidylinositol
(Chukwuma, Ibrahim, Rat Streptozocin- induced Untreated, non- Sciatic nerve – Decreased
and Islam 2016) diabetic rat Plasma – Unchanged
(Loy et al. 1990) Rat Streptozocin- induced Untreated, non- Superior – Decreased
diabetic rat cervical ganglion
(Greene et al. 1987) Rat Biobreeding Biobreeding diabetic Sciatic nerve – Decreased
given insulin
(Palmano, Whiting, and Rabbit Alloxan induced Untreated non- Retina – Deceased
Hawthorne 1977) diabetic rabbit
(Zhu and Eichberg 1990) Rabbit Alloxan induced diabetic Kidney – Unchanged
(mild or severe) Left ventricle of
heart,
Atrioventricular
node region,
Aqueous humor,
Retinal
choriocapillaris,
Cornea
Alloxan induced diabetic Aortic myointima – Decreased
(mild, 19
days exposure)
Alloxan induced diabetic – Unchanged
(mild, 2
months exposure)
Alloxan induced diabetic – Increased
(severe, 2
months exposure)
(continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1633

Table 4. Continued.
Ref Species Model of diabetes Control Location D-Chiro-inositol Myo-inositol
Alloxan induced diabetic Retinal – Decreased
(severe diabetes, 2 pigmented
months exposure) epithelium
Alloxan induced diabetic Serum – Decreased
(mild and severe)
(Mayer and Rat Goto-Kakizaki Wistar rat controls Urine Decreased Increased
Tomlinson 1983)
(Greene, De Jesus, and Rhesus monkeys Spontaneously diabetic Non- Urine Decreased –
Winegrad 1975) diabetic monkey
(Kennington et al. 1990) Rhesus monkeys Spontaneously diabetic Urine Decreased Increased
(Solomonia et al. 2010) Rat Streptozocin induced Untreated rat Urine Increased Increased
Mouse db/db mice Wild-type mice Urine Increased Increased
Ref: Study reference. Inositols measured are non-lipid-bound forms unless otherwise stated.

Table 5. Case-control studies of IPGs in non-pregnant diabetes compared with non-diabetic controls.
Ref Type of diabetic Analysis method Location P-IPG A-IPG
(Asplin, Galasko, and Type 2 diabetes PDH phosphatase assay Muscle Decreased –
Larner 1993) of P-IPG fraction Hemodialysate Decreased Unchanged
cAMP dependent Urine (24 h output) Decreased Decreased
protein kinase assay of
A-IPG fraction
D-chiro-inositol and Hemodialysate D-chiro-inositol/ myo-inositol ratio decreased
myo-inositol in P-IPG Muscle D-chiro-inositol/ myo-inositol ratio decreased
fraction by HPLC
(Kennington et al. 1990) Type 2 diabetes Myo-inositol and D- Muscle biopsy before Decrease in D-chiro- Increased in myo-
chiro-inositol content in insulin administration inositol content inositol content
combined P-IPG and A- Muscle biopsy 15 and
IPG fractions analyzed 20 min into
by GC euglycemia
insulinemic
clamp study
(Kunjara et al. 1999) Types 1 and 2 diabetes PDH phosphatase assay Urine Unchanged Increased
of P-IPG fraction
(creatinine
normalized),
Lipogeneses assay of
A-IPG fraction
(creatinine
normalized)
Ref: Study reference.

with PCOS [2000 mg myo-inositol (Artini et al. 2013; PCOS patients (Formuso, Stracquadanio, and Ciotta 2015)
Genazzani et al. 2012)], in obese and overweight pregnant (vide-infra). Meanwhile, scyllo-inositol and epi-inositol
women [4000 mg (Dʼ Anna et al. 2015; Santamaria, Di show promise for neurological function. Scyllo-inositol
Benedetto, et al. 2016) myo-inositol], and in women with showed effectiveness in-vitro and in-vivo as potential
GDM with no insulin therapy [4000 mg myo-inositol Alzheimer’s disease treatment, decreasing neuronal tox-
(Corrado et al. 2011; Fraticelli et al. 2018)]. However, no icity, cognitive deficits and the aggregation of amyloid b
change in insulin resistance as assessed by HOMA-IR was peptides and increasing long-term potentiation (Ma et al.,
observed among pregnant women with GDM treated with 2012; Ma et al., 2012). In rats, scyllo-inositol also showed
D-chiro-inositol [500 mg D-chiro-inositol or a combination anti-convulsive effects and maybe useful in treating epi-
of 1100 myo-inositol/27.6 mg D-chiro-inositol (Fraticelli lepsy (Nozadze et al. 2011), while epi-inositol reduced
et al. 2018)]. However, differences in the statistical power of anxiety (Bersudsky et al. 1999; Einat and Belmaker 2001).
these studies, and in the methods used, make compari- Some inositol isomers may also influence the effects of
sons difficult. other inositols (Strieleman et al., 1992; Wentzel et al. 2001;
So far, only trials of supplements containing myo-inositol Salman et al. 1999; Strieleman and Metzger 1993;
(without other inositols) suggested efficacy in reducing risk Strieleman et al., 1992). Scyllo-inositol, for example, in the
of GDM (Zheng et al. 2015). Treatment with D-chiro-inosi- developing rat conceptus, inhibited myo-inositol uptake
tol [500 mg] alone showed no effect on GDM prevention, and the synthesis myo-inositol derived phosphoinositide
while combined myo-inositol [1100 mg] and D-chiro-inositol and phosphoinositide-phosphate (PIP) (Strieleman et al.,
[27.6 mg] treatment has shown no consistent positive effect 1992; Wentzel et al. 2001; Salman et al. 1999; Strieleman
(Farren et al. 2017; Celentano et al. 2020). and Metzger 1993; Strieleman et al., 1992), and these
Unlike in GDM, both myo-inositol and D-chiro-inositol changes were associated with increased structural abnor-
treatments were able to significantly improve endocrine malities and deceased crown rump length and somite
and metabolic parameters in non-pregnant overweight number (Strieleman et al., 1992).
Table 6. IPGs in pregnant women with and without diabetes. 1634

Ref Pregnant Population Sample (gestational age) Method of assay P-IPG A-IPG
Uncomplicated pregnancy compared with non-pregnant controls

(Suzuki et al. 2014) Non-diabetic pregnancies Urine 24 h output Extraction of IPG fractions followed by Increased compared to –
(26 to 37 weeks) bioactivity assay non-pregnant
Pregnant women with preexisting diabetes (or diabetes not specified) compared with matched non-diabetic pregnant controls of similar gestational age†

(Suzuki et al. 2014) Type 1 diabetes treated with insulin Maternal urine 24 h output Extraction and bioactivity assay Similar to controls Similar to
(31 to 38 weeks) controls
(Sacks et al. 2012) Type 2 diabetes Maternal urine (spot collection any time Polyclonal antibody assay, Increased –
O. C. WATKINS ET AL.

point before 30 weeks) creatinine normalized


Maternal urine Similar to controls –
(spot collection after 30 weeks)
(Scioscia, Gumaa, et al. 2007)
Diabetes Endothelium of term placental vessels Immuno-histochemical staining Decreased –
(type not specified)
Pregnant women with GDM compared with matched non-diabetic pregnant controls of similar gestational age†

(Zeitler, Wu, and GDM Maternal urine 24 h output Extraction of IPG fractions followed Increased –
Handwerger 1991) (30.2 ± 6.3 (SD) weeks) by bioactivity assay
(Sacks et al. 2012) GDM Maternal urine (spot collection 20-35 weeks) Polyclonal antibody assay, Increased –
Maternal urine (spot collection after 35 weeks) creatinine normalized Similar to controls –
(Hayashi, Hasegawa, GDM Maternal urine Extraction of IPG fractions followed – Decreased
and Tomita 1976) (spot collection, gestational age not available) by bioactivity assay, creatinine
normalized
Ref: Study reference. †All studies are cross sectional. GA: Gestational age. GDM: gestational diabetes mellitus. SD: standard deviations.

Table 7. P-IPG in pre-eclampsia (PE) compared to controls with an uncomplicated pregnancy.


Ref Location Gestational age (weeks) Measured by P-IPG in pre-eclampsia
(463) Urine (spot collection) PE: 32.0 ± 4.5, Polyclonal antibody assay, Increased
Control 31.4 ± 3.5 creatinine normalized
(464) Urine (spot collection) Urine collected every 4 weeks Polyclonal antibody assay, Increased
after booking creatinine normalized
(Best et al. 1946) Urine (catheter urine sample At delivery Polyclonal antibody assay Increased
collected at the start of
the cesarean section)
Amniotic fluid Increased
Peripheral vein Extraction and No change
Umbilical vein bioactivity assay No change
Umbilical artery No change
Uterine vein Decreased
(Suzuki et al. 2014) Urine (24 h output) PE: 26 to 37, Extraction and Increased
Placenta Control: 31 to 38 bioactivity assay Increased
(Hegsted et al. 1973) Cultured placenta incubated Post delivery Extraction and Decreased placental P-IPG
with insulin bioactivity assay after insulin exposure
(Chu and Hegsted 1980) Cultured placenta incubated Post delivery Extraction and Decreased placental P-IPG
with insulin bioactivity assay after insulin exposure
Ref: Study reference.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1635

Figure 1. Synthesis, catabolism, excretion, isomerization and derivatization of inositols.


The synthesis of methylated inositol derivatives has so far only been found in plants and whether this process also occurs in mammals is controversial.

Inositol derivatization Wells 1965). Meanwhile, an enzymatic assay study which


Inositols can be derivatized with lipids, phosphates, sugars, compared water-soluble and total acid hydrolyzable inositol
proteins and other compounds to form a wide range of bio- in human placenta found 77% of the inositol in the water-
active molecules (Figures 1 and 2). A lack of suitable analyt- soluble form (Islam, Selvam, et al. 2019).
ical techniques means that little is known about the relative It should be noted that although most quantified water
abundance of different derivatives. One gas chromatography soluble inositol will originate from underivatized inositol,
study compared water soluble myo-inositol (aqueous extract, some will be due to the cleavage of other water-soluble inosi-
probably mainly underivatized) to lipid-bound inositol tol derivatives during processing. The degree to which this
(organic extraction followed by acidic hydrolysis) (Wells, occurs is not known, but the amount of cleavage will depend
Pittman, and Wells 1965). This study demonstrated that in on the methods used, with the harsher methods used to pre-
the brain and kidney most inositol was found in the water- pare samples for gas chromatography likely to cause more
soluble form (83 and 87% respectively), while in the liver cleavage than other methods. The degree of cleavage will also
most inositol was lipid-bound (94%) (Wells, Pittman, and depend on the inositol derivative involved. For example, the
1636 O. C. WATKINS ET AL.

Figure 2. Inositol metabolism leads to the formation of diverse bioactive derivatives many of which interact with insulin signaling pathways. Inositol derivatives
containing lipids are shown in gold while those that do not are shown in green. Other compounds are shown in black. Blue arrows and descriptions show selected
signaling or metabolic processes affected by inositol or inositol derivatives. Abbreviations: Arachidonic acid (AA), Diacylglycerol (DAG), Glycosyl-phosphatidyl-inosi-
tols (GPI), Inositol phosphoglycans (IPGs), Lysophosphatidic acid (LPA), lyso-phosphotidyl inositol (LPI), Phosphoinositide phosphates (PIPs).

phosphate-ester bond in inositol phosphates will be more sen- to form galactose (Parthasarathy, Parthasarathy, and Vadnal
sitive to cleavage than the ether bond of pinitol (D-chiro- 1997). ISYNA1 is found in all eukaryotes, while IMPA1 is
inositol-O-methyl ether) or the glycosidic bonds of inositol found in all Animalia (Zerbino et al. 2018). Although both
glycans and inositol phosphoglycans (IPGs). Thus, the are ubiquitously expressed in mammalian tissues, ISYNA1 is
amount of inositol quantified will depend on extraction, proc- particularly highly expressed in human placenta and testis,
essing and quantification methods used and results cannot while IMPA1 is especially highly expressed in the testis
easily be compared between studies. (Fagerberg et al. 2014; Guan, Dai, and Shechter 2003).
Labeled myo-inositol and D-chiro-inositol injected into rats Despite such expression patterns, the kidneys remain the pre-
generally remained as free underivatized inositols (94% and 83.4% dominant site of myo-inositol synthesis (approximately 4 g/
respectively), with substantially les s incorporated into inositol day in humans (Clements Jr 1979)), with appreciable amounts
phosphates (1.8% and 16.4%), or phospholipids (3.2% and 0.2%), also synthesized by the brain [30 mmoles/kg wet weight/h
when assessed in all tissues combined after 72 h, suggesting inosi- (Stewart, Sherman, and Harris 1970)] and testis [316 mmoles/
tol derivatization is isomer dependent (Pak et al. 1998). kg wet weight/hr (Stewart, Sherman, and Harris 1970;
Eisenberg and Bolden 1963; Middleton and Setchell 1972;
Voglmayr and White 1971; Hauser and Finelli 1963)], con-
Inositol synthesis, iomerism and catabolism sistent with these tissues’ high inositol content (Table 1).
Myo-inositol synthesis
Myo-inositol can be endogenously synthesized from glucose Myo-inositol epimerization to D-chiro-inositol
by inositol-3-phosphate synthase (ISYNA1) and inositol-1- Myo-inositol is thought to be converted into D-chiro-inositol
monophosphatase (IMPA1) (Noventa et al. 2016; Holub by myo-inositol-D-chiro-inositol-1-epimerase. Epimerization
1986) (Figure 1). These enzymes are relatively specific to was evaluated in-vivo via the conversion of injected radio-iso-
inositol metabolism, although IMPA1 can also hydrolyze gal- tope-labeled [3H]-myo-inositol into [3H]-D-chiro-inositol in
actose 1-phosphate (an intermediate of galactose metabolism) non-diabetic rats (Pak et al. 1992). The relative amount of
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1637

isotope label found as D-chiro-inositol varied between tissues, identification of the inositols involved (Lin, Ma, Gopalan,
ranging from 0.7% in the heart, to 2.2% in liver. Much higher et al. 2009). This hypothesis was based on research which
relative amounts of D-chiro-inositol were observed in urine suggested that mice cannot convert myo-inositol into D-
(36%) and blood (60.4%) (Pak et al. 1992), which reflect the chiro-inositol and instead rely on dietary D-chiro-inositol
combined whole body tissue conversion rates. Labeling of (Lin, Ma, Gopalan, et al. 2009). D-chiro-inositol was found
other inositol isomers including scyllo-inositol, neo-inositol, to fall rapidly in the plasma, stools and urine of mice fed a
epi-inositol, and mucoinositol was less than 0.06%. myo-inositol containing, but pinitol and D-chiro-inositol
Myoinositol epimerase activity appears altered in several deficient diet (Lin, Ma, Gopalan, et al. 2009). Moreover,
conditions of insulin resistance, but the direction of change mice fed the deficient diet who were fed deuterium-labeled
depends on the condition and tissue studied. Ovarian thecal water or injected intraperitoneally with [2H6]-myoinositol
cells from patients with PCOS showed decreased myo-inosi- produced urine that contained deuterium-labeled myo-inosi-
tol:chiro-inositol ratios, and increased in-vitro epimerase tol, but no deuterium labeled D-chiro-inositol.
activity (Heimark, McAllister, and Larner 2014; Carlomagno, However, such evidence alone is not strong enough to sug-
Unfer, and Roseff 2011). In contrast, the muscle, liver, and gest that the myoinositol epimerase has been mis-identified.
fat of Goto-Kakizaki diabetic rats showed increased in-vivo Instead these results could also be explained by low endogen-
myo-inositol:chiro-inositol ratios and decreased in-vivo epi- ous D-chiro-inositol synthesis compared to dietary intake, the
merase activity (Sun et al. 2002; Pak et al. 1998). sensitivity limits of stable isotope based analysis, differences
We cannot rule out that differences between these studies in mouse inositol metabolism compared to other species, or
could possibly be due to methodological issues, but this does to the metabolic consequences a pinitol and D-chiro-inositol-
not seem likely. In both cases, in-vivo epimerase activity was deficient diet for 15 weeks prior to the measurement of the
quantified by measuring the conversion of myoinositol to chiro- outcome of interest. However, further research on the activity
inositol by HPLC. The ovarian study measured the conversion of this epimerase in different tissues and conditions is needed
of non-labeled isotopes using an Ag/AgCl electrochemical before strong conclusions can be drawn.
detector, with peak identity confirmed by GCMS and by com-
parison to known standards of myo-inositol and D-chiro-inosi- The synthesis of other inositol isomers
tol. In contrast, the rat study measured the conversion of Myo-inositol is converted into scyllo-inositol by myo-ino-
radiolabeled inositols using a radiolabel detector, a more sensi- sose-2 (Sherman, Stewart, Kurien, et al. 1968; Sherman,
tive technique which automatically adjusts for recovery losses. Stewart, Kurien, et al. 1968; Hipps, Holland, and Sherman
Peak identity was confirmed by GCMS and by comparison to 1977; Hipps, Ackermann, and Sherman 1982). Myo-inosose-
known standards of myo-inositol, D-chiro-inositol, scyllo-inosi- 2 is expressed in a variety of rat and rabbit tissues, but is
tol, neo-inositol, epi-inositol, and mucoinositol. particularly high in brain, testis and kidneys (Sherman,
It is more likely that the observed differences are due to Stewart, Kurien, et al. 1968; Sherman, Stewart, Kurien, et al.
the tissues and disorder studied. Treatment with intramus- 1968), suggesting an important role for scyllo-inositol in
cular insulin increased epimerase activity in the livers of these tissues. However, whether this is also the case in
Sprague Dawley rats suggesting that epimerase activity likely humans remains to be established. Neo-inositol is synthe-
depends on insulin availability and insulin sensitivity (Sun sized from D-mannose 6-phosphate, but the process is not
et al. 2002; Pak et al. 1998). The muscle, liver, and fat of well understood (Sherman, Goodwin, and Gunnell 1971).
Goto-Kakizaki diabetic rats are likely more insulin resistant
than controls, which could lead to decreased epimerase
Myo-inositol catabolism and excretion
activity. In contrast, in PCOS the ovaries, unlike other tis-
Myo-inositol is catabolized by myo-inositol oxygenase
sues do not become insulin resistant, but are still exposed to
(MIOX) into D-glucuronic acid (Charalampous 1959; Arner
increased insulin leading to increased epimerase activity
et al. 2001). D-glucuronic acid can then be converted into
(Carlomagno, Unfer, and Roseff 2011).
D-xylulose 5-phosphate, which enters the pentose phosphate
Insulin treatment also increased the conversion of total
pathway (Charalampous 1959; Arner et al. 2001). The kid-
[3H]myo-inositol to [3H]chiro-inositol in a rat fibroblast cell
ney is the primary organ for myo-inositol catabolism
line that expressed the human insulin receptor (Pak et al.
(Holub 1986), with proximal tubular epithelial cells being
1993). However, the increase in radio-label incorporation was
the main site of MIOX expression (Arner et al. 2006). The
mainly observed in the acid hydrolyzed lipid fraction, suggest-
kidneys play a major role in regulating inositol excretion
ing either that epimerization occurred in phospholipids rather
with about 95% of filtered inositol being reabsorbed in iso-
than free inositol or that the insulin-promoted increase in
lated perfused dog kidney (Troyer et al. 1986). Patients with
newly synthesized D-chiro-inositol was rapidly converted into
chronic renal failure demonstrate a systemic build-up of
lipids. However, these results may be artefacts brought about
inositol which resolves following renal transplantation,
by acid treatment and further research is needed.
showing that healthy kidney function is required for inositol
excretion (Clements Jr 1979). Nervous tissue can also catab-
Controversy about epimerase activity olize myo-inositol and MIOX is highly expressed in the sci-
One study has proposed that the function of the myoinositol atic nerve, pigmented epithelium and lens epithelium
epimerase in the previously described rat, ovarian and fibro- suggesting that local inositol content needs to be closely
blast studies had been misidentified, due to the mis- regulated for optimal nervous function (Arner et al. 2006).
1638 O. C. WATKINS ET AL.

Inositol transport proteins Davanzo et al. 2001). Neutrophils in bipolar I disorder, but
not bipolar II disorder, expressed higher SMIT mRNA than
Inositol is hydrophilic and requires transporters to cross
controls (Willmroth et al. 2007). These findings suggest
membranes. Intracellular inositol concentrations are there-
specific regional changes in inositol transport rather than
fore dependent on the expression and activity of transport-
general physiological deficiency in mood disorders
ers. The high levels of inositol in the brain and the female
(Willmroth et al. 2007; Frey et al. 1998; Moore et al. 1999;
reproductive organs are thought to be due to high inositol
Moore et al. 2000; Silverstone, McGrath, and Kim 2005).
import, rather than high local inositol synthesis (Thomas,
Mood stabilizers lithium, valproate and carbamazepine
Mills, and Potter 2016; Berry et al. 1999; Lewin et al. 1982; Di
Daniel et al. 2009; Bourgeois, Coady, and Lapointe 2005; inhibit SMIT activity in astrocyte cultures, and decrease
Michaelis et al., 1993; Frej, Otto, and Williams 2017). Sodium/ brain inositol levels in bipolar disorder, suggesting a possible
Myo-Inositol Transporters (SMIT) are expressed in multiple mode of action for these medications (Calker and Belmaker,
tissues among Animalia (Zerbino et al. 2018; Schneider 2015), 2000; Silverstone, McGrath, and Kim 2005; Harwood 2005;
while proton (Hþ)/Myo-Inositol symporters (HMIT) are found Lubrich and van Calker 1999; Calker and Belmaker, 2000;
in all living organisms and are closely related to the GLUT Wolfson et al. 2000).
family of sugar transporters (Schneider 2015; Mueckler and In the diabetic rat model of acute stage streptozocin-
Thorens 2013). SMIT and HMIT are symporters, which induced hyperglycemia SMIT expression increased in the
require a sodium or proton gradient respectively to pump hippocampus, while SMIT-1 knockout mice display reduced
inositol into cells. The affinity of different inositol isomers to inositol in the frontal cortex and hippocampus (Yamashita
each transporter is summarized in Table 2. et al. 1998). During human pregnancy, protein expression of
Regulation of inositol transport is important for maintaining SMIT2 and HMIT in the placenta were downregulated with
cell osmolality. SMIT-1 is upregulated by intracellular hyperton- increasing maternal glycemia and this is thought to contrib-
icity and downregulated by hypotonicity (Schneider 2015). ute to lower placental inositol in GDM compared to controls
Inositol transport is also affected by volume sensitive organic (Pillai et al. 2020). Such changes may be directly attributable
osmolyte anion channels, but the underlying mechanisms are to glucose since glucose treatment of placental explants in-
poorly understood (Strange et al. 1994; Isaacks et al. 1999; vitro downregulated SMIT2 mRNA expression (Pillai et al.
Jackson and Strange 1993). Other inositol transporters are 2020). Understanding the regulation of transporter activity
found in plants, fungi and yeasts, but these have not yet been is therefore important for understanding inositol-related
described in animals and humans (Schneider 2015). pathology and potential side-effects of inositol supplements
Competition between inositol isomers and other saccha- outside the target tissue.
rides for inositol transporters affects the relative uptake of
each inositol isomer. For example, the uptake of myo-inosi- Oral intake and absorption of inositols
tol by SMIT-1 is inhibited by scyllo-inositol in isolated
bovine cardiac sarcolemmal vesicles (Rubin and Hale 1993), Inositol hexaphosphate (phytate) is an important dietary
and by scyllo-inositol and a range of other saccharides in source of myo-inositol found in cereals and legumes
human embryonic HEK293 kidney cell-line (Fenili 2010). (Schlemmer et al. 2009). High fiber diets are rich in phytates
This raises the possibility that a divergence from a normal and it has been suggested that the health benefits of such
ratio of inositol isomers and glucose in the circulation or diets may be due to inositol (Prynne et al. 2010). Phytate is
renal filtrate could impact local inositol uptake and supply. broken down by intestinal microbes to release underivatized
inositol, which is then taken up by inositol transporters in
Inositol transport in pathological conditions the small intestine (Grases et al. 2001; Steer and Gibson
Disrupted inositol transport has been observed in multiple 2002). Experiments in rats have suggested that SMIT-2 in
pathological conditions, mostly involving the central nervous particular maybe involved in intestinal inositol uptake
system. For example, particular single nucleotide polymor- (Aouameur et al. 2007). Some studies hypothesized that
phisms within HMIT are associated with Parkinson’s disease mammals may also absorb phytate directly, but more recent
(Satake et al. 2009; Gao et al. 2012). Meanwhile, the SMIT-1 work in HeLa cells suggests that most physiological phytate
gene, found on chromosome 21 (Berry et al. 1995) and is synthesized in-situ (Letcher, Schell, and Irvine 2008).
likely over-expressed in Down syndrome (trisomy 21) Phytate was classically thought to decrease the dietary bio-
(Schneider 2015; Berry et al. 1995; Fruen and Lester 1991; availability of zinc, iron and copper ions, but recent studies
Guo et al. 1997), may contribute to inositol elevation in suggest that phytate does not usually affect mineral status in
brain, cerebrospinal fluid, and amniotic fluid (Berry et al. humans (Grases et al. 2001). Dietary phytate confers protec-
1999; Lamar et al. 2011; Santamaria et al. 2014) and an tion against kidney stones, colon, lung and mammary can-
increased risk of Alzheimer’s disease (Lamar et al. 2011). cer, and has both anti-oxidant and hypocholesterolaemic
Depressive symptoms are associated with decreased effects (Grases et al. 2001). However, it is unclear whether
myo-inositol in the frontal cortex of the brain (Willmroth these effects are due to phytate itself or due to increased
et al. 2007; Frey et al. 1998; Moore et al. 1999; Moore et al. inositol availability (Grases et al. 2001). Dietary inositol may
2000; Silverstone, McGrath, and Kim 2005; Coupland et al. also originate from lipid-bound inositols, pinitol or 6-
2005), while increased inositol in the cingulate cortex was b-galactinol (Holub 1986; Croze and Soulage 2013). Lipid-
observed in children with mania (Davanzo et al. 2003; bound inositols are particularly important, making up 56%
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1639

Figure 3. The availability of phosphoinositides (PI), phosphoinositide phosphates (PIP) and inositol phosphates, and therefore the activity of many signaling proc-
esses, is tightly regulated by metabolism. Phosphates may be selectively and specifically added to PI by kinases, or removed by phosphatases, to produce PIP such
as PIP1 and PIP2. These PIPs are cleaved by lipases to form inositol phosphates and diacylglycerol (DAG).

of the inositol in a typical American diet (Holub 1986; Phosphoinositide phosphates (PIP) and inositol
Croze and Soulage 2013). phosphates
Phosphoinositide phosphates (PIPs) and inositol phosphates
Inositol derivatives are influential and highly energetic signaling compounds
that regulate diverse processes (Croze and Soulage 2013;
Phosphoinositides (PI) Balla 2013; Bitsanis et al. 2005). The availability of these
Inositols can be derivatized with phosphoglycerol and either transient compounds is rapidly and tightly controlled by the
one or two fatty acids to form lyso-phosphoinositides (LPI; activity of kinases, phosphatases and lipases (illustrated for
also known as lyso-phosphatidylinositols) and phosphoinosi- PIP1 and PIP2 in Figure 3), enabling cells to rapidly
tides (PI; also known as phosphatidylinositol) respectively respond to constantly changing environments (Farese 1983).
(Traynor-Kaplan et al. 2017). PIPs are produced when the inositol component of PI and
PIs are generally found at relatively low concentrations LPI lipids is phosphorylated by kinases (Hansen 2015; Czech
making up only 2–12% of the total phospholipids in mam- 2000). Multiple phosphates may be selectively and specifically
malian tissues (White 1973; D’Souza and Epand 2014), but added in a variety of configurations and the phosphate moi-
these lipids are vitally important in maintaining cell archi- eties of PIPs can be further phosphorylated to form high
tecture, membrane function and dynamics (Di Paolo and De energy pyro-phosphates (Hansen 2015; Czech 2000). PIPs can
Camilli 2006; Shewan, Eastburn, and Mostov 2011). be hydrolyzed by phospholipase C (PLC) to produce a wide
PI species are enriched in poly-unsaturated fatty acids range of inositol phosphates, pyro-phosphates and diacylgly-
(PUFA), particularly at the SN2 position and 70% of all PI cerols (DAG), (Hansen 2015; Czech 2000). Inositol phos-
species contain both arachidonic acid (an important phates can also be produced by direct phosphorylation of
omega-6 PUFA) and oleic acid (Traynor-Kaplan et al. inositol (as evident in slime molds and plants) (Letcher,
2017; D’Souza and Epand 2014). PI metabolism therefore Schell, and Irvine 2008), or from the further phosphorylation
regulates the cellular storage and availability of PUFA and of existing inositol triphosphates (in all organisms) (Letcher,
the synthesis of PUFA derived signaling molecules such as Schell, and Irvine 2008; Shears 2015). The relative amount of
eicosanoids (Traynor-Kaplan et al. 2017; D’Souza and PIPs and inositol phosphates compared to PI in biological
Epand 2014). Myo-inositol, D-chiro-inositol, scyllo-inositol systems is difficult to quantify, because these molecules occur
and muco-inositol have all been found in PI in mammals, at very low concentrations and are very susceptible to degrad-
but the relative abundance of each isomer in such lipids ation during processing (Wenk et al. 2003; Fukami and
and the biological importance of this variation remains Takenawa 1989; Pettitt et al. 2006).
unknown (Fenili et al. 2007; Freedman et al. 2014; Behuria PIPs and inositol phosphates act as powerful and versatile
et al. 2018). signals that mediate processes as varied as cell proliferation,
1640 O. C. WATKINS ET AL.

apoptosis, metabolism and motility (Hansen 2015; Balla GPIs and GPI-anchored proteins are particularly import-
2013; Berridge and Irvine 1989). Individual PIPs and inositol ant in signal transduction and vesicular trafficking (Paulick
phosphates, have distinct bioactivities, but most provide and Bertozzi 2008). At a cellular level, membrane GPIs in
phosphate-ester based energetic currency for kinases (Shears lipid rafts and caveolae are important for lipid raft function,
2015) and many regulate Ca2þ signaling pathways (Hansen potocytosis and the regulation of Kþ and Ca2þ transport
2015; Czech 2000). Cleavage of PIP2 for example forms (Reeves, Thomas, and Smart 2012; Noble, Zhang, and Wray
inositol 1,4,5- trisphosphate (IP3), which stimulates protein 2006), while cleavage of GPI-anchored proteins by phospho-
kinase C, inducing a cascade of kinase regulated changes lipases releases a range of signaling proteins (Low 2000).
(Van Sande et al. 2006; Sayers and Hanyaloglu 2018; GPIs are also involved in many key physiological functions
Berridge 2009). Interestingly, scyllo-inositol phosphates were of the body, as well as pathophysiological processes. GPIs in
found to act as agonists for myo-inositol-IP3 receptors, sug- sperm are essential for egg fertilization, while GPIs synthe-
gesting that inositol phosphates containing different isomers sized by macrophages modulate immune cell function
might have different, or even antagonistic activities (Wilcox (Kondoh et al. 2005; Patrussi et al. 2013). During infection,
et al. 1998; Brautigan et al. 2005; Lampe and Potter 1993). GPIs produced by protozoan parasites such as Plasmodium
Many PIPs and inositol phosphates act as second messen- and Trypanosoma, induce the pathological production of
gers in hormone signaling pathways (Yamazaki, Zawalich, cytokines, chemokines and nitric oxide (NO) by the host
and Zawalich 2010; Blazer-Yost and Nofziger 2005; Manna eliciting clinical symptoms such as hypoglycemia, acidosis or
and Jain 2013). For instance, the binding of insulin to tyro- anemia (Debierre-Grockiego and Schwarz 2010). Several
sine kinase receptors activates phosphoinositide 3 kinase GPIs are also currently under investigation as malaria vac-
(PI3K), that phosphorylates PIP2 to form PIP3 which acti- cine candidates (Malik et al. 2020). The generation of abnor-
vates protein kinase B (PKB) causing a cascade of kinase mal GPI-anchored proteins has also been observed in prion
regulated effects (Lizcano and Alessi 2002). disease (Simons and Ehehalt 2002).
Inositol phosphates may show contrasting effects on insu-
lin pathways as part of homeostatic regulation. For example, Inositol phosphoglycans (IPGs) and inositol glycans
diphosphoinositol pentakisphosphate (IP7) increases insulin The lipid moiety of GPIs may be cleaved off by phospholip-
secretion, but impairs insulin signaling (Rajasekaran et al. ase C or D (PLC, PLD) to form inositol phosphoglycans
2018; Barker and Berggren 2013; Nagamatsu and Ohara- (IPGs) and inositol glycans (Huang et al. 1993; Larner,
Imaizumi 2007). In pancreatic b-cells, glucose increases the Brautigan, and Thorner 2010; Croze and Soulage 2013;
synthesis of IP7 by hexakisphosphate kinase1(IP6K1) from Kunjara et al. 1999; Nestler and Unfer 2015). Historically it
phytate (IP6). Increased IP7 then increases insulin secretion has been difficult to differentiate between IPGs and inositol
enabling IP7 to regulate first phase insulin release glycans, and these classes are often not distinguished in pub-
(Rajasekaran et al. 2018; Barker and Berggren 2013; lished work. IPGs and inositol glycans commonly act as
Nagamatsu and Ohara-Imaizumi 2007). However, IP7 itself insulin second messengers, but they can also act as insulin
appears to suppress PKB activity, resulting in reduced insu- mimics independently from insulin, whilst also having non-
lin signaling (Kim et al. 2019). Indeed a reduction in the insulin related effects (Larner, Brautigan, and Thorner 2010;
amount of IP7 via the knockdown of IP6K1 resulted in an M€ uller et al. 1998). These molecules are thought to regulate
increase in PKB signaling in many in-vitro experiments, enzymes by acting as catalytic chelators, coordinating the
while a total knockout of IP6K1 in mice was associated with traffic of Mn2þ and Zn2þ within cells, enabling them to
reduced insulin resistance and a phenotype resistant to control kinase activity and a cascade of downstream effects
high-fat diet-induced obesity and diabetes (Kim et al. 2019; (McLean et al. 2008).
Mackenzie and Elliott 2014). This creates an in-built auto- As well as having local effects, IPGs and inositol glycans
crine homeostatic mechanism where the direct effects of IP7 may be released into the circulation as signals to other tis-
on suppressing PKB counter-balances those of IP7-induced sues (McLean et al. 2008; Alvarez et al. 1991; Turner,
insulin secretion which would induce PKB activation and Chakraborty, and d’Alarcao 2005). The origin of IPGs and
ensures responses are timely, transient and highly sensitive inositol glycans in the circulation is still unclear, but the
to changes in the environment. process appears to be regulated by insulin (Scioscia 2017;
Scioscia et al. 2013; Kunjara, Greenbaum, et al. 2000).
Glucose consumption was associated with a spike in circu-
Glycosylphosphatidylinositols (GPIs) lating IPG (Baillargeon et al. 2006), while IPG release into
Glycans can be attached to PIs containing myo-inositol or muscle could be induced by the administration of insulin
D-chiro-inositol to form glycosylphosphatidylinositols during euglycemic-hyperinsulinemic-clamp studies
(GPIs) (Balla 2013; Tsai, Liu, and Seeberger 2012; Pak and (Kennington et al. 1990). Furthermore, IPG release by cul-
Larner 1992). GPIs impact diverse cellular processes and can tured placental trophoblast microvillous membranes could
interact with intracellular targets, or be transported across be induced by the addition of insulin to culture media
the cell membrane by transporters such as Glut2 to act as a (Scioscia et al. 2006; Scioscia et al. 2008).
“hormone” at a distance (Mariggi o et al. 2006). GPIs can be Insulin activates phospholipase C and D, so it has been
attached to proteins which enables these proteins to be suggested that increases in IPG and inositol glycans are due
anchored into membranes (Paulick and Bertozzi 2008). to increased cleavage of GPI, enabling IPGs to act as an
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1641

insulin second messenger (Kennington et al. 1990; Pak et al. Methylated inositol derivatives such as pinitol, sequoyitol and
1993; Goel and Azev 2009; Nascimento et al. 2006). bornesitol are well known plant secondary metabolites
However, IPG availability will likely also be regulated by (Owczarczyk-Saczonek et al. 2018), but the origin of mamma-
GPI availability, IPG transport and IPG metabolism and lit- lian pinitol is uncertain and could be dietary or endogenously
tle is known about how these processes are altered by insu- synthesized (Lin, Ma, Gopalan, et al. 2009).
lin. Experiments in primary rat hepatocytes demonstrated Another compound extracted from a malarial parasite P-
that radiolabeled IPGs can be transported into cells through IPG fraction was found to contain myo-inositol, phos-
an energy-dependent IPG transporter, but nothing else is phorus, galactosamine, glucosamine, and glucose (Elased
known about the transport of these molecules (Alvarez et al. et al. 2001). Meanwhile the makeup of A-IPGs is particularly
1991). IPGs may also affect cellular function by interacting unclear with some appearing to contain myo-inositol, glu-
with external cell surface receptors, with fluorescently- cosamine, galactose and ethanolamine (Larner, Brautigan,
labeled IPG stimulating lipogenesis in a rat adipocyte cell and Thorner 2010).
line, despite an inability to enter the cell (Turner,
Chakraborty, and d’Alarcao 2005).
IPGs influence glucose and lipid metabolism
IPGs alter glucose and lipid metabolism in similar ways to
P-IPGs and A-IPGs insulin, leading these molecules to be classified as insulin
The structure of most IPGs and inositol glycans is unknown mimics. For example, both P-IPG and A-IPG fractions and
(Larner, Brautigan, and Thorner 2010; M€ uller et al. 1998) so INS-2 (a synthetic P-IPG) stimulated the incorporation of
these molecules are classified based on their biochemical prop- labeled glucose into glycogen in rat diaphragm muscles,
erties and our ability to separate them into two fractions. The hepatoma cells, erythroleukemia cells and rat primary adipo-
first fraction (P-IPGs) elutes at pH 2.0 and activates pyruvate cytes (Huang et al. 1993; Larner et al. 2003; Lazar et al.
dehydrogenase (an enzyme that links the glycolysis metabolic 1994; Lawrence, Guinovart, and Larner 1977). Furthermore,
pathway to the citric acid cycle via the production of acetyl- P-IPG fractions purified from human plasma or bovine liver
CoA), while the second fraction (A-IPGs) elutes at pH 1.3 and and synthetic INS-2 stimulated glucose oxidation and lipo-
inhibits protein kinase A (an enzyme that modulates glucose genesis in a variety of model systems (Larner, Brautigan,
and lipid metabolism) (Larner, Brautigan, and Thorner 2010). and Thorner 2010; Kunjara, Greenbaum, et al. 2000; Elased
These IPG fractions likely contain both IPGs and inositol gly- et al. 2001; Larner et al. 2003; Galasko et al. 1996).
cans, but little is known about the relative amounts of each. Moreover, the A-IPG fraction tested in a variety of assays
Generally, the levels of P-IPG and A-IPG have been quantified also increased lipid synthesis by decreasing the amount of
by testing these fractions using enzymatic assays (e.g. for pyru- fatty acids directed toward b oxidation by activating acetyl-
vate dehydrogenase activation activity), or by measuring the CoA carboxylase (ACC) (Huang et al. 1993; Kunjara et al.
amount of inositol in these fractions by techniques such as Gas 1999) and lowered lipolysis by activating cAMP-phospho-
Chromatography Mass Spectrometry (GCMS) (Paine et al. diesterase (Larner, Brautigan, and Thorner 2010; Kunjara
2006). An antibody-mediated assay is also available, but only et al. 1999; Scioscia 2017; Caro et al. 1997; Scioscia, Gumaa,
for a pregnancy-specific P-IPG, and this test only works in and Rademacher 2009; Witters and Kemp 1992).
urine and not plasma or serum samples (Paine et al. 2003). P-IPG can also stimulate the activity of several other
Hence, the development of more discriminatory tests are sorely regulatory molecules within the insulin signaling pathway
needed to better characterize IPGs and inositol glycans. including Phosphatidyl Inositol 3-Kinase (PI3K), Protein
P-IPGs and A-IPGs were classically thought to contain D- Kinase B (PKB) and Insulin Receptor Substrate (IRS) pro-
chiro-inositol and myo-inositol respectively, but current evi- teins (Larner, Brautigan, and Thorner 2010; Kunjara et al.
dence suggests that both classes contain a range of inositol 1999; Scioscia 2017; Varela-Nieto, Le on, and Caro 1996;
and inositol phosphate isomers bound to a range of glycans Burton, Scioscia, and Rademacher 2011) and thus could
and phosphoglycans, and that many of these compounds potentially enhance insulin actions, as well as act as insulin
have different effects even when they are from the same mimics in their own right. INS-2, for example was just as
fraction (Asplin, Galasko, and Larner 1993; Elased et al. effective as insulin in stimulating testosterone production by
2001). P-IPG fractions from human hemodialysate have a D- human ovarian thecal cells in-vitro, and the effect of insulin
chiro-inositol:myo-inositol ratio of 1.19, whilst those from could be blocked by an anti-INS-2 polyclonal antibody sug-
skeletal muscle extracts have a ratio of 9.42 (Asplin, Galasko, gesting that INS-2 is critical to this signaling pathway
and Larner 1993) suggesting that IPG composition varies (Nestler et al. 1998).
between biological fluids and tissue types. IPGs can also show anti-insulin-like effects. For example
The composition and structures of some IPGs have been the A-IPG fraction was found to inhibit adenylate cyclase
investigated more closely. For instance, one compound named (which is normally activated by insulin) and protein kinase
INS-2 was isolated from the P-IPG fraction of bovine liver and A (which normally stimulates insulin secretion) in multiple
was found to contain galactosamine, pinitol and Mn2þ assays (Larner, Brautigan, and Thorner 2010; Kunjara et al.
(Larner, Brautigan, and Thorner 2010; Larner et al. 2003)). 1999; Scioscia 2017; Caro et al. 1997; Scioscia, Gumaa, and
The structure of this molecule was subsequently confirmed by Rademacher 2009; Villalba, Kelly, and Mato 1988; Scioscia,
the synthesis of an analogue with identical biological properties Gumaa, et al. 2007). The A-IPG fraction could also decrease
(Larner, Brautigan, and Thorner 2010; Larner et al. 2003). glucose oxidation by antagonizing the in-vitro stimulation of
1642 O. C. WATKINS ET AL.

the PDH phosphatase by P-IPG (Kunjara et al. 1999; Chu and Hegsted 1980). In many rat experiments, myo-
Kunjara, Greenbaum, et al. 2000). IPGs also modulate the inositol deficiency was associated with increased adipose tis-
actions of other hormones besides insulin, with A-IPG sue lipid mobilization and increased hepatic triacylglycerol
inhibiting leptin release from cultured non-pregnant rat adi- (TAG) and cholesterol ester (CE) accumulation while myo-
pocytes (Kunjara, Greenbaum, et al. 2000). IPGs are there- inositol supplementation reversed these effects (Hayashi
fore important regulators of both glucose and lipid et al. 1978; Andersen and Holub, 1980; Burton and Wells
metabolism through insulin-dependent and independent 1976; Best et al. 1946; Gavin and McHenry 1941).
mechanisms (Nestler et al. 1998). Alterations to lipid metabolism appeared fatty acid specific,
and in rats fed inositol-deficient diets liver phospholipids
contained less linoleic acid whilst TAGs contained more pal-
Synthetic GPIs and IPGs and the relationship between
mitoleic acid (Andersen and Holub 1976; Andersen and
structure and function
Holub, 1980). Increased lipid storage in adipose tissue,
Since the isolation and structural elucidation of natural IPGs
rather than ectopically in liver, muscle or other organs is
has been difficult, chemists have produced a range of syn-
known to associate with decreased cardiometabolic adversity,
thetic GPIs, IPGs and inositol glycans in order to better
suggesting that this myo-inositol effect may be beneficial
understand structure-function relationships and identify pos-
(Hayashi et al. 1978; Gaggini, Saponaro, and Gastaldelli
sible drug candidates (Larner, Brautigan, and Thorner 2010;
2015). However, overall increased obesity could itself lead to
Goel and Azev 2009; Suzuki et al. 2014). While many of
negative outcomes and even alter responses to myo-inositol.
these candidates showed insulin mimetic activity in various
assays, the strength of the effect, and whether the effect was
on glucose, glycogen or lipid metabolism varied by molecu- Differential lipid storage responses to myo-inositol
lar structure (Larner, Brautigan, and Thorner 2010; Goel with obesity
and Azev 2009; Suzuki et al. 2014). These effects were The effects of myo-inositol on adipose tissue appear to be
reviewed by Goel et.al. (Goel and Azev 2009), who found opposite in obese rodent models compared to non-obese
that high insulin-like activity was more often found for models. Myo-inositol supplementation of a non-pregnant
compounds containing a cyclic phosphate on the inositol, an high fat diet obese adult mouse model was associated with
unacylated amino group on the second sugar, and a reduced fat accretion in white adipose tissue and a partial
phosphonate, phosphate or sulfate on one or more of the normalization of plasma leptin concentrations (Croze,
mannose residues (Goel and Azev 2009). Hence, it seems Gelo€en, and Soulage 2015). Similarly, myo-inositol supple-
likely that natural GPIs and IPGs will play a wide range of mentation in obese PCOS patients, was found to signifi-
roles in glucose and lipid metabolism and insulin signaling. cantly decrease body mass index (BMI) and circulating
However, these will not be fully understood until the effects leptin concentrations (Gerli et al. 2007). However, it is
of individual IPGs rather than broad class effects can be reli- unclear whether this reduction in adipose tissue fat accretion
ably quantified. is protective (from obesity-related pathology), or whether it
is harmful (by increasing ectopic lipid accretion). It is not
known why obesity alters the effect of myo-inositol on adi-
Inositols influence lipid metabolism, transport
pose tissue. Adipocytes from obese populations tend to be
and storage
more insulin resistant and have low liposynthetic capacity
Inositol and its derivatives regulate lipid mobilization, trans- and high lipolytic capacity (Zhang and Zhang 2010), and
port and storage, but effects vary depending on the organ thus might be less likely to respond to insulin-dependent
and model studied. There is some evidence that myo-inosi- inositol effects while being more likely to release free fatty
tol supplementation reduces circulating triglycerides, and acids. Adipose tissue in obese populations also shows more
total and LDL cholesterol levels in patients with metabolic pro-inflammatory activity (Zhang and Zhang 2010), which
diseases such as diabetes, GDM, PCOS and metabolic syn- may be altered by inositols. Indeed other studies have sug-
drome (Tabrizi et al. 2018). Myo-inositol supplementation gested a role for inositol in many immune and inflammatory
appears to alter body wide lipid distribution and, in general, processes (Krystal 2000; Miller, Chamberlain, and Cooke
causes more lipids to be stored in adipose tissue and less to 2008). Hence, obesity modifies the responses of adipose tis-
be stored ectopically in the liver and other areas, but this sue to myo-inositol, and understanding how this occurs will
varies depending on the model studied as discussed below enable the development of strategies to enhance the benefits
(Hayashi et al. 1978). of myo-inositol supplementation in obese populations.

Lipid storage and myo-inositol in animal models Myo-inositol status and lipid mobilization during preg-
Various animal models have been utilized to examine myo- nancy and lactation
inositol’s effect on lipid storage and body-wide distribution. Myo-inositol and lipid mobilization have also been specific-
Myo-inositol deficiency in gerbils caused a decrease in ally investigated in the context of pregnancy and lactation.
plasma lipids and lipoproteins, but increased lipid storage in Rats given a myo-inositol deficient diet during pregnancy
large lipid droplets in intestinal mucosal cells leading to the until 49 days after the birth, developed fatty liver only dur-
development of intestinal lipodystrophy (Hegsted et al. 1973; ing lactation and these changes were reversed by treatment
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1643

with inositol or by the cessation of lactation (Burton and esters into the blood stream and increased hepatic uptake of
Wells 1977). This suggests that myo-inositol deficiency is plasma cholesterol and cholesterol biliary secretion (Burgess
more dangerous during periods of massive lipid mobiliza- et al. 2003; Stamler et al. 2000). Plasma from rabbits treated
tion from adipose tissue, such as during lactation (Oben with PI also showed lower LCAT activity within 10 minutes of
et al. 2010; Vernon 2005). The livers of these myo-inositol- PI injection. Since PI will not be converted into myo-inositol
deficient lactating rats showed increased numbers of intra- in this short period, it seems likely that PI, rather than unes-
cellular lipid droplets, which were larger and contained terified inositol, is responsible for altered cholesterol metabol-
more cholesterol-ester and TAG, but less cholesterol and PI ism. Inositols also appear to regulate the synthesis and
(Burton and Wells 1977). Interestingly, no difference in lip- hydrolysis of PI (Strieleman et al., 1992; Wentzel et al. 2001;
ids was seen in kidney or intestinal tissues, suggesting a Salman et al. 1999). Scyllo-inositol for example inhibited the
liver-specific effect (Burton and Wells 1977). Lactating rats incorporation of myo-inositol into PI and the hydrolysis of
supplemented with inositol and given radiolabeled palmitic
PIP into myo-inositol phosphates in the developing rat concep-
acid showed decreased radiolabeled liver TAG, but increased
tus (McPhee, Downes, and Lowe 1991). D-chiro-inositol mean-
radiolabeled serum TAG compared to animals given an
while, was found to be a potent inhibitor of mycobacterial PI
inositol-deficient diet (Burton and Wells 1979). Together
synthase (Salman et al. 1999). Overall, how inositols and their
these findings again suggest inositol selectively modulates
derivatives are involved in lipid metabolism in different species,
lipid transfer and storage around the body producing differ-
ent effects in different organs and different effects depending metabolic conditions, tissue types, and various stages of devel-
on the population studied. opment is still not well understood and further research is
necessary. The effects of inositol supplementation on lipid
metabolism in GDM pregnancy will be discussed in section
Mechanisms of myo-inositol regulation of lipid mobiliza- “Inositol and fetal development.”
tion and distribution
Myo-inositol-deficient rats showed increased hepatic fatty
acid synthetase and acetyl-CoA carboxylase (ACC) activity, The anti-oxidant effects of inositol
which could explain the increase in hepatic lipogenic activity
(Beach and Flick 1982). Myo-inositol deficiency in yeasts Inositols can act as anti-oxidants and reactive oxygen species
also increased ACC activity, resulting in an accumulation of (ROS) scavengers (Valluru and Van den Ende 2011; Hu,
neutral lipids and this effect was reversed when the defi- Chen, and Lin 1995). In Jian carp, myo-inositol was shown to
ciency was rectified by myo-inositol treatment (Hayashi, increase enzymatic anti-oxidant capacity by altering the activ-
Hasegawa, and Tomita 1976). In contrast, mouse adipocyte ity of catalase, glutathione peroxidase and glutathione reduc-
cells treated with myo-inositol in-vitro showed increased tase superoxide dismutase and glutathione-S-transferase (Jiang
lipid accumulation due to increased fatty acid synthase et al. 2009). Furthermore one small study also suggested that
expression and reduced lipolysis (Kim, Han, and Kim 2014). there is an increase in thiol oxidation and oxidative stress in
Collectively, these studies suggest that myo-inositol may the follicular fluid of patients with PCOS compared to con-
potentially reduce ectopic fat deposition by mobilizing lipid trols and that this oxidation was reduced by the administra-
stores in the liver, which is an ectopic site, and increasing tion of D-chiro-inositol (De et al. 2012; Dona et al. 2012).
adipose tissue stores, although this needs to be validated in Uncomplicated pregnancy, pre-eclampsia, diabetic pregnancy,
further studies. GDM and diabetes in non-pregnant populations are also all
In addition, myo-inositol effects may interact with the auto- associated with increased oxidative stress in general (Wentzel
nomic nervous system. Plasma adrenalin levels were higher in et al. 2001; Burton and Jauniaux 2004; Jenkins et al. 2000;
myo-inositol-deficient rats, suggesting that myo-inositol may
Eriksson and Borg 1991; Eriksson and Borg 1993; Wentzel,
affect the autonomic nervous system and hormonal regulation
Welsh, and Eriksson 1999; Trocino et al. 1995; Newsholme
(Hayashi et al. 1978). Moreover, administration of sympathetic
et al. 2007), but further research is needed to determine how
nervous system blockers to myo-inositol-deficient rats inhibited
this is related to inositol dysregulation.
hepatic lipid deposition and increased serum free fatty acids
Inositols are important for male fertility, testicular func-
(Holub 1986; Hayashi et al. 1978), indicating that sympathetic
tion and spermatogenesis (Steegers-Theunissen, Groenen,
nervous activity also modulates myo-inositol effects.
Inositol may also impact cholesterol and lipoprotein metab- and Beemster 2002; Condorelli et al. 2017) and a small clin-
olism (Hayashi et al. 1978; Burton and Wells 1977). Myo-inosi- ical study showed that myo-inositol supplementation could
tol-deficient rats showed increased plasma hormone-sensitive be used to treat idiopathic male infertility (Calogero et al.
lipase activity in epididymal adipose tissues, which would 2015). The biology behind this is not yet known, but it has
increase the local breakdown of circulating lipoproteins and been suggested that the anti-oxidant effects of inositol may
increase lipid uptake (Hayashi et al. 1978). Myo-inositol defi- play an important role (Colone et al. 2010). In particular,
cient rats also showed reduced plasma lecithin–cholesterol acyl- in-vitro myo-inositol treatment of spermatozoa reduced ROS
transferase (LCAT) activity suggesting decreased cholesterol activity and mitochondrial cristae damage and reduced the
ester and lipoprotein formation (Burton and Wells 1977; Wells number of spermatozoa covered with amorphous fibrous
and Hogan 1968). Additionally, intravenous injection of PI material, which gives an excessive viscosity to the seminal
into rabbits inhibited the transfer of hepatocyte cholesteryl fluid that increases subfertility (Colone et al. 2010).
1644 O. C. WATKINS ET AL.

Inositol in diabetes mellitus decreased activity in glycogen phosphorylase in skeletal


muscle (Ortmeyer, Huang, et al. 1993). This suggests D-
Inositol bioavailability is altered in diabetes
chiro-inositol itself could be involved in the regulation of
Circulating and urinary D-chiro-inositol and myo-inositol glycogen synthesis, which is known to be impaired in type 2
levels are altered in adult type 1 and type 2 diabetes popula- diabetes (Ashcroft et al. 2017; Cline et al. 1999). In humans,
tions compared with non-diabetic controls (Table 3), with urinary D-chiro-inositol was also strongly correlated with
most studies reporting decreased urinary D-chiro-inositol fasting plasma glucose, glycated hemoglobin and urinary
and increased urinary myo-inositol in both types of diabetes. glucose, and the urinary increase in D-chiro-inositol with
However, two studies on overweight and obese populations diabetes was reversed by insulin treatment (Ostlund et al.
found an increase in both urinary D-chiro-inositol and 1993). Although this suggests that increased D-chiro-inositol
myo-inositol (Ostlund et al. 1993; Jung et al. 2005). This dif- excretion may cause or be caused by insulin deficiency or by
ference was discussed by Larner et.al. 2010, who noted that hyperglycemia, it does not exclude the possibility that
D-chiro-inositol excretion also increased with increasing increased D-chiro-inositol excretion is an adaptive or pro-
obesity and insulin resistance in PCOS. A similar increase is tective response against diabetes-induced complications or
also observed in GDM as discussed in section “Maternal could result from diabetes-related secondary pathology.
inositols in an uncomplicated pregnancy and lactation.”
Taken together, it appears that diabetes, obesity and PCOS Tissue inositol content in diabetes. In humans, how tissue
may each have different and independent associations with inositol content is changed with diabetes is not known. A
circulating and urinary inositol concentrations. few studies (Asplin, Galasko, and Larner 1993; Kennington
et al. 1990) measured the myo-inositol and D-chiro-inositol
content in IPG fractions in human muscle tissue (see Table
Obesity alters the relationship between inositols and dia- 5), but IPGs represent only a small fraction of the total
betes. The differences in systemic inositol concentrations inositol present in cells and biological systems, and IPG
between obese and normal weight diabetes populations is also quantification cannot show if diabetes is associated with
apparent in animal studies (Table 4). Studies using rodent mod- general tissue inositol deficiency. In animal studies strong
els of diabetes associated with obesity such as streptozocin- evidence for myo-inositol depletion in diabetes is only pre-
treated rats and db/db mice, showed increased urinary D-chiro- sent for nervous tissue (Table 4). Further studies are needed
inositol and myo-inositol compared with non-diabetes controls to determine how inositol metabolism is changed in both
(Kawa, Przybylski, and Taylor 2003). In contrast, those from type 1 and type 2 diabetes and how this is altered by insulin,
the non-obese Goto-Kakizaki diabetic rat model showed oral hypoglycemic or lifestyle/dietary treatments. Findings
decreased urinary D-chiro-inositol and increased myo-inositol will depend on how well inositol isomers are separately
compared with controls (Suzuki et al. 1991). These findings fur- quantified and what inositol derivatives are included in the
ther support the idea that obesity may modify the associations extraction and analysis methods.
of inositol dysregulation with the diabetic state.
However, most previous human and animal studies did
not adequately adjust for BMI or bodyweight as a covariate, IPGS and diabetes
nor use weight-matched controls. Many studies also defined IPGs are a diverse group of compounds, which can regulate
their study populations by insulin-dependence which makes glycemic and lipidomic pathways (Larner, Brautigan, and
it difficult to discern the associations of inositol alterations Thorner 2010), and are altered in diabetes (Table 5).
with different subtypes of diabetes or treatments. Many of Urinary and tissue P-IPGs are generally decreased in type 2
the patients were also taking oral hypoglycemic agents, with diabetes (Asplin, Galasko, and Larner 1993; Kunjara et al.
or without insulin, but these populations were not analyzed 1999; Kennington et al. 1990), whilst urinary and tissue A-
separately. Further research will be necessary to determine IPGs are increased, but findings varied depending on the
how inositol bioavailability and action is altered in diabetes population studied and the analysis method (Table 5). In
in different patient populations, the direction of causality one study, the urinary P-IPG:A-IPG ratio was decreased in
and to determine whether urinary or circulatory inositols diabetes and the ratio was negatively correlated with a rise
could be used as a tool to screen for, monitor treatment and in systolic blood pressure, BMI and hemoglobin-A1c
assess progression of diabetes. (HbA1c), which are all markers of worsening metabolic syn-
drome (Kunjara et al. 1999). P-IPG has insulin-like effects
Inositol correlations with degree of insulin resistance. (Kunjara et al. 1999; Scioscia 2017; Scioscia, Gumaa, and
Changes in systemic and local inositol levels in diabetes are Rademacher 2009), so a decrease in P-IPG may promote
strongly correlated with disease severity and insulin resist- diabetogenic effects. A-IPG in contrast, may direct metabol-
ance. For example, in spontaneously diabetic rhesus mon- ism away from glucose oxidation and toward energy conser-
keys, the urinary D-chiro-inositol excretion rate is directly vation and lipid storage promoting a diabetic phenotype
correlated with insulin-mediated glucose disposal rates and (Kunjara et al. 1999; Scioscia 2017; Kunjara, Greenbaum,
glucose tolerance (Ortmeyer, Huang, et al. 1993). Urinary et al. 2000; Scioscia, Gumaa, and Rademacher 2009).
D-chiro-inositol was also correlated with increased glycogen However, P-IPG and A-IPG fractions both contain many
synthetase activity in skeletal muscle and adipose tissue, and different myo-inositol, D-chiro-inositol and pinitol
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1645

containing IPGs with a range of bioactivities. Until these PCOS. Furthermore, this review did not adequately differen-
can be separately analyzed, and their individual effects quan- tiate between the use of different types of inositol interven-
tified, it will be difficult to deepen our understanding of the tions, although most utilized myo-inositol.
role of these molecules in diabetes. Trials including D-chiro-inositol supplementation are
IPG concentrations display sex differences. Urinary A- scarce. One small trial involving twenty men with type 2
IPG is five-fold higher in healthy female urine than healthy diabetes showed that the administration of both myo-inosi-
male urine samples and there is a decrease in the P-IPG:A- tol and D-chiro-inositol in combination for three months,
IPG ratio in women compared with men (Kunjara, lowered fasting blood glucose and HbA1c levels compared
Greenbaum, et al. 2000), yet most studies do not take these to pretrial baseline (Pintaudi, Di Vieste, and Bonomo 2016).
sex differences into account. Further studies accounting for A separate 24 weeks’ long RCT involving 26 overweight
sex, type of diabetes, diabetic treatment and BMI are needed patients with type 1 diabetes tested the effect of D-chiro-
to understand how IPGs are altered in different tissues and inositol (1 g, plus 400 mcg folic acid daily) compared to a
bodily fluids in type 1 and type 2 diabetes. folic acid control (Maurizi et al. 2017). A significant reduc-
tion in HbA1c level was seen in the treated group compared
with control, but no significant reduction in BMI or insulin
Alterations in inositols in diabetes: pathological
requirements was observed (Maurizi et al. 2017).
or adaptive?
Several studies of patients with type 2 diabetes (Kim et al.
Although inositols participate in numerous insulin, glucose 2005; Kang et al. 2006; Kim et al. 2012; Kim et al. 2007) and
and lipid metabolic pathways, (Yamazaki, Zawalich, and non-diabetic adults (Hernandez-Mijares et al. 2013), sug-
Zawalich 2010; Manna and Jain 2013; Coustan 2013) it is gested that pinitol (3-O-methyl-D-chiro-inositol) can also
not known if these inositol perturbations have an etiological improve glycemic control. However, small studies in non-
role in any diabetic features, or if they are a consequence of diabetic older subjects (66 ± 8 years) (Campbell et al. 2004)
the pathology. These inositol-related changes could even be and non-diabetic obese subjects (Davis et al. 2000), sug-
protective. For example, myo-inositol-derived PIP3 enhances gested that pinitol treatment had no impact on fasting glu-
the transport of glucose into cells by stimulating the trans- cose, insulin-mediated glucose disposal, or plasma lipids.
location of GLUT4 to the cell membrane (Paul and Brady Therefore, whilst these initial small studies show promise
2015). One simple perspective is that a decrease in tissue and whilst there has been no mention of serious adverse
myo-inositol and consequently PIP3 could therefore effects, larger randomized controlled double-blind trials are
decrease glucose uptake and promote hyperglycemia, contri- required to determine whether treatment with any inositol
buting to diabetes pathology. However, a decrease in tissue isomer or inositol derivative could be used to reduce dia-
myo-inositol could also result in lower intracellular glucose betes risk, or to complement current established treatments
levels, protecting specific cells from glucose overload in the for diabetes and other conditions of insulin dysregulation.
context of hyperinsulinemic hyperglycemia. The association
between inositol perturbations and diabetes is also likely tis-
Inositols as treatment for diabetes in animal models
sue-specific and influenced by many factors such as type of
and possible mechanisms of action
diabetes, BMI or ethnicity.
D-chiro-inositol treatment lowered postprandial glycemia in
streptozocin-induced hyperglycemic rats, wild-type rats and
Inositols as treatment for diabetes in humans
rhesus monkeys with varying degrees of spontaneous insulin
In general, inositol supplementation appears to reduce gly- resistance (Ortmeyer, Huang, et al. 1993; Hansen and
cemia and increase insulin sensitivity, suggesting that inosi- Bodkin 1986). Furthermore, in obese insulin-resistant mon-
tol supplementation could suppress diabetic features even in keys, myo-inositol or D-chiro-inositol supplementation
the absence of inositol deficiency. A systematic review and decreased postprandial plasma glucose concentrations, while
meta-analysis that included 20 RCTs with a total of 1239 myo-inositol also decreased urinary glucose (Ortmeyer
subjects with different types of insulin resistant conditions, 1996). Meanwhile, the supplementation of streptozotocin
which studied the effects of inositol on glucose homeostasis, and high fat diet induced diabetic rats with myo-inositol
showed that inositol treatment decreased fasting plasma glu- decreased fasting glycemia, HOMA-IR and plasma insulin
cose (mean difference [MD]  0.44 mmol/l, 95% CI 0.65, compared with diabetic controls (Antony et al. 2017).
0.23), 2 h plasma glucose after a 75 g oral glucose load Supplementation of diabetic rats with myo-inositol has
(MD 0.69 mmol/l, 95% CI 1.14, 0.23), the incidence of also been associated with an improvement in several other
abnormal glucose tolerance (relative risk [RR] 0.28, 95% CI diabetic-associated changes including glomerular filtration
0.12, 0.66), fasting insulin (MD 38.49 pmol/l, 95% CI rate, serum urea, creatinine and arachidonic acid, urinary
52.63, 24.36) and the Homeostatic Model Assessment of albumin, IgG excretion, endoneural blood flow and motor
Insulin Resistance (HOMA-IR) (MD 1.96, 95% CI 2.62, nerve conduction velocity (Antony et al. 2017; Carlomagno
1.30) (Mi~ nambres et al. 2019). However, this review and Unfer 2011; Pugliese et al. 1990; Coppey et al. 2002;
included an extremely heterogeneous set of studies and did Solomonia et al. 2010; Mayer and Tomlinson 1983; Yue
not analyze the effects of inositol in diabetes separately from et al. 1989). In obese insulin-resistant monkeys, neither D-
the effects of other insulin resistant conditions such as chiro-inositol nor myo-inositol altered post-prandial
1646 O. C. WATKINS ET AL.

circulating insulin levels, suggesting that inositol may have (Stewart, Sherman, and Harris 1970), catabolism (Arner
an effect on downstream insulin pathways (Ortmeyer 1996). et al. 2006) and transport (Berry et al. 1999) processes are
In streptozotocin and high fat diet induced diabetic rats, particularly active in nervous tissue, this tissue is unlikely to
myo-inositol treatment increased the adipose tissue expres- be representative of changes occurring in other tissues
sion of PPARc and significantly upregulated GLUT4 and in diabetes.
insulin receptor signaling molecules, leading to the sugges-
tion that myo-inositol acts as a PPARc agonist (Antony Altered transporter protein activity. Tissue inositol content
et al. 2017). is regulated by inositol transporter proteins with varying
Injected intravenously, both A-IPG and P-IPG fractions affinities for different inositol isomers. These transporters’
from various natural sources (Elased et al. 2001; Larner different isomeric specificity (Table 2) may partly explain
et al. 2003) and synthetic INS-2 (Larner et al. 2003) could the different D-chiro-inositol and myo-inositol profiles in
also dose-dependently reduce hyperglycemia in streptozocin- diabetes. Meanwhile, at a systemic level, altered intestinal or
induced diabetic rats (Larner et al. 2003), db/db mice, and kidney transport could lead to altered inositol absorption
ob/ob mice (Elased et al. 2001), suggesting that IPGs may be and excretion, and hence, systemic inositol concentrations.
partly responsible for the insulin like effects of inosi- Skeletal muscle SMIT-2 activity is upregulated by insulin, as
tol treatment. evidenced by an 18-fold increase in D-chiro-inositol trans-
Myo-inositol may also decrease glycemia by reducing port in insulin-treated rat skeletal muscle cells that were
dietary glucose absorption in rats (Chukwuma, Ibrahim, and transiently transfected with human SMIT-2 (Lin, Ma,
Islam 2016). Myo-inositol was found to inhibit duodenal Gopalan, et al. 2009). In contrast, tumor necrosis factor a
glucose absorption, reduce post-prandial glucose excursions,
(TNFa), a critical factor in the pathophysiology of diabetes
delay gastric emptying and accelerate intestinal transit in
mellitus (Navarro-Gonzalez et al. 2009), caused a decrease in
both normal and diabetic (fructose-fed and streptozocin-
SMIT-1 mRNA expression and activity in adipocytes (Yorek,
injected) rats (Chukwuma, Ibrahim, and Islam 2016). These
Dunlap, and Lowe 1998). Differential alterations in locally
effects were observed both in ex-vivo studies where isolated
regulated inositol transporter activity could therefore lead to
rat jejunum were treated with increasing concentrations of
varying disruptions in inositol content in tissues.
myo-inositol and in-vivo in rats given oral myo-inositol co-
administrated with glucose (Chukwuma, Ibrahim, and
Dysregulated inositol metabolism. Inositol concentrations in
Islam 2016).
diabetes are locally controlled by the relative balance of
inositol synthesis, epimerization, derivatization and catabol-
Proposed mechanisms underlying inositol dysregulation ism. Glucose increased ISYNA1 expression in HepG2
in diabetes (human liver-derived) cell line, suggesting that diabetes may
increase hepatic myo-inositol synthesis (Guan, Dai, and
Myo-inositol depletion by increased excretion. It has been
Shechter 2003). Furthermore, myo-inositol catabolism
suggested that systemic myo-inositol deficiency could result
appears to be promoted by upregulation of MIOX in human
from increased urinary inositol excretion due to increased
competition with glucose for transporters during renal tubu- and porcine kidney cells (HK-2 or LLC-PK1) treated with
lar reabsorption of the glomerular filtrate (Daughaday and high glucose (Zhan et al. 2015; Xie et al. 2010), and by
Larner 1954; Kennington et al. 1990; Baillargeon et al. increased MIOX expression in the kidneys of mice fed a
2006), resulting in reduced inositol reabsorption (Asplin, high fat diet (Tominaga et al. 2016). The production of
Galasko, and Larner 1993; Thomas, Mills, and Potter 2016; many IPGs could also be increased in diabetes, since the
Bourgeois, Coady, and Lapointe 2005). However, hypergly- hydrolysis of membrane proteins, including GPI-anchored
cemia is not always associated with increased urinary inosi- proteins from which IPGs are released, by phospholipase C
tol excretion, nor with systematic inositol deficiency or is activated by transforming growth factor (TGF)-b, a factor
universally decreased tissue inositol (Table 4), and there is which is over-expressed in diabetes (Ziyadeh 2004; Vivien
variation between isomers, suggesting alterations in circulat- et al. 1993).
ing inositols in diabetes are due to more complex processes Myo-inositol to D-chiro-inositol epimerase is insulin sen-
(Loy et al. 1990). sitive with evidence of diminished conversion in-vivo in the
Strong evidence for myo-inositol depletion in diabetes is insulin resistant Goto-Kakizaki (GK) rat model of type 2
so far only evident for nervous tissue (Table 4). Myo-inositol diabetes, resulting in increased myo-inositol, but decreased
is consistently depleted in the nervous tissue of diabetic rats D-chiro-inositol in insulin sensitive tissues such as the liver,
and rabbits across studies (Greene et al. 1987; Loy et al. fat, muscle and blood, but not in the non-insulin-sensitive
1990; Palmano, Whiting, and Hawthorne 1977; Zhu and spleen, kidney, intestine, heart and brain (Sun et al. 2002;
Eichberg 1990; Greene, De Jesus, and Winegrad 1975; Pak et al. 1998). It has been proposed that a deficit of D-
Greene and Mackway 1986), while human retinal pigment chiro-inositol could impair the synthesis of IPGs containing
epithelial cells incubated in high glucose showed decreased D-chiro-inositol, explaining the observed D-chiro-inositol-
myo-inositol content and decreased PI synthesis (Thomas IPG deficiency in skeletal muscle, hemodialysate and urine
et al. 1994; Thomas et al. 1993; Del Monte et al. 1991; of patients with type 2 diabetes (Asplin, Galasko, and Larner
Nakamura et al. 1992). However, since inositol synthesis 1993). However, it is unclear whether D-chiro-inositol
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1647

availability is the rate limiting factor in D-chiro-inositol A meta-analysis of nine RCTs involving 496 women with
IPG synthesis. PCOS (Unfer et al. 2017) showed that inositol treatment
PI/PIP/Inositol-phosphate signaling pathways, which (myo-inositol or chiro-inositol) significantly decreased fasting
regulate many insulin-dependent processes, are also dysregu- insulin (Standardized Mean Difference (SMD) 1.021 mU/mL,
lated in diabetes. SHIP-2 for example, inhibits peripheral 95% CI: 1.791 to 0.251, P ¼ 0.009) and HOMA-IR
insulin signaling, by catalyzing the conversion of PIP3 to (0.585, 1.145 to 0.025, P ¼ 0.041). Interestingly, in obese
PIP2 (Soeda et al. 2010) and SHIP-2 knockout mice showed PCOS patients, myo-inositol administration appeared more
increased liver and skeletal muscle insulin sensitivity (Soeda effective in reducing insulin resistance (area under the curve
et al. 2010). SHIP-2 protein is significantly increased in the of OGTT) in those with higher fasting plasma insulin levels,
quadriceps muscle, epididymal fat tissue and brain, but not suggesting that inositol treatment may be most effective in
in the liver, of diabetic db/db mice relative to controls those who are more insulin resistant (Genazzani et al. 2012).
(Soeda et al. 2010; Hori et al. 2002). Furthermore, mutations Meanwhile, a separate meta-analysis of ten RCTs involv-
in SHIP-2 are associated with diabetes and the metabolic ing 362 women with PCOS showed that inositol supplemen-
syndrome in population studies (Ishida et al. 2006; Kagawa tation (myo-inositol or D-chiro-inositol) was associated with
et al. 2005; Marion et al. 2002), which may explain why significantly improved ovulation rate and increased fre-
some individuals have increased susceptibility to develop- quency of menstruation compared with placebo (Pundir
ing diabetes. et al. 2018). Favorable effects of myo-inositol on ovaries and
gametes could also potentially improve assisted reproduction
Other mechanisms. Receptors and effectors of inositol com- technology outcomes (Nestler and Unfer 2015; Bevilacqua
pounds and their derivatives are also dysregulated in dia- et al. 2015) and supplementation reduced the likelihood of
betes. For example, insulin-induced activation of glycerol-3- PCOS mothers developing GDM in one small study (D’anna
phosphate acyltransferase by D-chiro-inositol-containing et al. 2012). Some studies have also suggested that combined
IPGs appeared defective in the adipocytes of type II diabetic D-chiro-inositol and myo-inositol treatment may be more
Goto-Kakizaki rats and was associated with altered lipid syn- effective than myo-inositol alone (Colazingari et al. 2013;
thesis (Farese et al. 1994). Meanwhile, renal type I inositol- Nordio and Proietti 2012). However, more definitive studies
1,4,5-trisphosphate receptor expression was reduced in the are needed before clinical recommendations can be made.
kidneys of diabetic rats and mice (Sharma et al. 1999) and a
polymorphism in this gene was found to be a risk factor for
type 1 diabetes in a genetic mapping study (Swedish cohort) Inositol dysregulation in polycystic ovary syndrome
(Roach et al. 2006). In PCOS, urinary D-chiro-inositol was inversely correlated
with insulin sensitivity (assessed using multiple methods)
Polycystic ovary syndrome and was found to be a strong independent predictor of per-
ipheral insulin resistance (Baillargeon et al. 2006; Baillargeon
Polycystic ovary syndrome (PCOS) afflicts 6-10% of women et al. 2010). However, this association with insulin resistance
worldwide and is the most common endocrine disorder in is unlikely to be unique to PCOS, since in a study on Greek
young women (Goodarzi et al. 2011). PCOS is characterized women, urinary D-chiro-inositol was found to be correlated
by amenorrhea or oligomenorrhoea, ovulatory dysfunction, with hyperinsulinemia, even after correction for BMI and
hyper-androgenism and polycystic ovaries (Ehrmann 2005). waist-to-hip ratio regardless of PCOS status (Baillargeon
Women with PCOS are more insulin resistant than matched et al. 2008). It is not yet clear whether plasma and urinary
controls (Ehrmann et al. 1999) and have an increased risk of inositols deviate from normal in PCOS. The same study on
obesity, hirsutism, cardiovascular disease, type 2 diabetes, Greek women, showed no significant differences in plasma
GDM, obstetric complications and infertility (Unfer et al. and urinary D-chiro-inositol or myo-inositol between PCOS
2017). The role of inositol in PCOS has been reviewed by patients and controls (Baillargeon et al. 2008). Meanwhile, a
others (Unfer et al. 2017; Carlomagno, Unfer, and Roseff separate study on American women by the same group,
2011; Nestler and Unfer 2015; Regidor et al. 2018), so this showed that obese PCOS patients had higher urinary con-
review will focus on how inositol may influence insulin sen- centrations and lower plasma concentrations of D-chiro-
sitivity and glucose and lipid metabolism in PCOS. inositol compared with non-obese non-PCOS women, while
myo-inositol does not appear altered (Baillargeon et al.
2006). However, these changes in inositol content were not
Treatment of PCOS with inositol supplements
significant when adjusted for BMI and/or insulin sensitivity
Myo-inositol and D-chiro-inositol supplementation separ- again suggesting that they may be related to differences in
ately and in combination have been shown to improve insu- BMI or insulin sensitivity rather than PCOS itself. Follicular
lin sensitivity, glycemia and androgen status, circulating fluid and serum concentrations of myo-inositol were corre-
lipid profiles, blood pressure, and anthropometry in PCOS lated with oocyte quality and ovulatory activity in women
(Unfer et al. 2012; Facchinetti et al. 2015; Unfer et al. 2017; undergoing IVF suggesting that myo-inositol deficiency
Pundir et al. 2018; Formuso, Stracquadanio, and Ciotta could lead to impaired fertility (Chiu et al. 2002). Overall,
2015; Cianci et al. 2015; Lagana, Barbaro, and Pizzo 2015; these findings suggest that inositol dysregulation in PCOS
Nestler et al. 1999; Iuorno et al. 2002). could be involved in the insulin resistance and infertility
1648 O. C. WATKINS ET AL.

issues associated with the syndrome. However, further work inositol in plants (Vernon and Bohnert 1992), there is not
is required to untangle how inositol is linked with PCOS, yet evidence that this occurs in animals (Lin, Ma, Gopalan,
insulin resistance and BMI. et al. 2009). It is therefore not yet known whether an
increase in D-chiroinositol caused by supplementation or
increased epimerase activity could increase pinitol contain-
Inositol and IPG regulation in PCOS. The precise mecha-
ing P-IPGs.
nisms of inositol involvement in regulating insulin sensitiv- Inositols and their derivatives clearly play important roles
ity in PCOS remains poorly understood. In non-PCOS
in various aspects of the pathophysiology of PCOS, but fur-
women, glucose consumption is associated with a spike in
ther research is necessary to disentangle these details. In
circulating P-IPG thought to be caused by an insulin-
particular, most studies did not examine the effects of BMI
induced cleavage of GPIs (Baillargeon et al. 2006). Several
and a lack of suitable controls means it is unclear whether
studies have shown that patients with PCOS show reduced
effects are due to obesity, insulin resistance or PCOS.
glucose-stimulated release of P-IPG into the circulation and
that this reduction has been correlated with decreased insu-
lin sensitivity (Cheang et al. 2008; Baillargeon et al. 2006; Inositol in pregnancy
Baillargeon et al. 2010; Baillargeon et al. 2008). Since P-IPG
acts as an insulin second messenger and insulin mimic, Pregnancy is associated with increasing maternal insulin
decreased P-IPG release likely contributes to insulin resist- resistance as gestation progresses, reducing uptake of glucose
ance and obesity in these women. However, these studies do and lipids by maternal tissues and mobilizing resources to
not adequately control for variance in obesity status and fur- promote fetal nutritional supply, especially in the latter half
ther research is necessary to draw conclusions. Interestingly, of pregnancy (Napso et al. 2018). To drive such maternal
although metformin therapy decreased post-prandial serum adaptation to pregnancy, pro-diabetic biological signals
insulin concentrations in obese women with PCOS, gly- including hormones, adipocytokines and exosomes are
cemia-induced release of P-IPG was not altered (Baillargeon released into the maternal circulation from the placenta,
et al. 2004). This finding may suggest that P-IPG secretion with some originating from the fetus (Scioscia 2017;
is regulated by factors that do not overlap with those Kunjara, Greenbaum, et al. 2000; Jayabalan et al. 2017). Our
affected by metformin and raises the prospect of synergism understanding of these processes remains incomplete, but
in metformin and inositol combination therapy in PCOS. inositol appears to play an important role (Scioscia 2017;
It was initially hypothesized that the increased urinary Kunjara, Greenbaum, et al. 2000; Jayabalan et al. 2017).
excretion of D-chiro-inositol led to reduced systemic D-
chiro-inositol and D-chiro-inositol containing P-IPG Maternal inositols in an uncomplicated pregnancy
(Nestler et al. 1999). If this was true it would suggest oral and lactation
D-chiro-inositol supplementation could restore reduced D-
chiro-inositol-containing P-IPGs availability in women with To date, we know very little about how maternal inositols
PCOS and improve some clinical features (Nestler et al. change during gestation and after delivery. One older study
1999). However, D-chiro-inositol supplementation in women reported that the maternal serum concentration of myo-
with PCOS did not increase glucose-stimulated release of P- inositol remains stable across gestation and is similar to that
IPG (Cheang et al. 2008). Furthermore, there is no evidence in non-pregnant women, at around 21–26 mmol/L (Quirk
that increased urinary D-chiro-inositol excretion results in and Bleasdale 1983). However, this study would not have
systemic D-chiro-inositol deficiency, or that D-chiro-inositol captured all the different derivatives of inositol nor all inosi-
availability is the limiting factor in P-IPG production. tol stereoisomers in the circulation. In urine, there is a two-
At the local tissue level, ovarian thecal cells from women fold increase in P-IPGs in the third trimester of uncompli-
with PCOS show increased chiro-inositol:myo-inositol ratios cated pregnancies compared with non-pregnant controls
and increased epimerase activity compared to controls, sug- (Kunjara, Greenbaum, et al. 2000), and at least one of these
gesting that in PCOS an overactive ovarian epimerase con- P-IPGs is unique to the pregnant state (Paine et al. 2003). A
verts more myo-inositol into D-chiro-inositol (Heimark, cross-sectional study showed that urinary P-IPGs rise
McAllister, and Larner 2014). Increased D-chiro-inositol between the 18th and 42nd week of pregnancy (Scioscia,
excretion in PCOS could therefore be due to increased ovar- Gumaa, et al. 2007), suggesting that IPGs could play a role
ian synthesis and such possibilities need to be investigated in mediating the changes in glucose and lipid metabolism
to ensure that supplementation studies do not worsen a pos- that occur over this time period (Scioscia, Gumaa, et al.
sible D-chiro-inositol oversupply. 2007). Moreover, urinary P-IPG levels are highest prior to
Future studies also need to consider the effects of D- delivery and are significantly higher during labor than in
chiro-inositol supplementation on pinitol containing IPGs. non-laboring women, suggesting that P-IPGs may also be
Treatment with INS-2, a pinitol containing P-IPG (Larner, associated with labor onset and progress (Paine et al. 2003).
Brautigan, and Thorner 2010; Larner et al. 2003), increased Unfortunately, IPG research in pregnancy is hindered by
testosterone synthesis in PCOS thecal cells, suggesting that limitations in analytical techniques for measuring different
an increase in pinitol based P-IPGs could contribute to the IPGs. Currently, IPGs can be quantified by separating the
hyperandrogenism seen in PCOS (Nestler et al. 1998). A-IPG and P-IPG fractions and testing fractions for enzym-
However, although pinitol is synthesized from D-Chiro- atic activity or for inositol content (Paine et al. 2006), but
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1649

this is laborious, nonspecific and will only quantify some inositol is altered or regulated as gestation progresses
and not all IPGs. There is an antibody-mediated P-IPG (Sussman and Matschinsky 1988).
assay that is specific for one pregnancy-specific P-IPG, but
it works for urine samples and doesn’t work well for plasma Inositol transfer and transport. There was a consistently
or serum samples (Paine et al. 2003). higher inositol concentration reported in term umbilical
Human breast milk contains a very large amount of un- artery blood (75 mM (HPLC) (Brusati et al. 2005)) that flows
derivatized inositol (Table 1), but this falls over the first from the fetus to the placenta, compared with term umbil-
6 weeks after birth (Pereira et al. 1990). However, whether ical venous blood (64 mM), which flows from the placenta
this decrease reflects declining maternal inositol after preg- back to the fetus (Toh et al. 1987; Santamaria, Di Benedetto,
nancy, or decreasing infant need for inositol is not known. et al. 2016; Campling and Nixon 1954), which is highly sug-
Thus, overall, the changes in maternal inositol biology gestive of fetal to placental transfer of inositol.
across gestation and after delivery remain poorly Moreover, a stable isotope transport study, involving the
characterized. maternal administration of 2H6-inositol 2 hours prior to
cesarean delivery, indicated that only a small fraction (10%
or less) of fetal circulating myo-inositol was derived from
Placental and fetal inositols in pregnancy transplacental maternal supply in late pregnancy (Staat et al.
Placental inositol. The amount of measured inositol in 2012). Since, the concentration of inositol in both venal and
human placental tissue is influenced by the method of arterial cord blood is much higher than maternal blood
extraction, sample processing and the assay method. By gas (26 mM (Brusati et al. 2005)), this suggests that the fetus syn-
chromatography, the human placenta was found to contain thesizes substantial amounts of inositol and that there is net
0.53 mmol/kg of un-derivatized inositol (Toh et al. 1987) fetal inositol export to the placenta.
and by enzymatic assay, to contain 2.7 mmol/kg of un-deriv- Inositol concentrations in umbilical cord blood obtained
atized and 3.5 mmol/kg total acid hydrolyzable inositol, by cordocentesis at mid-gestation (125 mM (Quirk and
Bleasdale 1983)) and from neonates born preterm (108 mM),
which includes all un-derivatized and derivatized forms
was significantly higher than at term (86 mM) (Pereira et al.
(Islam, Selvam, et al. 2019) (Table 1). These amounts are
1990) suggesting a decline in fetal circulating inositol with
similar to those reported in rat placenta (0.7–1.7 mmol/kg
advancing gestation. In rabbits and kittens circulating inosi-
(Battaglia et al. 1961)) and rat liver (0.57 mmol/kg
tol falls rapidly in the first month post-delivery reaching
(Palmano, Whiting, and Hawthorne 1977)), but much lower
equilibrium after 28 days (Campling and Nixon 1954).
than rat brain or kidney (Table 1) (Palmano, Whiting, and
Taken together, these studies suggests that the fetus may
Hawthorne 1977). Little is known about the types or pro-
export declining amounts of inositol to the placenta with
portions of inositol isomers in placenta, but current studies
advancing gestation. It is not known whether this decline is
suggest myo-inositol is the predominant form (Islam,
due to inositol being more important for fetal-placental
Selvam, et al. 2019). development early in pregnancy or whether this decrease
acts as an important regulatory signal in maternal-placental-
Fetal inositol. Substantial synthesis and metabolism of myo- fetal communication.
inositol occurs in fetal tissues suggesting an important role
for inositol in fetal development. In sheep and goats, fetal Inositol transporters SMIT-1 (Berry et al. 1995), SMIT-2
circulating inositol is substantially higher than that of adults (Roll et al. 2002) and the SMIT signal transcription factor
(Table 1D), whilst fetal CNS, kidney and skeletal muscle TonEBP are highly expressed in the placenta (Shin et al.
inositol levels are at the upper end of corresponding adult 2012; Trama et al. 2000), suggesting placental inositol trans-
tissues, whereas fetal liver and heart inositol levels are gener- port between the maternal-placental-fetal compartments is
ally lower than in adults (Table 1A). important. Naþ concentrations are generally higher outside
The human fetal lung and liver also showed high IMPA1 the cell than inside the cell due to the action of the Na/K-
activity (myo-inositol synthesis) (Quirk and Bleasdale 1983) ATPase, so SMIT (a sodium-inositol symporter) likely trans-
and in rabbit fetal kidney both ISYNA1 (myo-inositol syn- ports inositol into the placental syncytiotrophoblast cell bar-
thesis) and MIOX (myo-inositol catabolism) were highly rier from both maternal and fetal compartments (Schneider
expressed (Bry and Hallman 1991). This is consistent with 2015; Johansson, Jansson, and Powell 2000). The proton gra-
the possibility of important roles for inositol in fetal devel- dient between the maternal-placental-fetal compartments is
opment. Relatively high inositol concentrations were also not constant throughout gestation, and in the presence of
found in human second trimester amniotic fluid (79.0 mM, other inositol efflux transporters, the net directionality of
GCMS), suggesting that the fetal kidney can excrete inositol inositol transport through HMIT (a proton-inositol sym-
(Santamaria, Di Benedetto, et al. 2016). The presence of porter) remains unclear (Schneider 2015; Strange et al. 1994;
inositol synthesis, metabolism and excretion indicates that Isaacks et al. 1999; Jackson and Strange 1993Ganapathy
the fetus can to some extent regulate fetal inositol availabil- et al., 2000). Overall, it would appear that the placenta is
ity. The inositol content of cranial and spinal neuroectoder- able to both synthesize and import inositol, suggesting an
mal tissue in rat embryos was found to double between day important role for inositol in placental function (see section
11 and 12, but little else is known about how fetal tissue “Placental and fetal inositols in pregnancy”). Interestingly,
1650 O. C. WATKINS ET AL.

SMIT mRNA expression was very high in the brains of rat fetal supply (Lewis, Wadsack, and Desoye 2018), myo-inosi-
embryos, but was gradually down-regulated nearer to birth tol actions in placenta likely have a significant impact on
and after birth suggesting that myo-inositol and its trans- fetal growth and development.
porter play an important role in early CNS development Inositol may also affect fetal growth through the facilita-
(Guo et al. 1997). In contrast, SMIT mRNA expression in tion of IGF-1 and IGF-2 signaling, which utilizes PI/PIP/
kidney was very weak in the embryonic stages, but increased inositol phosphate signaling pathways (Siddle 2011).
significantly after birth in the rat (Guo et al. 1997), a period Maternal and fetal circulating IGF-1 and IGF-2 are strongly
of development that is equivalent to the in-utero develop- associated with the promotion of placental nutrient trans-
ment occurring in the third trimester of human pregnancy. port and fetal size (Jansson et al. 2013; Baumann et al.
This suggests that myo-inositol plays a key role in the devel- 2014). Increases in birthweight occur with increasing glucose
oping fetus, particularly the CNS and kidneys, but that role and amino acid transport to the fetus, and this is likely
might change as gestation progresses. mediated by IGF-1, since IGF-1 treatment was associated
with increased placental glucose and amino acid transporter
(GLUT1, SNAT2) expression in multiple placental in-vitro
Inositol and fetal development models (Jansson et al. 2013; Baumann et al. 2014). However,
Inositol appears critical for healthy fetal development. Myo- IGF also decreased lipoprotein lipase activity in placental vil-
inositol levels were increased in the urine of both large-for- lous explants in-vitro (Magnusson-Olsson et al. 2006), which
gestational-age (LGA; birthweight above 90th percentile) could lead to decreased maternal to fetal fatty acid transfer
and small-for-gestational-age (SGA; birthweight below 10th if the same occurred in-vivo. Dysregulation in inositol con-
percentile) human newborns (Marincola et al. 2015; tent or metabolism would be expected to alter IGF signaling
Barberini et al. 2014) and increased in intrauterine growth and could therefore have significant impact on fetal growth,
restricted (IUGR) piglets (Nissen et al. 2011), which suggests with the direction of growth-change dependent on which
that inositol dysregulation is associated with both extremes nutritional supply pathways are most impacted.
of the fetal growth spectrum. However, the mechanistic Inositol derivatives also affect the placental secretion of
pathways linking LGA and SGA with inositol dysregulation hormones that modulate fetal growth. In particular, inositol
remains speculative. Excessive urinary inositol excretion phosphates are involved in inducing the release from placental
could be a sign of inositol over production in the fetus, or syncytiotrophoblasts of human placental lactogen (hPL; see
conversely, reflect a pathological inositol depletion through section “Placental phosphoinositides”), which has anti-insulin
increased excretion. Since inositols are important regulators properties (Zeitler and Handwerger 1985; Zeitler, Wu, and
of glucose and lipid metabolism, one could speculate that Handwerger 1991; Zeitler, Markoff, and Handwerger 1983;
fetal modulation of its own endogenous inositol production Handwerger and Freemark 2000) and is one of the key hor-
and bio-availability could therefore be used as a signaling mones involved in maternal adaptation to pregnancy. As pre-
mechanism to tailor nutritional access from the maternal viously discussed (section “Inositols influence lipid
metabolism, transport and storage”), both A-IPG and myo-
and placental compartments. In cases of LGA and SGA such
inositol appear to decrease adipocyte leptin release in non-
inositol-mediated mechanisms are postulated to either be
pregnant in-vitro and in-vivo rat models (Croze, Gelo€en, and
malfunctioning leading to pathology, or used as a compensa-
Soulage 2015; Kunjara, Greenbaum, et al. 2000). If a similar
tory mechanism to try and optimize nutritional supply in
effect of reduced leptin release from the placenta and adipose
the presence of pathology, such as placental insufficiency or
tissue occurs in pregnancy, it could potentially suppress
hyperglycemia.
mobilization of glucose, amino acids and lipids to the fetus
For example, we postulate that alterations in inositol sig-
for growth (Tessier, Ferraro, and Gruslin 2013). Inositol also
nals to the placenta may regulate transplacental supply of
appears to be important for CNS development. Maternal cir-
fatty acids and lipids for growth. Myo-inositol treatment of
culatory myo-inositol in the lowest decile has also been associ-
human placental explants in-vitro alters the processing of
ated with spina bifida in humans (Groenen, Merkus, et al.
fatty acids into lipids, with myo-inositol displaying fatty
2003), although no association was seen with amniotic fluid
acid-specific effects (Watkins et al. 2019). In particular,
inositol levels (Groenen, Merkus, et al. 2003). There are also
myo-inositol increased the incorporation of isotope-labeled
several studies reporting associations between myo-inositol
oleic acid into triacylglycerols, phosphatidylcholines, phos-
deficiency and dysmorphogenesis in rodents (Cogram et al.
phatidylethanolamines and decreased the incorporation of
2002; Steegers-Theunissen, Groenen, and Beemster 2002;
docosahexaenoic acid (DHA) into triacylglycerols in placen-
Copp and Greene 2010; Baker et al. 1990) which will be dis-
tal explants (Watkins et al. 2019). This is of importance,
cussed in detail in section “Placental phosphoinositides.”
because the amount of each fatty acid available for fetal sup-
ply will depend on the balance between the different lipid
forms incorporating the fatty acid; more ready supply if the Hypothesis: Inositol acts as a communication signal
fatty acid is found in transportable forms (e.g. non-esterified across the maternal-placental-fetal axis to regulate fetal
and lysophospholipid), or in labile and accessible storage growth and development
forms (e.g. glycolipids), but less fetal supply if in longer We postulate that levels of inositol in pregnancy, whether of
term storage (e.g. structural phospholipids). Since placental maternal, fetal or placental origin, act as biological signals
lipid processing is key to regulating transplacental fatty acid that can regulate maternal and placental glucose and lipid
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1651

Figure 4. Hypothesis: Inositol, acts as a communication signal across the maternal-placental-fetal axis to regulate maternal glycemia and lipidemia, placental func-
tion and fetal physiology and therefore fetal growth and development. We also suggest that much of this inositol is of fetal origin and is likely regulated by factors
such as fetal growth potential and growth velocity, forming a feedback loop and enabling the fetus to control its nutritional supply according to its needs, to regu-
late its own size/growth and development. However, inositol regulation becomes dysfunctional in pregnancies complicated by disorders such as GDM leading to
disorders of fetal growth and maternal insulin resistance. 1 Increased excretion into the amniotic fluid decreases fetal inositol. 2 Increased excretion into maternal
urine decreases maternal inositol. 3 Placental inositol impacts placental function. In particular, inositol and inositol derivatives alter the balance of placental nutri-
ent import, metabolism, storage and export and therefore fetal nutrient supply. Inositol derivatives also affect placental signaling since they act as second messen-
gers for hormones such as insulin and insulin like growth factor (IGF) and impact the synthesis and secretion of bioactive lipid mediators and hormones such as
human placental lactogen. 4 Inositol regulates maternal lipidemia by altering the balance of lipid mobilization between different types of adipose tissue, but the
direction of this inter-adipose-depot redistribution appears strongly affected by BMI. 5 Inositol and its derivatives can regulate maternal glycemia directly or by
affecting maternal insulin sensitivity.

metabolism, endocrine activity and fetal nutrient supply. As glycogen, glucose and lipid metabolic pathways, which in
a result, inositol and its derivatives are able to regulate fetal other organs are mainly regulated by insulin (Larner,
growth and development. Details of the direction of inositol Brautigan, and Thorner 2010; Kunjara et al. 1999; Scioscia
exchange between the maternal, placental and fetal compart- 2017). Uniquely, the placenta is not reliant on insulin for
ments and the main foci of inositol action in maternal and glucose uptake, a feature that preserves continuous rapid
fetal tissues, and whether maternal inositol supplementation maternal-fetal transfer of glucose, which is essential for fetal
can address defects in these physiological mechanisms survival (Lager and Powell 2012). The role of IPGs in facili-
remain to be established. How defects in these processes tating anaerobic glycolysis is probably particularly important
may lead to macrosomia in GDM is addressed in section early in gestation, to provide an alternative to aerobic glu-
“Dysregulated maternal-placental-fetal lipid metabolism, adi- cose oxidation in a low oxygen environment prior to deep
posity and fetal growth in gestational diabetes” and in trophoblast invasion, which begins at the end of the first tri-
Figure 4. mester, and prior to development of an effective hemocho-
rial placenta for optimal oxygen and nutrient transfer to the
fetus (Scioscia et al. 2011). Later on in pregnancy, P-IPGs
Placental and fetal IPGs
may play an important physiological role in the regulation
Healthy human placenta contains more P-IPG than any of insulin resistance in order to maintain optimal fetal nutri-
other studied organ (25-fold more than liver), but contains tional supply (Larner 2002; Kunjara, Greenbaum, et al. 2000;
no or little detectable A-IPG (Scioscia 2017; Scioscia et al. Scioscia, Gumaa, and Rademacher 2009). Placental P-IPG
2013; Kunjara, Greenbaum, et al. 2000). P-IPG holds a crit- could alter placental function locally, or be released into the
ical role in the placenta and is involved in the regulation of maternal or fetal circulation where it could alter maternal or
1652 O. C. WATKINS ET AL.

fetal metabolism (Scioscia et al. 2008; , Schofield, and Placental Phosphoinositides


Rademacher 2003).
Phosphoinositide (PI) lipids act as important signaling mole-
Even though placental P-IPGs can regulate placental
cules in the placenta and are a major store of essential poly-
functions without insulin involvement, insulin may still pro-
unsaturated fatty acids (PUFA) within the fetal-placental
mote placental P-IPG actions by effecting P-IPG synthesis
unit (Bayon et al. 1993). A placental phosphatidylinositol
and transport. For example, in-vitro studies showed that
synthase (CDIPT) has previously been isolated and charac-
insulin stimulation caused more P-IPGs to be released by
terized, suggesting that the placenta can synthesize phos-
maternal-facing syncytiotrophoblast microvillous membrane
phoinositide lipids (Antonsson 1994). Arachidonic acid (AA,
isolates (Scioscia et al. 2008). This increase in P-IPG is an omega-6 fatty acid; 20:4) for example, comprises 33% of
thought to be due to the insulin induced activation of pla- the fatty acids held in placental PI (Bitsanis et al. 2005).
cental GPI-specific phospholipase D (GPI-PLD) which Such storage reservoirs are important, because the fetal-pla-
cleaves IPG from precursor GPIs (Scioscia et al. 2008; cental unit cannot synthesize PUFA due to a lack of desatur-
Deborde, Schofield, and Rademacher 2003). ase enzymes (Bayon et al. 1993). PUFAs can be released
However, IPG synthesis will also be affected by the avail- from placental phospholipids by phospholipase A (PLA)
ability of GPI precursors and GPI-PLD and nothing is (Schrey et al. 1992; Schrey, Read, and Steer 1987; Petit et al.
known about how this may be affected by insulin. 1989), a process vital for fetal PUFA supply and brain devel-
Even though placental syncytiotrophoblast microvillous opment (Crawford 2000), and for the synthesis of many
membranes contain active GPI-PLD enzyme (Deborde, PUFA-derived signaling molecules essential for fetal and pla-
Schofield, and Rademacher 2003), no GPI-PLD mRNA has cental development and labor such as prostaglandins and
been found in BeWo cells (trophoblast-like cell line) or in leukotrienes (Holub 1986; Walsh and Parisi 1986; Boone,
two placental cDNA libraries (Deborde, Schofield, and Currie, and Leung 1993).
Rademacher 2003). This suggests GPI-PLD is not synthe-
sized by the placenta and is instead of maternal or fetal ori-
gin (Deborde, Schofield, and Rademacher 2003). Large Inositol and arachidonic acid in dysmorphogenesis. Myo-inosi-
amounts of GPI-PLD are exported into the circulation by tol’s role in preventing fetal anomalies appears to be par-
the liver outside of pregnancy, suggesting that placental GPI tially mediated via its effect on AA metabolism and
cleavage maybe regulated by the maternal or fetal liver prostaglandin synthesis. It is well-established that dysmor-
(Low 2000). phogenesis, particularly fetal neural tube defects, are associ-
We do not know if placental GPI is of maternal, placental ated with poorly controlled preexisting diabetes in humans
or fetal origin. However, Deborde et al. has suggested that (Wentzel and Eriksson 2020). Similar defects are also
the structure of placental syncytiotrophoblast may regulate observed in streptozocin-induced diabetic pregnant rat and
mouse embryos incubated with excess glucose (Wentzel
how easily GPI-PLD can access GPIs stored in caveolae
et al. 2001; Sussman and Matschinsky 1988; Baker et al.
membrane invaginations and that this may alter IPG avail-
1990; Hashimoto et al. 1990). Such embryos also displayed
ability in various pregnancy complications (Deborde,
an associated decrease in inositol content, particularly in the
Schofield, and Rademacher 2003; Parpal, Gustavsson, and
neuroectoderm (Steegers-Theunissen, Groenen, and
Strålfors 1995; Anderson et al. 1992).
Beemster 2002; Wentzel et al. 2001; Sussman and
Although some IPGs appear to be synthesized by the pla-
Matschinsky 1988; Baker et al. 1990; Hashimoto et al. 1990),
centa, one pregnancy-specific P-IPG which can be detected
and indeed, the addition of either prostaglandin E2 or myo-
by an antibody assay appears to be formed by the fetus. In
inositol prevented the neural tube defects (Zeitler, Wu, and
uncomplicated pregnancies, this pregnancy-specific P-IPG
Handwerger 1991; Baker et al. 1990). Separately, in the non-
was found to be several hundred times more concentrated diabetic curly tail mice model, D-chiro-inositol and myo-
in amniotic fluid than in maternal urine (Scioscia 2017; inositol supplementation in-vitro was found to reduce the
Paine et al. 2006; Scioscia et al. 2011). Furthermore in pla- frequency of spina bifida (Cogram et al. 2002).
centa, this pregnancy-specific P-IPG was predominantly The protective effect of myo-inositol was reversed in-vivo
localized in the syncytiotrophoblast, endothelium of placen- by the addition of compounds that inhibited the synthesis of
tal vessels and extravillous trophoblast in the basal plate, prostaglandins from AA (Baker et al. 1990). This suggests
with only a little immunostaining within the villous stroma that inositol plays an important role by providing adequate
(Scioscia et al. 2013). If this pregnancy specific P-IPG is of AA for prostaglandin synthesis necessary for healthy CNS
fetal origin, it could potentially act as a signal to the mother development although the nature of this role is not known
by which the fetus can regulate its own nutritional supply (Cogram et al. 2002; Baker et al. 1990).
according to its needs. However, evidence for this postula- Inositol might increase the availability of AA by acting as
tion is still scarce and needs to be explored further. a structural base for greater AA storage in the form of PI.
Interestingly amniotic fluid P-IPG was decreased in the third Alternatively, inositol might influence the metabolism and
trimester compared to earlier in pregnancy (Scioscia, release of AA from a range of phospholipids through regu-
Gumaa, et al. 2007) possibly suggesting that this P-IPG is lating lipid synthesis and lysis. For example, cleavage of
more important early in pregnancy, or that a decrease in inositol containing PIP2 stimulates protein kinase C and
this P-IPG is an important signal in late pregnancy. activates PLA2, the phospholipase that cleaves PUFA from
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1653

phospholipids (Wentzel et al. 2001). Inositol could also Gestational diabetes


affect the downstream synthesis of prostaglandins. However,
Gestational diabetes (GDM) is glucose intolerance first rec-
further studies are needed to confirm the inositol-related
ognized during pregnancy, which is associated with
mechanisms on AA release and prostaglandin synthesis.
increased risk of short term and long term adverse outcomes
Rat embryos incubated in high glucose concentrations
for both mother and child (Sacks et al. 2012). Both hyper-
and rat embryos from mothers with streptozocin-induced
glycemia and lipid dysregulation are postulated to be
diabetes showed decreased expression of cytosolic PLA2
involved in its pathophysiology (Gallo, Barrett, and Nitert
(Wentzel et al. 2001) and cyclooxygenase COX-2 (the
2017). GDM is associated with increased risk of hypertensive
enzyme catalyzing the synthesis of prostaglandins from AA;
disorders of pregnancy, preterm delivery and cesarean sec-
(Wentzel, Welsh, and Eriksson 1999)) compared with
tion delivery and babies are at risk of numerous negative
respective controls on embryonic day E10 but not E11 of
consequences including neonatal hypoglycemia, jaundice
gestation. This suggests a narrow gestational window for
and perinatal death (Metzger et al. 2008; Crowther et al.
phospholipid cleavage, AA release and prostaglandin synthe-
2005; Mitanchez et al. 2015). GDM is also associated with
sis to impact CNS development. However, this change has
fetal macrosomia which is linked with increased risk of
yet to be linked to inositol. If it is, then the timing of inosi-
shoulder dystocia and birth trauma (Metzger et al. 2008;
tol supplementation in pregnancy to prevent defects would Crowther et al. 2005; Mitanchez et al. 2015). In the longer
be critical. term offspring are at an increased risk of major chronic
Interestingly, incubation of mouse embryos with scyllo- health conditions including obesity, type 2 diabetes, meta-
inositol caused similar defects as incubation with glucose, bolic syndrome and cardiovascular disease later in life, while
which suggests that scyllo-inositol competes as a ligand with women who had GDM are 7 times more likely to later
myo-inositol in early development to disrupt the important develop type 2 diabetes than controls (Mitanchez et al. 2015;
role played by myo-inositol (Steegers-Theunissen, Groenen, Bellamy et al. 2009; Ruchat, Hivert, and Bouchard 2013; Ma
and Beemster 2002; Wentzel et al. 2001; Baker et al. 1990). et al. 2015).
Pregnancy is a pro-diabetogenic state and is associated
Interactions of inositol derivatives and lipids in maternal- with rising maternal insulin resistance, increased maternal
placental-fetal physiology. The inter-relationships between glucose intolerance and an elevation of circulating non-
placental inositol derivatives, AA availability, maternal endo- esterified fatty acids, most prominently in the third trimester
crine regulation by the placenta and fetal growth are com- (Scioscia 2017; Kunjara, Greenbaum, et al. 2000; Jayabalan
plex. In-vitro AA treatment in various models of placental et al. 2017). These changes are essential for providing the
explants, dispersed placental cells and cultured term human fetus with adequate nutrients, but individual hereditary and
syncytiotrophoblast, stimulated the hydrolysis of PIPs by environmental vulnerabilities can lead to accentuated
phospholipase C (Zeitler and Handwerger 1985; Zeitler, Wu, changes that result in GDM and related complications in
and Handwerger 1991; Zeitler, Markoff, and Handwerger some women. It has been postulated that women who are
1983Petit et al., 1989). This caused the release of placental already sub-clinically metabolically challenged preconception
inositol phosphates, which then induced a rapid release of may be unable to mount an adequate insulin response in
human placental lactogen (hPL) (Zeitler and Handwerger pregnancy, resulting in GDM (Catalano et al. 2003). Since
1985; Zeitler, Wu, and Handwerger 1991; Zeitler, Markoff, inositols play an influential role in pregnancy and in dia-
and Handwerger 1983Petit et al., 1989). This process is betes, it is important to understand the interactions between
important especially in the third trimester of pregnancy, GDM and inositol metabolism and action, in order to fully
because hPL changes the metabolic state of the mother to exploit inositol’s potential as an intervention in this context.
facilitate mobilization of glucose and fatty acids for fetal
nutrition (Zeitler, Wu, and Handwerger 1991; Handwerger
and Freemark 2000). Inositol phosphates are also crucial in Relationship between gestational diabetes and inositol
regulating placental lipid metabolism. For example, PIP2 Alterations in the level of inositols in biological fluids are
activates placental phospholipase D, which hydrolyzes phos- apparent many weeks prior to the clinical detection of
pholipids to produce phosphatidic acid, a bioactive lipid hyperglycemia and GDM diagnosis (Murphy et al. 2016).
which modulates kinase signaling (Vinggaard and Hansen When assessed during early pregnancy, women who would
1995; Liscovitch et al. 2000; Wang et al. 2006). subsequently manifest the signs of GDM later in pregnancy
Thus, in pregnancy, inositols, PI lipids, PIPs and inositol displayed increased first trimester urinary D-chiro-inositol
phosphates play critical roles in regulating paracrine, endo- and myo-inositol (Murphy et al. 2016) and increased second
crine, intracellular and intercellular signaling, to influence trimester amniotic fluid myo-inositol (Santamaria, Di
maternal and fetal physiology, and fetal nutritional provi- Benedetto, et al. 2016), compared to women who did not
sion. Given the diversity of inositol involvement in various subsequently develop GDM. Such findings suggest that dys-
biological pathways of maternal-placental-fetal physiology, regulated inositol metabolism and excretion in the mother
some form of inositol dysregulation is likely involved in the or the fetal-placental unit predates the development of
pathophysiological processes of pregnancy complications, hyperglycemia, and may be involved in the generation of
and conversely, great potential for inositol-related interven- pathological events leading to GDM development. Since
tions to aid in preventing or managing these complications. similar increases in urinary D-chiro-inositol and myo-
1654 O. C. WATKINS ET AL.

inositol were also observed in non-pregnant overweight and systematic increase in P-IPG to try to counter insulin resist-
obese diabetic adults (Ostlund et al. 1993; Hong et al. 2012), ance, or reflect pathological increased urinary excretion,
and in obese diabetic rodent models (Table 3), it has been which could in turn reduce systemic P-IPG and promote
postulated that a similar pathological pathway of inositol insulin resistance.
dysregulation may be shared by GDM and adult diabetes Even though early pregnancy urinary P-IPGs are higher
that is driven by obesity. in diabetic pregnancies, women with preexisting type 2 dia-
Currently, there is little evidence to suggest how inositol betes and GDM do not show the normal ontogenic rise in
content and actions in the maternal and fetal tissues, are urinary P-IPG with advancing gestation (Kunjara,
altered in GDM, nor how they may alter the risk of GDM Greenbaum, et al. 2000; Scioscia, Gumaa, and Rademacher
development. Our data suggests that placental inositol con- 2009). Therefore, by the late third trimester, urinary P-IPG
tent is significantly reduced in GDM pregnancies compared levels in diabetic and non-diabetic groups are similar (Table
with non-GDM cases (Pillai et al. 2020) and work is cur- 6) (Kunjara, Greenbaum, et al. 2000; Scioscia, Gumaa, and
rently on-going to establish the underlying mechanisms for Rademacher 2009). This could suggest that GDM pregnan-
this difference. However, the concurrent rise of both amni- cies go through the P-IPG and glycemic-related metabolic
otic fluid and maternal urinary inositol during pregnancy adaptations associated with later pregnancy at much earlier
suggests that GDM does not conform to the theory of a gen- gestations. If true, this could increase fetal nutritional supply
eral diabetic inositol deficiency caused by increased urinary over a longer period and possibly contribute to excessive
inositol excretion. More SMIT-1 protein was found in GDM fetal growth and adiposity in GDM (Scioscia et al. 2011;
placenta compared to non-GDM placenta, with expression Scioscia, Gumaa, and Rademacher 2009).
changes localized to the trophoblast suggesting that
increased inositol transport into the placenta occur in GDM A-IPGs in pregnancy and GDM. In the third trimester,
(Mejia, Reynolds, and Arroyo 2016). In contrast, our studies urinary A-IPG was found to be lower in GDM than in non-
showed a decline in IMPA1 (myo-inositol synthesis), SMIT- GDM controls and correlated negatively with glycemic con-
2 and HMIT expression, associated with reductions in pla- trol, but positively with BMI (Scioscia, Gumaa, et al. 2007).
cental inositol content, with increasing maternal fasting gly- This study also found a trend suggestive of an association
cemia (Pillai et al. 2020). However, many other inositol- between urinary A-IPG and birth weight. Since A-IPG in-
related factors are likely altered in GDM, and further work vitro influences many glucose, lipid and insulin-related
is needed to understand how inositol alterations within the metabolic processes, in particular stimulating the synthesis
maternal-placental-fetal axis relates to GDM of lipids and glycogen, while lowering lipolysis (D’Anna and
pathophysiology. Santamaria 2018; D’Oria et al. 2017; Malvasi et al. 2014;
Pintaudi, Di Vieste, and Bonomo 2016; D’Anna et al., 2013;
P-IPGs in pregnancy and GDM. P-IPGs may play an Matarrelli et al. 2013), it is plausible that A-IPG may play a
important physiological role in the regulation of insulin role in the pathophysiology of glucose intolerance and fetal
resistance in pregnancy (Larner 2002; Kunjara, Greenbaum, macrosomia. However, in-vivo mechanistic studies are lack-
et al. 2000; Scioscia, Gumaa, and Rademacher 2009). ing and we cannot exclude the possibility that A-IPG may
Urinary P-IPGs measured by PDH assay were found to only be an associated indicator of glucose intolerance.
physiologically increase with increasing gestation among Furthermore, since tissue A-IPG has not yet been quantified
non-diabetic women (Kunjara, Greenbaum, et al. 2000; in GDM we cannot tell whether low maternal urinary
Scioscia, Gumaa, and Rademacher 2009). Since insulin A-IPG reflects low systemic A-IPG or decreased excretion.
resistance also rises in late pregnancy, and because of the A-IPGs have been reported to inhibit leptin release from
link between urinary IPGs and insulin resistance in the non- adipocytes, increase lipid synthesis and decrease the oxida-
pregnant state, it has been suggested that P-IPGs are likely tive disposal of glucose in a variety of cell types (Kunjara,
involved in the development of insulin resistance in the Greenbaum, et al. 2000; Scioscia, Gumaa, et al. 2007;
pregnant state (Larner 2002; Kunjara, Greenbaum, et al. Kunjara, Greenbaum, et al. 2000). However, the action of A-
2000; Scioscia, Gumaa, and Rademacher 2009). IPGs in pregnancy remains unknown and it is difficult to
Urinary P-IPGs in women with preexisting type 2 dia- predict how these molecules may be involved in GDM.
betes and those who subsequently developed GDM, were
elevated in the first two trimesters of pregnancy compared Ipgs as biomarkers of GDM. Whether urinary or serum ino-
with non-diabetic pregnancy (Kunjara, Greenbaum, et al. sitols or inositol derivatives such as IPGs could represent
2000; Scioscia, Gumaa, and Rademacher 2009). Furthermore, useful biomarkers to identify in early pregnancy women
in one small study of women with GDM, urinary P-IPG lev- who are at significant risk of developing GDM later in preg-
els were found to correlate with antenatal glycemia in the nancy remains to be explored. Examining if inositol bio-
third trimester and with birthweight, but these correlations markers could improve predictive power on top of that
were not observed in the control group (Scioscia, Gumaa, provided by conventional clinical risk assessments using fac-
et al. 2007). However, the amount of P-IPG in maternal tis- tors such as previous history of GDM, previous fetal macro-
sues and plasma in pregnancies complicated by diabetes somia, high BMI and family history of diabetes. Preventive
compared to uncomplicated pregnancies is not known. measures could be instituted early and targeted at those who
Increased urinary P-IPG may either reflect an adaptive are most likely to benefit.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1655

Prevention and treatment of gestational diabetes significantly decreased in overweight and obese pregnant
with inositols women (DʼAnna et al. 2015; Santamaria, Di Benedetto, et al.
2016), but not those with a family history of type 2 diabetes
Inositol supplementation is a promising intervention for the
(D’Anna et al., 2013). However, these studies were open
prevention and treatment of GDM. A recent meta-analysis
label and were all conducted by the same research group on
(Zhang et al. 2019) of 5 RCTs (Farren et al. 2017; D’Anna
a homogeneous population. Thus, it remains uncertain if
et al., 2013; Matarrelli et al. 2013; Dʼ Anna et al. 2015;
findings can be generalized to other populations.
Santamaria, Di Benedetto, et al. 2016) with a total of 927
One study also analyzed the effect of myo-inositol treat-
pregnant women with metabolic risk factors, found that the
ment on lipids in pregnancy (Malvasi et al. 2014). In this
daily supplementation of pregnant women with 2  4 g myo-
small trial (24 treatment, 24 control), the combination of
inositol from early pregnancy reduced the incidence of
myo-inositol, D-chiro-inositol, folic acid and manganese
GDM (RR 0.43, 95%CI 0.21–0.89), and preterm delivery
given to non-obese pregnant women was associated with
(RR 0.36, 0.17–0.73). Other meta-analyses of these studies
reduced glycemia, as well as total cholesterol, low-density
generally agreed with these findings (Crawford et al. 2015;
lipoproteins (LDL), high-density lipoproteins (HDL), and
Zheng et al. 2015; Santamaria et al. 2018; Vitagliano et al.
triglycerides compared with control. However, it is unclear
2019). These RCTs were mainly conducted in Caucasian
whether these changes are due to inositol or to other treat-
women who had risk factors for developing GDM namely
ment components.
obesity/overweight (Dʼ Anna et al. 2015; Santamaria, Di Despite these promising findings, all reviews to date have
Benedetto, et al. 2016), a family history of type 2 diabetes concluded there is insufficient data to properly evaluate
(D’Anna et al., 2013), or an impaired fasting glucose in the whether the use of myo-inositol during pregnancy is effica-
first trimester (Matarrelli et al. 2013), with supplementation cious in reducing the risk of GDM development. In particu-
with myo-inositol mostly beginning from the late first tri- lar, the populations studied were too small and lacked
mester onwards. In a separate study, myo-inositol treatment diversity, and many studies were open-label (unblinded) and
starting preconception in women with PCOS (n ¼ 83) was thus exposed to bias. Furthermore, there is a lack of data on
also found to reduce the risk of developing GDM (17.45% neonatal outcomes including perinatal mortality and serious
vs 54%, odds ratio 2.4, 95% CI, 1.3–4.4) and this difference infant morbidity and longer-term offspring health. Also,
remained significant even after adjusting for covariates (age, there is insufficient data to determine whether myo-inositol
BMI, parity, family history of diabetes) (D’anna et al. 2012). is a better treatment option than D-chiro-inositol or inositol
Two studies have looked at the effects of D-chiro-inositol combinations, or whether these molecules have interactive
combined with myo-inositol. One small randomized control effects (Celentano et al. 2020).
trial on non-obese Italian pregnant women (n ¼ 157) with Further evaluation in large-scale, multicentre, double-
elevated fasting glucose in the first or early second trimester, blinded RCTs are necessary to prove the efficacy of inositols
who were supplemented with either myo-inositol (4000 mg), in reducing the risk of GDM, macrosomia and preterm
D-chiro-inositol (500 mg), combined myo- and D-chiro- delivery. Furthermore, these trials will need to include preg-
inositol (1100/27.6 mg) or placebo, showed that all the inosi- nant women of different ethnicities and more varied risk
tol treatments significantly decreased fasting and post-pran- factors, with examination of different doses of myo-inositol
dial glycemia and reduced the risk of developing GDM and other inositol isomers in comparison with placebo. An
(Celentano et al. 2020). Non-inferiority analysis demon- example of such a trial is the NiPPeR study (Godfrey et al.
strated the largest benefit was found in the myo-inositol 2017) (clinicaltrials.gov NCT02509988) which included
group, although dosage differences make comparison diffi- women of multiple ethnicities with both low and high meta-
cult (Celentano et al. 2020). In contrast, another study in bolic risks from UK, Singapore and New Zealand. The inter-
Irish women (n ¼ 240) with a family history of diabetes vention is a nutritional formulation containing myo-inositol,
found that a combination of myo-inositol 1100 mg and D- probiotics and additional micronutrients, commenced prior
chiro-inositol 27.6 mg/day did not lower GDM incidence to conception and continued throughout pregnancy, and
compared to controls (Farren et al. 2017). results are expected to be published later in 2020. There is
One meta-analysis (Zhang et al. 2019) showed that myo- also another smaller on-going clinical trial in the UK
inositol had no substantial impact on the OGTT 2-h post- (EMmY study, ISRCTN48872100) (Amaefule et al. 2018) of
glucose load response or on neonatal birthweight and mac- myo-inositol supplementation starting in early pregnancy in
rosomia risk (birthweight >4 kg). However, the meta-ana- women at high risk of GDM development. With completion
lysis was likely underpowered to demonstrate modulation of of these trials a clearer picture will emerge on the efficacy of
fetal size because of the low prevalence of macrosomia inositol supplementation in reducing pregnancy risks, the
(Zhang et al. 2019). In contrast, the Santamaria et al. (2018) safety of the intervention, and whether specific subgroups
meta-study which focused only on a subset of three RCTs in would benefit more than others.
an Italian population at risk of GDM (high BMI or family
history of type 2 diabetes n ¼ 595), showed that myo-inositol
significantly decreased GDM diagnosis, fasting and post- Inositol use after GDM diagnosis. Upon the diagnosis of
prandial glycemia, preterm birth and macrosomia. The effect GDM, maternal hyperglycemia can be controlled by a var-
of myo-inositol on insulin resistance in these Italian studies iety of interventions, usually starting with diet control, exer-
appears to be population dependent with HOMA-IR cise and lifestyle changes, then adding in therapies such as
1656 O. C. WATKINS ET AL.

metformin and/or insulin if required (Falavigna et al. 2012; no apparent effect on fetal size (Plows et al. 2017). In con-
Sweeting et al. 2016). Two randomized open-label trials trast, in a high fat diet mouse model of GDM, myo-inositol
have tested the effect of inositol in women already diag- treatment did not affect maternal glycemia or fat accretion
nosed with GDM. One trial on 69 women with GDM found (Plows et al. 2020), but did increase maternal adipose tissue
that myo-inositol (4000 mg) improved insulin resistance mRNA expression of Ir, Irs1, Akt2, and Pck1, which are all
(lower HOMA-IR) and increased circulating adiponectin key members of the insulin signaling pathway, and demon-
(Corrado et al. 2011). Another trial (n ¼ 80) tested the effect strated an associated decrease in offspring birthweight
4000 mg myo-inositol or 500 mg D-chiro-inositol or 1100/ (Plows et al. 2020). This suggests that inositol might have a
27.6 mg myo/D-chiro-inositol (Fraticelli et al. 2018). This pro-insulin or an insulin-like effect in adipose tissue result-
study found that insulin resistance was significantly ing in decreased maternal lipid mobilization and lipid trans-
decreased in women given myo-inositol, and that groups on fer for fetal growth. Myo-inositol may also alter lipid supply
myo-inositol-containing interventions (but not the chiro- to the fetus by affecting placental lipids metabolism as dis-
inositol alone group) were less likely to require insulin cussed in section “Inositol and fetal development.”
therapy, and delivered newborns with significantly lower Current management of GDM does not involve lipid
birthweights. A third observational study also found that monitoring nor treatment of dyslipidemia, and lipid lower-
women with GDM treated with 1.2 g myo-inositol were less ing drugs are generally contraindicated in pregnancy. Myo-
likely to require insulin treatment later in pregnancy but inositol and its derivatives, as naturally-occurring com-
lacked a contemporaneous control group for valid compari- pounds, are promising candidates to address this metabolic
son (Lubin et al. 2016). Collectively, these studies suggest dysfunction in GDM since they can alter both glucose and
that myo-inositol could potentially be considered as first lipid metabolism. This venture must firstly involve develop-
line GDM therapy, or as adjuvant therapy with insulin to ment of a much deeper understanding of how inositols
reduce insulin dosage and reduce the risk of hypoglycemia influence lipid metabolism in maternal tissues and the pla-
associated with this treatment, although this remains to be centa, and how they may influence maternal-fetal lipid
clinically evaluated. transfer. Then, further clinical studies focusing on lipid and
adiposity outcomes in pregnancy are necessary to determine
whether myo-inositol or any other inositol isomers or deriv-
Dysregulated maternal-placental-fetal lipid metabolism, atives could be an effective treatment for the anomalous lipi-
adiposity and fetal growth in gestational diabetes domic features of GDM.
According to Pederson, fetal macrosomia is a result of the
anabolic and lipogenic effect of fetal hypersecretion of pan-
Postulated actions of inositol in gestational diabetes
creatic insulin, which is stimulated by increased maternal-
fetal glucose transfer in maternal diabetes (Pedersen 1952). The mechanisms by which myo-inositol supplementation
However, GDM cases with apparently satisfactory glycemic may reduce the risk of GDM and fetal macrosomia are not
control, still show an increased risk of fetal macrosomia, understood (Coustan 2013). Diabetes is classically hypothe-
and neonates even if appropriately-grown-for-gestational-age sized to be a condition of myo-inositol deficiency (see above
as judged by birthweight, still show increased neonatal adi- 4.5). However, there has been no clear evidence for a gen-
posity in body composition measures (Poston et al. 2015; eral systemic inositol deficiency in GDM women that would
Zhuo, Wang, and Yu 2014). Furthermore, maternal circulat- require correction (Table 6, section “Relationship between
ing TAGs and free fatty acids, have been positively corre- gestational diabetes and inositol”). Current findings instead
lated with neonatal weight and fat mass in well-controlled suggest a complex situation where inositol perturbations
GDM, but not in non-GDM pregnancies (Schaefer-Graf may be caused by ill-defined alterations in synthesis, trans-
et al. 2008; Schaefer et al. 2011). Hence, it has been postu- port and metabolism, and may possibly differ between tissue
lated that dysregulated lipid metabolism across the mater- types. Furthermore, these alterations could be either patho-
nal-placental-fetal axis, on top of glycemia, may contribute logical leading to the development of GDM, or be an adap-
to the development of excessive fetal growth and adiposity tive response to the GDM condition to minimize its impact.
in GDM (Herrera and Ortega-Senovilla 2010). Furthermore, Therefore, there is not yet a full picture of how these various
it is already well documented that in GDM there is reduced processes would be affected by maternal inositol supplemen-
maternal-placental-fetal transfer of the essential long chain tation and what the net effects would be in different individ-
(LC)-PUFAs (Herrera and Ortega-Senovilla 2018; Larque uals. However, here we have drawn together threads of our
et al. 2014; Delhaes et al. 2018; I et al. 2002; Fraser et al. current understanding (Figure 4) upon which new know-
1990; Schwartz et al. 1994) and it is likely that further ledge can be added.
abnormalities of lipid regulation will be described with on- Myo-inositol supplementation has been shown to
going research. improve insulin resistance and glycemic and lipid status in
Inositol treatment of animal models of GDM appears to various metabolic conditions in the non-pregnant state and
alter maternal lipid metabolism, but effects vary depending may, in theory, also act on maternal tissues during preg-
on the model studied. Myo-inositol treatment in the genetic- nancy to improve maternal metabolic health (Croze and
ally-modified db/þ mouse model of GDM reduced maternal Soulage 2013; Coustan 2013; Zhang et al. 2019). However,
intra-abdominal, gonadal and perirenal adiposity, but had pregnancy involves not just the mother, but also the
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1657

placenta and the fetus, and any intervention needs to con- Another consideration is how concurrent insulin treat-
sider the potential risks and benefits on all three. ment, during GDM management, may influence the effects
Inositol and inositol derivatives are thought to play a role of on-going inositol supplementation. In-vitro, insulin can
in the action or release of many hormones that affect mater- stimulate cultured non-GDM placental trophoblast microvil-
nal metabolism including insulin, thyroid stimulating hor- lous membranes to release the insulin mimic P-IPGs
mone, leptin and human placental lactogen (Van Sande (Scioscia et al. 2008), potentially promoting further insulin-
et al. 2006; Sayers and Hanyaloglu 2018; Berridge 2009; like actions in placenta and mother. In insulin-treated
Zeitler et al., 1991; Handwerger and Freemark 2000; Perez- GDM, there are more insulin receptors in trophoblasts
Perez et al. 2016; Hauguel-de Mouzon, Lepercq, and found at the end of pregnancy than in diet-treated GDM.
Catalano 2006). We postulate that alterations in placental Therefore, insulin treatment could potentially increase both
inositol content, could alter fetal nutritional supply by placental P-IPG-insulin-mimic release and increase placental
releasing endocrine signals that affect maternal metabolism responsivity to insulin/insulin mimics through receptor up-
or by directly changing placental glucose, lipid and amino regulation, in addition to the direct effects of insulin treat-
acid metabolism and transport. Observational data from the ment on maternal tissues to lower glycemia (Scioscia et al.
GUSTO mother-offspring cohort (Chu et al. 2020) and from 2008; Desoye and Hauguel-de Mouzon 2007).
a separate smaller study (Pillai, et al. 2020) showed that a How these different actions of inositol and inositol deriv-
high placental inositol content is associated with a suppres- atives, and the actions of various hormones interact and
sion of the fetal growth-promoting and pro-adipogenic work together to bring about maternal adaptations to preg-
effects of maternal glycemia. nancy and optimize fetal growth is not well studied to date.
Whether maternal inositol supplementation can increase Neither is it understood how inositol supplementation and
placental inositol content to favorably modulate fetal growth pathological states like GDM alter these actions.
and adiposity remains to be established. Another important consideration is that myo-inositol
If myo-inositol supplementation can lower maternal supplementation is currently being proposed as a preventive
blood glucose and suppress mobilization of lipids to the measure for GDM. Such a strategy would involve myo-inosi-
fetus, then this could reduce excessive nutrient transfer to tol exposure of many pregnancies which would not ultim-
the fetus, reducing macrosomia risk, but if taken to an ately develop GDM. Thus, any potential regulatory changes
extreme this may possibly result in fetal nutritional insuffi- or harms brought about by inositol treatment during preg-
ciency and growth restriction. Furthermore, fetal poly-unsat- nancy needs to be properly evaluated and understood in
urated fatty acid (PUFA) supply is already decreased in both uncomplicated and at-risk pregnancies, before wide-
GDM and this may be worsened if general fatty acid transfer spread implementation of programs for GDM prevention
is decreased further. and treatment with myo-inositol supplementation.
Myo-inositol supplementation may also alter how the pla-
centa metabolizes and transports nutrients. For example,
Pre-eclampsia
inositol availability will impact IGF signaling, which requires
the conversion of PIP2 into PIP3 (Section “Inositol and fetal Pre-eclampsia is characterized by the onset of new hyperten-
development”). Increased IGF signaling in GDM is thought sion or exacerbation of preexisting hypertension, accompa-
to increase placental glucose and amino acid transport and nied by significant proteinuria or multi-system disorder
therefore fetal growth (Jansson et al. 2013; Baumann et al. during pregnancy, with the pathophysiology arising from
2014; Stanirowski et al. 2017). However, it is not clear abnormal placental implantation and endothelial dysfunction
whether inositol supplementation would increase or decrease (Cheng and Wang 2009). It is a major cause of maternal
IGF signaling, since whilst increased PIP2 would increase mortality and morbidity worldwide and is associated with
IGF signaling, increased PIP3 would decrease IGF signaling. intrauterine growth restriction (IUGR) and long-term health
Our studies of labeled fatty acid uptake by human pla- issues for both mother and baby (Cheng and Wang 2009;
cental explants from non-GDM pregnancies showed that Ghulmiyyah and Sibai 2012). Pre-eclampsia is also a state of
myo-inositol treatment in-vitro reduced the incorporation of heightened insulin resistance, and patients with pre-eclamp-
the LC-PUFA, DHA, into lipids, but increased the incorpor- sia show many similar metabolic changes to those with
ation of the mono-unsaturated fatty acid, oleic acid, into lip- GDM (Cheng and Wang 2009; Ghulmiyyah and Sibai 2012).
ids (Watkins et al. 2019). This suggests that myo-inositol In addition, women previously exposed to a pre-eclamptic
acts differently on PUFA compared to non-PUFA. In a best- pregnancy carry an increased risk of future diabetes and car-
case scenario, reduced incorporation of PUFA into placental diovascular disease, while the associated hyperinsulinemia
lipids could make available more un-esterified PUFA to be and hyperandrogenism of pregnancy may persist for up to
transported to the fetus to alleviate fetal PUFA deficiency. 17 years after delivery (Kaaja et al. 1999; Sampson et al.
By the same logic, increased incorporation of non-PUFA 2004; Seely and Solomon 2003).
into lipids could decrease the transport of non-PUFA and Inositol regulation and processing are also altered in pre-
decrease the risk of GDM-associated macrosomia. However, eclampsia, although inositol’s possible role in pre-eclampsia
further studies are needed to investigate the response of etiology remains unclear (Table 7 and 8). Compared with
non-GDM and GDM placenta to inositol treatment and to control placentas, placentas from pre-eclamptic pregnancies
explore how this may impact fetal growth. showed higher inositol content, reduced expression of
1658 O. C. WATKINS ET AL.

#Urine
SMIT-1 protein and increased expression of the SMIT signal

A-IPG
transcription factor TonEBP (Lee et al. 2014). The former
two alterations in pre-eclampsia-affected placenta are in
contrast to those reported in GDM-affected placentas, hint-
ing at pathophysiological divergence, despite the apparent

the circulation
Insulin induced
metabolic similarities of the two conditions described earlier.

P-IPG release

the placenta
# Release from
# Release into
Like GDM, however, maternal urinary, placental and amni-
otic fluid P-IPGs are higher in pre-eclampsia cases than in
uncomplicated pregnancies (Paine et al. 2006; Williams et al.
2007; Dawonauth et al. 2014). Furthermore, maternal urin-
ary P-IPG rises steeply in the weeks prior to the clinical
onset of signs of pre-eclampsia, and this increase was linked
with exacerbated insulin resistance (Williams et al. 2007;

#Hemo-dialysate

"Amniotic fluid
Paine et al. 2010), just as in GDM. Nevertheless, longitu-

P-IPG
dinal studies measuring maternal urinary P-IPG from the

"Placenta,
#Placenta
#muscle

"Urine,
#Urine

"Urine

"Urine

"Urine
first trimester onwards (spot urine collection, normalized to
creatinine) (Scioscia et al. 2011; Paine et al. 2010) showed P-
IPG levels could not reliably predict pre-eclampsia, because
healthy women also had brief sporadic episodes of increased
P-IPG excretion for unknown reasons. It is to be noted that

D-Chiro-inositol
glucose consumption is known to cause a spike in circulat-
ing P-IPG in the non-pregnant state (Baillargeon et al. 2006)

#Plasma
"Urine,

"Ovary
#Urine

"Urine
and it seems likely that the P-IPG content of individual
urine or plasma samples will be influenced by previous food
consumption. If this is the case, the brief sporadic episodes
of increased P-IPG excretion seen in the study described
above could be caused by dietary factors and future studies

"Placenta inositol
# Nervous tissue

" Amniotic fluid


should take this into account, or set out to perform assess-
Myo-inositol

ments in the fasting state.

# Placenta
# Ovary

A key limitation of existing studies lies in the quantifica-


" Urine

"Urine

"urine
tion methods for P-IPG. The increase in urinary P-IPG is
larger when quantified by an antibody assay that detects
Table 8. Overview of inositol dysregulation associated with conditions involving insulin resistance.

only the pregnancy-specific P-IPG (Paine et al. 2006;


Williams et al. 2007; Dawonauth et al. 2014), than when
using the (less specific) PDH phosphatase activity assay that
Myo-inositol !

epimerase

measures a variety of P-IPGs (Kunjara, Greenbaum, et al.


D-Chiro-
inositol

2000). This suggests that the pregnancy specific P-IPG may


#Muscle
" Ovary
#Liver

be largely responsible for urinary P-IPG increase in pre-


#Fat

eclampsia. Of note, pre-eclampsia was not associated with


an increased amount of general serum P-IPGs as measured
by pyruvate dehydrogenase assay (Paine et al. 2006). Since
the antibody assay does not work well in plasma or serum,
Inositol transporters

" Placental SMIT-1

#Placental SMIT-1

it is difficult to confirm if circulating pregnancy-specific P-


IPG fraction is similarly increased.
Like in uncomplicated pregnancies, in pre-eclampsia
there is more P-IPG in amniotic fluid than in corresponding
maternal urine (Scioscia et al. 2011). However, in pre-
eclamptic pregnancies, but apparently not in uncomplicated
pregnancies, there are also higher amounts of P-IPG in the
umbilical arteries and vein relative to maternal uterine vein
Polycystic ovary syndrome

Uncomplicated pregnancy

and peripheral vein (Scioscia et al. 2011). This suggests that


Diabetes (type 1/ type 2)

Pregnancy with large or


small for gestational

the increased maternal urinary, placental and amniotic fluid


Condition or disorder

Gestational diabetes

Preexisting diabetes

P-IPG observed in pre-eclampsia is likely of fetal origin


in pregnancy

(Scioscia et al. 2011). Since P-IPG can act as an insulin-like


Pre-eclampsia
age infant

signal this could represent an example of fetal-maternal


communication, possibly as an adaptive response to the
increased maternal insulin resistance. Alternatively, P-IPG
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1659

produced by the fetus may play a role in the evolving patho- Myo-inositol and D-chiro-inositol were also found to
physiology as subsequently discussed. increase mitogen-activated protein kinase (MAPK) phos-
As mentioned earlier, placental IPGs may be cleaved and phorylation (D’Oria et al. 2017; Brancho et al. 2003). MAPK
released in response to insulin by placental GPI-PLD phosphorylation and activity are upregulated in both the
(Scioscia et al. 2006; Scioscia et al. 2008; Deborde, Schofield, placenta and umbilical cord in pre-eclampsia (Afroze et al.
and Rademacher 2003). However, although placental P-IPG 2016). MAPK regulates diverse cellular processes, including
appears increased in pre-eclampsia (Kunjara, Greenbaum, placental development and trophoblast invasion and growth
et al. 2000), insulin-induced release of P-IPG is decreased in (Adams et al. 2000; Li et al. 2003), but whether the conse-
pre-eclamptic placental explants in-vitro (Scioscia et al. 2006; quences of increased MAPK activation are protective or
Scioscia et al. 2008; Deborde, Schofield, and Rademacher pathological in pre-elcampsia is unclear (Adams et al. 2000;
2003). The net effect of alterations in maternal, placental Li et al. 2003; Peters et al. 2000).
and fetal inositol biology in pre-eclampsia is therefore Overall, it is not yet clear how systemic and local inositol
unclear (Scioscia et al. 2006; Scioscia et al. 2008). concentrations are altered in pre-eclampsia, nor how this
Furthermore, GPI-PLD protein expression at delivery is may alter maternal insulin resistance (D’Oria et al. 2017). It
similar in placentas from both uncomplicated and pre- is thus, too early to suggest if inositols or their derivatives
eclamptic pregnancies (Deborde, Schofield, and Rademacher could be potential therapeutic targets in pre-eclampsia.
2003). This suggests that increased placental P-IPG could be
due to increased synthesis or import, rather than to altera-
tions in GPI cleavage, or to the increased insulin-resistance Conclusion
of pre-eclampsia placenta (Scioscia et al. 2006; Scioscia et al.
Inositols and their derivatives are highly bioactive molecules
2008; Deborde, Schofield, and Rademacher 2003). However,
that influence glycemia regulation, insulin signaling and
the degree to which the placenta may become insulin resist-
lipid metabolism. Optimal inositol intake, metabolism, trans-
ant in pre-eclampsia is not well understood (Desoye and
port and excretion are necessary for health, and inositol dys-
Hauguel-de Mouzon 2007; Challier, Hauguel, and
regulation is associated with conditions involving insulin
Desmaizieres 1986).
resistance, dysglycemia and deranged lipidemia, including
It has been postulated that P-IPGs may play a role in the
those occurring during pregnancy such as gestational dia-
generation of pre-eclampsia pathology (Kunjara et al. 1999;
betes and pre-eclampsia (Table 8).
Scioscia et al. 2011). Some P-IPGs have insulin-like effects,
Inositols participate in maternal-placental-fetal cross talk
so the increase in P-IPG could alter maternal physiology to
and play an important regulatory role in the adaptation of
further compromise nutrient mobilization to the fetus result-
maternal physiology to nurture a healthy pregnancy. Thus,
ing in fetal growth restriction (Kunjara et al. 1999; Scioscia
inositol supplements could be promising interventions for
et al. 2011). P-IPGs also increase glycogen synthesis in rat
the prevention and treatment of a range of metabolic disor-
and bovine liver by activating pyruvate dehydrogenase phos-
ders and in adverse conditions in pregnancy such as GDM.
phatase, glycogen synthase phosphatase, and glycerol-3-
However, their effects need to be better understood in order
phosphate acyltransferase (Varela-Nieto, Le on, and Caro
to ensure they are used safely and effectively, targeting rele-
1996). It has therefore been suggested that increased placen-
vant populations.
tal P-IPGs could be responsible for the observed increase in
placental glycogen synthesis and glycogen synthase activity
in pre-eclampsia (Deborde, Schofield, and Rademacher
Author contributions and Acknowledgements
2003). However, IPGs have a range of bioactivities (Kunjara,
Greenbaum, et al. 2000), and the precise role of the preg- Manuscript was written by Watkins O.C., and Chan S.Y.
nancy-specific P-IPG, and other IPGs in the dysregulation with advice and editing provided by Yong H.E.J. and
of maternal-fetal physiology remains to be elucidated. Sharma N. We acknowledge Celes Maria Catherine Dado,
Insulin signaling though the PI3K signaling pathway was Samantha Grace Loon Magadia, Preben Selvam, Victoria K.
decreased in pre-eclampsia in the placenta compared to con- Cracknell and Reshma Appukuttan Pillai for their support
trols (Scioscia et al. 2006). These changes decreased PIP3 in preparing this manuscript.
and PKB activation which in turn are thought to cause
endothelial dysfunction and insulin resistance (D’Oria et al.,
2017). Human endothelial cells (HUVECs), exposed in vitro Disclosure statement
to myo-inositol or D-chiro-inositol, showed increased PKB This research is supported by a Clinician Scientist Award awarded to
phosphorylation on Ser473, reflective of increased activation. Chan S.Y. from the Singapore National Medical Research Council
Furthermore, the magnitude of changes induced by inositol (NMRC/CSA-INV/0010/2016), by the National University of
treatment were greater than could be induced by insulin ( Singapore, National University Health System Singapore and the
ASTAR Singapore Institute for Clinical Sciences. SYC is part of an
D’Oria et al. 2017). These findings suggest that inositol may
academic consortium that has received research funding from Nestec
be able to combat the pre-eclampsia-related defects in PI3K and is a co-inventor on pending patents, which covers the use of inosi-
signaling, suggesting that increased inositol in pre-eclampsia tol in human health applications. The other authors have no financial
could be adaptive. or personal conflict of interest to declare.
1660 O. C. WATKINS ET AL.

ORCID Artini, P. G., O. Di Berardino, F. Papini, A. Genazzani, G. Simi, M.


Ruggiero, and V. Cela. 2013. Endocrine and clinical effects of myo-
Oliver C. Watkins http://orcid.org/0000-0001-8202-6412 inositol administration in polycystic ovary syndrome. A randomized
Hannah E. J. Yong http://orcid.org/0000-0002-8814-752X study. Gynecological Endocrinology 29 (4):375–379. doi: 10.3109/
Neha Sharma http://orcid.org/0000-0002-4287-5994 09513590.2012.743020.
Shiao-Yng Chan http://orcid.org/0000-0002-3530-3023 Ashcroft, F. M., M. Rohm, A. Clark, and M. F. Brereton. 2017. Is type
2 diabetes a glycogen storage disease of pancreatic b cells? Cell
Metabolism 26 (1):17–23. doi: 10.1016/j.cmet.2017.05.014.
References Asplin, I., G. Galasko, and J. Larner. 1993. Chiro-inositol deficiency
and insulin resistance: A comparison of the chiro-inositol-and the
Adams, R. H., A. Porras, G. Alonso, M. Jones, K. Vintersten, S. Panelli,
myo-inositol-containing insulin mediators isolated from urine,
A. Valladares, L. Perez, R. Klein, and A. R. Nebreda. 2000. Essential
hemodialysate, and muscle of control and type II diabetic subjects.
role of p38a MAP kinase in placental but not embryonic cardiovas-
Proceedings of the National Academy of Sciences 90 (13):5924–8. doi:
cular development. Molecular Cell 6 (1):109–116. doi: 10.1016/
10.1073/pnas.90.13.5924.
S1097-2765(05)00014-6.
Baillargeon, J. P., E. Diamanti-Kandarakis, R. E. Ostlund, T.
Afroze, S. H., R. R. Kalagiri, M. Reyes, J. D. Zimmerman, M. R.
Apridonidze, M. J. Iuorno, and J. E. Nestler. 2006. Altered D-chiro-
Beeram, N. Drever, D. C. Zawieja, T. J. Kuehl, and M. N. Uddin.
inositol urinary clearance in women with polycystic ovary syndrome.
2016. Apoptotic and stress signaling markers are augmented in pree-
Diabetes Care 29 (2):300–5. doi: 10.2337/diacare.29.02.06.dc05-1070.
clamptic placenta and umbilical cord. BBA Clinical 6:25–30. doi: 10.
Baillargeon, J.-P., J. E. Nestler, R. E. Ostlund, T. Apridonidze, and E.
1016/j.bbacli.2016.05.003.
Diamanti-Kandarakis. 2008. Greek hyperinsulinemic women, with
Alvarez, J., J. Sanchez-Arias, A. Guadano, F. Estevez, I. Varela, J. Feliu,
or without polycystic ovary syndrome, display altered inositols
and J. Mato. 1991. Transport in isolated rat hepatocytes of the phos-
metabolism. Human Reproduction 23 (6):1439–46. doi: 10.1093/
pho-oligosaccharide that mimics insulin action. Effects of adrenalec-
humrep/den097.
tomy and glucocorticoid treatment. Biochemical Journal 274 (2): Baillargeon, J.-P., M. J. Iuorno, D. J. Jakubowicz, T. Apridonidze, N.
369–74. doi: 10.1042/bj2740369. He, and J. E. Nestler. 2004. Metformin therapy increases insulin-
Amaefule, C. E., Z. Drymoussi, J. Dodds, L. Sweeney, E. Pizzo, J. Daru, stimulated release of D-chiro-inositol-containing inositolphosphogly-
J. Robson, L. Poston, A. Khalil, J. Myers, et al. 2018. Effectiveness can mediator in women with polycystic ovary syndrome. The
and acceptability of myo-inositol nutritional supplement in the pre- Journal of Clinical Endocrinology and Metabolism 89 (1):242–9. doi:
vention of gestational diabetes (EMmY): A protocol for a rando- 10.1210/jc.2003-030437.
mised, placebo-controlled, double-blind pilot trial. BMJ Open 8 (9): Baillargeon, J. P., M. J. Iuorno, T. Apridonidze, and J. E. Nestler. 2010.
e022831., doi: 10.1136/bmjopen-2018-022831. Uncoupling between insulin and release of a D-chiro-inositol–con-
Andersen, D., and B. Holub. 1976. The influence of dietary inositol on taining inositolphosphoglycan mediator of insulin action in obese
glyceride composition and synthesis in livers of rats fed different women with polycystic ovary syndrome. Metabolic Syndrome and
fats. The Journal of Nutrition 106 (4):529–36. doi: 10.1093/jn/106.4. Related Disorders 8 (2):127–36. doi: 10.1089/met.2009.0052.
529. Baker, L., R. Piddington, A. Goldman, J. Egler, and J. Moehring. 1990.
Andersen, D., and B. Holub. 1980. Myo-inositol-responsive liver lipid Myo-inositol and prostaglandins reverse the glucose inhibition of
accumulation in the rat. The Journal of Nutrition 110 (3):488–95. neural tube fusion in cultured mouse embryos. Diabetologia 33 (10):
doi: 10.1093/jn/110.3.488. 593–596. doi: 10.1007/BF00400202.
Andersen, D., and B. Holub. 1980. The relative response of hepatic lip- Balla, T. 2013. Phosphoinositides: Tiny lipids with giant impact on cell
ids in the rat to graded levels of dietary myo-inositol and other lipo- regulation. Physiological Reviews 93 (3):1019–137. doi: 10.1152/phys-
tropes. The Journal of Nutrition 110 (3):496–504. doi: 10.1093/jn/ rev.00028.2012.
110.3.496. Barberini, L., A. Noto, C. Fattuoni, D. Grapov, A. Casanova, G. Fenu,
Anderson, R. G., B. A. Kamen, K. G. Rothberg, and S. W. Lacey. 1992. M. Gaviano, R. Carboni, G. Ottonello, M. Crisafulli, et al. 2014.
Potocytosis: Sequestration and transport of small molecules by cav- Urinary metabolomics (GC-MS) reveals that low and high birth
eolae. Science255 (5043):410–412. doi: 10.1126/science.1310359. weight infants share elevated inositol concentrations at birth. The
Antonsson, B. E. 1994. Purification and characterization of phosphati- Journal of Maternal-Fetal & Neonatal Medicine 27 (sup2):20–26.,
dylinositol synthase from human placenta. Biochemical Journal 297 doi: 10.3109/14767058.2014.954786.
(3):517–522. doi: 10.1042/bj2970517. Barker, C. J., and P.-O. Berggren. 2013. New horizons in cellular regu-
Antony, P. J., G. R. Gandhi, A. Stalin, K. Balakrishna, E. Toppo, K. lation by inositol polyphosphates: Insights from the pancreatic
Sivasankaran, S. Ignacimuthu, and N. A. Al-Dhabi. 2017. b-cell. Pharmacological Reviews 65 (2):641–69. doi: 10.1124/pr.112.
Myoinositol ameliorates high-fat diet and streptozotocin-induced 006775.
diabetes in rats through promoting insulin receptor signaling. Barker, D. 2004. The developmental origins of adult disease. Journal of
Biomedicine & Pharmacotherapy ¼ Biomedecine & Pharmacotherapie the American College of Nutrition 23 (sup6):588S–595S. doi: 10.
88:1098–113. doi: 10.1016/j.biopha.2017.01.170. 1080/07315724.2004.10719428.
Aouameur, R., S. Da Cal, P. Bissonnette, M. J. Coady, and J.-Y. Battaglia, F. C., G. Meschia, J. N. Blechner, and D. H. Barron. 1961.
Lapointe. 2007. SMIT2 mediates all myo-inositol uptake in apical The free myo-inositol concentration of adult and fetal tissues of sev-
membranes of rat small intestine. American Journal of Physiology. eral species. Quarterly Journal of Experimental Physiology and
Gastrointestinal and Liver Physiology 293 (6):G1300–G1307. doi: 10. Cognate Medical Sciences 46:188–193. doi: 10.1113/expphysiol.1961.
1152/ajpgi.00422.2007. sp001532.
Arner, R. J., K. S. Prabhu, J. T. Thompson, G. R. Hildenbrandt, A. D. Baumann, M. U., H. Schneider, A. Malek, V. Palta, D. V. Surbek, R.
Liken, and C. C. Reddy. 2001. Myo-Inositol oxygenase: Molecular Sager, S. Zamudio, and N. P. Illsley. 2014. Regulation of human
cloning and expression of a unique enzyme that oxidizes myo-inosi- trophoblast GLUT1 glucose transporter by insulin-like growth factor
tol and D-chiro-inositol. The Biochemical Journal 360 (Pt 2): I (IGF-I). PLoS One 9 (8):e106037. doi: 10.1371/journal.pone.
313–320. doi: 10.1042/bj3600313. 0106037.
Arner, R. J., K. S. Prabhu, V. Krishnan, M. C. Johnson, and C. C. Bayon, Y., M. Croset, V. Chirouze, J. Tayot, and M. Lagarde. 1993.
Reddy. 2006. Expression of myo-inositol oxygenase in tissues sus- Phospholipid molecular species from human placenta lipids. Lipids
ceptible to diabetic complications. Biochemical and Biophysical 28 (7):631–636. doi: 10.1007/BF02536058.
Research Communications 339 (3):816–820. doi: 10.1016/j.bbrc.2005. Beach, D. C., and P. K. Flick. 1982. Early effect of myo-inositol defi-
11.090. ciency on fatty acid synthetic enzymes of rat liver. Biochimica et
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1661

Biophysica Acta (BBA) - Lipids and Lipid Metabolism 711 (3):452–9. Brown, J., J. Crawford Tineke, and J. Alsweiler. 2015. Myo-inositol for
doi: 10.1016/0005-2760(82)90059-5. preventing gestational diabetes – planning of reveiw. Cochrane
Behuria, B.,. S. Ghimere, S. Choudhary, M. Agrawal, R. P. Bodroth, Database Syst Rev 4
and T. V. G. Rao. 2018. Possible enhancement of phosphatidyl Brusati, V., M. Jozwik, M. J
ozwik, C. Teng, C. Paolini, A. M. Marconi,
inositol (PI) depends on its metabolic pathway components as and F. C. Battaglia. 2005. Fetal and maternal non-glucose carbohy-
second messengers: Evidence from the growth and pi levels of nor- drates and polyols concentrations in normal human pregnancies at
mal, PI grown, and UV exposed Saccharomyces cerevisiae. Global term. Pediatric Research 58 (4):700–704. doi: 10.1203/01.PDR.
Journal of Biology. Agriculture & Health Sciences 7:26–31. 0000180549.86614.73.
Bellamy, L., J. P. Casas, A. D. Hingorani, and D. Williams. 2009. Type Bry, K., and M. Hallman. 1991. Perinatal Development of inositol syn-
2 diabetes mellitus after gestational diabetes: A systematic review thesis and catabolism in rabbit kidney. Neonatology 60 (3-4):
and meta-analysis. The Lancet 373 (9677):1773–1779. doi: 10.1016/ 249–257. doi: 10.1159/000243416.
S0140-6736(09)60731-5. Burgess, J. W., J. Boucher, T. A. Neville, P. Rouillard, C. Stamler, S.
Berridge, M. J. 2009. Inositol trisphosphate and calcium signalling Zachariah, and D. L. Sparks. 2003. Phosphatidylinositol promotes
mechanisms. Biochimica et Biophysica Acta (Bba) - Molecular Cell cholesterol transport and excretion. Journal of Lipid Research 44 (7):
Research 1793 (6):933–40. doi: 10.1016/j.bbamcr.2008.10.005. 1355–63. doi: 10.1194/jlr.M300062-JLR200.
Berridge, M. J., and R. F. Irvine. 1989. Inositol phosphates and cell sig- Burton, G. J., and E. Jauniaux. 2004. Placental oxidative stress: From
nalling. Nature 341 (6239):197–205. doi: 10.1038/341197a0. miscarriage to preeclampsia. Journal of the Society for Gynecologic
Berry, G. T., J. J. Mallee, H. M. Kwon, J. S. Rim, W. R. Mulla, M. Investigation 11 (6):342–52. doi: 10.1016/j.jsgi.2004.03.003.
Muenke, and N. B. Spinner. 1995. The human osmoregulatory Burton, G. J., M. Scioscia, and T. W. Rademacher. 2011. Endometrial
Naþ/myo-inositol cotransporter gene (SLC5A3): molecular cloning secretions: Creating a stimulatory microenvironment within the
and localization to chromosome 21. Genomics 25 (2):507–513. doi: human early placenta and implications for the aetiopathogenesis of
10.1016/0888-7543(95)80052-N. preeclampsia. Journal of Reproductive Immunology 89 (2):118–25.
Berry, G. T., Z. J. Wang, S. F. Dreha, B. M. Finucane, and R. A. doi: 10.1016/j.jri.2011.02.005.
Zimmerman. 1999. In vivo brain myo-inositol levels in children Burton, L. E., and W. W. Wells. 1976. Myo-inositol metabolism during
with Down syndrome. The Journal of Pediatrics 135 (1):94–97. doi: lactation and development in the rat. The prevention of lactation-
10.1016/S0022-3476(99)70334-3. induced fatty liver by dietary myo-inositol. The Journal of Nutrition
Bersudsky, Y., H. Einat, Z. Stahl, and R. H. Belmaker. 1999. Epi-inosi- 106 (11):1617–28. doi: 10.1093/jn/106.11.1617.
tol and inositol depletion: Two new treatment approaches in affect- Burton, L. E., and W. W. Wells. 1977. Characterization of the lacta-
ive disorder. Current Psychiatry Reports 1 (2):141–147. doi: 10.1007/ tion-dependent fatty liver in myo-inositol deficient rats. The Journal
s11920-999-0023-z. of Nutrition 107 (10):1871–83. doi: 10.1093/jn/107.10.1871.
Best, C., C. Lucas, J. M. Patterson, and J. H. Ridout. 1946. The influ- Burton, L. E., and W. W. Wells. 1979. Myo-inositol deficiency: Studies
ence of biotin upon the relative lipotropic effects of choline and on the mechanism of lactation-dependent fatty liver formation in
inositol. The Biochemical Journal 40 (3):368–73. the rat. The Journal of Nutrition 109 (8):1483–91. doi: 10.1093/jn/
Bevilacqua, A., G. Carlomagno, S. Gerli, M. Montanino Oliva, P. 109.8.1483.
Devroey, A. Lanzone, C. Soulange, F. Facchinetti, G. Carlo Di Van Calker, D, and R. H. Belmaker. 2000. The high affinity inositol
Renzo, M. Bizzarri, et al. 2015. Results from the International transport system-implications for the pathophysiology and treatment
Consensus Conference on myo-inositol and D-chiro-inositol in of bipolar disorder. Bipolar Disorders 2 (2):102–7. doi:10.1034/j.
Obstetrics and Gynecology–assisted reproduction technology. 1399-5618.2000.020203.x. PMC: 11252649
Gynecological Endocrinology 31 (6):441–6. doi: 10.3109/09513590. Calker, D., and R. H. Belmaker. 2000. The high affinity inositol trans-
2015.1006616. port system–implications for the pathophysiology and treatment of
Bitsanis, D., M. A. Crawford, T. Moodley, H. Holmsen, K. bipolar disorder. Bipolar Disorders 2 (2):102–7. doi: 10.1034/j.1399-
Ghebremeskel, and O. Djahanbakhch. 2005. Arachidonic acid pre- 5618.2000.020203.x.
dominates in the membrane phosphoglycerides of the early and Calogero, A., G. Gullo, S. La Vignera, R. Condorelli, and A. Vaiarelli.
term human placenta. The Journal of Nutrition 135 (11):2566–71. 2015. Myoinositol improves sperm parameters and serum reproduct-
doi: 10.1093/jn/135.11.2566. ive hormones in patients with idiopathic infertility: A prospective
Bizzarri, M., and G. Carlomagno. 2014. Inositol: History of an effective double-blind randomized placebo-controlled study. Andrology 3 (3):
therapy for polycystic ovary syndrome. European Review for Medical 491–5. doi: 10.1111/andr.12025.
and Pharmacological Sciences 18 (13):1896–903. Campbell, W. W., M. D. Haub, J. D. Fluckey, R. E. Ostlund, Jr J. P.
Blazer-Yost, B. L., and C. Nofziger. 2005. Phosphoinositide lipid second Thyfault, H. Morse-Carrithers, M. W. Hulver, and Z. K. Birge. 2004.
messengers: New paradigms for transepithelial signal transduction. Pinitol supplementation does not affect insulin-mediated glucose
Pflugers Archiv: European Journal of Physiology 450 (2):75–82. doi: metabolism and muscle insulin receptor content and phosphoryl-
10.1007/s00424-004-1371-5. ation in older humans. The Journal of Nutrition 134 (11):2998–3003.
Boone, D. L., W. D. Currie, and P. C. K. Leung. 1993. Arachidonic doi: 10.1093/jn/134.11.2998.
acid and cell signalling in the ovary and placenta. Prostaglandins, Campling, J., and D. Nixon. 1954. The inositol content of foetal blood
Leukotrienes and Essential Fatty Acids 48 (1):79–87. doi: 10.1016/ and foetal fluids. The Journal of Physiology 126 (1):71–80. doi: 10.
0952-3278(93)90013-M. 1113/jphysiol.1954.sp005192.
Bourgeois, F., M. J. Coady, and J.-Y. Lapointe. 2005. Determination of Carlomagno, G., and V. Unfer. 2011. Inositol safety: Clinical evidences.
transport stoichiometry for two cation-coupled myo-inositol European Review for Medical and Pharmacological Sciences 15 (8):
cotransporters: SMIT2 and HMIT. The Journal of Physiology 563 (Pt 931–6.
2):333–343. doi: 10.1113/jphysiol.2004.076679. Carlomagno, G., V. Unfer, and S. Roseff. 2011. The D-chiro-inositol
Brancho, D., N. Tanaka, A. Jaeschke, J.-J. Ventura, N. Kelkar, Y. paradox in the ovary. Fertility and Sterility 95 (8):2515–2516. doi:
Tanaka, M. Kyuuma, T. Takeshita, R. A. Flavell, and R. J. Davis. 10.1016/j.fertnstert.2011.05.027.
2003. Mechanism of p38 MAP kinase activation in vivo. Genes & Caro, H. N., S. Kunjara, T. Rademacher, Y. Leon, D. Jones, M. Avila,
Development 17 (16):1969–1978. doi: 10.1101/gad.1107303. and I. Varela-Nieto. 1997. Isolation and partial characterisation of
Brautigan, D. L., M. Brown, S. Grindrod, G. Chinigo, A. Kruszewski, insulin-mimetic inositol phosphoglycans from human liver.
S. M. Lukasik, J. H. Bushweller, M. Horal, S. Keller, S. Tamura, Biochemical and Molecular Medicine 61 (2):214–28. doi: 10.1006/
et al. 2005. Allosteric activation of protein phosphatase 2C by d- bmme.1997.2607.
chiro-inositol  galactosamine, a putative mediator mimetic of insu- Catalano, P. M., J. P. Kirwan, S. Haugel-de Mouzon, and J. King. 2003.
lin action. Biochemistry 44 (33):11067–73. doi: 10.1021/bi0508845. Gestational diabetes and insulin resistance: Role in short-and long-
1662 O. C. WATKINS ET AL.

term implications for mother and fetus. The Journal of Nutrition randomized controlled trial. Archives of Gynecology and Obstetrics
133 (5 Suppl 2):1674S–1683S. doi: 10.1093/jn/133.5.1674S. 288 (6):1405–11. doi: 10.1007/s00404-013-2855-3.
Cavalli, P., G. Tonni, E. Grosso, and C. Poggiani. 2011. Effects of inosi- Colone, M., G. Marelli, V. Unfer, G. Bozzuto, A. Molinari, and A.
tol supplementation in a cohort of mothers at risk of producing an Stringaro. 2010. Inositol activity in oligoasthenoteratospermia–an
NTD pregnancy. Birth Defects Research Part A: Clinical and in vitro study. European Review for Medical and Pharmacological
Molecular Teratology 91 (11):962–965. doi: 10.1002/bdra.22853. Sciences 14 (10):891–6.
Celentano, C.,. B. Matarrelli, G. Pavone, E. Vitacolonna, P. A. Mattei, Condorelli, R. A., S. La Vignera, L. M. Mongioı, S. G. Vitale, A. S.
V. Berghella, and M. Liberati. 2020. The influence of different inosi- Lagana, L. Cimino, and A. E. Calogero. 2017. A. Myo-inositol as a
tol stereoisomers supplementation in pregnancy on maternal gesta- male fertility molecule: Speed them up. European Review for Medical
tional diabetes mellitus and fetal outcomes in high-risk patients: A and Pharmacological Sciences 21 (2 Suppl):30–35.
randomized controlled trial. The Journal of Maternal-Fetal & Copp, A. J., and N. D. Greene. 2010. Genetics and development of
Neonatal Medicine 33 (5):743–751. doi: 10.1080/14767058.2018. neural tube defects. The Journal of Pathology 220 (2):217–230. doi:
1500545. 10.1002/path.2643.
Ceriello, A., D. Giugliano, A. Quatraro, and F. D’Onofrio. 1988. Does a Coppey, L. J., J. S. Gellett, E. P. Davidson, J. A. Dunlap, and M. A.
common mechanism induce diverse complications of diabetes? Yorek. 2002. Effect of treating streptozotocin-induced diabetic rats
Diabetes Care 11 (4):372–3. doi: 10.2337/diacare.11.4.372. with sorbinil, myo-inositol or aminoguanidine on endoneurial blood
Challier, J., S. Hauguel, and V. Desmaizieres. 1986. Effect of insulin on flow, motor nerve conduction velocity and vascular function of epi-
glucose uptake and metabolism in the human placenta. The Journal neurial arterioles of the sciatic nerve. International Journal of
of Clinical Endocrinology and Metabolism 62 (5):803–807. doi: 10. Experimental Diabetes Research 3 (1):21–36. doi: 10.1080/
1210/jcem-62-5-803. 15604280212525.
Charalampous, F. C. 1959. Biochemical studies on inositol. Journal of Corrado, F., R. D’Anna, G. Di Vieste, D. Giordano, B. Pintaudi, A.
Biological Chemistry 234 (2):220–7. Santamaria, and A. Di Benedetto. 2011. The effect of myoinositol
Cheang, K. I., J. P. Baillargeon, P. A. Essah, R. E. Ostlund, T. supplementation on insulin resistance in patients with gestational
Apridonize, L. Islam, and J. E. Nestler. 2008. Insulin-stimulated diabetes. Diabetic Medicine 28 (8):972–975. doi: 10.1111/j.1464-5491.
release of D-chiro-inositol-containing inositolphosphoglycan medi- 2011.03284.x.
ator correlates with insulin sensitivity in women with polycystic Coupland, N. J., C. J. Ogilvie, K. M. Hegadoren, P. Seres, C. C.
ovary syndrome . Metabolism: clinical and Experimental 57 (10): Hanstock, and P. S. Allen. 2005. Decreased prefrontal Myo-inositol
1390–7. doi: 10.1016/j.metabol.2008.05.008. in major depressive disorder. Biological Psychiatry 57 (12):1526–34.
Cheng, M.-H., and P.-H. Wang. 2009. Placentation abnormalities in doi: 10.1016/j.biopsych.2005.02.027.
the pathophysiology of preeclampsia. Expert Review of Molecular Coustan, D. R. 2013. Can a dietary supplement prevent gestational dia-
Diagnostics 9 (1):37–49. doi: 10.1586/14737159.9.1.37. betes mellitus? Diabetes Care 36 (4):777–9. doi: 10.2337/dc12-2505.
Chiu, T. T., M. S. Rogers, E. L. Law, C. M. Briton-Jones, L. Cheung, Crawford, M. 2000. Placental delivery of arachidonic and docosahexae-
and C. J. Haines. 2002. Follicular fluid and serum concentrations of noic acids: implications for the lipid nutrition of preterm infants.
myo-inositol in patients undergoing IVF: Relationship with oocyte The American Journal of Clinical Nutrition 71 (1 Suppl):275S–284S.
quality. Human Reproduction 17 (6):1591–6. doi: 10.1093/humrep/ doi: 10.1093/ajcn/71.1.275S.
17.6.1591. Crawford, T. J., C. A. Crowther, J. Alsweiler, and J. Brown. 2015.
Chu, A. H., Tint, M. T. Chang, H. F. Wong, G. Yuan, W. L. Tull, D. Antenatal dietary supplementation with myo-inositol in women dur-
Nijagal, B. Narayana, V. K. Meikle, and P. J. Chang Kt. 2020. High ing pregnancy for preventing gestational diabetes. Cochrane Database
placental inositol content associated with suppressed pro-adipogenic of Systematic Reviews
effects of maternal glycaemia in offspring: The GUSTO cohort. Crowther, C. A., J. E. Hiller, J. R. Moss, A. J. McPhee, W. S. Jeffries,
International Journal of Obesity 1–11. and J. S. Robinson. 2005. Australian carbohydrate intolerance study
Chu, S. H. W., and D. Hegsted. 1980. Myo-inositol deficiency in ger- in pregnant women trial G. Effect of treatment of gestational dia-
bils: Comparative study of the intestinal lipodystrophy in Meriones betes mellitus on pregnancy outcomes. New England Journal of
unguiculatus and Meriones libycus. The Journal of Nutrition 110 (6): Medicine 352 (24):2477–2486. doi: 10.1056/NEJMoa042973.
1209–16. doi: 10.1093/jn/110.6.1209. Croze, M. L., A. Gelo€en, and C. O. Soulage. 2015. Abnormalities in
Chukwuma, C. I., M. A. Ibrahim, and M. S. Islam. 2016. Myo-inositol myo-inositol metabolism associated with type 2 diabetes in mice fed
inhibits intestinal glucose absorption and promotes muscle glucose a high-fat diet: Benefits of a dietary myo-inositol supplementation.
uptake: A dual approach study. Journal of Physiology and British Journal of Nutrition 113 (12):1862–75. doi: 10.1017/
Biochemistry 72 (4):791–801. doi: 10.1007/s13105-016-0517-1. S000711451500121X.
Cianci, A., M. Panella, M. Fichera, C. Falduzzi, M. Bartolo, and S. Croze, M. L., and C. O. Soulage. 2013. Potential role and therapeutic
Caruso. 2015. D-chiro-Inositol and alpha lipoic acid treatment of interests of myo-inositol in metabolic diseases. Biochimie 95 (10):
metabolic and menses disorders in women with PCOS. 1811–27. doi: 10.1016/j.biochi.2013.05.011.
Gynecological Endocrinology 31 (6):483–6. doi: 10.3109/09513590. Czech, M. P. 2000. PIP2 and PIP3: Complex roles at the cell surface.
2015.1014784. Cell 100 (6):603–6. doi: 10.1016/S0092-8674(00)80696-0.
Clements Jr, R. 1979. The metabolism of myo-inositol by the human D’Anna, R., and A. Santamaria. 2018. Myo-inositol supplementation in
kidney. Journal of Laboratory and Clinical Medicine 93:210–9. gestational diabetes. Nutrition and diet in maternal diabetes,
Cline, G. W., K. F. Petersen, M. Krssak, J. Shen, R. S. Hundal, Z. 229–35.Springer.
Trajanoski, S. Inzucchi, A. Dresner, D. L. Rothman, and G. I. D’anna, R., V. Di Benedetto, P. Rizzo, E. Raffone, M. Interdonato, F.
Shulman. 1999. Impaired glucose transport as a cause of decreased Corrado, and A. Di Benedetto. 2012. Myo-inositol may prevent ges-
insulin-stimulated muscle glycogen synthesis in type 2 diabetes. The tational diabetes in PCOS women. Gynecological Endocrinology 28
New England Journal of Medicine 341 (4):240–6. doi: 10.1056/ (6):440–2. doi: 10.3109/09513590.2011.633665.
NEJM199907223410404. D’Oria, R., L. Laviola, M. Scioscia, F. Fascilla, S. Bettocchi, and F.
Cogram, P., S. Tesh, J. Tesh, A. Wade, G. Allan, N. D. Greene, and Giorgino. 2017. PKB/Akt phosphorylation in human umbilical vein
A. J. Copp. 2002. D-chiro-inositol is more effective than myo-inosi- endothelial cells is highly induced by myo-inositol and D-chiro
tol in preventing folate-resistant mouse neural tube defects. Human inositol: Novel potential insights in the pathogenesis of preeclamp-
Reproduction (Oxford, England) 17 (9):2451–2458. doi: 10.1093/ sia. Pregnancy Hypertension: An International Journal of Women’s
humrep/17.9.2451. Cardiovascular Health 7:56. doi: 10.1016/j.preghy.2016.10.005.
Colazingari, S., M. Treglia, R. Najjar, and A. Bevilacqua. 2013. The Dʼ Anna, R., A. Di Benedetto, A. Scilipoti, A. Santamaria, M. L.
combined therapy myo-inositol plus D-chiro-inositol, rather than D- Interdonato, E. Petrella, I. Neri, B. Pintaudi, F. Corrado, and F.
chiro-inositol, is able to improve IVF outcomes: Results from a Facchinetti. 2015. Myo-inositol supplementation for prevention of
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1663

gestational diabetes in obese pregnant women: A randomized con- 2009. Evaluation of expression and function of the Hþ/myo-inositol
trolled trial. Obstetrics and Gynecology 126 (2):310–315. doi: 10. transporter HMIT. BMC Cell Biology 10 (1):54. doi: 10.1186/1471-
1097/AOG.0000000000000958. 2121-10-54.
D’Anna, R., A. Scilipoti, D. Giordano, C. Caruso, M. L. Cannata, M. L. Di Paolo, G., and P. De Camilli. 2006. Phosphoinositides in cell regula-
Interdonato, F. Corrado, and A. Di Benedetto. 2013. Di Benedetto tion and membrane dynamics. Nature 443 (7112):651–7. doi: 10.
A. myo-Inositol supplementation and onset of gestational diabetes 1038/nature05185.
mellitus in pregnant women with a family history of type 2 diabetes: Dona, G., C. Sabbadin, C. Fiore, M. Bragadin, F. L. Giorgino, E.
A prospective, randomized, placebo-controlled study. Diabetes Care Ragazzi, C. G. Bordin, and L. Armanini. 2012. D. Inositol adminis-
36 (4):854–857. doi: 10.2337/dc12-1371. tration reduces oxidative stress in erythrocytes of patients with poly-
Daughaday, W. H., and J. Larner. 1954. The renal excretion of inositol cystic ovary syndrome. European Journal of Endocrinology 166:
in normal and diabetic human beings. The Journal of Clinical 703–10.
Investigation 33 (3):326–32. doi: 10.1172/JCI102901. D’Oria, R., L. Laviola, F. Giorgino, V. Unfer, S. Bettocchi, and M.
Davanzo, P., K. Yue, M. A. Thomas, T. Belin, J. Mintz, T. Scioscia. 2017. PKB/Akt and MAPK/ERK phosphorylation is highly
Venkatraman, E. Santoro, S. Barnett, and J. McCracken. 2003. induced by inositols: Novel potential insights in endothelial dysfunc-
Proton magnetic resonance spectroscopy of bipolar disorder versus tion in preeclampsia. Pregnancy Hypertension 10:107–112. doi: 10.
intermittent explosive disorder in children and adolescents. The 1016/j.preghy.2017.07.001.
American Journal of Psychiatry 160 (8):1442–52. doi: 10.1176/appi. D’Souza, K., and R. M. Epand. 2014. Enrichment of phosphatidylinosi-
ajp.160.8.1442. tols with specific acyl chains. Biochimica et Biophysica Acta (BBA) -
Davanzo, P., M. A. Thomas, K. Yue, T. Oshiro, T. Belin, M. Strober, Biomembranes 1838 (6):1501–8. doi: 10.1016/j.bbamem.2013.10.003.
and J. McCracken. 2001. Decreased anterior cingulate myo-inositol/ Ehrmann, D. A. 2005. Polycystic ovary syndrome. The New England
creatine spectroscopy resonance with lithium treatment in children Journal of Medicine 352 (12):1223–36. doi: 10.1056/NEJMra041536.
with bipolar disorder. Neuropsychopharmacology: Official Publication Ehrmann, D. A., R. B. Barnes, R. L. Rosenfield, M. K. Cavaghan, and J.
of the American College of Neuropsychopharmacology 24 (4):359–69. Imperial. 1999. Prevalence of impaired glucose tolerance and dia-
doi: 10.1016/S0893-133X(00)00207-4. betes in women with polycystic ovary syndrome. Diabetes Care 22
Davis, A., M. Christiansen, J. F. Horowitz, S. Klein, M. K. Hellerstein, (1):141–6. doi: 10.2337/diacare.22.1.141.
and R. E. Ostlund. 2000. Effect of pinitol treatment on insulin action Einat, H., and R. Belmaker. 2001. The effects of inositol treatment in
in subjects with insulin resistance. Diabetes Care 23 (7):1000–5. doi: animal models of psychiatric disorders. Journal of Affective Disorders
10.2337/diacare.23.7.1000. 62 (1-2):113–121. doi: 10.1016/S0165-0327(00)00355-4.
Dawonauth, L., L. Rademacher, A. D. L’Omelette, S. Jankee, M. Y. L. Eisenberg, F., Jr, and A. H. Bolden. 1963. Biosynthesis of inositol in rat
K. Yan, R. B. Jeeawoody, and T. W. Rademacher. 2014. Urinary testis homogenate. Biochemical and Biophysical Research
inositol phosphoglycan-P type: Near patient test to detect pree- Communications 12 (1):72–77. doi: 10.1016/0006-291X(63)90416-9.
clampsia prior to clinical onset of the disease. A study on 416 preg- Elased, K. M., K. Gumaa, J. De Souza, H. Rahmoune, J. H. Playfair,
nant Mauritian women. Journal of Reproductive Immunology 101-
and T. Rademacher. 2001. Reversal of type 2 diabetes in mice by
102:148–152. doi: 10.1016/j.jri.2013.06.001.
products of malaria parasites: II. Role of inositol phosphoglycans
De, V. L., A. M. La, V. Cappelli, A. Stendardi, R. Focarelli, M.
(IPGs). Molecular Genetics and Metabolism 73 (3):248–58. doi: 10.
Musacchio, and P. Piomboni. 2012. Evaluation of the treatment with
1006/mgme.2001.3186.
D-chiro-inositol on levels of oxidative stress in PCOS patients.
Eriksson, U. J., and L. H. Borg. 1993. Diabetes and embryonic malfor-
Minerva Ginecologica 64:531–8.
mations: Role of substrate-induced free-oxygen radical production
Debierre-Grockiego, F., and R. T. Schwarz. 2010. Immunological reac-
for dysmorphogenesis in cultured rat embryos. Diabetes 42 (3):
tions in response to apicomplexan glycosylphosphatidylinositolsF
411–9. doi: 10.2337/diab.42.3.411.
Debierre-Grockiego and R T Schwarz. Glycobiology 20 (7):801–11.
Eriksson, U., and L. Borg. 1991. Protection by free oxygen radical scav-
doi: 10.1093/glycob/cwq038.
enging enzymes against glucose-induced embryonic malformations
Deborde, S., J. Schofield, and T. Rademacher. 2003. Placental GPI-PLD
is of maternal origin and its GPI substrate is absent from placentae in vitro. Diabetologia 34 (5):325–31. doi: 10.1007/BF00405004.
Facchinetti, F., M. Bizzarri, S. Benvenga, R. D’Anna, A. Lanzone, C.
of pregnancies associated with pre-eclampsia. Journal of
Reproductive Immunology 59 (2):277–294. doi: 10.1016/S0165- Soulage, D. G. C. Renzo, M. Hod, P. Cavalli, and T. T. Chiu. 2015.
0378(03)00054-8. Results from the International Consensus Conference on Myo-inosi-
Del Monte, M. A., R. Rabbani, T. C. Diaz, S. A. Lattimer, J. Nakamura, tol and d-chiro-inositol in Obstetrics and Gynecology: The link
M. C. Brennan, and D. A. Greene. 1991. Sorbitol, myo-inositol, and between metabolic syndrome and PCOS. European Journal of
rod outer segment phagocytosis in cultured hRPE cells exposed to Obstetrics & Gynecology and Reproductive Biology 195:72–6.
glucose: In vitro model of myo-inositol depletion hypothesis of dia- Fagerberg, L., B. M. Hallstr€ om, P. Oksvold, C. Kampf, D. Djureinovic,
betic complications. Diabetes 40 (10):1335–45. doi: 10.2337/diabetes. J. Odeberg, M. Habuka, S. Tahmasebpoor, A. Danielsson, K.
40.10.1335. Edlund, et al. 2014. Analysis of the human tissue-specific expression
Delhaes, F., S. A. Giza, T. Koreman, G. Eastabrook, C. A. McKenzie, S. by genome-wide integration of transcriptomics and antibody-based
Bedell, T. R. H. Regnault, and B. de Vrijer. 2018. Altered maternal proteomics. Molecular & Cellular Proteomics: Proteomics 13 (2):
and placental lipid metabolism and fetal fat development in obesity: 397–406. doi: 10.1074/mcp.M113.035600.
Current knowledge and advances in non-invasive assessment. Falavigna, M., M. I. Schmidt, J. Trujillo, L. F. Alves, E. R. Wendland,
Placenta 69:118–124. doi: 10.1016/j.placenta.2018.05.011. M. R. Torloni, S. Colagiuri, and B. B. Duncan. 2012. Effectiveness of
Desoye, G., and S. Hauguel-de Mouzon. 2007. The human placenta in gestational diabetes treatment: A systematic review with quality of
gestational diabetes mellitus: The insulin and cytokine network. evidence assessment. Diabetes Research and Clinical Practice 98 (3):
Diabetes Care 30 (Supplement 2):S120–S126. doi: 10.2337/dc07-s203. 396–405. doi: 10.1016/j.diabres.2012.09.002.
Dessı, A., and V. Fanos. 2013. Myoinositol: A new marker of intrauter- Farese, R. V. 1983. The phosphatidate-phosphoinositide cycle: An
ine growth restriction? Journal of Obstetrics and Gynaecology: intracellular messenger system in the action of hormones and neu-
Gynaecology 33 (8):776–80. doi: 10.3109/01443615.2013.831046. rotransmitters. Metabolism 32 (6):628–41. doi: 10.1016/0026-
Di Cianni, G., R. Miccoli, L. Volpe, C. Lencioni, and S. Del Prato. 0495(83)90035-5.
2003. Intermediate metabolism in normal pregnancy and in gesta- Farese, R. V., M. Standaert, K. Yamada, L. Huang, C. Zhang, D.
tional diabetes. Diabetes/Metabolism Research and Reviews 19 (4): Cooper, Z. Wang, Y. Yang, S. Suzuki, and T. Toyota. 1994. Insulin-
259–270. doi: 10.1002/dmrr.390. induced activation of glycerol-3-phosphate acyltransferase by a
Di Daniel, E., M. H. Mok, E. Mead, C. Mutinelli, E. Zambello, L. L. chiro-inositol-containing insulin mediator is defective in adipocytes
Caberlotto, T. J. Pell, C. J. Langmead, A. J. Shah, G. Duddy, et al. of insulin-resistant, type II diabetic, Goto-Kakizaki rats. Proceedings
1664 O. C. WATKINS ET AL.

of the National Academy of Sciences 91 (23):11040–4. doi: 10.1073/ Garcia-Perez, A., and M. B. Burg. 1990. Importance of organic osmo-
pnas.91.23.11040. lytes for osmoregulation by renal medullary cells. Hypertension16
Farren, M., N. Daly, A. McKeating, B. Kinsley, M. J. Turner, and S. (6):595–602. doi: 10.1161/01.HYP.16.6.595.
Daly. 2017. The prevention of gestational diabetes mellitus with Gateva, A., V. Unfer, and Z. Kamenov. 2018. The use of inositol (s)
antenatal oral inositol supplementation: A randomized controlled isomers in the management of polycystic ovary syndrome: A com-
trial. Diabetes Care 40 (6):759–763. doi: 10.2337/dc16-2449. prehensive review. Gynecological Endocrinology 34 (7):545–6. doi: 10.
Fenili, D. 2010. Characterization of inositol transporters as a method 1080/09513590.2017.1421632.
for drug delivery to the central nervous system. Gavin, G., and E. McHenry. 1941. Inositol: A lipotropic factor. Journal
Fenili, D., M. Brown, R. Rappaport, and J. McLaurin. 2007. Properties of Biological Chemistry 139
of scyllo–inositol as a therapeutic treatment of AD-like pathology. Genazzani, A. D., A. Prati, S. Santagni, F. Ricchieri, E. Chierchia, E.
Journal of Molecular Medicine (Berlin, Germany) 85 (6):603–11. doi: Rattighieri, A. Campedelli, T. Simoncini, and P. G. Artini. 2012.
10.1007/s00109-007-0156-7. Differential insulin response to myo-inositol administration in obese
Fisher, S. K., J. E. Novak, and B. W. Agranoff. 2002. Inositol and polycystic ovary syndrome patients. Gynecological Endocrinology 28
higher inositol phosphates in neural tissues: Homeostasis, metabol- (12):969–973. doi: 10.3109/09513590.2012.685205.
ism and functional significance. Journal of Neurochemistry 82 (4): Gerli, S., E. Papaleo, A. Ferrari, D. Renzo, and G. Randomized. 2007.
736–754. doi: 10.1046/j.1471-4159.2002.01041.x. Double blind placebo-controlled trial: Effects of myo-inositol on
Formuso, C., M. Stracquadanio, and L. Ciotta. 2015. Myo-inositol vs. ovarian function and metabolic factors in women with PCOS.
D-chiro inositol in PCOS treatment. Minerva Ginecologica 67 (4): European Review for Medical and Pharmacological Sciences 11:
321–325. 347–54.
Fraser, D., S. Weitzman, J. Leiberman, and E. Eschwege. 1990. Factors Ghulmiyyah, L., and B. Sibai. 2012. Maternal mortality from pree-
influencing birth weight in newborns of diabetic and non-diabetic clampsia/eclampsia. Seminars in Perinatology 36 (1):56–59. doi: 10.
women a population based study. European Journal of Epidemiology 1053/j.semperi.2011.09.011.
6 (4):427–431. doi: 10.1007/BF00151720. Giordano, D., F. Corrado, A. Santamaria, S. Quattrone, B. Pintaudi, A.
Fraticelli, F.,. C. Celentano, I. A. Zecca, G. Di Vieste, B. Pintaudi, M. Di Benedetto, and R. D’Anna. 2011. Effects of myo-inositol supple-
Liberati, M. Franzago, M. Di Nicola, and E. Vitacolonna. 2018. mentation in postmenopausal women with metabolic syndrome: A
Effect of inositol stereoisomers at different dosages in gestational perspective, randomized, placebo-controlled study. Menopause18 (1):
diabetes: An open-label, parallel, randomized controlled trial. Acta 102–104. doi: 10.1097/gme.0b013e3181e8e1b1.
Diabetologica 55 (8):805–812. doi: 10.1007/s00592-018-1157-4. Godfrey, K. M., W. Cutfield, S. Y. Chan, P. N. Baker, and Y. S. Chong.
Freedman, N. D., R. Sinha, S. C. Moore, A. J. Cross, J. N. Sampson, S. 2017. Nutritional intervention preconception and during pregnancy
Boca, N. E. Caporaso, and W.-Y. Huang. 2014. Metabolites of
to maintain healthy glucose metabolism and offspring health
tobacco smoking and colorectal cancer risk. Carcinogenesis 35 (7):
(“NiPPeR”): Study protocol for a randomised controlled trial. Trials
1516–22. doi: 10.1093/carcin/bgu071.
18 (1):131. doi: 10.1186/s13063-017-1875-x.
Frej, A. D., G. P. Otto, and R. S. Williams. 2017. Tipping the scales:
Goel, M., and V. N. Azev. 2009. The biological activity of structurally
Lessons from simple model systems on inositol imbalance in neuro-
defined inositol glycans.
logical disorders. European Journal of Cell Biology 96 (2):154–163.
Goodarzi, M. O., D. A. Dumesic, G. Chazenbalk, and R. Azziz. 2011.
doi: 10.1016/j.ejcb.2017.01.007.
Polycystic ovary syndrome: Etiology, pathogenesis and diagnosis.
Frey, R., D. Metzler, P. Fischer, A. Heiden, J. Scharfetter, E. Moser,
Nature Reviews. Endocrinology 7 (4):219–31. doi: 10.1038/nrendo.
and S. Kasper. 1998. Myo-inositol in depressive and healthy subjects
2010.217.
determined by frontal 1H-magnetic resonance spectroscopy at 1.5
Grases, F., Simonet, B. M. Vucenik, I. Prieto, R. M. Costa,-Bauza, A.
tesla. Journal of Psychiatric Research 32 (6):411–20. doi: 10.1016/
March, J. G. Shamsuddin. and A. M. 2001. Absorption and excre-
S0022-3956(98)00033-8.
Fruen, B., and B. Lester. 1991. High-affinity [3 H] inositol uptake by tion of orally administered inositol hexaphosphate (IP6 or phytate)
dissociated brain cells and cultured fibroblasts from fetal mice. in humans. BioFactors 15 (1):53–61. doi: 10.1002/biof.5520150105.
Neurochemical Research 16 (8):913–918. doi: 10.1007/BF00965541. Greene, D. A., P. V. De Jesus, Jr., and A. I. Winegrad. 1975. Effects of
Fukami, K., and T. Takenawa. 1989. Quantitative changes in polyphos- insulin and dietary myoinositol on impaired peripheral motor nerve
phoinositides 1, 2-diacylglycerol and inositol 1, 4, 5-trisphosphate by conduction velocity in acute streptozotocin diabetes. The Journal of
platelet-derived growth factor and prostaglandin F2 alpha. The Clinical Investigation 55 (6):1326–36. doi: 10.1172/JCI108052.
Journal of Biological Chemistry 264 (25):14985–9. Greene, D., and A. Mackway. 1986. Decreased myo-inositol content
Gaggini, M., C. Saponaro, and A. Gastaldelli. 2015. Not all fats are cre- and Naþ-Kþ-ATPase activity in superior cervical ganglion of STZ-
ated equal: Adipose vs. ectopic fat, implication in cardiometabolic diabetic rat and prevention by aldose reductase inhibition. Diabetes
diseases. Hormone Molecular Biology and Clinical Investigation 22 35 (10):1106–8. doi: 10.2337/diab.35.10.1106.
(1):7–18. doi: 10.1515/hmbci-2015-0006. Greene, D., S. Chakrabarti, S. Lattimer, and A. Sima. 1987. Role of
Galasko, G.,. S. Abe, K. Lilley, C. Zhang, and J. Larner. 1996. sorbitol accumulation and myo-inositol depletion in paranodal swel-
Circulating factors and insulin resistance. II. The action of the novel ling of large myelinated nerve fibers in the insulin-deficient spon-
myo-inositol cyclic 1,2-inositol phosphate phosphoglycan insulin taneously diabetic bio-breeding rat. Reversal by insulin replacement,
antagonist from human plasma in regulating pyruvate dehydrogen- an aldose reductase inhibitor, and myo-inositol. The Journal of
ase phosphatase. The Journal of Clinical Endocrinology and Clinical Investigation 79 (5):1479–85. doi: 10.1172/JCI112977.
Metabolism 81 (3):1051–7. doi: 10.1210/jcem.81.3.8772575. Groenen, P. M., H. M. Merkus, F. C. Sweep, R. A. Wevers, F. S.
Gallo, L. A., H. L. Barrett, and M. D. Nitert. 2017. Review: Placental Janssen, and R. P. Steegers-Theunissen. 2003. Kinetics of myo-inosi-
transport and metabolism of energy substrates in maternal obesity tol loading in women of reproductive age. Annals of Clinical
and diabetes. Placenta 54:59–67. doi: 10.1016/j.placenta.2016.12.006. Biochemistry: International Journal of Laboratory Medicine 40 (1):
Ganapathy, V., P. D. Prasad, M. E. Ganapathy, and F. H. Leibach. 79–85. doi: 10.1258/000456303321016213.
2000. Placental transporters relevant to drug distribution across the Groenen, P. M., P. G. Peer, R. A. Wevers, D. W. Swinkels, B. Franke,
maternal-fetal interface. The Journal of Pharmacology and E. C. Mariman, and R. P. Steegers-Theunissen. 2003. myo-inositol,
Experimental Therapeutics 294 (2):413–420. glucose, and zinc status is associated with the risk of offspring with
Gao J, Nalls MA, Shi M, Joubert BR, Hernandez DG, Huang X, spina bifida. American Journal of Obstetrics and Gynecology 189 (6):
Hollenbeck A, Singleton AB, Chen H. 2012. An exploratory analysis 1713–1719. doi: 10.1016/S0002-9378(03)00807-X.
on gene-environment interactions for Parkinson disease. Groenen, P. M., R. A. Wevers, F. S. Janssen, J. H. Tuerlings, J. M.
Neurobiology of Aging, 33:2528.e1.e2521–2528.e2526 doi: 10.1016/j. Merkus, and R. P. Steegers-Theunissen. 2003. Are myo-inositol, glu-
neurobiolaging.2012.06.007. cose and zinc concentrations in amniotic fluid of fetuses with spina
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1665

bifida different from controls? Early Human Development 71 (1): Hipps, P. P., K. E. Ackermann, and W. R. Sherman. 1982. Inositol epi-
1–8. doi: 10.1016/S0378-3782(02)00040-3. merase - Inosose reductase from bovine brain. Methods in enzym-
Guan, G.,. P. Dai, and I. Shechter. 2003. cDNA cloning and gene ology. Vol 89, 593–8. Academic Press.
expression analysis of human myo-inositol 1-phosphate synthase. Hipps, P. P., W. H. Holland, and W. R. Sherman. 1977.
Archives of Biochemistry and Biophysics 417 (2):251–259. doi: 10. Interconversion of myo- and scyllo-inositol with simultaneous for-
1016/S0003-9861(03)00388-6. mation of neo-inositol by an NADP þ dependent epimerase from
Guo, W., S. Shimada, H. Tajiri, A. Yamauchi, T. Yamashita, S. Okada, bovine brain. Biochemical and Biophysical Research Communications
and M. Tohyama. 1997. Developmental regulation of Naþ/myo- 77 (1):340–346. doi: 10.1016/S0006-291X(77)80202-7.
inositol cotransporter gene expression. Molecular Brain Research 51 Holub, B. J. 1986. Metabolism and function of myo-inositol and inosi-
(1-2):91–96. doi: 10.1016/S0169-328X(97)00220-9. tol phospholipids. Annual Review of Nutrition 6:563–97. doi: 10.
Handwerger, S., and M. Freemark. 2000. The roles of placental growth 1146/annurev.nu.06.070186.003023.
hormone and placental lactogen in the regulation of human fetal Holub, B. J. The nutritional significance, metabolism, and function of
growth and development. Journal of Pediatric Endocrinology & myo-inositol and phosphatidylinositol in health and disease. Advances
Metabolism: JPEM 13 (4):343–356. doi: 10.1515/JPEM.2000.13.4.343. in nutritional research, 1982, 107–41. Springer.
Hansen, B., and N. Bodkin. 1986. Heterogeneity of insulin responses: Hong, J. H., H. W. Jang, Y. E. Kang, J. H. Lee, K. S. Kim, H. J. Kim,
Phases leading to type 2 (non-insulin-dependent) diabetes mellitus K. R. Park, and B. J. Ku. 2012. Urinary chiro-and myo-inositol levels
in the rhesus monkey. Diabetologia 29 (10):713–9. doi: 10.1007/ as a biological marker for type 2 diabetes mellitus. Disease Markers
BF00870281. 33 (4):193–199. doi: 10.1155/2012/734718.
Hansen, S. B. 2015. Lipid agonism: The PIP 2 paradigm of ligand-gated Hori, H.,. T. Sasaoka, H. Ishihara, T. Wada, S. Murakami, M. Ishiki,
ion channels. Biochimica et Biophysica Acta (BBA)-Molecular and and M. Kobayashi. 2002. Association of SH2-containing inositol
Cell Biology of Lipids 1851:620–8. phosphatase 2 with the insulin resistance of diabetic db/db mice.
Harwood, A. J. 2005. Lithium and bipolar mood disorder: The inositol- Diabetes 51 (8):2387–94. doi: 10.2337/diabetes.51.8.2387.
depletion hypothesis revisited. Molecular Psychiatry 10 (1):117–26. Hu, M. L., Y. K. Chen, and Y. F. Lin. 1995. The antioxidant and
doi: 10.1038/sj.mp.4001618. prooxidant activity of some B vitamins and vitamin-like compounds.
Hashimoto, M., S. Akazawa, M. Akazawa, M. Akashi, H. Yamamoto, Chemico-Biological Interactions 97 (1):63–73. doi: 10.1016/0009-
Y. Maeda, Y. Yamaguchi, H. Yamasaki, D. Tahara, and T. 2797(95)03608-8.
Nakanishi. 1990. Effects of hyperglycaemia on sorbitol and myo- Huang, L., M. Fonteles, D. Houston, C. Zhang, and J. Larner. 1993.
inositol contents of cultured embryos: Treatment with aldose reduc- Chiroinositol deficiency and insulin resistance. III. Acute glycogenic
tase inhibitor and myo-inositol supplementation. Diabetologia 33 and hypoglycemic effects of two inositol phosphoglycan insulin
(10):597–602. doi: 10.1007/BF00400203. mediators in normal and streptozotocin-diabetic rats in vivo.
Hauguel-de Mouzon, S., J. Lepercq, and P. Catalano. 2006. The known Endocrinology 132 (2):652–7. doi: 10.1210/endo.132.2.8425485.
and unknown of leptin in pregnancy. American Journal of Obstetrics I, E., H. d. Valk, B. Moll, E. t. Braak, and V. G. 2002. Macrosomia
and Gynecology 194 (6):1537–1545. doi: 10.1016/j.ajog.2005.06.064. despite good glycaemic control in Type I diabetic pregnancy; results
Hauser, G., and V. N. Finelli. 1963. The biosynthesis of free and phos- of a nationwide study in The Netherlands. Diabetologia 45 (11):
phatide myo-inositol from glucose by mammalian tissue slices. 1484–9. doi:10.1007/s00125-002-0958-7.
Journal of Biological Chemistry 238:64. Isaacks, R. E., A. S. Bender, C. Y. Kim, Y. F. Shi, and M. D.
H€aussinger, D. 1998. Osmoregulation of liver cell function: signalling, Norenberg. 1999. Effect of osmolality and anion channel inhibitors
osmolytes and cell heterogeneity. Cell Volume Regulation. Vol 123, ; on myo-inositol efflux in cultured astrocytes. Journal of Neuroscience
185–204 Karger Publishers. Research 57 (6):866–871. doi: 10.1002/(SICI)1097-
Hayashi, E., R. Hasegawa, and T. Tomita. 1976. Accumulation of neu- 4547(19990915)57:6<866::AID-JNR12>3.0.CO;2-K.
tral lipids in Saccharomyces carlsbergensis by myo-inositol defi- Ishida, S., A. Funakoshi, K. Miyasaka, H. Shimokata, F. Ando, and S.
ciency and its mechanism. Reciprocal regulation of yeast acetyl-CoA Takiguchi. 2006. Association of SH-2 containing inositol 50 -phos-
carboxylase by fructose bisphosphate and citrate. The Journal of phatase 2 gene polymorphisms and hyperglycemia. Pancreas 33:
Biological Chemistry 251 (18):5759–69. 63–7. doi: 10.1097/01.mpa.0000222317.82231.16.
Hayashi, E., T. Maeda, R. Hasegawa, and T. Tomita. 1978. The effect Islam, M. O., P. Selvam, R. Appukuttan Pillai, O. C. Watkins, and S. Y.
of myo-inositol deficiency on lipid metabolism in rats: III. The Chan. 2019. An enzymatic assay for quantification of inositol in
mechanism of an enhancement in lipolysis due to myo-inositol defi- human term placental tissue. Analytical Biochemistry 586:113409.
ciency in rats. Biochimica et Biophysica Acta 531 (2):197–205. doi: doi: 10.1016/j.ab.2019.113409.
10.1016/0005-2760(78)90143-1. Iuorno, M., Maria, J. Jakubowicz, M. Daniela, J. Baillargeon, M. Jean-
Hegsted, D., K. Hayes, A. Gallagher, and H. Hanford. 1973. Inositol Patrice, D. B. Pamela, Gunn, M. Ronald, D. Allan, P. Nestler, G,
deficiency: An intestinal lipodystrophy in the gerbil. The Journal of et al. 2002. Effects of D-chiro-inositol in lean women with the poly-
Nutrition 103 (2):302–7. doi: 10.1093/jn/103.2.302. cystic ovary syndrome. Endocrine Practice: Official Journal of the
Heimark, D., J. McAllister, and J. Larner. 2014. Decreased myo-inositol American College of Endocrinology and the American Association of
to chiro-inositol (M/C) ratios and increased M/C epimerase activity Clinical Endocrinologists 8 (6):417–23. doi: 10.4158/EP.8.6.417.
in PCOS theca cells demonstrate increased insulin sensitivity com- Jackson, P. S., and K. Strange. 1993. Volume-sensitive anion channels
pared to controls. Endocrine Journal 61 (2):111–7. doi: 10.1507/ mediate swelling-activated inositol and taurine efflux. American
endocrj.EJ13-0423. Journal of Physiology-Cell Physiology 265 (6):C1489–C1500. doi: 10.
Hernandez-Mijares, A., C. Ba~ nuls, J. E. Peris, N. Monz
o, A. Jover, L. 1152/ajpcell.1993.265.6.C1489.
Bellod, V. M. Victor, and M. Rocha. 2013. A single acute dose of Jansson, N., F. J. Rosario, F. Gaccioli, S. Lager, H. N. Jones, S. Roos, T.
pinitol from a naturally-occurring food ingredient decreases hyper- Jansson, and T. L. Powell. 2013. Activation of placental mTOR sig-
glycaemia and circulating insulin levels in healthy subjects. Food naling and amino acid transporters in obese women giving birth to
Chemistry 141 (2):1267–72. doi: 10.1016/j.foodchem.2013.04.042. large babies. The Journal of Clinical Endocrinology & Metabolism 98
Herrera, E., and H. Ortega-Senovilla. 2010. Disturbances in lipid (1):105–113. doi: 10.1210/jc.2012-2667.
metabolism in diabetic pregnancy–are these the cause of the prob- Jayabalan, N., S. Nair, Z. Nuzhat, G. E. Rice, F. A. Zu~ niga, L. Sobrevia,
lem? Best Practice & Research Clinical Endocrinology & Metabolism A. Leiva, C. Sanhueza, J. A. Gutierrez, M. Lappas, et al. 2017. Cross
24 (4):515–525. doi: 10.1016/j.beem.2010.05.006. talk between adipose tissue and placenta in obese and gestational
Herrera, E., and H. Ortega-Senovilla. 2018. Implications of lipids in diabetes mellitus pregnancies via exosomes. Frontiers in
neonatal body weight and fat mass in gestational diabetic mothers Endocrinology 8:239. doi: 10.3389/fendo.2017.00239.
and non-diabetic controls. Current Diabetes Reports 18 (2):7. doi: Jenkins, C., R. Wilson, J. Roberts, H. Miller, J. H. McKillop, and J. J.
10.1007/s11892-018-0978-4. Walker. 2000. Antioxidants: Their role in pregnancy and
1666 O. C. WATKINS ET AL.

miscarriage. Antioxidants & Redox Signaling 2 (3):623–8. doi: 10. Kondoh, G., H. Tojo, Y. Nakatani, N. Komazawa, C. Murata, K.
1089/15230860050192369. Yamagata, Y. Maeda, T. Kinoshita, M. Okabe, R. Taguchi, et al.
Jiang, W. D., L. Feng, Y. Liu, J. Jiang, and X. Q. Zhou. 2009. Myo- 2005. Angiotensin-converting enzyme is a GPI-anchored protein
inositol prevents oxidative damage, inhibits oxygen radical gener- releasing factor crucial for fertilization. Nature Medicine 11 (2):
ation and increases antioxidant enzyme activities of juvenile Jian 160–6. doi: 10.1038/nm1179.
carp (Cyprinus carpio var. Jian. Aquaculture Research 40 (15): Krystal, G. 2000. Lipid phosphatases in the immune system. Seminars
1770–6. doi: 10.1111/j.1365-2109.2009.02283.x. in Immunology 12 (4):397–403. doi: 10.1006/smim.2000.0222.
Johansson, M., T. Jansson, and T. Powell. 2000. Naþ-Kþ-ATPase is Kunjara, S., A. L. Greenbaum, D.-Y. Wang, H. N. Caro, P. McLean,
distributed to microvillous and basal membrane of the syncytiotro- C. W. Redman, and T. W. Rademacher. 2000. Inositol phosphogly-
phoblast in human placenta. American Journal of Physiology- cans and signal transduction systems in pregnancy in preeclampsia
Regulatory, Integrative and Comparative Physiology 279 (1): and diabetes: Evidence for a significant regulatory role in pree-
R287–R294. doi: 10.1152/ajpregu.2000.279.1.R287. clampsia at placental and systemic levels. Molecular Genetics and
Jung, T.-S., J.-R. Hahm, J.-J. Kim, J.-H. Jung, M.-Y. Kang, S.-W. Moon, Metabolism 69 (2):144–58. doi: 10.1006/mgme.2000.2964.
K.-W. Lee, H.-C. Kim, J.-D. Lee, J.-H. Kim, et al. 2005. Kunjara, S., D. Y. Wang, A. L. Greenbaum, P. McLean, A. Kurtz, and
Determination of urinary myo-/chiro-inositol ratios from Korean T. W. Rademacher. 1999. Inositol phosphoglycans in diabetes and
diabetes patients. Yonsei Medical Journal 46 (4):532–8. doi: 10.3349/ obesity: Urinary levels of IPG A-type and IPG P-type, and relation-
ymj.2005.46.4.532. ship to pathophysiological changes. Molecular Genetics and
Kaaja, R., H. Laivuori, M. Laakso, M. J. Tikkanen, and O. Ylikorkala. Metabolism 68 (4):488–502. doi: 10.1006/mgme.1999.2936.
1999. Evidence of a state of increased insulin resistance in pree- Kunjara, S., D. Y. Wang, P. McLean, A. L. Greenbaum, and T. W.
clampsia. Metabolism 48 (7):892–896. doi: 10.1016/S0026- Rademacher. 2000. Inositol phosphoglycans and the regulation of
0495(99)90225-1. the secretion of leptin: In vitro effects on leptin release from adipo-
Kagawa, S., T. Sasaoka, S. Yaguchi, H. Ishihara, H. Tsuneki, S. cytes and the relationship to obesity. Molecular Genetics and
Murakami, K. Fukui, T. Wada, S. Kobayashi, I. Kimura, et al. 2005. Metabolism 70 (1):61–68. doi: 10.1006/mgme.2000.2988.
Impact of SRC homology 2-containing inositol 50 -phosphatase 2 Lagana, A. S., L. Barbaro, and A. Pizzo. 2015. Evaluation of ovarian
gene polymorphisms detected in a Japanese population on insulin function and metabolic factors in women affected by polycystic
signaling. The Journal of Clinical Endocrinology & Metabolism 90 ovary syndrome after treatment with D-Chiro-Inositol. Archives of
(5):2911–9., doi: 10.1210/jc.2004-1724. Gynecology and Obstetrics 291 (5):1181–6. doi: 10.1007/s00404-014-
Kalra, B., S. Kalra, and J. Sharma. 2016. The inositols and polycystic 3552-6.
ovary syndrome. Indian Journal of Endocrinology and Metabolism 20 Lager, S., and T. L. Powell. 2012. Regulation of nutrient transport
across the placenta. Journal of Pregnancy 2012:1–14. doi: 10.1155/
(5):720–724. doi: 10.4103/2230-8210.189231.
2012/179827.
Kang, M.J., J.I. Kim, S.Y. Yoon, J. C. Kim, and I.J. Cha. 2006. Pinitol
Lamar, M., C. M. L. Foy, F. Beacher, E. Daly, M. Poppe, N. Archer, V.
from soybeans reduces postprandial blood glucose in patients with
Prasher, K. C. Murphy, R. G. Morris, A. Simmons, et al. 2011.
type 2 diabetes mellitus. Journal of Medicinal Food 9 (2):182–6. doi:
Down syndrome with and without dementia: An in vivo proton
10.1089/jmf.2006.9.182.
magnetic resonance spectroscopy study with implications for
Kawa, J. M., R. Przybylski, and C. G. Taylor. 2003. Urinary chiro-inosi-
Alzheimer’s disease. Neuroimage 57 (1):63–8. doi: 10.1016/j.neuro-
tol and myo-inositol excretion is elevated in the diabetic db/db
image.2011.03.073.
mouse and streptozotocin diabetic rat. Experimental Biology and
Lampe, D., and B. V. L. Potter. 1993. Synthesis of 2-fluoro-2-deoxy-
Medicine (Maywood, N.J.) 228 (8):907–14. doi: 10.1177/
myo-inositol 1,4,5-trisphosphate and scyllo-inositol 1,2,4-trisphos-
153537020322800806.
phate, novel analogues of the second messenger myo-inositol 1,4,5-
Kennington, A. S., C. R. Hill, J. Craig, C. Bogardus, I. Raz, H. K.
trisphosphate. Tetrahedron Letters 34 (14):2365–8. doi: 10.1016/
Ortmeyer, B. C. Hansen, G. Romero, and J. Larner. 1990. Low urin-
S0040-4039(00)77615-5.
ary chiro-inositol excretion in non-insulin-dependent diabetes melli- Larner, J. 2002. D-chiro-inositol–its functional role in insulin action
tus. New England Journal of Medicine 323 (6):373–8. doi: 10.1056/ and its deficit in insulin resistance. Journal of Diabetes Research 3:
NEJM199008093230603. 47–60.
Kennington, O. A. 1992. Insulin mediators and their inositol Larner, J., D. L. Brautigan, and M. O. Thorner. 2010. D-chiro-inositol
components. glycans in insulin signaling and insulin resistance. Molecular
Kim, H. J., K. S. Park, S. K. Lee, K. W. Min, K. A. Han, Y. K. Kim, Medicine 16 (11-12):543–52. doi: 10.2119/molmed.2010.00107.
and B. J. Ku. 2012. Effects of pinitol on glycemic control, insulin Larner, J., J. D. Price, D. Heimark, L. Smith, G. Rule, T. Piccariello,
resistance and adipocytokine levels in patients with type 2 diabetes M. C. Fonteles, C. Pontes, D. Vale, and L. Huang. 2003. Isolation,
mellitus. Annals of Nutrition and Metabolism 60 (1):1–5. doi: 10. structure, synthesis, and bioactivity of a novel putative insulin medi-
1159/000334834. ator. A galactosamine chiro-inositol pseudo-disaccharide Mn2þ che-
Kim, J. N., S. N. Han, and H. K. Kim. 2014. Phytic acid and myo- late with insulin-like activity. Journal of Medicinal Chemistry 46
inositol support adipocyte differentiation and improve insulin sensi- (15):3283–91. doi: 10.1021/jm030071j.
tivity in 3T3-L1 cells. Nutrition Research 34 (8):723–31. doi: 10. Larque, E., A. Pagan, M. T. Prieto, J. E. Blanco, A. Gil-Sanchez, M.
1016/j.nutres.2014.07.015. Zornoza-Moreno, M. Ruiz-Palacios, A. Gazquez, H. Demmelmair,
Kim, J., E. Dare, S. S. Rajasekaran, S. H. Ryu, P.-O. Berggren, and C. J. J. J. Parrilla, et al. 2014. Placental fatty acid transfer: A key factor in
Barker. 2019. Inositol pyrophosphates and Akt/PKB: Is the pancre- fetal growth. Annals of Nutrition & Metabolism 64 (3-4):247–253.,
atic b-cell the exception to the rule? Cellular Signalling 58:131–6. doi: 10.1159/000365028.
doi: 10.1016/j.cellsig.2019.02.003. Lawrence, J., J. Guinovart, and J. Larner. 1977. Activation of rat adipo-
Kim, J.-I., J. Kim, M.-J. Kang, M.-S. Lee, J.-J. Kim, and I.-J. Cha. 2005. cyte glycogen synthase by insulins. The Journal of Biological
Effects of pinitol isolated from soybeans on glycaemic control and Chemistry 252 (2):444–50.
cardiovascular risk factors in Korean patients with type II diabetes Lazar, D. F., J. J. Knez, M. E. Medof, P. Cuatrecasas, and A. R. Saltiel.
mellitus: A randomized controlled study. European Journal of 1994. Stimulation of glycogen synthesis by insulin in human
Clinical Nutrition 59 (3):456–458. doi: 10.1038/sj.ejcn.1602081. erythroleukemia cells requires the synthesis of glycosyl-phosphatidy-
Kim, M. J., K. H. Yoo, J. H. Kim, Y. T. Seo, B. W. Ha, J. H. Kho, linositol. Proceedings of the National Academy of Sciences 91 (21):
Y. G. Shin, and C. H. Chung. 2007. Effect of pinitol on glucose 9665–9. doi: 10.1073/pnas.91.21.9665.
metabolism and adipocytokines in uncontrolled type 2 diabetes. Lee, G., M. Price, C. Mejia, H. Galan, and J. Arroyo. 2014. Increased
Diabetes Research and Clinical Practice 77 (3):S247–S251. doi: 10. trophoblast expression of NFAT5/TonEBP in pre-eclamptic placen-
1016/j.diabres.2007.01.066. tas and hyperosmolar-treated BeWo cells. European Journal of
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1667

Obstetrics & Gynecology and Reproductive Biology 183:37–43. doi: malaria. ACS Chemical Biology 15 (1):171–8. doi: 10.1021/acschem-
10.1016/j.ejogrb.2014.10.002. bio.9b00739.
Letcher, A. J., M. J. Schell, and R. F. Irvine. 2008. Do mammals make Malvasi, A., F. Casciaro, M. Minervini, I. Kosmas, O. Mynbaev, E.
all their own inositol hexakisphosphate? Biochemical Journal 416 (2): Pacella, V. M. Condesnitt, A. Creanza, D. Renzo, and G. Tinelli.
263–70. doi: 10.1042/BJ20081417. 2014. A. Myo-inositol, D-chiro-inositol, folic acid and manganese in
Lewin, L. M., Y. Yannai, S. Melmed, and M. Weiss. 1982. myo-inositol second trimester of pregnancy: A preliminary investigation.
in the reproductive tract of the female rat. The International Journal European Review for Medical and Pharmacological Sciences. 18:
of Biochemistry 14 (2):147–150. doi: 10.1016/0020-711X(82)90154-9. 270–274.
Lewis, R. M., C. Wadsack, and G. Desoye. 2018. Placental fatty acid Manna, P., and S. K. Jain. 2013. PIP3 but not PIP2 increases GLUT4
transfer. Current Opinion in Clinical Nutrition and Metabolic Care surface expression and glucose metabolism mediated by AKT/PKCf/
21 (2):78–82. doi: 10.1097/MCO.0000000000000443. k phosphorylation in 3T3L1 adipocytes. Molecular and Cellular
Li, H.-Y., S.-P. Chang, C.-C. Yuan, H.-T. Chao, H.-T. Ng, and Y.-J. Biochemistry 381 (1-2):291–9. doi: 10.1007/s11010-013-1714-7.
Sung. 2003. Induction of p38 mitogen-activated protein kinase- Mariggio, S., C. Iurisci, J. Sebastia, J. Patton-Vogt, and D. Corda. 2006.
mediated apoptosis is involved in outgrowth of trophoblast cells on Molecular characterization of a glycerophosphoinositol transporter
endometrial epithelial cells in a model of human trophoblast-endo- in mammalian cells. FEBS Letters 580 (30):6789–96. doi: 10.1016/j.
metrial interactions. Biology of Reproduction 69 (5):1515–1524. doi: febslet.2006.11.039.
10.1095/biolreprod.103.015669. Marincola, F. C., A. Dessi, M. G. Pattumelli, S. Corbu, C. Ossicini, S.
Lin, X., L. Ma, C. Gopalan, and R. E. Ostlund. 2009. D-chiro-Inositol Ciccarelli, R. Agostino, M. Mussap, and V. Fanos. 2015. 1H NMR-
is absorbed but not synthesised in rodents. British Journal of based urine metabolic profile of IUGR, LGA, and AGA newborns in
Nutrition 102 (10):1426–1434. doi: 10.1017/S0007114509990456. the first week of life. Clinica Chimica Acta; International Journal of
Lin, X., L. Ma, R. L. Fitzgerald, and R. E. Ostlund. 2009. Human Clinical Chemistry 451:28–34. doi: 10.1016/j.cca.2015.08.008.
sodium/inositol cotransporter 2 (SMIT2) transports inositols but not Marion, E., P. J. Kaisaki, V. Pouillon, C. Gueydan, J. C. Levy, A.
glucose in L6 cells. Archives of Biochemistry and Biophysics 481 (2): Bodson, G. Krzentowski, J.-C. Daubresse, J. Mockel, J. Behrends,
197–201. doi: 10.1016/j.abb.2008.11.008. et al. 2002. The gene INPPL1, encoding the lipid phosphatase
Liscovitch, M., M. Czarny, G. Fiucci, and T. Xiaoqing. 2000. SHIP2, is a candidate for type 2 diabetes in rat and man. Diabetes
Phospholipase D: Molecular and cell biology of a novel gene family. 51 (7):2012–7., doi: 10.2337/diabetes.51.7.2012.
Biochemical Journal 345 (3):401–415. doi: 10.1042/bj3450401. Matarrelli, B., E. Vitacolonna, M. D’angelo, G. Pavone, P. A. Mattei,
Lizcano, J. M., and D. R. Alessi. 2002. The insulin signalling pathway. M. Liberati, and C. Celentano. 2013. Effect of dietary myo-inositol
Current Biology: CB 12 (7):R236–R238. doi: 10.1016/S0960-
supplementation in pregnancy on the incidence of maternal gesta-
9822(02)00777-7.
tional diabetes mellitus and fetal outcomes: A randomized controlled
Low, M. G. 2000. Structure and function of GPI-specific phospholi-
trial. The Journal of Maternal-Fetal & Neonatal Medicine26 (10):
pases. In PNH and the GPI-Linked Proteins, eds. N. S. Young and J.
967–972. doi: 10.3109/14767058.2013.766691.
Moss, 239–68. San Diego: Academic Press.
Maurizi, A. R., M. Menduni, R. Del Toro, S. Kyanvash, D. Maggi, C.
Loy, A., K. G. Lurie, A. Ghosh, J. M. Wilson, L. C. MacGregor, and
Guglielmi, A. L. Pantano, G. Defeudis, E. Fioriti, S. Manfrini, et al.
F. M. Matschinsky. 1990. Diabetes and the myo-inositol paradox.
2017. A pilot study of d-chiro-inositol plus folic acid in overweight
Diabetes 39 (10):1305–12. doi: 10.2337/diab.39.10.1305.
patients with type 1 diabetes. Acta Diabetologica 54 (4):361–5. doi:
Lubin, V., R. Shojai, P. Darmon, and E. Cosson. 2016. A pilot study of
gestational diabetes mellitus not controlled by diet alone: First-line 10.1007/s00592-016-0954-x.
Mayer, J., and D. Tomlinson. 1983. Prevention of defects of axonal
medical treatment with myoinositol may limit the need for insulin.
transport and nerve conduction velocity by oral administration of
Diabetes & Metabolism 42 (3):192–195. doi: 10.1016/j.diabet.2016.01.
005. myo-inositol or an aldose reductase inhibitor in streptozotocin-dia-
Lubrich, B., and D. van Calker. 1999. Inhibition of the high affinity betic rats. Diabetologia 25 (5):433–8. doi: 10.1007/BF00282524.
Myo-inositol transport system: A common mechanism of action of McLean, P., S. Kunjara, A. L. Greenbaum, K. Gumaa, J. L opez-Prados,
antibipolar drugs? Neuropsychopharmacology: Official Publication of M. Martin-Lomas, and T. W. Rademacher. 2008. Reciprocal control
the American College of Neuropsychopharmacology 21 (4):519–29. of pyruvate dehydrogenase kinase and phosphatase by inositol phos-
doi: 10.1016/S0893-133X(99)00037-8. phoglycans dynamic state set by “push-pull” system. The Journal of
Ma, K., L. A. M. Thomason, and J. McLaurin. 2012. scyllo-Inositol, Biological Chemistry 283 (48):33428–36. doi: 10.1074/jbc.
Preclinical, and Clinical Data for Alzheimer’s Disease. In: Michaelis M801781200.
EK, Michaelis ML, eds. Advances in Pharmacology. Vol 64: McPhee, F., C. Downes, and G. Lowe. 1991. Studies of inositol ana-
Academic Press; 177–212. logues as inhibitors of the phosphoinositide pathway, and incorpor-
Ma, K., L. A. Thomason, and J. McLaurin. 2012. Scyllo-inositol, preclin- ation of 2-deoxy-2-fluoro-myo-inositol to give analogues of
ical, and clinical data for Alzheimer’s disease. Advances in pharma- phosphatidylinositol intermediates. Biochemical Journal 277 (2):
cology. Vol 64, 177–212. Elsevier. 407–12. doi: 10.1042/bj2770407.
Ma, R. C., G. E. Tutino, K. A. Lillycrop, M. A. Hanson, and W. H. Mejia, C. A., P. R. Reynolds, and J. A. Arroyo. 2016. Placental expres-
Tam. 2015. Maternal diabetes, gestational diabetes and the role of sion of NFAT5, SLC5A3, and aldose reductase in gestational dia-
epigenetics in their long term effects on offspring. Progress in betes mellitus. The FASEB Journal 30 (851):855–851.
Biophysics and Molecular Biology 118 (1-2):55–68. doi: 10.1016/j. Metzger, B. E., L. P. Lowe, A. R. Dyer, E. R. Trimble, U. Chaovarindr,
pbiomolbio.2015.02.010. D. R. Coustan, D. R. Hadden, D. R. McCance, M. Hod, H. D.
Mackenzie, R. W., and B. T. Elliott. 2014. Akt/PKB activation and insu- McIntyre, et al. 2008. Hyperglycemia and adverse pregnancy out-
lin signaling: A novel insulin signaling pathway in the treatment of comes. The New England Journal of Medicine 358 (19):1991–2002.
type 2 diabetes. Diabetes, metabolic syndrome and obesity. Targets doi: 10.1056/NEJMoa0707943.
and Therapy 7:55. Michaelis, T., G. Helms, K. D. Merboldt, W. H€anicke, H. Bruhn, and J.
Magnusson-Olsson, A. L., B. Hamark, A. Ericsson, M. Wennergren, T. Frahm. 1993. Identification of scyllo-inositol in proton NMR spectra
Jansson, and T. L. Powell. 2006. Gestational and hormonal regula- of human brain in vivo. NMR in Biomedicine 6 (1):105–109. doi: 10.
tion of human placental lipoprotein lipase. Journal of Lipid Research 1002/nbm.1940060116.
47 (11):2551–2561. doi: 10.1194/jlr.M600098-JLR200. Michaelis, T., G. Helms, K.-D. Merboldt, W. H€anicke, H. Bruhn, and J.
Malik, A., F. Steinbeis, M. A. Carillo, P. H. Seeberger, B. Lepenies, and Frahm. 1993. Identification of scyllo-inositol in proton NMR spectra
D. Varon Silva. 2020. Immunological evaluation of synthetic glyco- of human brain in vivo. NMR in Biomedicine 6 (1):105–109. doi: 10.
sylphosphatidylinositol glycoconjugates as vaccine candidates against 1002/nbm.1940060116.
1668 O. C. WATKINS ET AL.

Middleton, A., and B. Setchell. 1972. The origin of inositol in the rete signal transduction system. The Journal of Clinical Endocrinology
testis fluid of the ram. Reproduction 30 (3):473–475. doi: 10.1530/jrf. and Metabolism 83 (6):2001–5. doi: 10.1210/jcem.83.6.4886.
0.0300473. Nestler, J. E., D. J. Jakubowicz, P. Reamer, R. D. Gunn, and G. Allan.
Miller, A. T., P. P. Chamberlain, and M. P. Cooke. 2008. Beyond IP3: 1999. Ovulatory and metabolic effects of D-chiro-inositol in the
Roles for higher order inositol phosphates in immune cell signaling. polycystic ovary syndrome. New England Journal of Medicine 340
Cell Cycle (Georgetown, Tex.) 7 (4):463–7. doi: 10.4161/cc.7.4.5518. (17):1314–20. doi: 10.1056/NEJM199904293401703.
Mi~
nambres, I., G. Cuixart, A. Gonçalves, and R. Corcoy. 2019. Effects Newsholme, P., E. Haber, S. Hirabara, E. Rebelato, J. Procopio, D.
of inositol on glucose homeostasis: Systematic review and meta-ana- Morgan, H. Oliveira-Emilio, A. R. Carpinelli, and R. Curi. 2007.
lysis of randomized controlled trials. Clinical Nutrition38 (3): Diabetes associated cell stress and dysfunction: Role of mitochon-
1146–52. doi: 10.1016/j.clnu.2018.06.957. drial and non-mitochondrial ROS production and activity. The
Mitanchez, D., C. Yzydorczyk, B. Siddeek, F. Boubred, M. Benahmed, Journal of Physiology 583 (Pt 1):9–24. doi: 10.1113/jphysiol.2007.
and U. Simeoni. 2015. The offspring of the diabetic mother - Short- 135871.
and long-term implications. Best Practice & Research Clinical Nielson, J. R., and J. P. Rutter. 2018. Lipid-mediated signals that regu-
Obstetrics & Gynaecology 29 (2):256–269. doi: 10.1016/j.bpobgyn. late mitochondrial biology. Journal of Biological Chemistry 001655
2014.08.004. Nissen, P. M., C. Nebel, N. Oksbjerg, and H. C. Bertram. 2011.
Moore, C. M., J. L. Breeze, S. A. Gruber, S. M. Babb, B. B. Frederick, Metabolomics reveals relationship between plasma inositols and
R. A. Villafuerte, A. L. Stoll, J. Hennen, D. A. Yurgelun-Todd, B. M. birth weight: Possible markers for fetal programming of type 2 dia-
Cohen, et al. 2000. Choline, myo-inositol and mood in bipolar dis- betes. Journal of Biomedicine and Biotechnology 2011:1–8. doi: 10.
order: A proton magnetic resonance spectroscopic imaging study of 1155/2011/378268.
the anterior cingulate cortex. Bipolar Disorders 2 (3 Pt 2):207–16. Noble, K., J. Zhang, and S. Wray. 2006. Lipid rafts, the sarcoplasmic
doi: 10.1034/j.1399-5618.2000.20302.x. reticulum and uterine calcium signalling: An integrated approach.
Moore, G. J., J. M. Bebchuk, J. K. Parrish, M. W. Faulk, C. L. Arfken, The Journal of Physiology 570 (Pt 1):29–35. doi: 10.1113/jphysiol.
J. Strahl-Bevacqua, and H. K. Manji. 1999. Temporal dissociation 2005.098475.
between lithium-induced changes in frontal lobe myo-inositol and Nordio, M., and E. Proietti. 2012. The combined therapy with myo-
clinical response in manic-depressive illness. The American Journal inositol and D-chiro-inositol reduces the risk of metabolic disease in
of Psychiatry 156 (12):1902–8. doi: 10.1176/ajp.156.12.1902. PCOS overweight patients compared to myo-inositol supplementa-
Mueckler, M., and B. Thorens. 2013. The SLC2 (GLUT) family of tion alone. European Review for Medical and Pharmacological
membrane transporters. Molecular Aspects of Medicine 34 (2-3): Sciences 16 (5):575–81.
121–138. doi: 10.1016/j.mam.2012.07.001. Noventa, M., A. Vitagliano, M. Quaranta, S. Borgato, B. Abdulrahim,
M€
uller, G., S. Wied, C. Piossek, A. Bauer, J. Bauer, and W. Frick. 1998. and S. Gizzo. 2016. Preventive and therapeutic role of dietary inosi-
Convergence and divergence of the signaling pathways for insulin tol supplementation in periconceptional period and during preg-
and phosphoinositolglycans. Molecular Medicine 4 (5):299–323. doi: nancy: A summary of evidences and future applications.
10.1007/BF03401738. Reproductive Sciences (Thousand Oaks, Calif.) 23 (3):278–88. doi: 10.
Murphy, A., A. Shamshirsaz, D. Markovic, R. Ostlund, and B. Koos. 1177/1933719115594018.
2016. Urinary excretion of myo-inositol and d-chiro-inositol in early Nozadze, M., E. Mikautadze, E. Lepsveridze, E. Mikeladze, N.
pregnancy is enhanced in gravidas with gestational diabetes mellitus. Kuchiashvili, T. Kiguradze, M. Kikvidze, and R. Solomonia. 2011.
Reproductive Sciences (Thousand Oaks, Calif.) 23 (3):365–371. doi: Anticonvulsant activities of myo-inositol and scyllo-inositol on pen-
10.1177/1933719115602767. tylenetetrazol induced seizures. Seizure20 (2):173–176. doi: 10.1016/j.
Muscogiuri, G., S. Palomba, A. S. Lagana, and F. Orio. 2016. Inositols seizure.2010.10.008.
in the treatment of insulin-mediated diseases. International Journal Oben, J. A., A. Mouralidarane, A.-M. Samuelsson, P. J. Matthews,
of Endocrinology 2016:1. doi: 10.1155/2016/6189820. M. L. Morgan, C. McKee, J. Soeda, D. S. Fernandez-Twinn, M. S.
Nagamatsu, S., and M. Ohara-Imaizumi. 2007. IP7 debut in insulin Martin-Gronert, S. E. Ozanne, et al. 2010. Maternal obesity during
release. Science318 (5854):1249–50. doi: 10.1126/science.1151361. pregnancy and lactation programs the development of offspring
Nakamura, J., M. A. Del Monte, D. Shewach, S. A. Lattimer, and D. A. non-alcoholic fatty liver disease in mice. Journal of Hepatology 52
Greene. 1992. Inhibition of phosphatidylinositol synthase by glucose (6):913–20. doi: 10.1016/j.jhep.2009.12.042.
in human retinal pigment epithelial cells. American Journal of Ortmeyer, H. K. 1996. Dietary myoinositol results in lower urine glu-
Physiology-Endocrinology and Metabolism 262 (4):E417–E426. doi: cose and in lower postprandial plasma glucose in obese insulin
10.1152/ajpendo.1992.262.4.E417. resistant rhesus monkeys. Obesity Research 4 (6):569–75. doi: 10.
Napso, T., H. E. J. Yong, J. Lopez-Tello, and A. N. Sferruzzi-Perri. 1002/j.1550-8528.1996.tb00271.x.
2018. The role of placental hormones in mediating maternal adapta- Ortmeyer, H. K., L. C. Huang, L. Zhang, B. C. Hansen, and J. Larner.
tions to support pregnancy and lactation. Frontiers in Physiology 9: 1993. Chiroinositol deficiency and insulin resistance. II. Acute
1091 doi: 10.3389/fphys.2018.01091. effects of D-chiroinositol administration in streptozotocin-diabetic
Nascimento, N. R., L. M. Lessa, M. R. Kerntopf, C. M. Sousa, R. S. rats, normal rats given a glucose load, and spontaneously insulin-
Alves, M. G. Queiroz, J. Price, D. B. Heimark, J. Larner, X. Du, resistant rhesus monkeys. Endocrinology 132 (2):646–51. doi: 10.
et al. 2006. Inositols prevent and reverse endothelial dysfunction in 1210/endo.132.2.8425484.
diabetic rat and rabbit vasculature metabolically and by scavenging Ortmeyer, H. K., N. Bodkin, K. Lilley, J. Larner, and B. C. Hansen.
superoxide. Proceedings of the National Academy of Sciences of the 1993. Chiroinositol deficiency and insulin resistance. I. Urinary
United States of America 103 (1):218–23. doi: 10.1073/pnas. excretion rate of chiroinositol is directly associated with insulin
0509779103. resistance in spontaneously diabetic rhesus monkeys. Endocrinology
Navarro-Gonzalez, J. F., A. Jarque, M. Muros, C. Mora, and J. Garcıa. 132 (2):640–5. doi: 10.1210/endo.132.2.8425483.
2009. Tumor necrosis factor-a as a therapeutic target for diabetic Ostlund, R. E., J. B. McGill, I. Herskowitz, D. M. Kipnis, J. V.
nephropathy. Cytokine & Growth Factor Reviews 20 (2):165–73. doi: Santiago, and W. R. Sherman. 1993. D-chiro-inositol metabolism in
10.1016/j.cytogfr.2009.02.005. diabetes mellitus. Proceedings of the National Academy of Sciences 90
Nestler, J. E., and V. Unfer. 2015. Reflections on inositol(s) for PCOS (21):9988–92. doi: 10.1073/pnas.90.21.9988.
therapy: Steps toward success. Gynecological Endocrinology31 (7): Owczarczyk-Saczonek, A., L. B. Lahuta, M. Ligor, W. Placek, R. J.
501–5. doi: 10.3109/09513590.2015.1054802. Gorecki, and B. Buszewski. 2018. The healing-promoting properties
Nestler, J. E., D. J. Jakubowicz, A. F. de Vargas, C. Brik, N. Quintero, of selected cyclitols—A review. Nutrients 10 (12):1891. doi: 10.3390/
and F. Medina. 1998. Insulin stimulates testosterone biosynthesis by nu10121891.
human thecal cells from women with polycystic ovary syndrome by Paine, M. A., M. Scioscia, P. J. Williams, K. Gumaa, C. H. Rodeck, and
activating its own receptor and using inositolglycan mediators as the T. W. Rademacher. 2010. Urinary inositol phosphoglycan P-type as
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1669

a marker for prediction of preeclampsia and novel implications for milk, formula for infants, and parenteral nutrition. The American
the pathophysiology of this disorder. Hypertension in Pregnancy 29 Journal of Clinical Nutrition 51 (4):589–593. doi: 10.1093/ajcn/51.4.
(4):375–384. doi: 10.3109/10641950903242667. 589.
Paine, M., C. Rodeck, P. Williams, and T. Rademacher. 2003. Possible Perez-Perez, A., P. Guadix, J. Maymo, J. L. Due~ nas, C. Varone, M.
involvement of inositol phosphoglycan-P in human parturition. Fernandez-Sanchez, and V. Sanchez-Margalet. 2016. Insulin and lep-
Journal of Reproductive Immunology 59 (2):267–75. doi: 10.1016/ tin signaling in placenta from gestational diabetic subjects. Hormone
S0165-0378(03)00053-6. and Metabolic Research ¼ Hormon- Und
Paine, M., M. Scioscia, K. A. Gumaa, C. Rodeck, and T. Rademacher. Stoffwechselforschung ¼ Hormones et Metabolisme 48 (1):62–69. doi:
2006. P-type inositol phosphoglycans in serum and amniotic fluid in 10.1055/s-0035-1559722.
active pre-eclampsia. Journal of Reproductive Immunology 69 (2): Peters, T., B. Chapman, M. Wolfe, and M. Soares. 2000. Placental lac-
165–79. doi: 10.1016/j.jri.2005.09.008. togen-I gene activation in differentiating trophoblast cells: Extrinsic
Pak, Y., and J. Larner. 1992. Identification and characterization of chi- and intrinsic regulation involving mitogen-activated protein kinase
roinositol-containing phospholipids from bovine liver. Biochemical signaling pathways. The Journal of Endocrinology 165 (2):443–456.
and Biophysical Research Communications 184 (2):1042–7. doi: 10. doi: 10.1677/joe.0.1650443.
1016/0006-291X(92)90696-I. Petit, A., G. Guillon, M. Tence, S. Jard, N. Gallo-Payet, D. Bellabarba,
Pak, Y., C. R. Paule, Y.-D. Bao, L. C. Huang, and J. Larner. 1993. J. G. Lehoux, and S. Belisle. 1989. Angiotensin II stimulates both
Insulin stimulates the biosynthesis of chiro-inositol-containing phos- inositol phosphate production and human placental lactogen release
pholipids in a rat fibroblast line expressing the human insulin recep- from human trophoblastic cells. The Journal of Clinical
tor. Proceedings of the National Academy of Sciences 90 (16): Endocrinology and Metabolism 69 (2):280–286. doi: 10.1210/jcem-69-
7759–7763. doi: 10.1073/pnas.90.16.7759. 2-280.
Pak, Y., L. C. Huang, K. J. Lilley, and J. Larner. 1992. In vivo conver- Pettitt, T. R., S. K. Dove, A. Lubben, S. D. Calaminus, and M. J.
sion of [3H]myoinositol to [3H]chiroinositol in rat tissues. Journal Wakelam. 2006. Analysis of intact phosphoinositides in biological
of Biological Chemistry 267:16904–16910. samples. Journal of Lipid Research 47 (7):1588–96. doi: 10.1194/jlr.
Pak, Y., Y. Hong, S. Kim, T. Piccariello, R. V. Farese, and J. Larner. D600004-JLR200.
1998. Vivo chiro-Inositol Metabolism in the Rat: A Defect in Philipps, L., S. Santhakumaran, C. Gale, E. Prior, K. Logan, M. Hyde,
chiroInositol Synthesis from myo-Inositol and an Increased and N. Modi. 2011. The diabetic pregnancy and offspring BMI in
Incorporation of chiro-[3H] lnositol into Phospholipid in the Goto- childhood: A systematic review and meta-analysis. Diabetologia 54
Kakizaki (GK) Rat. Molecules & Cells, 8. Springer Science & (8):1957–1966. doi: 10.1007/s00125-011-2180-y.
Business Media BV. Pillai, R. A., Islam M. O., P. Selvam, N. Sharma, A. H. Chu, O. C.
Palmano, K., P. H. Whiting, and J. N. Hawthorne. 1977. Free and lipid
Watkins, K. M. Godfrey, R. M. Lewis, and S. Y. Chan 2020.
myo-inositol in tissues from rats with acute and less severe strepto-
Placental inositol reduced in gestational diabetes as glucose alters
zotocin-induced diabetes. The Biochemical Journal 167 (1):229–35.
inositol transporters and IMPA1 enzyme expression. JCEM. In press
doi: 10.1042/bj1670229.
Pintaudi, B., G. Di Vieste, and M. Bonomo. 2016. The effectiveness of
Papaleo, E., V. Unfer, J. Baillargeon, and T. Chiu. 2009. Contribution
myo-inositol and D-chiro inositol treatment in type 2 diabetes.
of myo-inositol to reproduction. European Journal of Obstetrics &
International Journal of Endocrinology 2016:1–5. doi: 10.1155/2016/
Gynecology and Reproductive Biology 147 (2):120–123. doi: 10.1016/j.
9132052.
ejogrb.2009.09.008.
Plows, J. F., F. Budin, R. A. Andersson, V. J. Mills, K. Mace, S. T.
Papaleo, E., V. Unfer, J.-P. Baillargeon, L. De Santis, F. Fusi, C.
Davidge, M. H. Vickers, P. N. Baker, I. Silva-Zolezzi, and J. L.
Brigante, G. Marelli, I. Cino, A. Redaelli, and A. Ferrari. 2007. Myo-
Stanley. 2017. The effects of myo-inositol and B and D vitamin sup-
inositol in patients with polycystic ovary syndrome: A novel method
plementation in the db/þ mouse model of gestational diabetes mel-
for ovulation induction. Gynecological Endocrinology 23 (12):700–3.
doi: 10.1080/09513590701672405. litus. Nutrients 9 (2):141. doi: 10.3390/nu9020141.
Parpal, S., J. Gustavsson, and P. Strålfors. 1995. Isolation of phosphoo- Plows, J., J. R. Nieves, F. Budin, K. Mace, C. Reynolds, M. Vickers, I.
ligosaccharide/phosphoinositol glycan from caveolae and cytosol of Silva-Zolezzi, P. Baker, and J. Stanley. 2020. The effects of myo-
insulin-stimulated cells. The Journal of Cell Biology 131 (1):125–135. inositol and probiotic supplementation in a high fat fed preclinical
doi: 10.1083/jcb.131.1.125. model of glucose intolerance in pregnancy. British Journal of
Parthasarathy, R., L. Parthasarathy, and R. Vadnal. 1997. Brain inositol Nutrition 123 (5):516–39. doi: 10.1017/S0007114519003039.
monophosphatase identified as a galactose 1-phosphatase. Brain Poston, L., R. Bell, H. Croker, A. C. Flynn, K. M. Godfrey, L. Goff, L.
Research 778 (1):99–106. doi: 10.1016/S0006-8993(97)01042-1. Hayes, N. Khazaezadeh, S. M. Nelson, E. Oteng-Ntim, et al. 2015.
Pascente, R., F. Frigerio, M. Rizzi, L. Porcu, M. Boido, J. Davids, M. Effect of a behavioural intervention in obese pregnant women (the
Zaben, D. Tolomeo, M. Filibian, W. P. Gray, et al. 2016. Cognitive UPBEAT study): A multicentre, randomised controlled trial. The
deficits and brain myo-Inositol are early biomarkers of epileptogene- Lancet. Diabetes & Endocrinology 3 (10):767–777. doi: 10.1016/
sis in a rat model of epilepsy. Neurobiology of Disease 93:146–155. S2213-8587(15)00227-2.
doi: 10.1016/j.nbd.2016.05.001. Prynne, C. J., A. McCarron, M. E. Wadsworth, and A. M. Stephen.
Patrussi, L., S. Mariggio, D. Corda, and C. Baldari. 2013. The glycero- 2010. Dietary fibre and phytate–a balancing act: Results from three
phosphoinositols: From lipid metabolites to modulators of T-cell time points in a British Birth Cohort. British Journal of Nutrition
signaling. Frontiers in Immunology 4 doi: 10.3389/fimmu.2013. 103 (2):274–80. doi: 10.1017/S0007114509991644.
00213. Pugliese, G., R. G. Tilton, A. Speedy, E. Santarelli, D. M. Eades, M. A.
Paul, C., and D. M. Brady. 2015. Inositol modulation of essential meta- Province, C. Kilo, W. R. Sherman, and J. R. Williamson. 1990.
bolic pathways of insulin resistance in metabolic syndrome, polycys- Modulation of hemodynamic and vascular filtration changes in dia-
tic ovarian syndrome, and type 2 diabetes. Enzyme (GS) 7:16. betic rats by dietary myo-inositol. Diabetes 39 (3):312–22. doi: 10.
Paulick, M. G., and C. R. Bertozzi. 2008. The 2337/diab.39.3.312.
Glycosylphosphatidylinositol Anchor: A Complex Membrane- Pundir, J., D. Psaroudakis, P. Savnur, P. Bhide, L. Sabatini, H. Teede,
Anchoring Structure for Proteins. Biochemistry 47 (27):6991–7000. A. Coomarasamy, and S. Thangaratinam. 2018. Inositol treatment of
doi: 10.1021/bi8006324. anovulation in women with polycystic ovary syndrome: A meta-ana-
Pedersen, J. 1952. Diabetes and pregnancy; blood sugar of newborn lysis of randomised trials. BJOG: An International Journal of
infants during fasting and glucose administration. Nordisk Medicin Obstetrics & Gynaecology 125 (3):299–308. doi: 10.1111/1471-0528.
47 (30):1049. 14754.
Pereira, G. R., L. Baker, J. Egler, L. Corcoran, and R. Chiavacci. 1990. Quirk, J. G., Jr., and J. E. Bleasdale. 1983. myo-Inositol homeostasis in
Serum myoinositol concentrations in premature infants fed human the human fetus. Obstetrics and Gynecology 62 (1):41–44.
1670 O. C. WATKINS ET AL.

Rajasekaran, S. S., J. Kim, G.-C. Gaboardi, J. Gromada, S. B. Shears, concentrations in women later developing gestational diabetes melli-
K. T. dos Santos, E. L. Nolasco, S. d S. Ferreira, C. Illies, M. K€
ohler, tus or pregnancy-induced hypertension. The Journal of Maternal-
et al. 2018. Inositol hexakisphosphate kinase 1 is a metabolic sensor Fetal & Neonatal Medicine 29 (14):2245–2247. doi: 10.3109/
in pancreatic b-cells. Cellular Signalling 46:120–8. doi: 10.1016/j.cell- 14767058.2015.1081886.
sig.2018.03.001. Santamaria, A., F. Corrado, M. L. Interdonato, G. Baviera, G.
Reeves, V. L., C. M. Thomas, and E. J. Smart. 2012. Lipid rafts, caveo- Carlomagno, P. Cavalli, V. Unfer, and R. D’Anna. 2014. Myo-inosi-
lae and GPI-linked proteins, 3–13. Caveolins and Caveolae: Springer. tol in Down syndrome amniotic fluid. A case-control study.
Regidor, P.-A., A. E. Schindler, B. Lesoine, and R. Druckman. 2018. Prenatal Diagnosis 34 (9):917–8. doi: 10.1002/pd.4398.
Management of women with PCOS using myo-inositol and folic Satake, W., Y. Nakabayashi, I. Mizuta, Y. Hirota, C. Ito, M. Kubo, T.
acid. New clinical data and review of the literature. Hormone Kawaguchi, T. Tsunoda, M. Watanabe, A. Takeda, et al. 2009.
Molecular Biology and Clinical Investigation 0 (0) doi: 10.1515/ Genome-wide association study identifies common variants at four
hmbci-2017-0067. loci as genetic risk factors for Parkinsons disease. Nature Genetics
Roach, J. C., K. Deutsch, S. Li, A. F. Siegel, L. M. Bekris, D. C. 41 (12):1303–1307. doi: 10.1038/ng.485.
Einhaus, C. M. Sheridan, G. Glusman, L. Hood, A. Lernmark, et al. Sayers, N., and A. C. Hanyaloglu. 2018. Intracellular follicle-stimulating
2006. Genetic mapping at 3-kilobase resolution reveals inositol hormone receptor trafficking and signaling. Frontiers in
1,4,5-triphosphate receptor 3 as a risk factor for type 1 diabetes in Endocrinology 9:653. doi: 10.3389/fendo.2018.00653.
Sweden. American Journal of Human Genetics 79 (4):614–27., doi: Schaefer, -Graf, U., Meitzner, K. Ortega,-Senovilla, H. Graf, K. Vetter,
10.1086/507876. K. Abou-Dakn, M. Herrera. and E. 2011. Differences in the implica-
Roll, P., A. Massacrier, S. Pereira, A. Robaglia-Schlupp, P. Cau, and P. tions of maternal lipids on fetal metabolism and growth between
Szepetowski. 2002. New human sodium/glucose cotransporter gene gestational diabetes mellitus and control pregnancies. Diabetic
(KST1): Identification, characterization, and mutation analysis in Medicine 28 (9):1053–1059. doi: 10.1111/j.1464-5491.2011.03346.x.
ICCA (infantile convulsions and choreoathetosis) and BFIC (benign Schaefer-Graf, U. M., K. Graf, I. Kulbacka, S. L. Kjos, J. Dudenhausen,
familial infantile convulsions) families. Gene 285 (1-2):141–148. doi: K. Vetter, and E. Herrera. 2008. Maternal lipids as strong determi-
10.1016/S0378-1119(02)00416-X. nants of fetal environment and growth in pregnancies with gesta-
Romero, G., and J. Larner. 1993. Insulin mediators and the mechanism tional diabetes mellitus. Diabetes Care 31 (9):1858–1863. doi: 10.
of insulin action. Advances in pharmacology. Vol 24, 21–50. Elsevier. 2337/dc08-0039.
Rubin, L. J., and C. C. Hale. 1993. Characterization of a Mg-dependent, Schlemmer, U., W. Frølich, R. M. Prieto, and F. Grases. 2009. Phytate
Na-inositol Co-transport process in cardiac sarcolemmal vesicles. in foods and significance for humans: Food sources, intake, process-
Journal of Molecular and Cellular Cardiology 25 (6):721–731. doi:
ing, bioavailability, protective role and analysis. Molecular Nutrition
10.1006/jmcc.1993.1084.
& Food Research 53 (S2):S330–S375. doi: 10.1002/mnfr.200900099.
Ruchat, S. M., M. F. Hivert, and L. Bouchard. 2013. Epigenetic pro-
Schneider, S. 2015. Inositol transport proteins. FEBS Letters 589 (10):
gramming of obesity and diabetes by in utero exposure to gesta-
1049–1058. doi: 10.1016/j.febslet.2015.03.012.
tional diabetes mellitus. Nutrition Reviews 71 (Suppl 1):S88–S94. doi:
Schrey, M. P., A. M. Read, and P. J. Steer. 1987. Stimulation of
10.1111/nure.12057.
phospholipid hydrolysis and arachidonic acid mobilization in
Sacks, D. A., D. R. Hadden, M. Maresh, C. Deerochanawong, A. R.
human uterine decidua cells by phorbol ester. The Biochemical
Dyer, B. E. Metzger, L. P. Lowe, D. R. Coustan, M. Hod, J. J. Oats,
Journal 246 (3):705–713. doi: 10.1042/bj2460705.
et al. 2012. Frequency of gestational diabetes mellitus at collaborat-
Schrey, M. P., J. R. Holt, P. A. Cornford, H. Monaghan, and F. Al-
ing centers based on IADPSG consensus panel-recommended crite-
Ubaidi. 1992. Human decidua is a target tissue for bradykinin and
ria: The Hyperglycemia and Adverse Pregnancy Outcome (HAPO)
kallikrein: Phosphoinositide hydrolysis accompanies arachidonic
Study. Diabetes Care 35 (3):526–528. doi: 10.2337/dc11-1641.
Salman, M., J. T. Lonsdale, G. S. Besra, and P. J. Brennan. 1999. acid release in uterine decidua cells in vitro. The Journal of clinical
Phosphatidylinositol synthesis in mycobacteria. Biochimica et Endocrinology and Metabolism 74 (2):426–435. doi: 10.1210/jcem.74.
Biophysica Acta 1436 (3):437–450. doi: 10.1016/S0005- 2.1309839.
2760(98)00151-9. Schwartz, R., P. A. Gruppuso, K. Petzold, D. Brambilla, V. Hiilesmaa,
Saltiel, A. R. 1990. Second messengers of insulin action. Diabetes Care and K. A. Teramo. 1994. Hyperinsulinemia and macrosomia in the
13 (3):244–56. doi: 10.2337/diacare.13.3.244. fetus of the diabetic mother. Diabetes Care 17 (7):640–648. doi: 10.
Sampson, M., M. Wolf, R. Thadhani, J. L. Ecker, E. W. Seely, C. Lam, 2337/diacare.17.7.640.
S. A. Karumanchi, V. P. Sukhatme, A. S. M. Shakir, A. Daftary, Scioscia, M. 2017. D-chiro inositol phosphoglycans in preeclampsia:
et al. 2004. Preeclampsia and future cardiovascular disease: Potential Where are we, where are we going? Journal of Reproductive
role of altered angiogenesis and insulin resistance. The Journal of Immunology 124:1–7. doi: 10.1016/j.jri.2017.09.010.
Clinical Endocrinology & Metabolism 89 (12):6239–6243. doi: 10. Scioscia, M., A. Pesci, G. Zamboni, B. Huppertz, and L. Resta. 2013.
1210/jc.2004-0548. D-chiro-inositol phosphoglycan expression in human placenta at
Santamaria, A., A. Alibrandi, A. Di Benedetto, B. Pintaudi, F. Corrado, term in diabetes. Archives of Gynecology and Obstetrics 288 (2):
F. Facchinetti, and R. D’Anna. 2018. Clinical and metabolic out- 459–60. doi: 10.1007/s00404-013-2729-8.
comes in pregnant women at risk for gestational diabetes mellitus Scioscia, M., K. Gumaa, and T. W. Rademacher. 2009. The link
supplemented with myo-inositol: A secondary analysis from 3 RCTs. between insulin resistance and preeclampsia: New perspectives.
American Journal of Obstetrics and Gynecology 219 (3): Journal of Reproductive Immunology 82 (2):100–5. doi: 10.1016/j.jri.
300.e1–300.e6. doi: 10.1016/j.ajog.2018.05.018. 2009.04.009.
Santamaria, A., A. Di Benedetto, E. Petrella, B. Pintaudi, F. Corrado, R. Scioscia, M., K. Gumaa, L. E. Selvaggi, C. H. Rodeck, and T. W.
D’Anna, I. Neri, and F. Facchinetti. 2016. Myo-inositol may prevent Rademacher. 2009. Increased inositol phosphoglycan P-type in the
gestational diabetes onset in overweight women: A randomized, con- second trimester in pregnant women with type 2 and gestational
trolled trial. The Journal of Maternal-Fetal & Neonatal Medicine 29 diabetes mellitus. Journal of Perinatal Medicine 37 (5):469–471. doi:
(19):3234–3237. doi: 10.3109/14767058.2015.1121478. 10.1515/JPM.2009.082.
Santamaria, A., D. Giordano, F. Corrado, B. Pintaudi, M. Interdonato, Scioscia, M., K. Gumaa, M. Whitten, L. E. Selvaggi, C. H. Rodeck, and
G. D. Vieste, A. D. Benedetto, and R. D’Anna. 2012. One-year T. W. Rademacher. 2007. Inositol phosphoglycan P-type in healthy
effects of myo-inositol supplementation in postmenopausal women and preeclamptic pregnancies. Journal of Reproductive Immunology
with metabolic syndrome. Climacteric15 (5):490–495. doi: 10.3109/ 76 (1-2):85–90. doi: 10.1016/j.jri.2007.03.016.
13697137.2011.631063. Scioscia, M., K. Gumaa, S. Kunjara, M. A. Paine, L. E. Selvaggi, C. H.
Santamaria, A., F. Corrado, G. Baviera, G. Carlomagno, V. Unfer, and Rodeck, and T. W. Rademacher. 2006. Insulin resistance in human
R. D’anna. 2016. Second trimester amniotic fluid myo-inositol preeclamptic placenta is mediated by serine phosphorylation of
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1671

insulin receptor substrate-1 and-2. The Journal of Clinical Silverstone, P. H., B. M. McGrath, and H. Kim. 2005. Bipolar disorder
Endocrinology & Metabolism 91 (2):709–17. doi: 10.1210/jc.2005- and myo-inositol: A review of the magnetic resonance spectroscopy
1965. findings. Bipolar Disorders 7 (1):1–10. doi: 10.1111/j.1399-5618.2004.
Scioscia, M., M. A. Paine, K. Gumaa, C. H. Rodeck, and T. W. 00174.x.
Rademacher. 2008. Release of inositol phosphoglycan P-type by the Simons, K., and R. Ehehalt. 2002. Cholesterol, lipid rafts, and disease.
human placenta following insulin stimulus: A multiple comparison The Journal of Clinical Investigation 110 (5):597–603. doi: 10.1172/
between preeclampsia, intrauterine growth restriction, and gesta- JCI16390.
tional hypertension. The Journal of Maternal-Fetal & Neonatal Soeda, Y., H. Tsuneki, H. Muranaka, N. Mori, S. Hosoh, Y. Ichihara, S.
Medicine 21 (8):581–5. doi: 10.1080/14767050802199934. Kagawa, X. Wang, N. Toyooka, Y. Takamura, et al. 2010. The inosi-
Scioscia, M., P. J. Williams, K. Gumaa, N. Fratelli, C. Zorzi, and T. W. tol phosphatase SHIP2 negatively regulates insulin/IGF-I actions
Rademacher. 2011. Inositol phosphoglycans and preeclampsia: From implicated in neuroprotection and memory function in mouse brain.
bench to bedside. Journal of Reproductive Immunology 89 (2): Molecular Endocrinology (Baltimore, Md.) 24 (10):1965–77. doi: 10.
173–177. doi: 10.1016/j.jri.2011.03.001. 1210/me.2010-0163.
Scioscia, M., S. Kunjara, K. Gumaa, A. M. Gomez Galan, P. McLean, Solomonia, R., E. Mikautadze, M. Nozadze, N. Kuchiashvili, E.
C. H. Rodeck, and T. W. Rademacher. 2007. Altered urinary release Lepsveridze, and T. Kiguradze. 2010. Myo-inositol treatment pre-
of inositol phosphoglycan A-type in women with gestational dia- vents biochemical changes triggered by kainate-induced status epi-
betes mellitus. Gynecologic and Obstetric Investigation 64 (4):217–23. lepticus. Neuroscience Letters 468 (3):277–81. doi: 10.1016/j.neulet.
doi: 10.1159/000106494. 2009.11.012.
Scioscia, M., S. Kunjara, K. Gumaa, P. McLean, C. Rodeck, and T. Staat, B. C., H. L. Galan, J. E. Harwood, G. Lee, A. M. Marconi, C. L.
Rademacher. 2007. Urinary excretion of inositol phosphoglycan P- Paolini, A. Cheung, and F. C. Battaglia. 2012. Transplacental supply
type in gestational diabetes mellitus. Diabetic Medicine 24 (11): of mannose and inositol in uncomplicated pregnancies using stable
1300–1304. doi: 10.1111/j.1464-5491.2007.02267.x. isotopes. The Journal of Clinical Endocrinology and Metabolism 97
Seaquist, E. R., and R. Gruetter. 1998. Identification of a high concen- (7):2497–2502. doi: 10.1210/jc.2011-1800.
tration of scyllo-inositol in the brain of a healthy human subject Stamler, C. J., D. Breznan, T. A. Neville, F. J. Viau, E. Camlioglu, and
using 1H- and 13C-NMR . Magnetic Resonance in Medicine 39 (2): D. L. Sparks. 2000. Phosphatidylinositol promotes cholesterol trans-
313–316. doi: 10.1002/mrm.1910390220. port in vivo. Journal of Lipid Research 41 (8):1214–21.
Seely, E. W., and C. G. Solomon. 2003. Insulin resistance and its Stanirowski, P. J., D. Szukiewicz, M. Pyzlak, N. Abdalla, W. Sawicki,
potential role in pregnancy-induced hypertension. The Journal of and K. Cendrowski. 2017. Impact of pre-gestational and gestational
Clinical Endocrinology and Metabolism 88 (6):2393–2398. doi: 10. diabetes mellitus on the expression of glucose transporters GLUT-1,
GLUT-4 and GLUT-9 in human term placenta. Endocrine 55 (3):
1210/jc.2003-030241.
799–808. doi: 10.1007/s12020-016-1202-4.
Shaldubina, A., R. Buccafusca, R. Johanson, G. Agam, R. Belmaker, G.
Steegers-Theunissen, R., P. Groenen, and P. Beemster. 2002.
Berry, and Y. Bersudsky. 2007. Behavioural phenotyping of sodium-
Involvement of Inositol in Reproduction. Nutrition Reviews 60 (3):
myo-inositol cotransporter heterozygous knockout mice with
80–87. doi: 10.1301/00296640260042748.
reduced brain inositol. Genes, Brain and Behavior 6 (3):253–259.
Steer, T. E., and G. R. Gibson. 2002. The microbiology of phytic acid
doi: 10.1111/j.1601-183X.2006.00253.x.
metabolism by gut bacteria and relevance for bowel cancer.
Sharma, K., L. Wang, Y. Zhu, A. DeGuzman, G.-Y. Cao, R. B. Lynn,
International Journal of Food Science and Technology 37 (7):783–90.
and S. K. Joseph. 1999. Renal type I inositol 1, 4, 5-trisphosphate
doi: 10.1046/j.1365-2621.2002.00616.x.
receptor is reduced in streptozotocin-induced diabetic rats and mice.
Stewart, M. A., W. R. Sherman, and J. T. Harris. 1970. Effects of gal-
American Journal of Physiology-Renal Physiology 276 (1):F54–F61. actose on levels of free myo-inositol in rat tissues. Annals of the
doi: 10.1152/ajprenal.1999.276.1.F54. New York Academy of Sciences 165:609–614.
Shears, S. B. 2015. Inositol pyrophosphates: Why so many phosphates? Stokes, C. E., and J. N. Hawthorne. 1987. Reduced phosphoinositide
Advances in Biological Regulation 57:203–16. doi: 10.1016/j.jbior. concentrations in anterior temporal cortex of Alzheimer-diseased
2014.09.015. brains. Journal of Neurochemistry 48 (4):1018–1021. doi: 10.1111/j.
Sherman, W. R., M. A. Stewart, M. M. Kurien, and S. L. Goodwin. 1471-4159.1987.tb05619.x.
1968. The measurement of myo-inositol, myo-inosose-2 and scyllo- Stokes, C. E., K. R. W. Gillon, and J. N. Hawthorne. 1983. Free and
inositol in mammalian tissues. Biochimica et Biophysica Acta total lipid myo-inositol concentrations decrease with age in human
(Bba) - General Subjects 158 (2):197–205. doi: 10.1016/0304- brain. Biochimica et Biophysica Acta (BBA) - Lipids and Lipid
4165(68)90131-1. Metabolism 753 (1):136–138. doi: 10.1016/0005-2760(83)90108-X.
Sherman, W. R., M. A. Stewart, P. C. Simpson, and S. L. Goodwin. Strange, K., F. Emma, A. Paredes, and R. Morrison. 1994.
1968. The identification of myo-inosose-2 and scyllo-inositol in Osmoregulatory changes in Myo-inositol content and Naþ/Myo-
mammalian tissues. Biochemistry 7 (2):819–824. doi: 10.1021/ inositol contransport in rat cortical astrocytes. Glia 12 (1):35–43.
bi00842a040. doi: 10.1002/glia.440120105.
Sherman, W. R., S. L. Goodwin, and K. D. Gunnell. 1971. neo-Inositol Strieleman, P. J., M. A. Connors, and B. E. Metzger. 1992.
in mammalian tissues. Identification, measurement, and enzymic Phosphoinositide metabolism in the developing conceptus. Effects of
synthesis from mannose 6-phosphate. Biochemistry 10 (19): hyperglycemia and scyllo-inositol in rat embryo culture. Diabetes 41
3491–3499. doi: 10.1021/bi00795a002. (8):989–997. doi: 10.2337/diab.41.8.989.
Shewan, A., D. J. Eastburn, and K. Mostov. 2011. Phosphoinositides in Strieleman, P., and B. E. Metzger. 1993. Glucose and scyllo-inositol
cell architecture. Cold Spring Harbor Perspectives in Biology 3 (8): impair phosphoinositide hydrolysis in the 10.5-day cultured rat con-
a004796. doi: 10.1101/cshperspect.a004796. ceptus: a role in dysmorphogenesis? Teratology 48 (3):267–278. doi:
Shimon, H., Y. Sobolev, M. Davidson, V. Haroutunian, R. H. 10.1002/tera.1420480310.
Belmaker, and G. Agam. 1998. Inositol levels are decreased in post- Strieleman, P., M. Connors, and B. E. Metzger. 1992. Phosphoinositide
mortem brain of schizophrenic patients. Biological Psychiatry 44 (6): metabolism in the developing conceptus: Effects of hyperglycemia
428–432. doi: 10.1016/S0006-3223(98)00071-7. and scyllo-inositol in rat embryo culture. Diabetes 41 (8):989–997.
Shin, J.-A., H.-M. Kwon, K.-H. Han, and H.-Y. Lee. 2012. TonEBP and doi: 10.2337/diab.41.8.989.
SMIT expression in human placenta. Anatomy & Cell Biology 45 Stull, A. J., J. P. Thyfault, M. D. Haub, R. E. Ostlund, Jr, and W. W.
(3):155–159. doi: 10.5115/acb.2012.45.3.155. Campbell. 2008. Relationships between urinary inositol excretions
Siddle, K. 2011. Signalling by insulin and IGF receptors: Supporting and whole-body glucose tolerance and skeletal muscle insulin recep-
acts and new players. Journal of Molecular Endocrinology 47 (1): tor phosphorylation. Metabolism 57 (11):1545–1551. doi: 10.1016/j.
R1–R10. doi: 10.1530/JME-11-0022. metabol.2008.06.009.
1672 O. C. WATKINS ET AL.

Sun, T-H, D. B. Heimark, T. Nguygen, J. L. Nadler, and J. Larner. tubular injury induced in obesity and diabetes. The Journal of
2002. Both myo-inositol to chiro-inositol epimerase activities and Biological Chemistry 291 (3):1348–67. doi: 10.1074/jbc.M115.698191.
chiro-inositol to myo-inositol ratios are decreased in tissues of GK Trama, J., Q. Lu, R. G. Hawley, and S. N. Ho. 2000. The NFAT-related
type 2 diabetic rats compared to Wistar controls. Biochemical and protein NFATL1 (TonEBP/NFAT5) is induced upon T cell activa-
Biophysical Research Communications 293 (3):1092–8. doi: 10.1016/ tion in a calcineurin-dependent manner. The Journal of Immunology
S0006-291X(02)00313-3. 165 (9):4884–4894. doi: 10.4049/jimmunol.165.9.4884.
Sussman, I., and F. M. Matschinsky. 1988. Diabetes affects sorbitol and Traynor-Kaplan, A., M. Kruse, E. J. Dickson, G. Dai, O. Vivas, H. Yu,
myo-inositol levels of neuroectodermal tissue during embryogenesis D. Whittington, and B. Hille. 2017. Fatty-acyl chain profiles of cellu-
in rat. Diabetes 37 (7):974–981. doi: 10.2337/diab.37.7.974. lar phosphoinositides. Biochimica et Biophysica Acta (Bba) -
Suzuki, S.,. C. Suzuki, Y. Hinokio, Y. Ishigaki, H. Katagiri, M. Kanzaki, Molecular and Cell Biology of Lipids 1862 (5):513–22. doi: 10.1016/j.
V. N. Azev, N. Chakraborty, and M. d’Alarcao. 2014. Insulin-mim- bbalip.2017.02.002.
icking bioactivities of acylated inositol glycans in several mouse Trocino, R. A., S. Akazawa, M. Ishibashi, K. Matsumoto, H. Matsuo,
models of diabetes with or without obesity. PloS One 9 (6):e100466. H. Yamamoto, S. Goto, Y. Urata, T. Kondo, and S. Nagataki. 1995.
doi: 10.1371/journal.pone.0100466. Significance of glutathione depletion and oxidative stress in early
Suzuki, S.,. H. Kawasaki, Y. Satoh, M. Ohtomo, M. Hirai, A. Hirai, S. embryogenesis in glucose-induced rat embryo culture. Diabetes 44
Hirai, M. Onoda, M. Matsumoto, Y. Hinokio, et al. 1994. Urinary (8):992–8. doi: 10.2337/diab.44.8.992.
chiro-inositol excretion is an index marker of insulin sensitivity in Troyer, D., D. Schwertz, J. Kreisberg, and M. A. Venkatachalam. 1986.
Japanese type II diabetes. Diabetes Care 17 (12):1465–8., doi: 10. Inositol phospholipid metabolism in the kidney. Annual Review of
2337/diacare.17.12.1465. Physiology 48:51–71. doi: 10.1146/annurev.ph.48.030186.000411.
Suzuki, S.,. Y. Taneda, S. Hirai, S. Abe, A. Sasaki, K-I Suzuki, and T. Tsai, Y.-H., X. Liu, and P. H. Seeberger. 2012. Chemical biology of glyco-
Toyota. 1991. Molecular mechanism of insulin resistance in spontan- sylphosphatidylinositol anchors. Angewandte Chemie (International ed.
eous diabetic GK (Goto-Kakizaki) rats. New Directions in Research In English) 51 (46):11438–56. doi: 10.1002/anie.201203912.
and Clinical Works for. Obesity and Diabetes Mellitus :197–203. Turner, D. I., N. Chakraborty, and M. d’Alarcao. 2005. A fluorescent
Sweeting, A. N., G. P. Ross, J. Hyett, L. Molyneaux, M. Constantino, inositol phosphate glycan stimulates lipogenesis in rat adipocytes by
A. J. Harding, and J. Wong. 2016. Gestational diabetes mellitus in extracellular activation alone. Bioorganic & Medicinal Chemistry
early pregnancy: Evidence for poor pregnancy outcomes despite Letters 15 (8):2023–5. doi: 10.1016/j.bmcl.2005.02.061.
treatment. Diabetes Care 39 (1):75–81. doi: 10.2337/dc15-0433. Uhl, O., H. Demmelmair, M. T. Segura, J. Florido, R. Rueda, C.
Tabrizi, R., V. Ostadmohammadi, K. B. Lankarani, P. Peymani, M. Campoy, and B. Koletzko. 2015. Effects of obesity and gestational
Akbari, F. Kolahdooz, and Z. Asemi. 2018. The effects of inositol diabetes mellitus on placental phospholipids. Diabetes Research and
Clinical Practice 109 (2):364–371. doi: 10.1016/j.diabres.2015.05.032.
supplementation on lipid profiles among patients with metabolic
Unfer, V., and G. Porcaro. 2014. Updates on the myo-inositol plus D-
diseases: A systematic review and meta-analysis of randomized con-
chiro-inositol combined therapy in polycystic ovary syndrome.
trolled trials. Lipids in Health and Disease 17 (1):123. doi: 10.1186/
Expert Review of Clinical Pharmacology 7 (5):623–31. doi: 10.1586/
s12944-018-0779-4.
17512433.2014.925795.
Taguchi, R., J. Yamazaki, Y. Tsutsui, and H. Ikezawa. 1997.
Unfer, V., F. Facchinetti, B. Orr u, B. Giordani, and J. Nestler. 2017.
Identification of chiro-inositol and its formation by isomerization of
Myo-inositol effects in women with PCOS: A meta-analysis of
myo-inositol during hydrolysis of glycosylphosphatidylinositol-anch-
randomized controlled trials. Endocrine Connections 6 (8):647–658.
ored proteins. Archives of Biochemistry and Biophysics 342 (1):
doi: 10.1530/EC-17-0243.
161–168. doi: 10.1006/abbi.1997.9991.
Unfer, V., G. Carlomagno, G. Dante, and F. Facchinetti. 2012. Effects
Tessier, D., Z. Ferraro, and A. Gruslin. 2013. Role of leptin in preg-
of myo-inositol in women with PCOS: A systematic review of
nancy: Consequences of maternal obesity. Placenta 34 (3):205–211.
randomized controlled trials. Gynecological Endocrinology: The
doi: 10.1016/j.placenta.2012.11.035. Official Journal of the International Society of Gynecological
Thomas, M. P., S. J. Mills, and B. V. L. Potter. 2016. The “Other” Endocrinology 28 (7):509–15. doi: 10.3109/09513590.2011.650660.
Inositols and Their Phosphates: Synthesis, Biology, and Medicine Valluru, R., and W. Van den Ende. 2011. Myo-inositol and beyond-
(with Recent Advances in myo-Inositol Chemistry). Angewandte emerging networks under stress. Plant Science: An International
Chemie (International ed. In English) 55 (5):1614–1650. doi: 10. Journal of Experimental Plant Biology 181 (4):387–400. doi: 10.1016/
1002/anie.201502227. j.plantsci.2011.07.009.
Thomas, T. P., E. L. Feldman, J. Nakamura, K. Kato, M. Lien, M. J. Van Sande, J., D. Dequanter, P. Lothaire, C. Massart, J. E. Dumont,
Stevens, and D. A. Greene. 1993. Ambient glucose and aldose reduc- and C. Erneux. 2006. Thyrotropin stimulates the generation of inosi-
tase-induced myo-inositol depletion modulate basal and carbachol- tol 1, 4, 5-trisphosphate in human thyroid cells. The Journal of
stimulated inositol phospholipid metabolism and diacylglycerol Clinical Endocrinology and Metabolism 91 (3):1099–107. doi: 10.
accumulation in human retinal pigment epithelial cells in culture. 1210/jc.2005-1324.
Proceedings of the National Academy of Sciences 90 (20):9712–6. doi: Varela-Nieto, I., Y. Le on, and H. N. Caro. 1996. Cell signalling by
10.1073/pnas.90.20.9712. inositol phosphoglycans from different species. Comparative
Thomas, T. P., F. Porcellati, K. Kato, M. J. Stevens, W. R. Sherman, Biochemistry and Physiology Part B: Biochemistry and Molecular
and D. A. Greene. 1994. Effects of glucose on sorbitol pathway acti- Biology 115 (2):223–41. doi: 10.1016/0305-0491(96)00087-9.
vation, cellular redox, and metabolism of myo-inositol, phosphoino- Vernon, D. M., and H. J. Bohnert. 1992. A novel methyl transferase
sitide, and diacylglycerol in cultured human retinal pigment induced by osmotic stress in the facultative halophyte
epithelial cells. The Journal of Clinical Investigation 93 (6):2718–24. Mesembryanthemum crystallinum. The EMBO Journal 11 (6):
doi: 10.1172/JCI117286. 2077–85. doi: 10.1002/j.1460-2075.1992.tb05266.x.
Thurston, J. H., W. R. Sherman, R. E. Hauhart, and R. F. Kloepper. Vernon, R. G. 2005. Lipid metabolism during lactation: A review of adipose
1989. myo-Inositol: A Newly Identified Nonnitrogenous tissue-liver interactions and the development of fatty liver. The Journal of
Osmoregulatory Molecule in Mammalian Brain. Pediatric Research Dairy Research 72 (4):460–9. doi: 10.1017/S0022029905001299.
26 (5):482–485. doi: 10.1203/00006450-198911000-00024. Villalba, M., K. L. Kelly, and J. M. Mato. 1988. Inhibition of cyclic AMP-
Toh, N., T. Inoue, H. Tanaka, and E. Kimoto. 1987. Polyol accumula- dependent protein kinase by the polar head group of an insulin-sensi-
tion in human placenta and umbilical cord. Biological Research in tive glycophospholipid. Biochimica et Biophysica Acta (Bba) - Molecular
Pregnancy and Perinatology 8 (1 1ST Half):13–15. Cell Research 968 (1):69–76. doi: 10.1016/0167-4889(88)90045-6.
Tominaga, T.,. R. K. Dutta, D. Joladarashi, T. Doi, J. K. Reddy, and Vinggaard, A. M., and H. S. Hansen. 1995. Characterization and partial
Y. S. Kanwar. 2016. Transcriptional and translational modulation of purification of phospholipase D from human placenta. Biochimica et
myo-inositol oxygenase (Miox) by fatty acids Implications in renal Biophysica Acta 1258 (2):169–176. doi: 10.1016/0005-2760(95)00121-R.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 1673

Vitagliano, A., G. Saccone, E. Cosmi, S. Visentin, F. Dessole, G. lithium, carbamazepine or valproic acid decreases inositol uptake at
Ambrosini, and V. Berghella. 2019. Inositol for the prevention of high inositol concentrations but increases it at low inositol concentra-
gestational diabetes: A systematic review and meta-analysis of tions. Brain Research 855 (1):158–61. doi: 10.1016/S0006-
randomized controlled trials. Archives of Gynecology and Obstetrics 8993(99)02371-9.
299 (1):55–68. doi: 10.1007/s00404-018-5005-0. Xie, P., L. Sun, P. J. Oates, S. K. Srivastava, and Y. S. Kanwar. 2010.
Vivien, D., E. Petitfrere, L. Martiny, H. Sartelet, P. Galera, B. Haye, Pathobiology of renal specific oxidoreductase/myo-inositol oxygenase
and J.-P. Pujol. 1993. IPG (inositolphosphate glycan) as a cellular in diabetic nephropathy: Its implications in tubulo-interstitial fibrosis.
signal for TGF-b1 modulation of chondrocyte cell cycle. Journal of American Journal of Physiology-Heart and Circulatory Physiology
Cellular Physiology 155 (3):437–44. doi: 10.1002/jcp.1041550302. Yamashita, T., M. Tamatani, M. Taniguchi, T. Takagi, T. Yoshimine,
Voglmayr, J., and I. White. 1971. Synthesis and metabolism of myoino- and M. Tohyama. 1998. Regulation of Naþ/myo-inositol cotrans-
sitol in testicular and ejaculated spermatozoa of the ram. Journal of porter gene expression in hyperglycemic rat hippocampus. Brain
Reproduction and Fertility 24 (1):29–37. doi: 10.1530/jrf.0.0240029. Research. Molecular Brain Research 57 (1):167–72. doi: 10.1016/
Walsh, S. W., and V. M. Parisi. 1986. The role of arachidonic acid metabo- S0169-328X(98)00078-3.
lites in preeclampsia. Paper presented at: Seminars in perinatology. Yamazaki, H., K. C. Zawalich, and W. S. Zawalich. 2010. Physiologic
Wang, X., S. P. Devaiah, W. Zhang, and R. Welti. 2006. Signaling func- implications of phosphoinositides and phospholipase C in the regu-
tions of phosphatidic acid. Progress in Lipid Research 45 (3): lation of insulin secretion. Journal of Nutritional Science and
250–278. doi: 10.1016/j.plipres.2006.01.005. Vitaminology 56 (1):1–8. doi: 10.3177/jnsv.56.1.
Watkins, O. C., M. O. Islam, P. Selvam, R. A. Pillai, A. Cazenave- Yorek, M. A., J. A. Dunlap, and W. L. Lowe. 1998. Opposing effects of
Gassiot, A. K. Bendt, N. Karnani, K. M. Godfrey, R. M. Lewis, tumour necrosis factor a and hyperosmolarity on Naþ/myo-inositol co-
M. R. Wenk, et al. 2019. Inositol alters 13C-labeled fatty acid metab- transporter mRNA levels and myo-inositol accumulation by 3T3-L1
olism in human placental explants. Journal of Endocrinology 243 (1): adipocytes. Biochemical Journal 336 (2):317–25. doi: 10.1042/bj3360317.
73–84., doi: 10.1530/JOE-19-0267. Yue, D. K., S. McLennan, E. Fisher, S. Heffernan, C. Capogreco, G. R.
Wells, I. C., and J. Hogan. 1968. Effects of dietary deficiencies of lipotropic Ross, and J. R. Turtle. 1989. Ascorbic acid metabolism and polyol path-
factors on plasma cholesterol esterification and tissue cholesterol in rats. way in diabetes. Diabetes 38 (2):257–61. doi: 10.2337/diab.38.2.257.
The Journal of Nutrition 95 (1):55–62. doi: 10.1093/jn/95.1.55. Zeitler, P., and S. Handwerger. 1985. Arachidonic acid stimulates phos-
Wells, W. W., T. A. Pittman, and H. J. Wells. 1965. Quantitative analysis phoinositide hydrolysis and human placental lactogen release in an
of myoinositol in rat tissue by gas-liquid chromatography. Analytical enriched fraction of placental cells. Molecular Pharmacology 28:549–554.
Biochemistry 10 (3):450–458. doi: 10.1016/0003-2697(65)90314-3. Zeitler, P., E. Markoff, and S. Handwerger. 1983. Characterization of
Wenk, M. R., L. Lucast, G. Di Paolo, A. J. Romanelli, S. F. Suchy, R. L. the synthesis and release of human placental lactogen and human
Nussbaum, G. W. Cline, G. I. Shulman, W. McMurray, and P. De chorionic gonadotropin by an enriched population of dispersed pla-
Camilli. 2003. Phosphoinositide profiling in complex lipid mixtures cental cells. The Journal of Clinical Endocrinology and Metabolism
using electrospray ionization mass spectrometry. Nature 57 (4):812–818. doi: 10.1210/jcem-57-4-812.
Biotechnology 21 (7):813–7. doi: 10.1038/nbt837. Zeitler, P., Y. Q. Wu, and S. Handwerger. 1991. Melittin stimulates
Wentzel, P., and U. J. Eriksson. 2020. Embryopathy and Diabetes. phosphoinositide hydrolysis and placental lactogen release:
Gestational Diabetes. Vol 28: Karger Publishers; 132–44. Arachidonic acid as a link between phospholipase A2 and phospho-
Wentzel, P., C. R. Wentzel, M. B. G€areskog, and U. J. Eriksson. 2001. lipase C signal-transduction pathways. Life Sciences 48 (21):
Induction of embryonic dysmorphogenesis by high glucose concen- 2089–2095. doi: 10.1016/0024-3205(91)90166-9.
tration, disturbed inositol metabolism, and inhibited protein kinase Zerbino, D. R., P. Achuthan, W. Akanni, M. R. Amode, D. Barrell, J.
C activity. Teratology 63 (5):193–201. doi: 10.1002/tera.1034. Bhai, K. Billis, C. Cummins, A. Gall, C. G. Gir on, et al. 2018.
Wentzel, P., N. Welsh, and U. J. Eriksson. 1999. Developmental dam- Ensembl 2018. Nucleic Acids Research 46 (D1):D754–D761. doi: 10.
age, increased lipid peroxidation, diminished cyclooxygenase-2 gene 1093/nar/gkx1098.
expression, and lowered prostaglandin E2 levels in rat embryos Zhan, M., I. M. Usman, L. Sun, and Y. S. Kanwar. 2015. Disruption of
exposed to a diabetic environment. Diabetes 48 (4):813–20. doi: 10. renal tubular mitochondrial quality control by Myo-inositol oxygen-
2337/diabetes.48.4.813. ase in diabetic kidney disease. Journal of the American Society of
Werner, E. F., P. Has, G. Tarabulsi, J. Lee, and A. Satin. 2016. Early Nephrology : JASN 26 (6):1304–21. doi: 10.1681/ASN.2014050457.
postpartum glucose testing in women with gestational diabetes mel- Zhang, H., and C. Zhang. 2010. Adipose “talks” to distant organs to
litus. Obstetrical & Gynecological Survey 71:697–699. doi: 10.1097/01. regulate insulin sensitivity and vascular function. Obesity (Silver
ogx.0000510809.66411.87. Spring. Md) 18:2071.
White, D. 1973. The phospholipid composition of mammalian tissues. Zhang, H., Y. Lv, Z. Li, L. Sun, and W. Guo. 2019. The efficacy of
Form and Function of Phospholipids 441–82. myo-inositol supplementation to prevent gestational diabetes onset:
Wilcox, R. A., W. U. Primrose, S. R. Nahorski, and R. A. J. Challiss. A meta-analysis of randomized controlled trials. The Journal of
1998. New developments in the molecular pharmacology of the Maternal-Fetal & Neonatal Medicine32 (13):2249–2255. doi: 10.
myo-inositol 1,4,5-trisphosphate receptor. Trends in Pharmacological 1080/14767058.2018.1428303.
Sciences 19 (11):467–75. doi: 10.1016/S0165-6147(98)01260-7. Zheng, X., Z. Liu, Y. Zhang, Y. Lin, J. Song, L. Zheng, and S. Lin.
Williams, P. J., K. Gumaa, M. Scioscia, C. W. Redman, and T. W. 2015. Relationship between myo-inositol supplementary and gesta-
Rademacher. 2007. Inositol phosphoglycan P-type in preeclampsia: a tional diabetes mellitus: A meta-analysis. Medicine 94 (42):e1604.
novel marker. Hypertension 49 (1):84–89. ? doi: 10.1161/01.HYP. doi: 10.1097/MD.0000000000001604.
0000251301.12357.ba. Zhu, X., and J. Eichberg. 1990. A myo-inositol pool utilized for phos-
Willmroth, F., T. Drieling, U. Lamla, M. Marcushen, H.-J. Wark, and phatidylinositol synthesis is depleted in sciatic nerve from rats with
D. Van Calker. 2007. Sodium-myo-inositol co-transporter (SMIT-1) streptozotocin-induced diabetes. Proceedings of the National
mRNA is increased in neutrophils of patients with bipolar I disorder Academy of Sciences 87 (24):9818–22. doi: 10.1073/pnas.87.24.9818.
and down-regulated under treatment with mood stabilizers. The Zhuo, Z.,. A. Wang, and H. Yu. 2014. Effect of metformin intervention
International Journal of Neuropsychopharmacology 10 (1):63–71. doi: during pregnancy on the gestational diabetes mellitus in women
10.1017/S1461145705006371. with polycystic ovary syndrome: a systematic review and meta-ana-
Witters, L., and B. Kemp. 1992. Insulin activation of acetyl-CoA carb- lysis. Journal of Diabetes Research 2014:381231. doi: 10.1155/2014/
oxylase accompanied by inhibition of the 5’-AMP-activated protein 381231.
kinase. The Journal of Biological Chemistry 267 (5):2864–7. Ziyadeh, F. N. 2004. Mediators of diabetic renal disease: The case for TGF-
Wolfson, M., Y. Bersudsky, E. Zinger, M. Simkin, R. Belmaker, and L. b as the major mediator. Journal of the American Society of Nephrology 15
Hertz. 2000. Chronic treatment of human astrocytoma cells with (90010):55S–S57. doi: 10.1097/01.ASN.0000093460.24823.5B.

You might also like