Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/7012492

Nutritional Influences on Implantation and Placental Development

Article  in  Nutrition Reviews · June 2006


DOI: 10.1301/nr.may.S12-S18 · Source: PubMed

CITATIONS READS
40 260

2 authors:

James C Cross Lindsay Anderson


The University of Calgary Government of British Columbia, Canada
182 PUBLICATIONS   12,903 CITATIONS    6 PUBLICATIONS   89 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Early pregnancy prediction of preterm birth <33 weeks View project

All content following this page was uploaded by James C Cross on 26 March 2018.

The user has requested enhancement of the downloaded file.


May 2006: (II)S12–S18

Nutritional Influences on Implantation and Placental


Development
James C. Cross, DVM, PhD, and Lindsay Mickelson, BSc

The placenta is critical for nourishing the fetus connect to the umbilical cord1,2; b) promoting blood flow
throughout pregnancy, and also produces hormones to the implantation site and intervillous space by produc-
that alter the metabolic functions of the mother. While ing angiogenic factors and promoting vasodilation3; 3)
the effects of nutrition on fetal development and long- producing metabolic hormones such as placental lacto-
term outcome have been very well documented, there gens and placental growth hormone,4,5 which alter insu-
are only a few reports based on studies in rat, sheep, lin production and promote insulin resistance in maternal
and guinea pigs on how specific nutrients or general tissues to increase glucose availability to the fetus, as
nutritional status affect the development of the blas-
well as leptin6 and ghrelin7; 4) accumulating glycogen in
tocyst, its implantation, and the subsequent placenta.
times of glucose surplus8 (Figure 1).
The data suggest that placental development is highly
adaptable and that many types of compensation are Each of these placental functions is highly regu-
possible for suboptimal nutrition. lated and undertaken by a specific compartment of the
placenta. The barrier covering the chorionic villi in the
Key words: amino acids, development, glucose, pla-
human and rodent placenta is composed of several cell
centa, trophoblast
© 2006 International Life Sciences Institute
layers, including a multinucleated syncytium of tro-
doi: 10.1301/nr.may.S12–S18 phoblast (syncytiotrophoblast) cells.2 Nutrient trans-
porters are highly expressed to facilitate and regulate
the transfer between the maternal and fetal circulatory
INTRODUCTION systems. A specialized subtype of trophoblast cell
invades outside of the confines of the villi (these cells
As mammals, the placenta is something that we are called extravillous cytotrophoblasts in humans and
can’t do without during our embryonic and fetal lives, trophoblast giant cells in rodents) to encounter uterine
but which we soon forget about after birth. Even for spiral arteries. They replace the endothelial cell lining
clinicians and researchers who are interested in under- of the arteries and therefore lead to the transition into
standing complications in fetal growth, the placenta is the trophoblast-lined blood vessels (hemochorial).3
often either not included in the analysis or is given The different placental hormones are produced by the
cursory examination—such as by simply weighing it. invasive trophoblast and the syncytiotrophoblast cells.
This treatment is unjustified, as the placenta is not simply Finally, glycogen accumulates in both villous and
a selective filter for nutrient transport to the growing extravillous trophoblast cells, but is most dramatic in
fetus. Rather, it plays several key roles in most mamma- the rodent placenta in cells designated as glycogen
lian species in regulating the nutritional status of both the trophoblast cells.8 They first start to appear in the
fetus and the mother by: a) forming a highly branched latter part of gestation within the middle layer of
villous structure that forms the surface area for nutrient placenta called the spongiotrophoblast (often also
and gas exchange and establishes its own circulation to called the junctional zone), which is analogous to the
cytotrophoblast columns in the human placenta. After
Dr. Cross and Ms. Mickelson are with the Genes appearing, they subsequently invade diffusely into the
and Development Research Group, University of Cal- interstitium of the decidual tissue of the uterus.
gary, Calgary, Alberta, Canada. The purpose of this review is to discuss the role of
Please address all correspondence to: Dr. James nutrition in regulating these developmental events. Al-
Cross, Genes and Development Research Group, Of-
though there is only limited direct experimental evi-
fice Room 2279 Health Sciences Centre, 3330 Hospi-
tal Drive NW, Calgary, Alberta Canada T2N 4N1; dence, it is clear that some specific nutrients and general
Phone: 403-220-6876; Fax: 403-270-0737; E-mail: nutritional status can play key roles in altering the de-
jcross@ucalgary.ca. velopmental trajectory of the placenta, effects that have

S12 Nutrition Reviews姞, Vol. 64, No. 5


Figure 1. Structures and essential functions of the human placenta.

direct consequences for the survival of the fetus and the elsewhere,2,8 but some of the key molecular pathways
well-being of the newborn. regulating placental development are summarized in Fig-
ure 2.
MOLECULAR MECHANISMS OF PLACENTAL There are several important themes that emerge
DEVELOPMENT from the work to date. First, the placenta normally
achieves the delicate balance of having the correct pro-
The cellular and molecular mechanisms underlying portions of the different cell types to achieve its func-
the development of the placenta are best understood in tions. Second, differentiation of the trophoblast compart-
the mouse as a result of studies of experimental embry- ments of the placenta is regulated by largely independent
ology, the ability to culture trophoblast stem cells, and molecular mechanisms (Figure 2). Third, primary defects
analysis of several mouse mutants that have defects in in one developmental process can lead to secondary
placental development usually resulting in embryonic changes in other parts. For example, as is observed in
mortality or intrauterine growth restriction. Whereas we retinoblastoma (Rb) mutant placentas, failure to properly
knew of only a few genes essential for placental devel- form villi can lead to secondary attempts to hypervascu-
opment a decade ago,9 we now know of approximately larize the villi that do form.10 This suggests that devel-
100. These subjects have been recently reviewed in detail opment is adaptable to environmental circumstances.

Figure 2. Developmental origins and molecular mechanisms underlying the differentiation of trophoblast cell types in the mouse
placenta.

Nutrition Reviews姞, Vol. 64, No. 5 S13


Indeed, the differentiation potential of trophoblast cells is EFFECT OF MATERNAL NUTRITIONAL
highly regulated by oxygen levels.11-13 Given this, it STATUS ON VILLOUS PLACENTAL
stands to reason that nutrients and nutritional status may DEVELOPMENT
also have similar effects.
While specific nutrients have not been evaluated for
effects on post-implantation placental development, the
EFFECT OF GLUCOSE, AMINO ACIDS, AND effect of feed restriction and overfeeding have been
FASTING ON BLASTOCYST DEVELOPMENT evaluated in rat, sheep, and guinea pig models. Curi-
ously, at a general level, while the responses in rats and
The progenitors of the placenta are established in the
sheep are similar, the guinea pig appears to show a
pre-implantation embryo. Many years of culturing mam-
different effect. In rats, protein restriction from the time
malian embryos have led to a significant understanding
of mating leads to intrauterine growth restriction (IUGR)
of their metabolism and of the optimal medium for
of approximately 10%, but the placentas show enhanced
embryos that allows them to reach the blastocyst stage
villous development.23 While the overall volume of the
and to successfully implant if transferred into a recipient
villous placenta (called the labyrinth in rodents) is not
mother.14 Blastocyst development and subsequent im- changed, the villous surface area is enhanced by about
plantation potential are reduced in diabetic mothers,15 15%, implying that branching had become more elabo-
and this effect can be mimicked by culturing embryos in rate. The underlying molecular mechanism is unclear.
high concentrations of D-glucose.16,17 Amino acid sup- Interestingly, though, the underlying vasculature does
plementation also affects development. Essential amino not undergo a proportional expansion. This implies that
acid supplementation during in vitro culture of pre- the trophoblast cells alter their development, perhaps in
implantation embryos enhances post-implantation fetal an attempt to compensate for the protein restriction.
development.14 In addition, a strong beneficial effect of IUGR may have occurred because the response was
nonessential amino acid supplementation is observed in inadequate or because, in the absence of a proportional
mouse embryos, leading to enhanced blastocyst forma- increase in vascularization, the uptake efficiency of the
tion18 and increased implantation rates.14,18 placenta may not have been sufficient.
The beneficial effect of the nonessential amino acids The effect of overall energy restriction on placental
can be observed in an in vitro model of implantation development and structure in rats has not been docu-
called blastocyst attachment and outgrowth. In the ab- mented, and this is significant since it is obviously
sence of amino acids, blastocysts do not attach but enter unclear if the ability of the placenta to compensate (or
a state of quiescence analogous to delayed implanta- what type of compensation occurs) is dependent on the
tion.19 With the addition of amino acids, the trophoblast specific type of nutrient deprivation. Maternal iron re-
cells change their adhesive characteristics and form out- striction before and during pregnancy also leads to an
growths. The effect is mediated by the mTOR signaling increase in villous surface area but, again, with a failure
system and can therefore be inhibited with rapamycin.19 in vascular adaptation.24 This model leads to anemia, and
Interestingly, blastocysts can be exposed to the attach- the response indicates that trophoblast-branching mor-
ment-promoting amino acid levels for as little as a phogenesis and differentiation can be regulated by tissue
critical 8-hour period. This narrow window implies that oxygenation. This is consistent with previous findings
amino acid sensing by blastocysts is a checkpoint mech- with knockout mice lacking hypoxia-inducible factor
anism for determining whether to continue to develop. If (HIF).11 Most other animal models of IUGR use feed
the blastocyst does not initiate attachment to the uterus, restriction rather than protein restriction.
the maternal-side window of implantation will close and In sheep, overall caloric intake has been tested as a
pregnancy will not be established. model and, interestingly, both feed restriction and over-
Another potential mechanism by which undernutri- feeding can lead to IUGR.25-28 The placenta in sheep is
tion may affect blastocyst development has recently been distributed over about 100 separate cotyledons, and nu-
suggested. Ghrelin is a hormone first described for its trient status can affect both the number of cotyledons that
ability to increase fat deposition by stimulating appetite form and their subsequent size and vascularity. Tropho-
and decreasing fat utilization.20 Its expression is stimu- blast proliferation25 and the expression of angiogenic
lated by fasting and insulin-induced hypoglycemia.20 factors27,28 are reduced by overfeeding during the first
The ghrelin receptor is also expressed by pre-implanta- and second trimesters, leading to smaller cotyledons that
tion embryos, and ghrelin treatment significantly reduces are relatively poorly vascularized. By focusing on shorter
the number of inner cell mass and trophectoderm cells in intervals, Wallace et al.26 have been able to show that
blastocysts,21 similar to the effect of a low-protein diet cotyledon number is most affected by overnourishment
during the period of blastocyst formation.22 during the first trimester, whereas final cotyledon size is

S14 Nutrition Reviews姞, Vol. 64, No. 5


most affected by nutritional status in the second and third needs to be expanded to leptin expression and particu-
trimesters. This coincides with the fact that cotyledons larly to the glucose transporters GLUT1 and GLUT3,
are formed during the first trimester, and implies that since both increases and decreases in their expression
once the number is established, the placenta has only a have been reported.37,38 However, in a food restriction
limited ability to compensate through increasing the size model of IUGR in rats, GLUT expression levels did not
of the remaining cotyledons. correlate with endogenous glucocorticoid levels, perhaps
In guinea pigs, a feed restriction model has been suggesting that glucocorticoids may not directly regulate
widely used as a model of IUGR. In this model, pregnant GLUT expression.43
dams are fed only 50% to 70% of the normal ad libidum
intake and birth weights are reduced by about 30%. By
term, the labyrinth layer of placenta is reduced in overall MODULATION OF PLACENTAL HORMONE
size,29,30 and the villous surface area is also dramatically PRODUCTION BY MATERNAL NUTRIENT
STATUS
reduced.30 The response of the placental structure is
significantly different than that in the rat models dis-
The placenta produces a number of metabolically
cussed above. It is not clear whether this reflects a
important hormones. The placenta in many mammalian
species difference, differences in response to protein
species has been reported to produce placental-specific
only versus total diet restriction, or is due to the fact that
members of the prolactin/growth hormone superfamily.
the guinea pig model involves a more significant chal-
lenge to the pregnancy. Insulin-like growth factor (IGF) In humans, both placental lactogen44 and placental
I and II expression, and the ratio of the IGFs to IGF- growth hormone45,46 are produced. In rodents, a placen-
binding proteins is reduced in the guinea pig model.31-34 tal growth hormone is not made but a huge cluster of
Moreover, IGF levels appear to be correlated with the over 25 prolactin-related genes that have been amplified
extent of villous branching in the labyrinth and are during evolution are expressed in the placenta, largely by
inversely correlated with the trophoblast barrier thick- trophoblast giant cells.47 The functions of most of the
ness.32,33 These observations are interesting, because prolactin-related genes are unknown. Four placental lac-
IGF-II mutant mice have defects in the extent of villous togen I (PL-I)-related proteins and a single PL-II all
branching in the labyrinth and the barrier thickness is work through the prolactin receptor,4 and as such can
increased.35,36 potentially mediate several important maternal adapta-
tions to pregnancy that are attributed to prolactin.48
These include mammary development and lactogenesis,
EFFECT OF GLUCOCORTICOIDS ON pancreatic islet hyperplasia and the associated increase in
PLACENTAL DEVELOPMENT AND FUNCTION
insulin production, as well as insulin resistance. There is
very little information about what regulates the synthesis
In rats, glucocorticoid treatment of pregnant dams
and release of the PLs. PL-I expression is suppressed by
during the second half of gestation results in IUGR. The
expression of several hormone and nutrient transporter progesterone.49 PL-II expression is reduced by IL-6,50
genes has been reported to be altered in this model.37-41 and has been found to be expressed in a circadian
Notably, though, placental weight is also dramatically rhythm.51 PL-II is suppressed by melatonin, which is
smaller in this model, and the decrease in placental size also expressed in the placenta.51
is associated with an increase in apoptotic cell death41 The placenta also produces leptin52 and ghrelin,20
that occurs in both the labyrinth and junctional zone, hormones that suppress and stimulate appetite, respec-
though the specific cell type(s) have not be defined.42 tively, and regulate other metabolic processes as
Therefore, the data on gene expression should be inter- well.6,20,53 While they no doubt contribute to metabolic
preted with caution, since it is not clear if the changes control during pregnancy, there are few insights into the
simply reflect a change in cell composition or a physio- specific function of the placentally derived hormones.
logical change in expression. Ain et al.41 have addressed Leptin is expressed by syncytiotrophoblast cells under
the point by conducting a microarray experiment focused the control of the Gcm1 transcription factor.6,39,54,55
on the family of prolactin-related hormone genes, and Leptin receptors are also expressed in the placenta,
then performing in situ hybridization to determine the suggesting an autocrine or negative feedback func-
relative number and spatial patterns of expressing cells. tion.39,40 Ghrelin expression in the placenta has only
It is evident from this work that at least some of the gene recently been described, but the site of the expression has
expression changes induced by glucocorticoids, includ- not been identified. Interestingly, while fasting leads to
ing the placental lactogens and IGF-II, are physiological increased levels of ghrelin in circulation during preg-
and due to cellular expression levels rather than to the nancy,56 placental expression is not altered by fasting in
pathological loss of cell types. This type of work clearly pregnant rats.7

Nutrition Reviews姞, Vol. 64, No. 5 S15


PLACENTAL DEPOSITS OF GLYCOGEN culature in the placenta. Mol Cell Endocrinol. 2002;
187:207–212.
In addition to transporting glucose from the maternal 4. Linzer DI, Fisher SJ. The placenta and the prolactin
family of hormones: regulation of the physiology of
to the fetal circulation, the placenta can also store it in the pregnancy. Mol Endocrinol. 1999;13:837– 840.
form of glycogen. Glycogen deposits are a common 5. Soares MJ, Muller H, Orwig KE, Peters TJ, Dai G.
pathological finding in different tissues, but placental The uteroplacental prolactin family and pregnancy.
accumulation occurs in all pregnancies and appears to be Biol Reprod. 1998;58:273–284.
highly regulated, which suggests that it may be physio- 6. Moschos S, Chan JL, Mantzoros CS. Leptin and
reproduction: a review. Fertil Steril. 2002;77:433-
logical. In mice, glycogen accumulation is limited to the
444.
latter half of gestation, and specifically occurs in a subset 7. Chanoine JP, Wong AC. Ghrelin gene expression is
of cells in the spongiotrophoblast layer after embryonic markedly higher in fetal pancreas compared with
day 12.5.57 Thereafter, the cells appear to massively fetal stomach: effect of maternal fasting. Endocri-
invade the wall of the uterus.57 The extent of glycogen nology. 2004;145:3813–3820.
8. Simmons DG, Cross JC. Determinants of tropho-
accumulation is increased in diabetic pregnancies,58,59
blast lineage and cell subtype specification in the
but is stimulated by insulin.60 By contrast, glycogen mouse placenta. Dev Biol. 2005;284:12–24.
content is reduced in IGF-II mutant placentas,61 suggest- 9. Cross JC, Werb Z, Fisher SJ. Implantation and the
ing that IGF-II may act in a paracrine manner within the placenta: key pieces of the development puzzle.
placenta to regulate glucose uptake and synthesis of Science. 1994;266:1508 –1518.
glycogen, perhaps through the insulin receptor. Gluca- 10. Wu L, de Bruin A, Saavedra HI, et al. Extra-embry-
onic function of Rb is essential for embryonic de-
gon has also been reported to diminish glycogen content velopment and viability. Nature. 2003;421:942–947.
in the placenta in one study,60 but in another study had 11. Adelman DM, Gertsenstein M, Nagy A, Simon MC,
no effect.62 While the ability of the placenta to store and Maltepe E. Placental cell fates are regulated in vivo
mobilize glycogen is interesting, it is unclear to what by HIF-mediated hypoxia responses. Genes Dev.
extent it contributes to the energy balance of the fetus 2000;14:3191–3203.
12. Caniggia I, Mostachfi H, Winter J, Gassmann M, Lye
and/or mother.
SJ, Kuliszewski M, Post M. Hypoxia-inducible fac-
tor-1 mediates the biological effects of oxygen on
CONCLUSION human trophoblast differentiation through TGF-
beta(3). J Clin Invest. 2000;105:577–587.
13. Genbacev O, Zhou Y, Ludlow JW, Fisher SJ. Reg-
The effects of specific nutrients and general nutri- ulation of human placental development by oxygen
tional status on placental development, structure, and tension. Science. 1997;277:1669 –1672.
function have been largely ignored by most researchers. 14. Lane M, Gardner DK. Increase in postimplantation
However, from the limited data that do exist, it is clear development of cultured mouse embryos by amino
acids and induction of fetal retardation and exen-
that placental development is highly regulated and also
cephaly by ammonium ions. J Reprod Fertil. 1994;
highly adaptable. In particular, villous morphogenesis 102:305–312.
can alter to change the maternal-fetal surface area and 15. Lea RG, McCracken JE, McIntyre SS, Smith W,
barrier thickness across which nutrients pass. In addition, Baird JD. Disturbed development of the preimplan-
the extent of vascularization within the villi is adaptable tation embryo in the insulin-dependent diabetic
and may be orchestrated by trophoblast cell expression BB/E rat. Diabetes. 1996;45:1463–1470.
16. Leunda-Casi A, De Hertogh R, Pampfer S. De-
of angiogenic factors. In addition to the development of creased expression of fibroblast growth factor-4
the basic structure, placental functions (endocrine, active and associated dysregulation of trophoblast differ-
nutrient transporters, and glycogen storage) are also entiation in mouse blastocysts exposed to high
highly adaptable and can change either in response to the D-glucose in vitro. Diabetologia. 2001;44:1318 –
maternal environment or to defects within the placenta 1325.
17. Leunda-Casi A, Genicot G, Donnay I, Pampfer S, De
itself.
Hertogh R. Increased cell death in mouse blasto-
cysts exposed to high D-glucose in vitro: implica-
REFERENCES tions of an oxidative stress and alterations in glu-
cose metabolism. Diabetologia. 2002;45:571–579.
1. Cross JC, Simmons DG, Watson ED. Chorioallan- 18. Gardner DK, Lane M. Amino acids and ammonium
toic morphogenesis and formation of the placental regulate mouse embryo development in culture.
villous tree. Ann N Y Acad Sci. 2003;995:84 –93. Biol Reprod. 1993;48:377–385.
2. Watson ED, Cross JC. Development of structures 19. Martin PM, Sutherland AE. Exogenous amino acids
and transport functions in the mouse placenta. regulate trophectoderm differentiation in the mouse
Physiology (Bethesda). 2005;20:180 –193. blastocyst through an mTOR-dependent pathway.
3. Cross JC, Hemberger M, Lu Y, Nozaki T, Whiteley K, Dev Biol. 2001;240:182–193.
Masutani M, Adamson SL. Trophoblast functions, 20. Tschop M, Smiley DL, Heiman ML. Ghrelin induces
angiogenesis and remodeling of the maternal vas- adiposity in rodents. Nature. 2000;407:908 –913.

S16 Nutrition Reviews姞, Vol. 64, No. 5


21. Kawamura K, Sato N, Fukuda J, et al. Ghrelin inhib- cental-specific IGF-II is a major modulator of pla-
its the development of mouse preimplantation em- cental and fetal growth. Nature. 2002;417:945–948.
bryos in vitro. Endocrinology. 2003;144:2623–2633. 37. Hahn T, Barth S, Graf R, et al. Placental glucose
22. Kwong WY, Wild AE, Roberts P, Willis AC, Fleming transporter expression is regulated by glucocorti-
TP. Maternal undernutrition during the preimplanta- coids. J Clin Endocrinol Metab. 1999;84:1445–
tion period of rat development causes blastocyst 1452.
abnormalities and programming of postnatal hyper- 38. Langdown ML, Sugden MC. Enhanced placental
tension. Development. 2000;127:4195– 4202. GLUT1 and GLUT3 expression in dexamethasone-
23. Doherty CB, Lewis RM, Sharkey A, Burton GJ. Pla- induced fetal growth retardation. Mol Cell Endocri-
cental composition and surface area but not vascu- nol. 2001;185:109 –117.
larization are altered by maternal protein restriction 39. Sugden MC, Langdown ML, Munns MJ, Holness
in the rat. Placenta. 2003;24:34 –38. MJ. Maternal glucocorticoid treatment modulates
24. Lewis RM, Doherty CB, James LA, Burton GJ, Hales placental leptin and leptin receptor expression and
CN. Effects of maternal iron restriction on placental materno-fetal leptin physiology during late preg-
vascularization in the rat. Placenta. 2001;22:534 – nancy, and elicits hypertension associated with hy-
539. perleptinaemia in the early-growth-retarded adult
25. Wallace JM, Aitken RP, Milne JS, Hay WW Jr. offspring. Eur J Endocrinol. 2001;145:529 –539.
Nutritionally mediated placental growth restriction 40. Smith JT, Waddell BJ. Leptin receptor expression in
in the growing adolescent: consequences for the the rat placenta: changes in ob-ra, ob-rb, and ob-re
fetus. Biol Reprod. 2004;71:1055–1062. with gestational age and suppression by glucocor-
26. Wallace JM, Bourke DA, Aitken RP, Cruickshank ticoids. Biol Reprod. 2002;67:1204 –1210.
MA. Switching maternal dietary intake at the end of 41. Ain R, Canham LN, Soares MJ. Dexamethasone-
the first trimester has profound effects on placental induced intrauterine growth restriction impacts the
development and fetal growth in adolescent ewes placental prolactin family, insulin-like growth fac-
carrying singleton fetuses. Biol Reprod. 1999;61: tor-II and the Akt signaling pathway. J Endocrinol.
101–110. 2005;185:253–263.
27. Reynolds LP, Borowicz PP, Vonnahme KA, et al. 42. Waddell BJ, Hisheh S, Dharmarajan AM, Burton PJ.
Placental angiogenesis in sheep models of compro- Apoptosis in rat placenta is zone-dependent and
stimulated by glucocorticoids. Biol Reprod. 2000;
mised pregnancy. J Physiol. 2005;565:43–58
63:1913–1917.
28. Redmer DA, Wallace JM, Reynolds LP. Effect of
43. Lesage J, Hahn D, Leonhardt M, Blondeau B, Bre-
nutrient intake during pregnancy on fetal and pla-
ant B, Dupouy JP. Maternal undernutrition during
cental growth and vascular development. Domest
late gestation-induced intrauterine growth restric-
Anim Endocrinol. 2004;27:199 –217.
tion in the rat is associated with impaired placental
29. Dwyer CM, Madgwick AJ, Crook AR, Stickland NC.
GLUT3 expression, but does not correlate with en-
The effect of maternal undernutrition on the growth
dogenous corticosterone levels. J Endocrinol. 2002;
and development of the guinea pig placenta. J Dev
174:37– 43.
Physiol. 1992;18:295–302.
44. Beckers JF, Zarrouk A, Batalha ES, Garbayo JM,
30. Roberts CT, Sohlstrom A, Kind KL, et al. Maternal Mester L, Szenci O. Endocrinology of pregnancy:
food restriction reduces the exchange surface area chorionic somatomammotropins and pregnancy-
and increases the barrier thickness of the placenta associated glycoproteins: review. Acta Vet Hung.
in the guinea-pig. Placenta. 2001;22:177–185. 1998;46:175–189.
31. Sohlstrom A, Katsman A, Kind KL, et al. Food re- 45. Su Y, Liebhaber SA, Cooke NE. The human growth
striction alters pregnancy-associated changes in hormone gene cluster locus control region supports
IGF and IGFBP in the guinea pig. Am J Physiol. position-independent pituitary- and placenta-spe-
1998;274:E410 –E416. cific expression in the transgenic mouse. J Biol
32. Roberts CT, Sohlstrom A, Kind KL, et al. Altered Chem. 2000;275:7902–7909.
placental structure induced by maternal food re- 46. Urbanek M, Russell JE, Cooke NE, Liebhaber SA.
striction in guinea pigs: a role for circulating IGF-II Functional characterization of the alternatively
and IGFBP-2 in the mother? Placenta. 2001; spliced, placental human growth hormone receptor.
22(suppl A):S77–S82. J Biol Chem. 1993;268:19025–19032.
33. Roberts CT, Kind KL, Earl RA, et al. Circulating 47. Wiemers DO, Shao LJ, Ain R, Dai G, Soares MJ. The
insulin-like growth factor (IGF)-I and IGF binding mouse prolactin gene family locus. Endocrinology.
proteins -1 and -3 and placental development in the 2003;144:313–325.
guinea-pig. Placenta. 2002;23:763–770. 48. Goffin V, Binart N, Touraine P, Kelly PA. Prolactin:
34. Olausson H, Sohlstrom A. Effects of food restriction the new biology of an old hormone. Annu Rev
and pregnancy on the expression of insulin-like Physiol. 2002;64:47– 67.
growth factors-I and -II in tIssues from guinea pigs. 49. Yamaguchi M, Endo H, Thordarson G, Ogren L,
J Endocrinol. 2003;179:437– 445. Talamantes F. Modulation of mouse placental lac-
35. Sibley CP, Coan PM, Ferguson-Smith AC, et al. togen-I secretion in vitro: effects of progesterone
Placental-specific insulin-like growth factor 2 (Igf2) and mouse placental lactogen-II. Endocrinology.
regulates the diffusional exchange characteristics of 1992;130:2897–2905.
the mouse placenta. Proc Natl Acad Sci U S A. 50. Yamaguchi M, Ogren L, Southard JN, Kurachi H,
2004;101:8204 – 8208. Miyake A, Talamantes F. Interleukin 6 inhibits
36. Constancia M, Hemberger M, Hughes J, et al. Pla- mouse placental lactogen II but not mouse placen-

Nutrition Reviews姞, Vol. 64, No. 5 S17


tal lactogen I secretion in vitro. Proc Natl Acad Sci U rats and in pregnancy. Obes Res. 2002;10:682–
S A. 1993;90:11905–11909. 687.
51. Lee CK, Moon DH, Shin CS, et al. Circadian expres- 57. Adamson SL, Lu Y, Whiteley KJ, et al. Interactions
sion of Mel1a and PL-II genes in placenta: effects of between trophoblast cells and the maternal and
melatonin on the PL-II gene expression in the rat fetal circulation in the mouse placenta. Dev Biol.
placenta. Mol Cell Endocrinol. 2003;200:57– 66. 2002;250:358 –373.
52. Pelleymounter MA, Cullen MJ, Baker MB, et al. 58. Padmanabhan R, Shafiullah M. Intrauterine growth
Effects of the obese gene product on body weight retardation in experimental diabetes: possible role
regulation in ob/ob mice. Science. 1995;269:540 – of the placenta. Arch Physiol Biochem. 2001;109:
543. 260 –271.
59. Barash V, Gutman A, Shafrir E. Fetal diabetes in rats
53. Prentice AM, Moore SE, Collinson AC, O’Connell
and its effect on placental glycogen. Diabetologia.
MA. Leptin and undernutrition. Nutr Rev. 2002;60:
1985;28:244 –249.
S56 –S87.
60. Goltzsch W, Bittner R, Bohme HJ, Hofmann E.
54. Yamada K, Ogawa H, Honda S, Harada N, Okazaki Effect of prenatal insulin and glucagon injection on
T. A GCM motif protein is involved in placenta- the glycogen content of rat placenta and fetal liver.
specific expression of human aromatase gene. Biomed Biochim Acta. 1987;46:619 – 622.
J Biol Chem. 1999;274:32279 –32286. 61. Lopez MF, Dikkes P, Zurakowski D, Villa-Komaroff
55. Lea RG, Howe D, Hannah LT, Bonneau O, Hunter L, L. Insulin-like growth factor II affects the appear-
Hoggard N. Placental leptin in normal, diabetic and ance and glycogen content of glycogen cells in the
fetal growth-retarded pregnancies. Mol Hum Re- murine placenta. Endocrinology. 1996;137:2100 –
prod. 2000;6:763–769. 2108.
56. Gualillo O, Caminos JE, Nogueiras R, et al. Effect of 62. Barash V, Shafrir E. Mobilization of placental glyco-
food restriction on ghrelin in normal-cycling female gen in diabetic rats. Placenta. 1990;11:515–521.

S18 Nutrition Reviews姞, Vol. 64, No. 5


View publication stats

You might also like