Pathophysiology of Type 2 Diabetes and The Impact of Altered Metabolic Interorgan Crosstalk

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 29

STATE-OF-THE-ART REVIEW

Pathophysiology of type 2 diabetes and the impact of


altered metabolic interorgan crosstalk
Jose Marcos Sanches1, Li Na Zhao1 , Albert Salehi1 , Claes B. Wollheim1,2 and
Philipp Kaldis1
€, Sweden
1 Department of Clinical Sciences, Lund University, Malmo
2 Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland

Keywords Diabetes is a complex and multifactorial disease that affects millions of


diabetes; free fatty acids; inflammation; people worldwide, reducing the quality of life significantly, and results in
interorgan crosstalk; liver; metabolites;
grave consequences for our health care system. In type 2 diabetes (T2D),
metabolomics; organokines; type 2 diabetes
the lack of b-cell compensatory mechanisms overcoming peripherally devel-
Correspondence oped insulin resistance is a paramount factor leading to disturbed blood
P. Kaldis, Department of Clinical Sciences, glucose levels and lipid metabolism. Impaired b-cell functions and insulin
Lund University, Box 50332, Malmo € SE-202 resistance have been studied extensively resulting in a good understanding
13, Sweden of these pathways but much less is known about interorgan crosstalk,
Tel: +4673 776 8101 which we define as signaling between tissues by secreted factors. Besides
E-mail: philipp.kaldis@med.lu.se
hormones and organokines, dysregulated blood glucose and long-lasting
hyperglycemia in T2D is associated with changes in metabolism with
(Received 21 May 2021, revised 14 October
2021, accepted 29 November 2021) metabolites from different tissues contributing to the development of this
disease. Recent data suggest that metabolites, such as lipids including free
doi:10.1111/febs.16306 fatty acids and amino acids, play important roles in the interorgan cross-
talk during the development of T2D. In general, metabolic remodeling
affects physiological homeostasis and impacts the development of T2D.
Hence, we highlight the importance of metabolic interorgan crosstalk in
this review to gain enhanced knowledge of the pathophysiology of T2D,
which may lead to new therapeutic approaches to treat this disease.

Abbreviations
AAT, alanine aminotransferase; ACC, acetyl-CoA carboxylase; ACL, ATP citrate lyase; AKT, also known as protein kinase B (PKB); ASC,
adaptor molecule apoptosis-associated speck-like protein containing a CARD; ATGL, adipose triglyceride lipase; CEPT1, choline/
ethanolaminephosphotransferase 1; ChREBP, carbohydrate response element binding protein; COX, cyclooxygenase; CPT1,
cholinephosphotransferase 1; CYP, cytochrome P450; DAG, diacylglycerol; DGAT1, diacylglycerol acyltransferase 1; DGAT2, Diacylglycerol
acyltransferase 2; DGK, Diacylglycerol kinase; EET, epoxyeicosatrienoic acids; EPT1, ethanolaminephosphotransferase 1; ER, endoplasmic
reticulum; F6P, fructose 6-phosphate; FAS, fatty acid synthase; FFAs, free fatty acids; G3P, glyceraldehyde 3-phosphate; G6P, glucose 6-
phosphate; GIP, gastric inhibitory peptide; Glcn6P, glucosamine 6-phosphate; GLP1, glucagon-like peptide-1; GLS, glutaminase; GS,
glutathione synthetase; GSDMD, gasdermin D; HETE, hydroxyeicosatetraenoic acids; HIS, hormone-sensitive lipase; IKKb, I-kappa-b-kinase;
IL, interleukin; IRS, insulin receptor substrate; JNK, c-Jun N-terminal kinase; LDH, lactate dehydrogenase; LOX, lipoxygenase; LT,
leukotriene; MCP-1, monocyte chemoattractant protein-1; MGAT1-MGAT3, monoacylglycerol acyltransferases; MOGAT2, monoacylglycerol
O-acyltransferase 2; NAD, nicotinamide adenine dinucleotide; NADPH, nicotinamide adenine dinucleotide phosphate; NF-kB, nuclear factor
kappa B; NLRP3, NOD-like receptor pyrin domain-containing protein 3, cryopyrin, or NALP3; PDK1, phosphoinositide-dependent kinase-1;
PG, prostaglandin; PI3K, phosphatidylinositol 3-kinase; PIP1, phosphatidylinositol 4-phosphate; PIP3, phosphatidylinositol (3,4,5)-
trisphosphate; PLA2, phospholipase A2; PLCs, phospholipases; RBP4, retinol binding protein 4; ROS, reactive oxygen species; SPT, serine
palmitoyl transferase; SREBP-1, sterol regulatory element binding transcription factor 1; TAG, triacylglyceride; TCA, tricarboxylic acid cycle;
TLR4, Toll-like receptor 4; TNF, tumor necrosis factor; cGCS, c-glutamylcysteine synthetase.

620 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and
distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

Introduction T2D, on the other hand, is a heterogeneous and multi-


factorial disease, which affects whole body physiology
Intermediary metabolism affects many functions in [12,21,22]. Among the regulators known to affect
cells, tissues, organs, and the entire organism. Here we metabolism by signaling to different tissues and there-
will focus on the metabolic interorgan crosstalk since fore important for T2D, are hormones (insulin, glucagon
it is important for the pathophysiology of many dis- [23,24], and glucagon-like peptide-1 [25]), organokines
eases [1–5]. In our view, metabolic interorgan crosstalk [adipokines/lipokines (e.g., leptin [26], adiponectin [27],
can be defined as the global effects of secreted factors resistin, Fgf21 [28], and adipsin [29])], and metabolites
from a target tissue that may trigger physiological (amino acids [23,30] like alanine [31,32], lipids, and
responses in one or several other tissues, affecting free fatty acids [32–34]).
homeostasis or the development of diseases. Neverthe- Although T2D most frequently develops in subjects
less, interorgan crosstalk is not only governed by with a family history of diabetes through the interplay
metabolites but also traditionally by hormones [6] and of risk genes expressed mainly in the b-cells and insu-
more recently by organokines (for a summary see lin resistance in target organs, there are many other
below) [7–10], which will not be discussed in detail forms with nongenetic etiology of hyperglycemia. We
here since this has been covered well in the literature. refer to an exhaustive review of the so-called atypical
Therefore, we envision at least three levels of regula- diabetes mellitus [35] and mention just a few of them
tion of interorgan crosstalk: hormones, organokines, here.
and metabolites. The regulation of interorgan crosstalk There are tight functional links between the exocrine
by metabolites in diabetes has not been discussed and endocrine pancreas, resulting in dysregulated pro-
extensively and this is the topic we will focus in this duction of pancreatic enzymes in T1D and T2D
review. [35,36]. Conversely, both acquired and inherited dis-
Since type 2 diabetes (T2D) is considered a complex eases of the exocrine pancreas often lead to diabetes by
and multifactorial metabolic disease that affects more affecting b-cell function. For example, iron overload
than 400 million people worldwide [11–15], we hypoth- that occurs in the hereditary conditions thalassemia
esize that one possible cause of T2D is alterations in and hemochromatosis is associated with progression of
the metabolic interorgan crosstalk. In this review, we b-cell dysfunction and T2D progression [35,37].
will focus especially on the pancreas, liver, adipose tis- While several enteroviruses have been connected to
sue and muscles as major metabolite producers and T1D, T2D has been associated with hepatitis C infec-
highlight selected metabolites, since these target tissues tion; in this case, the initiating event is insulin resis-
and organs play essential roles in glucose and lipid tance in the liver and other target tissues of insulin.
metabolism, as well as in insulin biology. The overrid- Antiviral therapy cures the defect in glucose homeosta-
ing control of metabolic homeostasis by the brain will sis [35]. Several endocrine disorders can lead to mild
not be addressed. Moreover, since T2D can be consid- as well as severe diabetes, among them acromegaly
ered as an inflammatory disease [13], we will summa- with increased plasma growth hormone concentrations
rize the association between metabolites that connect and Cushing’s disease characterized by overproduction
inflammation to T2D. of ACTH [35]. Like glucagon, growth hormones raise
Diabetes mellitus is a devastating chronic metabolic blood glucose and counteract the actions of insulin
disorder characterized by dysregulated insulin secretion and its overproduction causes insulin resistance both
with simultaneously reduced anabolic action on its tar- at the receptor and postreceptor level. Through
get tissues (insulin resistance), leading to altered increased lipolysis, growth hormones promote hepatic
metabolism, reflected among others by hyperglycemia glucose production [35]. Hyperplasia of the adrenal
and dyslipidemia [16,17]. Diabetes mellitus can be cells can also lead to the disease. Drugs like cortisol
divided into type 1 diabetes (T1D) and T2D. Based on and other glucocorticoids alter blood glucose control
clinical parameters, diabetes has been further subdi- by the induction of hepatic gluconeogenic enzymes
vided into five clusters, of which four correspond to and insulin resistance at the postreceptor signaling
T2D [18]. Diabetes reduces life quality through its level as well as b-cell dysfunction [38,39]. Hyperglyce-
many organ complications [19]. T1D, according to the mia is often observed with glucocorticoid therapy,
current hypotheses, is caused by an autoimmune which may result in persistent diabetes in patients with
destruction (through CD4+ and CD8+ T cells and mac- susceptibility to the disease.
rophages) of b-cells, leading to a complete loss of insu- Diabetes is a multifactorial disease where insulin
lin production, although in a small number of patients resistance and increased circulating FFAs collaborate
there is an idiopathic destruction of the b-cells [14,20]. with hyperglycemia, hyperinsulinemia, fat accumulation,

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 621
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

oxidative stress, and inflammation in the liver as well LECT2 expression is correlated with fatty liver
as other tissues [40–42]. As a consequence, steatosis is [10,55,56]. Some hepatokines seem to provide benefits
detected in 50% of diabetic patients. In addition, for the regulation of metabolism in T2D.
increased levels of FFAs suppress adipose tissue lipoly- The adipokine, adiponectin, contributes to insulin
sis by insulin, whereas elevated FFAs in the liver lead sensitivity in the liver through the activation of AMP-
to impaired hepatic fatty acid oxidation and insulin activated kinase (AMPK) and also suppresses gluco-
resistance [40]. T2D and NAFLD are likely bidirec- neogenesis (Fig. 1) [57]. Apart from the liver, adipo-
tionally related since NAFLD relates to the develop- nectin acts in muscles activating AMPK, increasing
ment of the insulin resistance [43]. the phosphorylation of ACC, glucose uptake, and
In this review, we will summarize relevant findings fatty acid oxidation [58,59]. In addition, this adipokine
concerning metabolic interorgan crosstalk connected contributes to the regulation of energy homeostasis
to the development and outcomes of T2D. [4,60]. Conversely, several adipokines have been associ-
ated with obesity and metabolic disorders, including
resistin [61,62], TNF-a [63–65], RBP4 [61,66], and
Organokines in type 2 diabetes
others, which impair the homeostasis and lead to com-
Hormones and organokines will not be discussed in plications in metabolic and inflammatory diseases
detail here but to understand the context of interorgan (Fig. 1) [4,61]. Related to T2D, leptin and resistin are
crosstalk, we provide a short summary of organokines. positively associated with insulin resistance, obesity,
Organokines are small secreted proteins produced by and induce a proinflammatory response [4,61,62,67].
target organ/tissues, such as the liver, adipose tissue, Skeletal muscles play an important role in T2D and
and muscles, and are classified as hepatokines, adipo- in the endocrine response through the production of
kines, and myokines, respectively. Organokines act in myokines which promote interorgan crosstalk and can
an endocrine, paracrine, and autocrine fashion, where also be considered as potential biomarkers for the dis-
their actions regulate several metabolic pathways, ease [3]. IL-6, usually associated with inflammatory
which affects the development of diseases. For exam- functions, is the best characterized myokine that plays
ple, hepatokines may act positively in homeostasis but different roles in certain tissues (Fig. 1). Treatment
dysregulation of hepatokines may lead to the develop- with IL-6 in a murine model suggested that insulin sig-
ment of the metabolic syndrome [44]. For example, naling through AKT was improved and reduced
fetuin inhibits the insulin receptor and consequently expression of gluconeogenic genes was observed as
triggers insulin resistance, which leads to inflammation well [68]. In adipose tissue, IL-6 was associated with
[7,44]. In accordance with this, fetuin-A as well as enhanced lipolysis and fat oxidation via AMPK activa-
fetuin-B are related to insulin sensitivity dysregulation tion [69]. IL-15 contributes to reduce visceral adipose
[45,46], impairment in glucose metabolism, and com- tissue and to enhance insulin action [70]. METRNL
plications in T2D [47,48]. In addition to fetuin-A and has been reported to contribute to energy expenditure
NEFA [44], other hepatokines have been linked to gly- through glucose and FFAs oxidation [71]. Thus, eluci-
cemic control, such as SMOC1 [49] and apolipoprotein dation of the roles of myokines, as well as other orga-
J, which improves glucose metabolism and insulin sen- nokines in the regulation of metabolic homeostasis,
sitivity in muscles [50]. holds promise for better understanding of metabolic
Another hepatokine that signals to the adipose tis- diseases such as diabetes.
sue is the selenoprotein P [51]. Selenoprotein P has
been suggested to cause insulin resistance in in vitro
The insulin signaling pathway in the
and in vivo experimental models [52,53], and when sele-
development of T2D
noprotein P levels are low, the levels of adiponectin
are high [51]. Evaluating the effects of FGF-21 in As mentioned earlier, T2D is a complex disease affect-
humans, high levels of FGF-21 were associated with ing the interplay between several metabolic pathways
obesity, NAFLD, and insulin resistance (Fig. 1) [44]. [72]. At physiological concentrations, glucose stimu-
In addition, high levels of sex hormone-binding globu- lates the b-cells to synthesize and release insulin,
lin have also been associated with T2D [54], as well as thereby promoting glucose uptake in muscle, adipose
hepassocin, LECT2, and retinol binding protein 4 tissue, liver, and brain. At the same time, insulin acts
(RBP4), which have been associated with impairment as an inhibitor for hepatic gluconeogenesis, as well as
in insulin signaling and glucose metabolism [10]. More- stimulating glycogen, protein synthesis, and lipogenesis
over, the secretion of LECT2 and RBP4 induces a while inhibiting lipolysis [73]. This illustrates that insu-
proinflammatory response in diabetic patients, and lin beyond its well-known glucose lowering action has

622 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

Fig. 1. Metabolic interorgan crosstalk in the development of T2D. Several metabolites from the adipose tissue (yellow), liver (orange),
pancreas (grey), and muscles (green) lead to signaling between tissues and this is connected to the development of type 2 diabetes.

also generalized hormonal effects, explaining why dia- pathways directly affects insulin’s functions. Lipogene-
betes involves several tissues. Insulin signal transduc- sis involves numerous mechanisms, including the
tion starts with the binding of the hormone to the induction of glucose uptake and glycolysis in the liver,
insulin receptor, which results in a series of phosphory- providing intermediates for the de novo synthesis of
lation events. Thus, intracellular protein substrates are fatty acids [75]. Insulin also induces the transcriptional
activated in order to trigger signaling cascades. Subse- activation of lipogenic genes, mediated by AKT2
quently, phosphatidylinositol 3-kinase (PI3K) induces which phosphorylates FOXO1 and SREBP1 [76]. We
the activation of protein kinase B (PKB), known as will touch upon these important pathways that are
AKT. With exception of hepatocytes, which mostly associated with insulin resistance and with the develop-
express the noninsulin-regulated glucose transporter 2 ment of T2D, especially in the context of the discussed
(GLUT2), GLUT4 is then translocated to the plasma metabolites.
membrane, with glycogen synthase and several
enzymes being activated through the action of insulin
Metabolic signature in type 2 diabetes
in target cells. The mitogen-activated protein kinase
pathway is also responsive to insulin signaling which Because of the impairment and imbalance in many
drives gene expression, protein translocation, and cell metabolic pathways in T2D, there is a need to under-
growth [74]. Overall, insulin is a primary driver for stand how amino acids, carbohydrates, nucleotides,
the regulation of carbohydrate, lipid, and protein carnitines, and lipid mediators are related to the devel-
metabolism, explaining why disbalance in metabolic opment and outcomes of T2D. Metabolomics

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 623
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

approaches are a great tool for the molecular charac- on providing a better understanding of how metabo-
terization of disease processes. However, a wide range lites can affect whole body physiology in the develop-
of metabolites were suggested as potential biomarkers ment of T2D. There is a wide range of lipids
leading to contradictory results, which questions the associated with diabetes including phospholipids, cera-
reproducibility of some of the earlier results. For mides, diacylglycerol, FFAs, and carnitine (Table 1).
example, increased levels of numerous amino acids Overall, after statistical meta-analysis, the most promi-
(AAs) have been correlated with IR and diabetes for nent lipids and acyl-carnitines in T2D are palmitic
decades [77–79]. In contrast, other studies have shown acid, myristic acid, stearic acid, linoleic acid, valeryl-
that many of these amino acids are decreased in diabe- carnitine (C5), and hexadecanoylcarnitine (C16),
tes [80–85]. Nonprospective and prospective studies respectively [86]. A recent lipidomics study on serum,
which analyzed plasma, serum, and/or urine in dia- muscle, and visceral adipose tissue of obese and T2D
betic subjects have shown that isoleucine, leucine, subjects emphasized changes in several lipid classes in
valine, tyrosine, phenylalanine, tryptophan [86–89], T2D, including increased deoxy-ceramides [104].
and the lysine metabolite 2-aminoadipic acid [90] were We have selected a few metabolites, which play
positively associated with T2D. Nevertheless, after a essential roles in the pathophysiology of T2D, that we
meta-analysis, the AAs that significantly associated will discuss in more detail (see below).
with T2D were isoleucine, leucine, tyrosine, phenylala-
nine, alanine, and glutamate [86]. Whether these corre-
Free fatty acids, diacylglycerol, and ceramides as
lations are cause or consequences of IR/T2D has not
key metabolites in the development of T2D
been clarified in detail. Thus, these contradictory data
require further analysis and additional technical The synthesis of fatty acids from nonlipid precursors
improvements of the metabolomic analyses including or ‘de novo lipogenesis’ is an enzyme coordinated path-
reproducible standards with which the samples can be way that occurs in the cytoplasm of hepatocytes and
spiked [89]. in adipocytes but FFAs can also be taken up from the
In general, the most prominent carbohydrates diet. Increased levels of glucose and ATP in cells pro-
altered in T2D are hexoses, glucose, mannose, and mote lipogenesis (Fig. 2) [105]. Following glycolysis,
fructose [86,91]. In addition, other carbohydrates have pyruvate enters the mitochondria and is converted into
been suggested as potential biomarkers in T2D acetyl-CoA through oxidative decarboxylation. Alter-
through their increased levels in biological samples. natively, pyruvate can also be converted into lactate in
These include lactate, 1,5-anhydroglucitol, galactose, the cytosol [106]. Subsequently, mitochondrial acetyl-
arabinose, inositol, trehalose, dihexose, phosphate, cit- CoA drives the tricarboxylic acid (TCA) cycle where it
rate, malate, 2-oxoglutaric acid, pyruvate, fumarate, is converted into citrate. High levels of insulin and glu-
cis-aconitate [86], and glucose-6-phosphate [89]. Since cose activate the transcription factors ChREBP and
the liver acts as a major metabolic organ in the human SREBP-1c, which leads to the transcription of lipo-
body, intermediate metabolites, such as glucose-6- genic genes, promoting de novo lipogenesis in the liver
phosphate, were suggested as a potential biomarker and the transport of citrate from mitochondria to the
produced by the liver in T2D (Table 1). In addition, cytosol. In the cytosol, citrate is converted into acetyl-
lactate was also detected at high concentrations in the CoA and oxaloacetate by the ATP-citrate lyase
liver. (ACLY) (Fig. 2). Acetyl-CoA carboxylase (ACC) car-
Besides carbohydrates, nucleotides play important boxylates acetyl-CoA to form malonyl-CoA. Finally,
metabolic roles, acting as cofactors [92] and they can malonyl-CoA (two carbons), acetyl-CoA (as a primer),
be linked to metabolic disorders [93]. Uridine, tri- as well as NADPH (reducing agent) are used to
methylamine, and glyoxylic acid, for example, are synthetize long-chain fatty acids by fatty acid synthase
upregulated in T2D [82,84,94–96]. Interestingly, not (FAS) catalysis (Fig. 2). The most prominent fatty
only in T2D but also in T1D, AMP, GMP, GTP, acid synthetized by FAS is palmitic acid (C16:0). Sub-
IMP, adenosine, guanosine, and inosine were found to sequently, palmitoleic acid is generated by stearoyl-
be increased [97,98]. CoA desaturase-1 and palmitoleic acid can be elon-
Lipids, as well as the lipid transporter carnitine (b- gated to oleoyl-CoA. The desaturation of stearic acid
hydroxy-c-trimethylammonium butyrate), which play leads to oleic acid (C18:0), which is the end product of
essential roles in fatty acid metabolism, have been de novo fatty acid synthesis [105,107].
associated with the development of diabetes [40,89,99– Increased levels of FFAs and deranged de novo lipo-
103]. Because of the relevance of lipid metabolism in genesis cause several metabolic disorders [40,105,108–
diabetes, especially in T2D, many studies have focused 110]. Although it is assumed that insulin resistance

624 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

Table 1. Metabolic profile associated with type 2 diabetes. Studies performed in (A) human samples and (B) animal samples. Amino acids,
carbohydrates, lipids, and carnitines detected in the blood (#), urine (&), liver (L), muscle (M), adipose tissue (AT), and islets (i) in T2D.

(A)

Amino acids Carbohydrates Lipids Carnitines

Cysteine # [81,84,110], & [94] Glucose # [86,110], & Phospholipids # [86,89], L/M/AT Propionylcarnitine # [89,301],
[86,91], L [89] [89] L [89]
Pantothenic acid # [81,84,110], & [94] Mannose # [86,110], & Ceramides i [101], L/M [133] Butyrylcarnitine # [89,301], L
[86,91] [89]
Creatine # [81,84,110], & [94] Fructose # [84,86], & Diacylglycerol M [302], L Valerylcarnitine # [86,89,301],
[86,91] [134,135] L [89]
Acetyl carnitine # [81,84,110], & [94] Lactate # [86], L [89] Lauric acid # [160] Octanoylcarnitine # [301], L
[89]
Butyryl carnitine # [81,84,110], & [94] 1 5-Anhydroglucitol # [86] Myristic acid # [160,303–305] Decanoylcarnitine # [89,301],
L [89]
Alanine # [80,81,83,85,306–308], & [82], L Galactose # [309] Pentadecanoic acid # [160,304] Tetradecenoyl # [301], L [89]
[89]
Glutamate # [80,81,83,85,306–308], & [82], Glucose-6-phosphate L 2-Pentadecenoic acid # [160] Hexadecanoylcarnitine #
L/M [89] [89] [86,89,301], L [89]
Glutamine # [80,81,83,85,306–308] & [82], Trehalose # [86] Palmitic acid # [84,160,303–305], Octadecanoylcarnitine # [301],
L [89] AT/L [89] L [89]
histidine # [80,81,83,85,306–308], & [82], dihexose # [110] Palmitoleic acid # [84,160,303– Arachidoylcarnitine # [301]
M [89] 305]
Isoleucine # [80,81,83,85,306–308], & [82], Malate # [86] Margaric acid # [160]
AT [89]
Leucine # [80,81,83,85,89,306–308] & [82] 2-Oxoglutaric acid # [86] 7-Methyl-6e-hexadecenoic acid #
[160]
Proline # [80,81,83,85,306–308], & [82], AT Arachidonic acid # [160], M [89]
[89]
Serine # [80,81,83,85,306–308], & [82] Stearic acid # [84,160,303]
Threonine # [80,81,83,85,306–308], & [82] Oleic acid # [84,160]
Lysine # [80,81,83,85,89,306–308], & [82], Linoleic acid # [160,303–305]
AT [89]
Tyrosine # [80,81,83,85,87–89,306–308], & Gamma linoleic acid #
[82,88], L/M [89] [160,304,305]
Valine # [80,81,83,85,306–308], & [82] 3e 9z 12z 15z-
Octadecatetraenoic acid # [160]
Phenylalanine # [80,81,83,85,87,88,306– Nonadecanoic acid #
308], & [82,88] [160,303,304]
Tryptophan # [80,81,83,85,87,88,306–308], Eicosanoid acid # [160,303,304]
& [82,88] AT [89]
Creatinine # [80,81,83,85,306–308], & [82], 11 14-Eicosadienoic acid # [160]
L [89]
2-Ketocaproic acid # [80,81,83,85,306– 5 8 11-Eicosatrienoic acid #
308], & [82] [160,304]
Pyroglutamic acid # [80,81,83,85,306–308], Eicosatetraenoic acid #
& [82] [160,304,305]
Choline # [80,81,83,85,306–308], & [82] eicosapentaenoic acid #
[160,303,305]
Ornithine # [80,81,83,85,306–308], & [82] 11z-Docosenoic acid # [160]
2-Aminoadipic acid # [90] Docosahexaenoic acid #
[160,304,305]

precedes the hypersecretion of insulin, considered as plasma lipids, in particular, palmitate, stimulate insu-
an adaptation to meet the increased insulin require- lin secretion even under fasting conditions [111]. Pal-
ments, there is evidence to the contrary. Increased mitate raises basal insulin secretion by promoting a

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 625
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

Table 1. (Continued).

(B)

Amino acids Lipids

Cysteine & [94] Ceramides L/M [133]


Pantothenic acid & [94] Diacylglycerol L [134,135]
Creatine & [94] Lauric acid # [160]
Acetyl carnitine & [94] Myristic acid # [160]
Butyryl carnitine & [94] Pentadecanoic acid # [160]
Alanine & [82] 2-Pentadecenoic acid # [160]
Glutamate & [82] Palmitic acid # [160]
Glutamine & [82] Palmitoleic acid # [160]
Histidine # [83] & [82] Margaric acid # [160]
Isoleucine # [83], & [82] 7-Methyl-6e-hexadecenoic acid # [160]
Leucine # [83] & [82] Arachidonic acid # [160]
Proline # [83], & [82] Stearic acid # [160]
Serine #, & [82] Oleic acid # [160]
Threonine # [83], & [82] Linoleic acid # [160]
Lysine # [83], & [82] Gamma linoleic acid # [160]
Tyrosine # [83], & [82] 3e 9z 12z 15z-Octadecatetraenoic acid # [160]
Valine # [83], & [82] Nonadecanoic acid # [160]
Phenylalanine # [83], & [82] Eicosanoid acid # [160]
Tryptophan # [83], & [82] 11 14-Eicosadienoic acid # [160]
Creatinine # [83], & [82] 5 8 11-Eicosatrienoic acid # [160]
2-Ketocaproic acid # [83], & [82] Eicosatetraenoic acid # [160]
Pyroglutamic acid # [83], & [82] Eicosapentaenoic acid # [160]
Choline # [83], & [82] 11z-Docosenoic acid # [160]
Ornithine # [83], & [82] Docosahexaenoic acid # [160]

mitochondrial proton leak in b-cells [112]. According (WAT) and in the liver [116–119], promoting the acti-
to this hypothesis, hyperinsulinism would in turn vation of NF-jB and consequently causing endoplas-
cause insulin resistance. Irrespective of the mechanism mic reticulum (ER) stress in metabolic organs and
of hyperinsulinemia, the consequence of insulin resis- immune cells, as well as inflammation; leading to insu-
tance is decreased glucose uptake in muscle and lin resistance [120,121]. Conversely, it has been sug-
increased production of liver glycogen, contributing to gested that insulin resistance is the initiating event, in
the development of T2D [2,12,40,41,86,113]. Fatty turn leading to inflammation [122]. Although the
acids such as palmitic acid, which is the major lipid molecular mechanisms related to fatty acids and insu-
produced through de novo lipogenesis, has been asso- lin resistance remain unclear, there is a tight associa-
ciated with the risk of T2D [86]. In addition to these tion of FFAs with the impairment of insulin secretion
data, palmitoleic acid, stearic acid, and oleic acid are and FFAs have long been considered a risk factor for
also considered markers associated with pathogenesis the development of T2D [123]. Taken together, fatty
of T2D through the de novo lipogenesis pathway acids act directly and indirectly in the development
[114]. Metabolic changes during the progression of and outcomes of T2D. De novo lipogenesis is a path-
T2D, such as increased free fatty acids, may activate way with a tight coupling to T2D, which is not only
other pathways that could contribute to the progres- due to the production of fatty acids but also due to
sion of T2D. For example, palmitic acid is a lipid fatty acids serving as substrates, playing a key role in
mediator that leads to a proinflammatory response synthesis of other lipids such as diacylglycerol (DAG)
through the toll-like receptor 4 (TLR4), causing toxic and ceramides [72].
effects in the islets, resulting in both impaired insulin Multiorgan involvement in T2D promotes increases
secretion and insulin resistance in the target organs in the production and release of lipid species
[115,116]. Moreover, saturated fatty acids, for exam- [100,124,125], carbohydrates [89,126], and amino acids
ple, palmitic acid, are responsible for the proinflam- [78,89,127]. Convincing evidence suggests that FFAs
matory response via TLR4 in white adipose tissue affect islets in many ways and exacerbates the

626 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

Fig. 2. Lipid metabolism and insulin resistance. High levels of glucose, insulin, and ATP promote the activation of the transcription factors
ChREBP and SREBP-1, which drive de novo lipogenesis. Mitochondrial citrate translocates to the cytosol, where it is converted into acetyl-
CoA and oxaloacetate by ACL. Then, malonyl-CoA is converted by FAS to form fatty acids. Thus, triglycerides can be esterified through
MOGAT2, DGAT2, and G3P and be stored in hepatocytes or adipose tissue. Also, TAG can be a major source for DAG synthesis. DAG can
be also synthesized from monoacylglycerol, phosphatidic, and phospholipids. DAG accumulation may inhibit the IRS adaptor protein, which
leads to the blockage of insulin signaling. Ceramides play an important role in insulin resistance through the inhibition of insulin signaling by
dephosphorylating AKT and blocking the action of AKT/PIP3-PDK1, which then affects the insulin receptor. JNK is activated by ER stress
caused by FFAs via TLR4 activation, leading to phosphorylation of IRS1 and IRS2 and this consequently results in insulin resistance. In
addition, ER stress can also activate JNK directly independently of TLR4. While de novo lipogenesis often happens in the liver, ceramides
can be synthesized in the liver, WAT, muscle, or islets. Nevertheless, these pathways are connected through interorgan crosstalk.

development of T2D. Since T2D has been considered liver, adipose tissue, islets, muscles, and probably also
as a multifactorial and complex disease [12,128], it is other organs. Fatty acids are ingested in the form of
reasonable to infer that T2D is a disease involving phospholipids and TAG and during ingestion the
numerous changes in metabolic pathways (Fig. 1). hydrolysis of these lipids are converted into mono-
This disease affects other tissues, such as the adipose and diacylglycerols, as well as FFAs. Short- and
tissue, muscle, and the liver [13,29,40,113,129], through medium-chain length FFAs may be observed in the
the production and distribution of metabolites via intestines and transported by serum albumin in the
blood circulation, leading to interorgan crosstalk. bloodstream and stored in the adipose tissue and liver
Free fatty acids can originate from the diet (15– [130]. Moreover, lipogenesis is another source of FFAs
20%), adipose tissue (~ 60%), and liver (15–25%). At where their synthesis is driven by the excess of carbo-
the same time, FFAs affect several organs including hydrates, triggering de novo lipogenesis, as explained

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 627
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

previously in this review. Lipogenesis occurs in several JNK/IKK-dependent serine phosphorylation of IRS1
cell types, but the liver, adipose tissue, and mammary leads to insulin resistance [145]. Besides that, abnor-
glands are the most active in this process [131,132]. mal levels of FFAs lead to cell stress due to lipotoxi-
In diabetic patients, several lipids species can accu- city inducing ER stress, ROS production, and
mulate in the liver and in the adipose tissue (Table 1). apoptosis [102]. This results in inflammation in adipo-
In the liver, phospholipids, ceramides, diacylglycerol, cytes [146] and causes chronic low-grade inflammation
and FFAs, such as palmitic acid, were increased in and insulin resistance, which contribute to the patho-
T2D [89,118,133–135]. Phospholipids and palmitic acid genesis of T2D [102].
were also found at high concentrations in the adipose The most widely held view is that insulin resistance
tissue [89], while phospholipids, ceramides, and arachi- leads to compensatory hypersecretion of the hormone
donic acid were elevated in T2D muscle samples from the b-cells, however, long term of exposure to
[89,133]. In addition, pancreatic islets displayed high FFAs at high levels results in b-cells dysfunction by
levels of ceramides in the diabetic condition [101]. lipotoxicity, which together with dysregulated blood
Interestingly, the total carnitine content in the human glucose control (glucotoxicity) eventually induce T2D
body is mostly stored in the heart and skeletal muscle [147]. Long-chain fatty acids, such as palmitate
[136]. Conversely, the levels of carnitines were most (C16:0) and stearate (18:0), and to a lesser extent myr-
prominent in the liver of T2D patients [89]. istate (C14:0) and laurate (C12:0), induce cytotoxicity
Elevated levels of FFAs released from the liver have in b-cells [148]. Chronic FFAs exposure of isolated
been linked to the development of T2D (Fig. 1) pancreatic islets at stimulatory glucose concentrations
[100,102,124,137]. T2D and NAFLD are likely bidirec- causes inhibition of glucose-stimulated insulin secre-
tionally related since NAFLD relates to the develop- tion (GSIS), altering gene expression, as well as pro-
ment of the insulin resistance [43] and up to 50% of moting apoptosis [149]. The b-cells are very sensitive
diabetic patients develop a fatty liver. The accumula- to ER stress and unfolded protein response with satu-
tion of FFAs in the liver is considerably increased in rated fatty acids inducing ER stress eventually leading
patients with T2D and NAFLD [138] often as a result to b-cells apoptosis [150]. Thus, chronic exposure of
of obesity. The liver is the metabolic center of the pancreatic islets to FFAs contributes to the develop-
human body, which plays essential roles in the detoxi- ment of T2D. Moreover, an in vitro study proposed
fication, digestion, and controlling many metabolic metabolic interorgan crosstalk between a fatty pan-
pathways and biological processes [139]. Because the creas and fatty liver, where the hepatokine fetuin-A
liver is functionally related to energy homeostasis and palmitate induced the expression of IL-6, CXCL8,
through lipid and glucose metabolism, and its links to and CCL2 in pancreatic adipocytes in a TLR4-
the gastrointestinal tract, disequilibrium of the liver dependent manner [2]. In the islets, the expression of
homeostasis and exposure to toxins can lead to liver IL-6 and CXCL8 was significantly higher after fetuin-
damage and diseases [113,140]. NAFLD is character- A and palmitate treatment, and when co-cultured with
ized by lipid deposition in the liver without alcohol macrophages, IL-1b was also released. Fetuin-A was
abuse or other etiologies related to viral or autoim- responsible for causing an impairment in glucose-
mune hepatitis [113]. The progression of NAFLD can induced insulin secretion (Fig. 1) [2]. Taken together,
lead to simple steatosis, nonalcoholic steatohepatitis this study suggests potential interorgan crosstalk
(NASH), liver fibrosis, cirrhosis, and eventually HCC between a fatty liver and pancreas leading to impair-
[42,141]. Conversely, due the lipid influx into the liver, ments in b-cells, with a fatty pancreas inducing a local
the liver contributes to increase in the levels of circu- inflammatory reaction [2].
lating lipid mediators that trigger metabolic crosstalk Regarding the adipose tissue, high levels of FFAs
and induce insulin resistance in other tissues [142]. promote its storage in adipocytes; however, increased
Stearic acid, for example, has been considered a pre- amount of fat in adipocytes may lead to cell and tissue
dictor of peripheral insulin resistance and glycemia in hypoxia and consequently to inflammation [132]. In
T2D [143]. Additional to insulin resistance that FFAs this context, due to the fat accumulation in the adi-
promote in several tissues, high levels of FFAs trigger pose tissue, TNF-a plays an important role through
several pathways that may collaborate to the out- the production and release by the adipose tissue during
comes of T2D. Increased levels of palmitate induce inflammation, increasing lipolysis and promoting insu-
the activation of the inflammasome and promote IL- lin resistance [151,152]. In addition, TNF-a acts in an
1b and IL-18 maturation and secretion, and conse- autocrine manner in the adipose tissue and reduces the
quently trigger a proinflammatory response [144]. The insulin sensitivity through the inhibition of IRS1 and
action of proinflammatory cytokines by inducing downregulating PPAR-c [153,154]. Due to increased

628 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

lipolysis promoted by adipocytes and high levels of promotes the expression of lipogenic genes, resulting
FFAs in the bloodstream, crosstalk between FFAs in in steatosis [160]. In this system, we consider FFA to
muscles, liver, and pancreas may lead to lipoapoptosis be the signaling molecule that drives crosstalk between
and organ failure [155]. Lipotoxicity in other tissues the liver, islets, WAT, and muscle [160]. Our study
and organs provokes several metabolic perturbations, indicates that defects in the liver may initiate the devel-
such as ROS production, leading to ER, lysosomal, opment of diabetes.
and mitochondrial stress, resulting in tissue damage Metabolic disbalance in the liver and muscles is
and loss of important functions of those target tissues often related to metabolic diseases and inflammation
and organs [156]. In fact, it is of interest in this con- [163,164], with DAG and ceramides playing important
text that T2D patients display impairment in mito- roles [165,166]. DAG accumulation in muscle and liver
chondrial activity in the deep subcutaneous adipose promotes the activation of PKC and blocks insulin
tissue, which leads to muscle insulin resistance and receptor activation, leading to insulin resistance [167–
increased substrate flux to the liver [157]. 169]. Abnormal lipid accumulation in the liver and
Skeletal muscle use glucose and FFAs as main meta- muscle correlates positively with insulin resistance
bolic energy source, and, since in T2D the flux of [170]. Conversely, insulin resistance seems to stimulate
TAG and FFAs is increased, the increased uptake of increased lipid deposition in these tissues [72]. Elevated
FFAs into myocytes is common, as well as their stor- concentrations of plasma FFAs result in fat accumula-
age as TAG [132,158]. The accumulation of FFAs in tion in the liver, WAT, and muscle by controlling the
myocytes, the synthesis of diacylglycerols and cera- amount of diacylglycerol (DAG), triglycerides (TAG),
mides, results in several consequences, including lipo- and long-chain acyl-CoA [171]. DAG is a precursor of
toxicity, inflammation, insulin resistance, and cell TAG, and by activation of protein kinase C (PKC)
damage [132]. In C2C12 myotubes, palmitate (C16:0) isoforms, modulates the phosphorylation of insulin
decreased the diameter of the myotubes and insulin pathway molecules [99]. The synthesis of DAG starts
signaling was affected [159], suggesting a direct effect with the catalysis of monoacylglycerol through monoa-
of FFAs on the physiology of muscle. cylglycerol acyltransferase, promoting the esterification
To illustrate how FFAs are involved in interorgan of monoacylglycerol (Fig. 2) [72,168]. On the other
crosstalk, we will now discuss one of our recent stud- hand, DAG can be synthesized by the hydrolysis of
ies. Using a mouse knockout model for cyclin- triacylglycerol and phospholipids by adipose triglycer-
dependent kinase 1 (Cdk1 cKO; Cdk1flox/flox Alb-Cre), ide lipase and phospholipases, respectively [72,168].
which is defective in hepatocyte proliferation and Another possibility to generate DAG is from phospha-
shows impairments in mitochondrial functions, our tidic acid where lipins (LPIN1 and LPIN3) mediate
group has uncovered impairments in fatty acid oxida- the dephosphorylation, consequently synthetizing
tion (FAO) and increased lipolysis, promoting high DAG (Fig. 2) [72]. In T2D, increased levels of DAG
levels of FFAs in the bloodstream, which caused promote the activation of PKCh and PKCe, phosphor-
enhanced insulin secretion by b-cells in young Cdk1 ylating the insulin receptor, and consequently leading
cKO mice [160]. Homeostatic levels of insulin in the to insulin resistance (Fig. 2) [167–169]. PKC isoforms
blood are essential for sugar metabolism, but at ele- have long been associated with insulin resistance
vated levels, classified as hyperinsulinemia or chronic [172,173] and their activation was also observed in dia-
hyperinsulinemia, insulin resistance develops [161,162]. betic rodent models [174,175]. Although increased
Hyperinsulinemia was observed in aged Cdk1 cKO levels of DAG were associated with insulin resistance,
mice associated with insulin resistance through reduced different DAG species may act in various ways regu-
INSRB expression as well as the reduction in insulin lating PKC activities. Nevertheless, the activation of
receptor autophosphorylation, high levels of glucose in PKCs through DAG accumulation in muscle and liver
the blood, decrease in glucose tolerance, and response modulates insulin sensitivity and seems to be an
to insulin [160]. Thus, in this mouse model, there is a important lipid mediator in the development of T2D
relationship between loss of hepatocyte proliferation [72,176,177].
and/or the Cdk1 gene and lipid metabolism, leading to Ceramides are sphingolipid-derived lipids that have
a diabetes-like phenotype. Mitochondrial dysfunction been linked to the pathogenesis of T1D and T2D
in hepatocytes results in excessive plasma FFAs that [101]. Although ceramides can play important roles in
are stored in the WAT leading to hyperinsulinemia. insulin resistance, not all ceramide species are associ-
Over time, Cdk1 cKO mice become insulin resistant ated with this condition and the development of T2D
and elevated blood glucose levels contribute to the [178]. Ceramides have been linked to muscle insulin
activation of the transcription factor LXR/RXR which resistance through their action downstream of IRS1 to

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 629
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

inhibit the phosphorylation and activation of PKB/ been described as potential biomarkers in T2D
Akt [179]. In vitro studies have shown that ceramides [86,96,199], and glutathione is an important scavenger
play important roles in insulin resistance in muscle of free radicals generated during oxidative stress [200],
cells [180,181], and there are data that link elevated we chose these pathways to highlight their important
levels of ceramides in muscles to T2D risk in roles in the development of T2D (Fig. 3).
men [182]. Thus, many more studies are needed to
investigate the relationship of lipid metabolism and
Lactate and alanine contribute to
insulin resistance in different tissues. Previous data
gluconeogenesis and the development of T2D
suggested that ceramides inhibit glucose uptake and
glycogenesis in muscle cells [183,184]. In an animal Lactate and alanine are a major source for gluconeo-
model of lipid infusion, ceramides were increased genesis [201]. The release of considerable amounts of
in muscles and the peripheral insulin sensitivity was lactate in plasma is the result of glycolysis in muscle
decreased [133]. and other tissues, and is inversely correlated with the
High levels of ceramides have been correlated with oxidative activity in mitochondria [202]. The mito-
inflammation driving insulin resistance, caused by the chondrial oxidative capacity is lower in T2D compared
reduction of adiponectin levels and the increased levels with healthy people [203] and the association between
of ceramide precursors [65,185,186]. Several studies high levels of lactate in the plasma with insulin resis-
have provided a better understanding on how ceramides tance has been observed [204] in T2D patients [205].
can affect insulin biology in cells and tissues [118,186– In addition, high concentrations of alanine were found
188]. Although the specific molecular mechanism of in diabetic patients including in the liver, blood, and
how ceramides trigger insulin resistance has not been urine [89,199,206]. There are two stereoisomers of lac-
elucidated, some studies have shown the association tate (2-hydroxypropanoate), L-lactate and D-lactate,
between insulin resistance and ceramides [101,186–194]. with L-lactate being prominent in physiological condi-
Ceramides promote the activation of phosphatase 2A tions through its production by glycolysis (Fig. 3)
[187,189], which dephosphorylates AKT [187,193], [207,208]. In the cytosol, pyruvate is reduced to L-
thereby blocking insulin-stimulated AKT attenuating lactate by lactate dehydrogenase (LDH) (Fig. 3) [209].
the translocation of the PIP3-PDK1 complex at the Evidence suggests that high concentrations of lactate
plasma membrane of target cells (Fig. 2) [191,194]. In lead to increased hepatic glucose production, which in
addition to these mechanisms, the accumulation of cer- association with the imbalance in lipid metabolism in
amides in membrane domains induces caspase activa- the liver triggers insulin resistance that may inevitably
tion leading to proinflammatory cytokine production contribute to the development of hyperglycemia and
and release, ROS generation, and cell death [195,196]. T2D [210]. Related to lactate metabolism, the high
Thus, ceramides are bioactive lipids that play essential concentrations of alanine in diabetic patients have
roles in cell signaling and also may affect the insulin been associated with the progression of T2D. The
pathway in target tissues. Intriguingly, the correlation reversible catalysis of alanine aminotransferase (AAT),
between ceramides and T2D needs further study since also known as glutamate pyruvate transaminase
ceramides may be induced by inflammation or could (GPT), promotes the transamination of L-alanine and
cause inflammation as a consequence of progression of a-ketoglutarate to convert them into pyruvate and glu-
T2D [65,101,185,197,198]. tamate. In addition, alanine can also stimulate gluco-
In general, lipids play essential roles in whole body neogenesis [211–213]. Being a glucogenic amino acid,
physiology as well as in the development of several alanine may act as an intermediate metabolite to form
diseases. In T2D, lipid mediators act through different glucose in the liver by the catalysis of alanine transam-
pathways and when the concentration of FFAs is high inase (ALT), with ALT using alanine and a-
and the synthesis of lipids is upregulated, metabolic ketoglutarate to form glutamate and pyruvate. Pyru-
changes occur that consequently affect physiology, vate then serves as a substrate for the gluconeogenesis
leading to cellular stress, ROS, and inflammation. pathway [211,212].
Thus, disorders of lipid metabolism play an important Since alanine induces gluconeogenesis in muscle
role in the development of T2D. In order to emphasize [211] as well as in liver [212,213], alanine may be a
the importance of a better understanding of the molec- potential biomarker for glucose synthesis in T2D.
ular mechanisms of metabolic pathways associated Overall, elevation of alanine seems to play a role in
with T2D, we will present next the pathways focusing the interorgan crosstalk in T2D. Alanine is produced
on lactate, alanine, glutamate, glutathione, and inflam- by ALT in a reaction including a-ketoglutarate, pyru-
mation. Since lactate, alanine, and glutamate have vate, and glutamate. From this reaction, it is clear that

630 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

Fig. 3. Lactate, alanine, glutamate, and glutathione metabolism. After glucose uptake in the liver and its oxidation through glycolysis,
pyruvate can be used to form lactate or alanine, both being major sources for gluconeogenesis. Glutamate is synthesized by different
pathways and serves as an intracellular as well as extracellular stimulus for insulin secretion, participating in incretin actions in b-cells.
Hyperglutamatemia is associated with the production of oxidative stress and leads to inflammation and insulin resistance. Altered
glutathione synthesis may play essential roles in the defense against proinflammatory cytokine expression and oxidative stress. On the
other hand, lipids, proinflammatory cytokines, and oxidative stress may block the synthesis of glutathione. The balance of these pathways
leading to oxidative stress and inflammation may determine the outcome of T2D.

alanine can be generated in all organs but larger 79 women using magnetic resonance imaging for fat
amounts are generated in organs with a high-energy content in the liver, and statistical correlations indi-
turnover. Liver and muscles may be considered major cated that liver and pancreas may communicate with
producers of several amino acids species where high each other [32]. Although not all individuals with T2D
concentrations are found in T2D (Table 1). Glutamate display hyperglucagonemia [214], which is related to
and tyrosine concentrations were reported to be ele- the secretory activity of a-cells in the pancreatic islets,
vated in the liver and muscles in patients under T2D increased levels of glucagon in the plasma have been
condition [89]. In the liver, alanine, glutamine, and cre- associated with the development of T2D [32]. A cohort
atinine were increased in comparison to other tissues study proposed that the glucagon–alanine index is
and organs, and histidine increased in the muscles [89]. associated with liver fat and this association was also
Adipose tissue also played an important role as major related to insulin resistance, indicating a risk factor for
producer of some amino acids, where the concentra- T2D [32]. Evaluating metabolism during liver regener-
tion of isoleucine, proline, lysine, and tryptophan was ation in a model with mitochondrial impairments in
more prominent [89]. hepatocytes, our group provided data that alanine pro-
An interaction between the fatty liver and pancreas duction from pyruvate is increased by the metabolic
was also shown through the liver-a-cell axis in a study flux through alanine transaminase [213]. Moreover,
that evaluated glucagon and alanine from plasma of elevated levels of a-ketoglutarate and glutamine were

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 631
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

observed, with increased concentrations of glutamine secretion, as well as in neuronal communication and
possibly related to a compensatory anabolic mecha- brain plasticity [223]. Glutamate is synthesized in sev-
nism in the context of mitochondrial impairment [215]. eral tissues, including the brain, skeletal muscle, liver,
Regarding muscles, T2D patients develop high sys- kidney, and pancreas, while acting both in intra- and
temic levels of amino acids and protein metabolic turn- extracellular signaling [223]. Glutamate can be gener-
over [216]. An interorgan crosstalk between the liver ated in the cytosol and in mitochondria (Fig. 3). In
and muscles suggests that the catabolism of alanine in the cytosol, glutamate is synthesized by using fructose-
the liver by ALT promotes an endocrine and meta- 6-phosphate (F6P) from the glycolysis pathway, where
bolic interaction that links hyperglycemia and muscle F6P and glutamine are converted by fructose-6-
atrophy in the diabetic condition [217]. ALT expres- phosphate aminotransferase; consequently forming
sion was upregulated in BKS-db/db mice with obesity glutamate. Moreover, through glutaminase, glutamine
and diabetes. Therefore, by silencing GPT and GPT2, can be deaminated to form glutamate (Fig. 3). In
the ALT isoforms, alanine tolerance was reversed, mitochondria, glutamate is formed by glutamate dehy-
linked to the lower glycemic response to alanine, as drogenase (GDH1 and GDH2), where a-ketoglutarate
well as polydipsia, glycosuria, and blood glucose levels is converted into glutamate (Fig. 3) [223]. Glutamate
were reduced by ALT silencing in the liver [217]. has been associated with glucose-induced insulin secre-
Moreover, the lack of ALT contributed to improve- tion for many years [224,225]. The synthesis of gluta-
ments in skeletal muscle mass in diabetic mice. The mate following glucose metabolism occurs either in
endocrine–liver–amino acid catabolic axis provides mitochondria of b-cells through GDH or in the cyto-
intriguing evidence that links hyperglycemia and mus- sol by transamination from aspartate [226–228]. In the
cle atrophy in T2D [217], providing a novel view former case, glutamate is transported to the cytosol by
regarding the metabolic interorgan crosstalk, which the glutamate carrier (GC1) [229]. Insulin exocytosis is
could serve as tools for new therapeutic approaches in primed following glutamate uptake into the insulin
T2D. secretory granules (Fig. 3). After glucose uptake into
Interestingly, alanine can be used as an intermediate b-cells, glutamate thus acts as a messenger molecule by
metabolite to form 1-deoxysphingolipids, which is an triggering insulin secretion [227,228]. Later, glutamate
atypical sphingolipid with toxic effects in neurons and was also linked to the potentiation of glucose-
b-cells. Sphingolipids have been associated with neuro- stimulated insulin secretion by the incretin hormones
logical diseases and T2D [218]. Their levels are also GLP1 and GIP. These gut hormones bind to their
increased in serum and visceral adipose tissue from respective receptors in the membrane of b-cells, gener-
T2D patients [104]. The synthesis of these sphingoli- ating cAMP and activating PKA [230,231]. This pro-
pids occurs when serine palmitoyltransferase condenses motes glutamate uptake by the insulin granules,
palmitoyl-CoA with alanine rather than serine [218]. facilitating insulin secretion [230,231]. Mechanistically,
In pancreatic a-cells, a study showed that a mixture of the high levels of glucose and consequently ATP pro-
amino acids (alanine, arginine, cysteine, and proline) duction lead to closure of the ATP-sensitive potassium
promoted increased levels of glucagon in the plasma in channel, which results in the depolarization of b-cells
female mice, as well as increased blood glucose and the opening of voltage-sensitive calcium channels.
through a glucagon receptor-dependent way [219]. Ala- The increase in cytosolic Ca2+ is the main trigger for
nine and other amino acids have been associated with insulin secretion [223]. Thus, intracellular glutamate
increases in a-cell proliferation [23,220,221]. Therefore, plays an important role in glucose- and incretin-
inhibition of glucagon signaling has been observed in stimulated insulin secretion [227,229]. VGLUT1 and
a-cell hyperplasia as well as hyperaminoacidemia VGLUT2 are the major glutamate transporters that
[23,220,222]. Thus, alanine seems to be a pivotal bio- are associated with the potentiation of insulin secretion
marker in pancreatic islet dysfunction, promoting met- [229].
abolic and structural changes that may participate in It is noteworthy that impaired b-cell responsiveness
the development of metabolic diseases, such as to incretin hormones is an early defect in T2D [231].
diabetes. Moreover, glutamate is cosecreted with insulin, caus-
ing high concentrations of glutamate in the extracellu-
lar space of the pancreatic islets. Therefore, the
Involvement of glutamate in health and disease
NMDA receptors in b-cells may be activated, which
Glutamate is a nonessential amino acid abundant in leads to depolarization and increases in cytosolic Ca2+,
the human body, which plays important roles in eventually resulting in b-cell dysfunction and death
metabolism, in the control of insulin and glucagon [232,233]. NMDA receptor antagonists have therefore

632 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

been suggested as treatment in T2D [234]. Concerning Type 2 diabetes as an inflammatory disease:
receptor-mediated effect of glutamate, it is worth men- metabolic association and outcomes
tioning that an inhibitory effect of glutamate on insu-
Several metabolites may cause inflammation during
lin secretion has also been associated with another
the progression of T2D. As we have already men-
membrane receptor, that is, GPRC5B [225].
tioned, high levels of FFAs, DAG, ceramides, and glu-
Obesity, aging, and T2D are characterized by insulin
tamate may collaborate with ROS and reactive
resistance, which not only affects glucose and lipid
nitrogen species production inducing a proinflamma-
homeostasis but also protein metabolism, resulting in
tory response in T2D [99,100,102,165–167,238,251]. In
diminished anabolic actions of insulin and increased
this section, we will summarize how metabolites trigger
plasma glutamate [235–237]. Hyperglutamatemia is
inflammation and how the proinflammatory response
associated with increased oxidative stress and inflam-
can affect the development of T2D (Fig. 4).
mation (Fig. 3), as is the case in diabetes [238–240].
High concentrations of free fatty acids have been
Proinflammatory factors and insulin resistance may
associated with increased plasma free radicals in
collaborate leading to cytotoxicity through high levels
humans [137], as well as ROS production in adipocytes
of glutamate [241]. Thus, more studies are needed in
through NADPH oxidase, which consequently resulted
order to uncover the effects of glutamate in the patho-
in proinflammatory cytokine production and release
physiology of T2D and its potential as a drug target.
from white adipose tissue [252]. ROS plays a key role
in signaling and inflammation [253]. The production
and release of proinflammatory cytokines may affect
Impairments in glutathione production may lead
not only the tissue where they were produced, such as
to disbalance of homeostasis
the adipose tissue and/or the liver, but also, by circu-
Glutathione (GSH) is a tripeptide synthesized from lating in the blood, may affect other tissues and lead
glutamate, cysteine, and glycine in the cytosol of all to an exacerbated proinflammatory response, cell
cell types, but especially in the liver which plays a death, and tissue damage [13].
major role in producing and exporting glutathione Lipid mediators, TNF-a, ROS, the activation of
[242]. First, c-glutamylcysteine is formed from gluta- Iҡba kinase b (IKKb) by hypoxia, as well as JNK
mate and cysteine by the c-glutamylcysteine synthetase induce the inhibition of IRS1 in adipose tissue and
(Fig. 3). Then, GSH is synthesized through GSH syn- liver (Fig. 4) [125,254,255]. Mechanistically, IKKb and
thetase which catalyzes c-glutamylcysteine and glycine JNK1 promote serine phosphorylation of IRS1 and
to form glutathione (Fig. 3) [243,244]. The amount of IRS2, driving the transcriptional activation of genes
GSH in the human body plays an important role in related to inflammation and consequently resulting in
many metabolic processes, especially since oxidative insulin resistance (Fig. 4) [255,256]. The NF-jB path-
stress triggers cellular and tissue damage via the way is also activated by high concentrations of FFAs
increased production of oxidizing free radicals. Several leading to proinflammatory cytokine production and
studies have demonstrated that reduced levels of GSH release, such as TNF-a, IL1b, IL-6, and MCP1
contribute to metabolic disorders [245]. Impairments (Fig. 4) [100]. Increased levels of circulating MCP1 are
and/or reduced levels of GSH are associated with associated with macrophage recruitment to tissues, in
aging, fibrotic diseases, liver diseases, and diabetes part mediating the inflammatory response [257].
mellitus [242,246]. In diabetes, nonglycemic mecha- Increased production of MCP1 by pancreatic islets
nisms are likely to alter GSH metabolism. For exam- during pathophysiological conditions has been associ-
ple, circulating fatty acids, high concentrations of ated with pancreatic b-cell dysfunction [258].
triglycerides, oxidative stress, and proinflammatory Free fatty acids can induce insulin resistance
cytokines may affect glutathione levels (Fig. 3) [247]. through different ways. First, FFAs are associated
In young mice deficient for Cdk1, an essential gene for with insulin resistance by increased lipid metabolism
hepatocyte proliferation and mitochondrial functions which leads to the inhibition of the insulin receptor
[213,248], oxidative stress activated FOXO1, which [259,260]. In addition, high concentrations of FFAs
leads to Pnpla2/ATGL expression resulting in high trigger ER stress in adipocytes [261], liver [262], and b-
concentrations of plasma FFAs [160]. Diabetic patient cells [263], and as a consequence, ER stress activates
samples display lower GSH concentrations [242,245– JNK leading to insulin resistance (Fig. 4) [264]. Not
247,249], possibly related to dyslipidemia and contrib- only are cytokines produced by NF-jB activation
uting to T2D complications [250]. Therefore, glutathi- stimulating JNK, which leads to insulin resistance, but
one metabolism in T2D is an important research area. they can also self-activate NF-jB in a feedback loop

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 633
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

Fig. 4. Metabolic connections in inflammation and insulin resistance. Several metabolites act as essential factors to trigger a
proinflammatory response. FFAs can promote the activation of NF-kB, induce ER stress, and consequently lead to ROS production. NF-kB
induces transcription factors that produce cytokines and chemokines, which leads to further activation of NF-kB. The release of cytokines
and chemokines induces the recruitment of macrophages and triggers a proinflammatory response. NF-kB, ROS, and lipid mediators, such
as ceramides, induce the activation of NLRP3 inflammasome and its oligomerization promotes the activation of caspase. Thus, caspase 1
promotes the cleavage of pro-IL-1b and pro-IL-18, and GDSMD. GDSMD microdomains form pores in the plasma membrane and IL-1b and
IL-18 are released, leading to a proinflammatory response. Hyperglycemia and hyperlipidemia can activate IKK which leads to insulin
resistance. Several factors may trigger insulin resistance including ceramides, DAG, ROS, ER stress, and NF-kB, which play important roles
in the blockage of insulin signaling. Proinflammatory stimuli or external factors can activate PLA2 and promote the synthesis of arachidonic
acid and consequently eicosanoid production, resulting in rapid release at the local site of inflammation.

(Fig. 4) [13]. This may trigger other proinflammatory production [116,124], as well as in the activation of the
pathways that consequently drive the detrimental out- inflammasome NLRP3 in obesity and T2D (Fig. 4)
comes in T2D. Liver and adipose tissue play an impor- [102]. Ceramides play roles in inflammation and insu-
tant role in the activation of proinflammatory lin resistance [186,194] and also activate the NLRP3
pathways through macrophages, for example, which inflammasome [197]. The NLRP3 inflammasome pro-
when activated, produce and release TNF, IL-6, and motes a proinflammatory response through the release
IL-1b [265–269]. IL-1b can be produced through the of IL-1b and IL-18 (Fig. 4) [273–275], which may be
expression of the proapoptotic receptor FAS in b-cells associated with an upregulated inflammation response
due to high concentrations of glucose [270,271]. The in diabetic patients.
activation of FAS and IL-1b contributes to glucose- In concert with the harmful effects of glucose in dia-
induced impairment in b-cells and may lead to cell betes, which lead to proinflammatory responses involv-
death [126,272]. FFAs also play a key role in IL-1b ing IL-1b transcription, production, and its release,

634 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

eicosanoids such as prostaglandin E2 can also induce Overall, the association between inflammation and
IL-1b production [276]. Prostaglandins and leukotri- T2D is convincing but is still under investigation.
enes are eicosanoid lipid mediators synthesized from From the data in the past and present literature
arachidonic acid, playing an important role in the emerges a close relationship between the inflammatory
acute and chronic inflammatory response. Eicosanoids process and the development of T2D, but how inflam-
are rapidly released by proinflammatory cytokines, mation promotes insulin resistance in target tissues, as
ROS, and external factors, resulting in pathological well as how the inflammation affects the outcomes of
consequences related to inflammation [277,278]. In T2D, needs to be further explored. Therefore, we
addition to their role in inflammation, arachidonic hypothesize that lipid metabolism plays important
acid and eicosanoids play important roles in tissue roles in the development of T2D through the synthesis
homeostasis, cellular signaling, and inducing vasodila- of lipid mediators related to inflammation and insulin
tation [279,280]. The synthesis of arachidonic acid can resistance. For example, diacylglycerol, ceramides, and
occur through the phospholipase A2 (PLA2) superfam- eicosanoids in high concentrations in cells and/or in
ily, where the enzymatic action on phospholipids of plasma may provoke changes in metabolism and affect
the membrane promotes its production (Fig. 4) several pathways, resulting in the progression of T2D.
[279,281]. In addition, arachidonic acid can also be In addition, changes in metabolism may also induce
formed via the activity of phospholipases C and D, proinflammatory responses through the production
which catalyze DAG production [282], as well as from and release of cytokines and chemokines, triggering
anandamides [283,284]. Eicosanoids are mainly gener- inflammation in different tissues and consequently pro-
ated from arachidonic acid by cyclooxygenase 1 or 2 voking tissue damage. Altogether, T2D can be consid-
(COX1/COX2), lipoxygenase (LOX), and cytochrome ered an inflammatory disease where several pathways
P450 (CYP) [285–288]. COX1 is expressed in most cell may lead to the inflammation process which is associ-
types and can be considered as a physiological enzyme ated with T2D development. Therefore, further studies
which is important for tissue homeostasis [286,287]. are needed in order to provide a better understanding
COX2 is required and is activated by proinflammatory of the immunometabolism in T2D.
processes and/or stimuli or tissue damage [289,290].
Through the catalysis of COX2 using arachidonic acid,
Conclusions and perspectives
the major eicosanoids synthesized are the prostaglan-
dins PGD2, PGE2, PGI2, PGF2a, and the thromboxane T2D is a disease that affects hundreds of millions of
TXA2 with their actions occurring through binding people and despite thorough research, we still do not
membrane receptors. In the context of T2D, it has fully understand this multifactorial disease. The dysre-
been demonstrated that high levels of glucose or pal- gulated secretory functions of islets and a reduced
mitic acid promote the expression of COX2 in human peripheral insulin action are central in T2D patho-
islets [291,292]. Palmitic acid also induces the expres- physiology. This has been studied extensively both
sion of the PGE2 receptor subtype 3, which is also in vitro using isolated islets and single b-cells as well as
upregulated in islets from T2D organ donors [293]. how the hormone insulin affects its target tissues
Inhibition of PGE2 synthesis or of its receptor pre- in vivo in animals and humans. One aspect that has
vents b-cell dysfunction and apoptosis [293]. Since drawn less attention is that liver and adipose tissue, in
COX is one of the enzymes responsible for eicosanoid addition to their well-documented properties of
synthesis, which can be associated with a proinflamma- increasing blood glucose and lipids, also release metab-
tory response, COX2 activation may also contribute olites and organokines that in turn affect the biology
to the development of inflammation in T2D. The of islets and insulin in a direct and indirect way. This
major eicosanoids derived from the LOX pathway is the main reason why we focused this review article
are hydroxyeicosatetraenoic acids (HETEs), such as on interorgan crosstalk and how this is involved in the
15(S)-HETE; 5(S)-HETE; 19(S)-HETE; 20-HETE; development and outcome of T2D. For example, as
()8-HETE; 12(S)-HETE; and also the leukotrienes the major organ for metabolism and energy produc-
LTA4, LTB4, LTC4, LTD4, and LTE4 [294,295]. The tion, the liver acts in several pathways in biological
LOX pathway is associated with the regulation of cyto- systems, contributing to the metabolic interorgan
kines and chemokines in several tissues, such as in islets crosstalk and triggering numerous responses in
and adipose tissue [296,297]. Previous data related 12- humans. Metabolites and hepatokines from the liver
LOX expression with hyperglycemia [298] and suggested affect metabolism of several tissues and may lead to
12-LOX to be involved (as well as 15-LOX) in insulin metabolic changes that drive ROS production, inflam-
resistance and adipose tissue inflammation [299,300]. mation, and cell death in other tissues, prompting the

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 635
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

development of T2D. The adipose tissue also plays an thank Maria Peleli for comments on the manuscript.
important role in the metabolic crosstalk since many The research work of PK and AS was supported by
metabolites produced in the adipose tissue contribute the Faculty of Medicine, Lund University, the Swedish
to obesity, inflammation, and T2D. Because of their Foundation for Strategic Research Dnr IRC15-0067,
participation in energy metabolism and positive effects and Strategic Research Area EXODIAB, Dnr 2009-
for liver and pancreas biology, muscle has also been 1039; PK was supported by the Swedish Research
under study for the past decades due to interaction Council (2021-01331) and the Swedish Cancer Society
with other tissues. In summary, there is a long road to (21 1566 Pj); AS was supported by the Swedish Research
go in order to determine how interorgan crosstalk Council, the Novo Nordic Foundation, the Swedish Dia-
affects the development and outcomes of T2D. betes Foundation, the Forget Foundation, and the
There are several limitations that affect the study of EFSD/Lilly Foundation; LNZ was supported by the
interorgan crosstalk: Crafoord Foundation and the IngaBritt och Arne Lund-
bergs Forskningsstiftelse LU2020-0013. The funders had
(1) Interorgan crosstalk is very difficult to study in
no role in study design, data collection and interpreta-
patients since biopsies from multiple organs are
tion, or the decision to submit the work for publication.
rarely available and when they are available, the
quantity of the biopsies only allows testing of sin-
gle parameters. Conflict of interest
(2) Ideally, interorgan crosstalk is investigated in an ani-
JMS, LNZ, and PK declare no conflict of interest. AS
mal system, which allows sampling of several organs
and CBW are founders of Abarceo Pharma, Malm€ o,
simultaneously and at any time of the experimental
Sweden, and hold stock in the company. CBW is a
setup. Nevertheless, one could argue that animals
member of its board.
(e.g., mice or rats) fundamentally differ from
humans, especially in the terms of metabolism.
(3) A further limitation is that there are ample data Author contributions
which associate metabolites (as well as protein and
PK conceptualized this review; JMS wrote the first
gene expression) with certain diseases or biological
draft of the manuscript and prepared the figures with
processes. Nevertheless, associations are not always
input from PK; JMS, LNZ, AS, CBW, and PK revised
reliable and they certainly do not allow us to
the manuscript; and all authors approved the final ver-
determine causality. Therefore, associations need
sion of this manuscript.
to be considered with a pinch of salt.
(4) In general, interorgan crosstalk is very difficult to
investigate since even when one knows the origin of References
the signal (metabolite, organokine, etc.), identifying
1 Chung HS & Choi KM (2020) Organokines in disease.
the target tissue(s) is arduous. It requires the collec- Adv Clin Chem 94, 261–321.
tion of a lot of data and to determine the causality 2 Gerst F, Wagner R, Kaiser G, Panse M, Heni M,
of the connections involved in the crosstalk. Machann J, Bongers MN, Sartorius T, Sipos B, Fend
In summary, metabolic interorgan crosstalk in T2D F et al. (2017) Metabolic crosstalk between fatty
is a field that needs to be much more explored in order pancreas and fatty liver: effects on local inflammation
and insulin secretion. Diabetologia 60, 2240–2251.
to provide a better understanding of the pathophysiol-
3 Severinsen MCK & Pedersen BK (2020) Muscle-organ
ogy behind metabolic disorders and development of
crosstalk: the emerging roles of myokines. Endocr Rev
T2D. Novel mouse models with sensors in several
41, 594–609.
organs could help to further investigate interorgan
4 Stern JH, Rutkowski JM & Scherer PE (2016)
crosstalk. Detailed knowledge of metabolic interorgan
Adiponectin, leptin, and fatty acids in the maintenance
crosstalk may help us to discover better treatments, of metabolic homeostasis through adipose tissue
provide better prognosis, and improve the quality of crosstalk. Cell Metab 23, 770–784.
life for the affected patients. 5 Romani P, Valcarcel-Jimenez L, Frezza C & Dupont S
(2021) Crosstalk between mechanotransduction and
metabolism. Nat Rev Mol Cell Biol 22, 22–38.
Acknowledgements
6 Petersen MC & Shulman GI (2018) Mechanisms of
We thank all present and past members of the Kaldis insulin action and insulin resistance. Physiol Rev 98,
laboratory for discussions, input, and support. We 2133–2223.

636 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

7 Choi KM (2016) The impact of organokines on insulin 23 Solloway M, Madjidi A, Gu C, Eastham-Anderson J,


resistance, inflammation, and atherosclerosis. Clarke H, Kljavin N, Zavala-Solorio J, Kates L,
Endocrinol Metab (Seoul) 31, 1–6. Friedman B, Brauer M et al. (2015) Glucagon couples
8 Funcke JB & Scherer PE (2019) Beyond adiponectin hepatic amino acid catabolism to mTOR-dependent
and leptin: adipose tissue-derived mediators of inter- regulation of a-cell mass. Cell Rep 12, 495–510.
organ communication. J Lipid Res 60, 1648–1684. 24 Knudsen JG, Hamilton A, Ramracheya R, Tarasov
9 de Oliveira dos Santos AR, de Oliveira Zanuso B, AI, Brereton M, Haythorne E, Chibalina MV, Spegel
Miola VFB, Barbalho SM, Santos Bueno PC, Flato P, Mulder H, Zhang Q et al. (2019) Dysregulation of
UAP, Detregiachi CRP, Buchaim DV, Buchaim RL, glucagon secretion by hyperglycemia-induced sodium-
Tofano RJ et al. (2021) Adipokines, myokines, and dependent reduction of ATP production. Cell Metab
hepatokines: crosstalk and metabolic repercussions. Int 29, 430–442.
J Mol Sci 22, 2639. 25 Holst JJ (2020) Incretin therapy for diabetes mellitus
10 Meex RCR & Watt MJ (2017) Hepatokines: linking type 2. Curr Opin Endocrinol Diabetes Obes 27, 2–10.
nonalcoholic fatty liver disease and insulin resistance. 26 Ravussin Y, Leibel RL & Ferrante AW (2014) A
Nat Rev Endocrinol 13, 509–520. missing link in body weight homeostasis: the catabolic
11 Adams SH, Hoppel CL, Lok KH, Zhao L, Wong SW, signal of the overfed state. Cell Metab 20, 565–572.
Minkler PE, Hwang DH, Newman JW & Timothy 27 Straub LG & Scherer PE (2019) Metabolic messengers:
Garvey W (2009) Plasma acylcarnitine profiles suggest adiponectin. Nat Metab 1, 334–339.
incomplete long-chain fatty acid beta-oxidation and 28 Han MS, Perry RJ, Camporez J-P, Scherer PE,
altered tricarboxylic acid cycle activity in type 2 diabetic Shulman GI, Gao G & Davis RJ (2021) A feed-
African-American women. J Nutr 139, 1073–1081. forward regulatory loop in adipose tissue promotes
12 Chatterjee S, Khunti K & Davies MJ (2017) Type 2 signaling by the hepatokine FGF21. Genes Dev 35,
diabetes. Lancet 389, 2239–2251. 133–146.
13 Donath MY & Shoelson SE (2011) Type 2 diabetes as 29 Lo J, Ljubicic S, Leibiger B, Kern M, Leibiger I,
an inflammatory disease. Nat Rev Immun 11, 98–107. Moede T, Kelly M, Chatterjee Bhowmick D, Murano
14 Eisenbarth GS (1986) Type I diabetes mellitus. A I, Cohen P et al. (2014) Adipsin is an adipokine that
chronic autoimmune disease. N Engl J Med 314, 1360– improves b cell function in diabetes. Cell 158, 41–53.
1368. 30 Holst JJ, Wewer Albrechtsen NJ, Pedersen J & Knop
15 Zheng Y, Ley SH & Hu FB (2018) Global aetiology FK (2017) Glucagon and amino acids are linked in a
and epidemiology of type 2 diabetes mellitus and its mutual feedback cycle: the liver-a-cell axis. Diabetes
complications. Nat Rev Endocrinol 14, 88–98. 66, 235–240.
16 Association AD (2014) Diagnosis and classification of 31 Winther-Sørensen M, Galsgaard KD, Santos A,
diabetes mellitus. Diabetes Care 37 (Suppl 1), S81–S90. Trammell SAJ, Sulek K, Kuhre RE, Pedersen J,
17 Mooradian AD (2009) Dyslipidemia in type 2 diabetes Andersen DB, Hassing AS, Dall M et al. (2020)
mellitus. Nat Clin Pract Endocrinol Metab 5, 150–159. Glucagon acutely regulates hepatic amino acid
18 Ahlqvist E, Storm P, K€ar€aj€am€aki A, Martinell M, catabolism and the effect may be disturbed by
Dorkhan M, Carlsson A, Vikman P, Prasad RB, Aly steatosis. Mol Metab 42, 101080.
DM, Almgren P et al. (2018) Novel subgroups of 32 Gar C, Haschka SJ, Kern-Matschilles S, Rauch B,
adult-onset diabetes and their association with Sacco V, Prehn C, Adamski J, Seissler J, Wewer
outcomes: a data-driven cluster analysis of six Albrechtsen NJ, Holst JJ et al. (2021) The liver-alpha
variables. Lancet Diabetes Endocrinol 6, 361–369. cell axis associates with liver fat and insulin resistance:
19 Hu FB, Satija A & Manson JE (2015) Curbing the a validation study in women with non-steatotic liver
diabetes pandemic: the need for global policy fat levels. Diabetologia 64, 512–520.
solutions. JAMA 313, 2319–2320. 33 Haber EP, Ximenes HM, Procopio J, Carvalho CR,
20 Foulis AK, McGill M & Farquharson MA (1991) Curi R & Carpinelli AR (2003) Pleiotropic effects of
Insulitis in type 1 (insulin-dependent) diabetes mellitus fatty acids on pancreatic beta-cells. J Cell Physiol 194,
in man-macrophages, lymphocytes, and interferon- 1–12.
gamma containing cells. J Pathol 165, 97–103. 34 Jimenez-Feltstrom J, Salehi A, Meidute Abaraviciene
21 Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy S, Henningsson R & Lundquist I (2011) Abnormally
MI, Brown MA & Yang J (2017) 10 years of GWAS decreased NO and augmented CO production in islets
discovery: biology, function, and translation. Am J of the leptin-deficient ob/ob mouse might contribute to
Hum Genet 101, 5–22. explain hyperinsulinemia and islet survival in leptin-
22 Rizza RA (2010) Pathogenesis of fasting and resistant type 2 obese diabetes. Regul Pept 170, 43–51.
postprandial hyperglycemia in type 2 diabetes: 35 Feingold KR. (2000) Atypical forms of diabetes. In
implications for therapy. Diabetes 59, 2697–2707. Endotext [Internet] (Feingold KR, Anawalt B, Boyce

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 637
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

A, Chrousos G, de Herder WW, Dhatariya K, 48 Meex R, Hoy A, Morris A, Brown R, Lo J, Burke M,


Dungan K, Hershman JM, Hofland J, Kalra S, et al. Goode R, Kingwell B, Kraakman M, Febbraio M et
eds). MDText.com, Inc., South Dartmouth, MA. al. (2015) Fetuin B is a secreted hepatocyte factor
Available from: https://www.ncbi.nlm.nih.gov/books/ linking steatosis to impaired glucose metabolism. Cell
NBK279128/ Metab 22, 1078–1089.
36 Hardt PD, Krauss A, Bretz L, Porsch-Ozc€ ur€
umez M, 49 Montgomery MK, Bayliss J, Devereux C, Bezawork-
Schnell-Kretschmer H, M€aser E, Bretzel RG, Zekorn Geleta A, Roberts D, Huang C, Schittenhelm RB,
T & Kl€ or HU (2000) Pancreatic exocrine function in Ryan A, Townley SL, Selth LA et al. (2020) SMOC1
patients with type 1 and type 2 diabetes mellitus. Acta is a glucose-responsive hepatokine and therapeutic
Diabetol 37, 105–110. target for glycemic control. Sci Trans Med 12,
37 Wang Z, Lai S-T, Xie LI, Zhao J-D, Ma N-Y, Zhu JI, eaaz8048.
Ren Z-G & Jiang G-L (2014) Metformin is associated 50 Seo JA, Kang M-C, Yang W-M, Hwang WM, Kim
with reduced risk of pancreatic cancer in patients with SS, Hong SH, Heo J-I, Vijyakumar A, Pereira de
type 2 diabetes mellitus: a systematic review and meta- Moura L, Uner A et al. (2020) Apolipoprotein J is a
analysis. Diabetes Res Clin Pract 106, 19–26. hepatokine regulating muscle glucose metabolism and
38 Scaroni C, Zilio M, Foti M & Boscaro M (2017) insulin sensitivity. Nat Commun 11, 2024.
Glucose metabolism abnormalities in cushing 51 Misu H, Ishikura K, Kurita S, Takeshita Y, Ota T,
syndrome: from molecular basis to clinical Saito Y, Takahashi K, Kaneko S & Takamura T
management. Endocr Rev 38, 189–219. (2012) Inverse correlation between serum levels of
39 Gremlich S, Roduit R & Thorens B (1997) selenoprotein P and adiponectin in patients with type 2
Dexamethasone induces posttranslational degradation diabetes. PLoS One 7, e34952.
of GLUT2 and inhibition of insulin secretion in 52 Misu H, Takamura T, Takayama H, Hayashi H,
isolated pancreatic beta cells. Comparison with the Matsuzawa-Nagata N, Kurita S, Ishikura K, Ando H,
effects of fatty acids. J Biol Chem 272, 3216–3222. Takeshita Y, Ota T et al. (2010) A liver-derived
40 Bedi O, Aggarwal S, Trehanpati N, Ramakrishna G & secretory protein, selenoprotein P, causes insulin
Krishan P (2019) Molecular and pathological events resistance. Cell Metab 12, 483–495.
involved in the pathogenesis of diabetes-associated 53 Yang SJ, Hwang SY, Choi HY, Yoo HJ, Seo JA, Kim
nonalcoholic fatty liver disease. J Clin Exp Hepatol 9, SG, Kim NH, Baik SH, Choi DS & Choi KM (2011)
607–618. Serum selenoprotein P levels in patients with type 2
41 Finck BN (2018) Targeting metabolism, insulin diabetes and prediabetes: implications for insulin
resistance, and diabetes to treat nonalcoholic resistance, inflammation, and atherosclerosis. J Clin
steatohepatitis. Diabetes 67, 2485–2493. Endocrinol Metab 96, E1325–E1329.
42 Buzzetti E, Pinzani M & Tsochatzis EA (2016) The 54 Perry JRB, Weedon MN, Langenberg C, Jackson AU,
multiple-hit pathogenesis of non-alcoholic fatty liver Lyssenko V, Sparsø T, Thorleifsson G, Grallert H,
disease (NAFLD). Metabolism 65, 1038–1048. Ferrucci L, Maggio M et al. (2010) Genetic evidence
43 Sung KC & Kim SH (2011) Interrelationship between that raised sex hormone binding globulin (SHBG)
fatty liver and insulin resistance in the development of levels reduce the risk of type 2 diabetes. Hum Mol
type 2 diabetes. J Clin Endocrinol Metab 96, 1093–1097. Genet 19, 535–544.
44 Stefan N & H€aring HU (2013) The role of hepatokines 55 Muise ES, Souza S, Chi AN, Tan Y, Zhao X, Liu F,
in metabolism. Nat Rev Endocrinol 9, 144–152. Dallas-yang Q, Wu M, Sarr T, Zhu L et al. (2013)
45 Pal D, Dasgupta S, Kundu R, Maitra S, Das G, Downstream signaling pathways in mouse adipose
Mukhopadhyay S, Ray S, Majumdar SS & tissues following acute in vivo administration of
Bhattacharya S (2012) Fetuin-A acts as an endogenous fibroblast growth factor 21. PLoS One 8, e73011.
ligand of TLR4 to promote lipid-induced insulin 56 Hwang H-J, Jung TW, Hong HC, Seo JA, Kim SG,
resistance. Nat Med 18, 1279–1285. Kim NH, Choi KM, Choi DS, Baik SH & Yoo HJ
46 Mukhopadhyay S & Bhattacharya S (2016) Plasma (2015) LECT2 induces atherosclerotic inflammatory
fetuin-A triggers inflammatory changes in reaction via CD209 receptor-mediated JNK
macrophages and adipocytes by acting as an adaptor phosphorylation in human endothelial cells.
protein between NEFA and TLR-4. Diabetologia 59, Metabolism 64, 1175–1182.
859–860. 57 Combs TP, Pajvani UB, Berg AH, Lin Y, Jelicks LA,
47 Xing W, Tan Y, Li K, Tian P, Tian F & Zhang H Laplante M, Nawrocki AR, Rajala MW, Parlow AF,
(2021) Upregulated hepatokine fetuin B aggravates Cheeseboro L et al. (2004) A transgenic mouse with a
myocardial ischemia/reperfusion injury through deletion in the collagenous domain of adiponectin
inhibiting insulin signaling in diabetic mice. J Mol Cell displays elevated circulating adiponectin and improved
Cardiol 151, 163–172. insulin sensitivity. Endocrinology 145, 367–383.

638 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

58 Tomas E, Tsao T-S, Saha AK, Murrey HE, Zhang Chamani E & Tavakoli T (2020) Characterization of
CC, Itani SI, Lodish HF & Ruderman NB (2002) the serum levels of Meteorin-like in patients with
Enhanced muscle fat oxidation and glucose transport inflammatory bowel disease and its association with
by ACRP30 globular domain: acetyl-CoA carboxylase inflammatory cytokines. Lipids Health Dis 19, 230.
inhibition and AMP-activated protein kinase 72 Yang Q, Vijayakumar A & Kahn BB (2018)
activation. Proc Natl Acad Sci USA 99, 16309–16313. Metabolites as regulators of insulin sensitivity and
59 Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki H, metabolism. Nat Rev Mol Cell Biol 19, 654–672.
Uchida S, Yamashita S, Noda M, Kita S, Ueki K et 73 Saltiel AR & Kahn CR (2001) Insulin signalling and
al. (2002) Adiponectin stimulates glucose utilization the regulation of glucose and lipid metabolism. Nature
and fatty-acid oxidation by activating AMP-activated 414, 799–806.
protein kinase. Nat Med 8, 1288–1295. 74 Niswender KD (2011) Basal insulin: physiology,
60 Kubota N, Yano W, Kubota T, Yamauchi T, Itoh S, pharmacology, and clinical implications. Postgrad Med
Kumagai H, Kozono H, Takamoto I, Okamoto S, 123, 17–26.
Shiuchi T et al. (2007) Adiponectin stimulates AMP- 75 Manning BD & Toker A (2017) AKT/PKB signaling:
activated protein kinase in the hypothalamus and navigating the network. Cell 169, 381–405.
increases food intake. Cell Metab 6, 55–68. 76 Gross DN, Wan M & Birnbaum MJ (2009) The role
61 Fasshauer M & Bl€ uher M (2015) Adipokines in health of FOXO in the regulation of metabolism. Curr Diab
and disease. Trends Pharmacol Sci 36:461–470. Rep 9, 208–214.
10.1016/j.tips.2015.04.014 77 Luetscher JA (1942) The metabolism of amino acids in
62 Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee diabetes mellitus. J Clin Invest 21, 275–279.
RR, Wright CM, Patel HR, Ahima RS & Lazar MA 78 Felig P, Marliss E & Cahill GF (1969) Plasma amino
(2001) The hormone resistin links obesity to diabetes. acid levels and insulin secretion in obesity. N Engl J
Nature 409, 307–312. Med 281, 811–816.
63 Dunmore SJ & Brown JE (2013) The role of 79 Sener A & Malaisse WJ (1980) L-leucine and a
adipokines in b-cell failure of type 2 diabetes. nonmetabolized analogue activate pancreatic islet
J Endocrinol 216, T37–T45. glutamate dehydrogenase. Nature 288:187–189.
64 Hotamisligil GS, Shargill NS & Spiegelman BM (1993) 80 Liu L, Wang M, Yang X, Bi M, Na L, Niu Y, Li Y &
Adipose expression of tumor necrosis factor-alpha: Sun C (2013) Fasting serum lipid and
direct role in obesity-linked insulin resistance. Science dehydroepiandrosterone sulfate as important
259, 87–91. metabolites for detecting isolated postchallenge
65 Peraldi P, Hotamisligil GS, Buurman WA, White MF diabetes: serum metabolomics via ultra-high-
& Spiegelman BM (1996) Tumor necrosis factor performance LC-MS. Clin Chem 59, 1338–1348.
(TNF)-alpha inhibits insulin signaling through 81 Bentley-Lewis R, Xiong G, Lee H, Yang A, Huynh J
stimulation of the p55 TNF receptor and activation of & Kim C (2014) Metabolomic analysis reveals amino
sphingomyelinase. J Biol Chem 271, 13018–13022. acid responses to an oral glucose tolerance test in
66 Bl€uher M (2012) Clinical relevance of adipokines. women with prior history of gestational diabetes
Diabetes Metab J 36, 317–327. mellitus. J Clin Transl Endocrinol 1, 38–43.
67 Verma S, Li S-H, Wang C-H, Fedak PWM, Li R-K, 82 Salek RM, Maguire ML, Bentley E, Rubtsov DV,
Weisel RD & Mickle DAG (2003) Resistin promotes Hough T, Cheeseman M, Nunez D, Sweatman BC,
endothelial cell activation: further evidence of adipokine- Haselden JN, Cox RD et al. (2007) A metabolomic
endothelial interaction. Circulation 108, 736–740. comparison of urinary changes in type 2 diabetes in
68 Peppler WT, Townsend LK, Meers GM, Panasevich mouse, rat, and human. Physiol Genomics 29, 99–108.
MR, MacPherson REK, Rector RS & Wright DC (2019) 83 Diao C, Zhao L, Guan M, Zheng Y, Chen M, Yang
Acute administration of IL-6 improves indices of hepatic Y, Lin L, Chen W & Gao H (2014) Systemic and
glucose and insulin homeostasis in lean and obese mice. characteristic metabolites in the serum of
Am J Physiol Gastrointest Liver Physiol 316, G166–G178. streptozotocin-induced diabetic rats at different stages
69 Pedersen BK & Febbraio MA (2008) Muscle as an as revealed by a (1)H-NMR based metabonomic
endocrine organ: focus on muscle-derived interleukin- approach. Mol BioSyst 10, 6866–6893.
6. Physiol Rev 88, 1379–1406. 84 Fiehn O, Garvey WT, Newman JW, Lok KH, Hoppel
70 Oh KJ, Lee DS, Kim WK, Han BS, Lee SC & Bae CL & Adams SH (2010) Plasma metabolomic profiles
KH (2016) Metabolic adaptation in obesity and type II reflective of glucose homeostasis in non-diabetic and
diabetes: myokines, adipokines and hepatokines. Int J type 2 diabetic obese African-American women. PLoS
Mol Sci 18, 8. One 5, e15234.
71 Gholamrezayi A, Mohamadinarab M, Rahbarinejad P, 85 Palmer ND, Stevens RD, Antinozzi PA, Anderson A,
Fallah S, Barez SR, Setayesh L, Moradi N, Fadaei R, Bergman RN, Wagenknecht LE, Newgard CB &

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 639
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

Bowden DW (2015) Metabolomic profile associated signature in type-2 diabetes mellitus and its
with insulin resistance and conversion to diabetes in pathophysiology. PLoS One 9, e85082.
the insulin resistance atherosclerosis study. J Clin 97 Dudzinska W (2014) Purine nucleotides and their
Endocrinol Metab 100, E463–E468. metabolites in patients with type 1 and 2 diabetes
86 Sun Y, Gao HY, Fan ZY, He Y & Yan YX (2020) mellitus. J Biomed Sci Eng 7, 38–44.
Metabolomics signatures in type 2 diabetes: a 98 Huang Q, Yin P, Wang J, Chen J, Kong H, Lu X &
systematic review and integrative analysis. J Clin Xu G (2011) Method for liver tissue metabolic
Endocrinol Metab 105, 1000–1008. profiling study and its application in type 2 diabetic
87 Tillin T, Hughes AD, Wang Q, W€ urtz P, Ala-Korpela rats based on ultra performance liquid
M, Sattar N, Forouhi NG, Godsland IF, Eastwood chromatography-mass spectrometry. J Chromatogr B
SV, McKeigue PM et al. (2015) Diabetes risk and Analyt Technol Biomed Life Sci 879, 961–967.
amino acid profiles: cross-sectional and prospective 99 Boden G (2011) Obesity, insulin resistance and free
analyses of ethnicity, amino acids and diabetes in a fatty acids. Curr Opin Endocrinol Diabetes Obes 18,
South Asian and European cohort from the SABRE 139–143.
(Southall And Brent REvisited) Study. Diabetologia 100 Boden G, She P, Mozzoli M, Cheung P, Gumireddy
58, 968–979. K, Reddy P, Xiang X, Luo Z & Ruderman N (2005)
88 Kaur P, Rizk N, Ibrahim S, Luo Y, Younes N, Perry Free fatty acids produce insulin resistance and activate
B, Dennis K, Zirie M, Luta G & Cheema AK (2013) the proinflammatory nuclear factor-kappaB pathway
Quantitative metabolomic and lipidomic profiling in rat liver. Diabetes 54, 3458–3465.
reveals aberrant amino acid metabolism in type 2 101 Galadari S, Rahman A, Pallichankandy S, Galadari A
diabetes. Mol BioSyst 9, 307–317. & Thayyullathil F (2013) Role of ceramide in diabetes
89 Diamanti K, Cavalli M, Pan G, Pereira MJ, Kumar C, mellitus: evidence and mechanisms. Lipids Health Dis
Skrtic S, Grabherr M, Riserus U, Eriksson JW, 12, 98.
Komorowski J et al. (2019) Intra- and inter-individual 102 Legrand-Poels S, Esser N, L’homme L, Scheen A,
metabolic profiling highlights carnitine and Paquot N & Piette J (2014) Free fatty acids as
lysophosphatidylcholine pathways as key molecular modulators of the NLRP3 inflammasome in obesity/
defects in type 2 diabetes. Sci Rep 9, 9653. type 2 diabetes. Biochem Pharmacol 92, 131–141.
90 Wang TJ, Ngo D, Psychogios N, Dejam A, Larson 103 Rutkowsky JM, Knotts TA, Ono-Moore KD, McCoin
MG, Vasan RS, Ghorbani A, O’Sullivan J, Cheng S, CS, Huang S, Schneider D, Singh S, Adams SH &
Rhee EP et al. (2013) 2-Aminoadipic acid is a Hwang DH (2014) Acylcarnitines activate
biomarker for diabetes risk. J Clin Invest 123, proinflammatory signaling pathways. Am J Physiol
4309–4317. Endocrinol Metab 306, E1378–E1387.
91 Zhang N, Geng F, Hu ZH, Liu B, Wang YQ, Liu JC, 104 Hannich JT, Loizides-Mangold U, Sinturel F,
Qi YH & Li LJ (2016) Preliminary study of urine Harayama T, Vandereycken B, Saini C, Gosselin P,
metabolism in type two diabetic patients based on GC- Brulhart-Meynet M-C, Robert M, Chanon S et al.
MS. Am J Transl Res 8, 2889–2896. (2020) Ether lipids, sphingolipids and toxic 1-
92 Park S, Sadanala KC & Kim EK (2015) A deoxyceramides as hallmarks for lean and obese type 2
metabolomic approach to understanding the metabolic diabetic patients. Acta Physiol (Oxf) 232, e13610.
link between obesity and diabetes. Mol Cells 38, 105 Ameer F, Scandiuzzi L, Hasnain S, Kalbacher H &
587–596. Zaidi N (2014) De novo lipogenesis in health and
93 Sparks DL, Doelle H & Chatterjee C (2014) disease. Metabolism 63, 895–902.
Circulating nucleotide levels in health and disease. 106 Feron O (2009) Pyruvate into lactate and back: from
Receptors Clin Invest 1, e344. the Warburg effect to symbiotic energy fuel exchange
94 Guan M, Xie L, Diao C, Wang NA, Hu W, Zheng Y, in cancer cells. Radiother Oncol 92, 329–333.
Jin L, Yan Z & Gao H (2013) Systemic perturbations 107 Dentin R, Girard J & Postic C (2005) Carbohydrate
of key metabolites in diabetic rats during the evolution responsive element binding protein (ChREBP) and
of diabetes studied by urine metabonomics. PLoS One sterol regulatory element binding protein-1c (SREBP-
8, e60409. 1c): two key regulators of glucose metabolism and
95 Nikiforova VJ, Giesbertz P, Wiemer J, Bethan B, lipid synthesis in liver. Biochimie 87, 81–86.
Looser R, Liebenberg V, Ruiz Noppinger P, Daniel H 108 Bugianesi E, McCullough AJ & Marchesini G (2005)
& Rein D (2014) Glyoxylate, a new marker metabolite Insulin resistance: a metabolic pathway to chronic
of type 2 diabetes. J Diabetes Res 2014, 685204. liver disease. Hepatology 42, 987–1000.
96 Padberg I, Peter E, Gonzalez-Maldonado S, Witt H, 109 Qian M, Hu H, Yao Y, Zhao D, Wang S, Pan C,
Mueller M, Weis T, Bethan B, Liebenberg V, Wiemer Duan X, Gao Y, Liu J, Zhang Y et al. (2020)
J, Katus HA et al. (2014) A new metabolomic Coordinated changes of gut microbiome and lipidome

640 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

differentiates nonalcoholic steatohepatitis (NASH) 122 Shimobayashi M, Albert V, Woelnerhanssen B, Frei


from isolated steatosis. Liver Int 40, 622–637. IC, Weissenberger D, Meyer-Gerspach AC, Clement
110 Suhre K, Meisinger C, D€ oring A, Altmaier E, Belcredi N, Moes S, Colombi M, Meier JA et al. (2018) Insulin
P, Gieger C, Chang D, Milburn MV, Gall WE, resistance causes inflammation in adipose tissue. J Clin
Weinberger KM et al. (2010) Metabolic footprint of Invest 128, 1538–1550.
diabetes: a multiplatform metabolomics study in an 123 Paolisso G, Tataranni PA, Foley JE, Bogardus C,
epidemiological setting. PLoS One 5, e13953. Howard BV & Ravussin E (1995) A high
111 Prentki M, Corkey BE & Madiraju SRM (2020) concentration of fasting plasma non-esterified fatty
Lipid-associated metabolic signalling networks in acids is a risk factor for the development of NIDDM.
pancreatic beta cell function. Diabetologia 63, 10–20. Diabetologia 38, 1213–1217.
112 Taddeo EP, Alsabeeh N, Baghdasarian S, Wikstrom 124 B€oni-Schnetzler M, Boller S, Debray S, Bouzakri K,
JD, Ritou E, Sereda S, Erion K, Li J, Stiles L, Abdulla Meier DT, Prazak R, Kerr-Conte J, Pattou F, Ehses
M et al. (2020) Mitochondrial proton leak regulated by JA, Schuit FC et al. (2009) Free fatty acids induce a
cyclophilin D elevates insulin secretion in islets at proinflammatory response in islets via the abundantly
nonstimulatory glucose levels. Diabetes 69, 131–145. expressed interleukin-1 receptor I. Endocrinology 150,
113 Hu M, Phan F, Bourron O, Ferre P & Foufelle F 5218–5229.
(2017) Steatosis and NASH in type 2 diabetes. 125 Ye J (2009) Emerging role of adipose tissue hypoxia in
Biochimie 143, 37–41. obesity and insulin resistance. Int J Obes 33, 54–66.
114 Imamura F, Fretts AM, Marklund M, Ardisson Korat 126 Maedler K, Sergeev P, Ris F, Oberholzer J, Joller-
AV, Yang W-S, Lankinen M, Qureshi W, Helmer C, Jemelka HI, Spinas GA, Kaiser N, Halban PA &
Chen T-A, Virtanen JK et al. (2020) Fatty acids in the Donath MY (2002) Glucose-induced beta cell
de novo lipogenesis pathway and incidence of type 2 production of IL-1beta contributes to glucotoxicity in
diabetes: a pooled analysis of prospective cohort human pancreatic islets. J Clin Invest 110, 851–860.
studies. PLoS Medicine 17, e1003102. 127 Floegel A, Stefan N, Yu Z, Muhlenbruch K, Drogan
115 Maedler K, Spinas GA, Dyntar D, Moritz W, Kaiser D, Joost H-G, Fritsche A, Haring H-U, Hrabe de
N & Donath MY (2001) Distinct effects of saturated Angelis M, Peters A et al. (2013) Identification of
and monounsaturated fatty acids on beta-cell turnover serum metabolites associated with risk of type 2
and function. Diabetes 50, 69–76. diabetes using a targeted metabolomic approach.
116 Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H & Diabetes 62, 639–648.
Flier JS (2006) TLR4 links innate immunity and fatty 128 Zimmet P, Alberti KG, Magliano DJ & Bennett PH
acid-induced insulin resistance. J Clin Invest 116, 3015– (2016) Diabetes mellitus statistics on prevalence and
3025. mortality: facts and fallacies. Nat Rev Endocrinol 12,
117 Chavez JA & Summers SA (2003) Characterizing the 616–622.
effects of saturated fatty acids on insulin signaling and 129 Birkenfeld AL & Shulman GI (2014) Nonalcoholic
ceramide and diacylglycerol accumulation in 3T3-L1 fatty liver disease, hepatic insulin resistance, and type
adipocytes and C2C12 myotubes. Arch Biochem 2 diabetes. Hepatology 59, 713–723.
Biophys 419, 101–109. 130 Ramırez M, Amate L & Gil A (2001) Absorption and
118 Holland WL, Bikman BT, Wang L-P, Yuguang G, distribution of dietary fatty acids from different
Sargent KM, Bulchand S, Knotts TA, Shui G, Clegg sources. Early Hum Dev 65 (Suppl), S95–S101.
DJ, Wenk MR et al. (2011) Lipid-induced insulin 131 Rui L (2014) Energy metabolism in the liver. Compr
resistance mediated by the proinflammatory receptor Physiol 4, 177–197.
TLR4 requires saturated fatty acid-induced ceramide 132 Sobczak AIS, Blindauer CA & Stewart AJ (2019)
biosynthesis in mice. J Clin Invest 121, 1858–1870. Changes in plasma free fatty acids associated with
119 Imamura F, Micha R, Wu JHY, de Oliveira Otto MC, type-2 diabetes. Nutrients 11, 2022.
Otite FO, Abioye AI & Mozaffarian D (2016) Effects 133 Holland WL, Brozinick JT, Wang L-P, Hawkins ED,
of saturated fat, polyunsaturated fat, monounsaturated Sargent KM, Liu Y, Narra K, Hoehn KL, Knotts TA,
fat, and carbohydrate on glucose-insulin homeostasis: Siesky A et al. (2007) Inhibition of ceramide synthesis
a systematic review and meta-analysis of randomised ameliorates glucocorticoid-, saturated-fat-, and
controlled feeding trials. PLoS Medicine 13, e1002087. obesity-induced insulin resistance. Cell Metab 5,
120 Hotamisligil GS & Davis RJ (2016) Cell signaling and 167–179.
stress responses. Cold Spring Harb Perspect Biol 8, 134 Choi CS, Savage DB, Kulkarni A, Yu XX, Liu Z-X,
a006072. Morino K, Kim S, Distefano A, Samuel VT, Neschen
121 Frakes AE & Dillin A (2017) The UPR ER: sensor S et al. (2007) Suppression of diacylglycerol
and coordinator of organismal homeostasis. Mol Cell acyltransferase-2 (DGAT2), but not DGAT1, with
66, 761–771. antisense oligonucleotides reverses diet-induced hepatic

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 641
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

steatosis and insulin resistance. J Biol Chem 282, beta-cell: the role of fatty acids. Clin Sci (Lond) 112,
22678–22688. 27–42.
135 Nagai Y, Yonemitsu S, Erion DM, Iwasaki T, Stark 149 Shimabukuro M, Zhou YT, Levi M & Unger RH
R, Weismann D et al. (2009) The role of peroxisome (1998) Fatty acid-induced beta cell apoptosis: a link
proliferator-activated receptor gamma coactivator-1 between obesity and diabetes. Proc Natl Acad Sci USA
beta in the pathogenesis of fructose-induced insulin 95, 2498–2502.
resistance. Cell Metab 9, 252–264. 150 Biden TJ, Boslem E, Chu KY & Sue N (2014)
136 Brass EP (1995) Pharmacokinetic considerations for Lipotoxic endoplasmic reticulum stress, b cell failure,
the therapeutic use of carnitine in hemodialysis and type 2 diabetes mellitus. Trends Endocrinol Metab
patients. Clin Ther 17, 176–185. 25, 389–398.
137 Paolisso G, Gambardella A, Tagliamonte MR, 151 Hotamisligil GS, Murray DL, Choy LN & Spiegelman
Saccomanno F, Salvatore T, Gualdiero P, D’Onofrio BM (1994) Tumor necrosis factor alpha inhibits
MV & Howard BV (1996) Does free fatty acid signaling from the insulin receptor. Proc Natl Acad Sci
infusion impair insulin action also through an increase USA 91, 4854–4858.
in oxidative stress? J Clin Endocrinol Metab 81, 152 Zhang HH, Halbleib M, Ahmad F, Manganiello VC
4244–4248. & Greenberg AS (2002) Tumor necrosis factor-alpha
138 Jensen MD, Caruso M, Heiling V & Miles JM (1989) stimulates lipolysis in differentiated human adipocytes
Insulin regulation of lipolysis in nondiabetic and through activation of extracellular signal-related kinase
IDDM subjects. Diabetes 38 , 1595–1601. and elevation of intracellular cAMP. Diabetes 51,
139 Tsung A, Geller DA (2011) Gross and cellular 2929–2935.
anatomy of the liver. In (Monga SPS. ed), pp. 3–6. 153 Dalziel B, Gosby AK, Richman RM, Bryson JM &
Molecular Pathology of Liver Diseases (MS, ed.). Caterson ID (2002) Association of the TNF-alpha -
Molecular Pathology Library, Springer, Molecular 308 G/A promoter polymorphism with insulin
Pathology Library Series, Boston, MA. resistance in obesity. Obes Res 10, 401–407.
140 Fausto N, Campbell JS & Riehle KJ (2006) Liver 154 Navarro-Gonzalez JF & Mora-Fernandez C (2008)
regeneration. Hepatology 43, S45–S53. The role of inflammatory cytokines in diabetic
141 Chalasani N, Younossi Z, Lavine JE, Charlton M, nephropathy. J Am Soc Nephrol 19, 433–442.
Cusi K, Rinella M, Harrison SA, Brunt EM & Sanyal 155 Kahn SE, Prigeon RL, Schwartz RS, Fujimoto WY,
AJ (2018) The diagnosis and management of Knopp RH, Brunzell JD et al. (2001) Obesity, body
nonalcoholic fatty liver disease: practice guidance from fat distribution, insulin sensitivity and Islet beta-cell
the American Association for the Study of Liver function as explanations for metabolic diversity.
Diseases. Hepatology 67, 328–357. J Nutr 131, 354S–S360.
142 
Gancheva S, Jelenik T, Alvarez-Hern andez E & 156 Engin AB (2017) What is lipotoxicity? Adv Exp Med
Roden M (2018) Interorgan metabolic crosstalk in Biol 960, 197–220.
human insulin resistance. Physiol Rev 98, 1371–1415. 157 Bodis K, Jelenik T, Lundbom J, Markgraf DF, Strom
143 Knebel B, Strassburger K, Szendroedi J, Kotzka J, A, Zaharia O-P, Karusheva Y, Burkart V, M€ ussig K,
Scheer M, Nowotny B, M€ ussig K, Lehr S, Pacini G, Kupriyanova Y et al. (2020) Expansion and impaired
Finner H et al. (2016) Specific metabolic profiles and mitochondrial efficiency of deep subcutaneous adipose
their relationship toinsulin resistance in recent-onset tissue in recent-onset type 2 diabetes. J Clin Endocrinol
type 1 and type 2 diabetes. J Clin Endocrinol Metab Metab 105, e1331–e1343.
101, 2130–2140. 158 Tumova J, Andel M & Trnka J (2016) Excess of free
144 Bonacina F, Baragetti A, Catapano AL & Norata GD fatty acids as a cause of metabolic dysfunction in
(2019) The interconnection between immuno- skeletal muscle. Physiol Res 65, 193–207.
metabolism, diabetes, and CKD. Curr Diab Rep 19, 21. 159 Bryner RW, Woodworth-Hobbs ME, Williamson DL
145 Capurso C & Capurso A (2012) From excess adiposity & Alway SE (2012) Docosahexaenoic acid protects
to insulin resistance: the role of free fatty acids. Vascul muscle cells from palmitate-induced atrophy. ISRN
Pharmacol 57, 91–97. Obes 2012, 647348.
146 Suganami T & Ogawa Y (2010) Adipose tissue 160 Ow JR, Caldez MJ, Zafer G, Foo JC, Li HY, Ghosh
macrophages: their role in adipose tissue remodeling. J S, Wollmann H, Cazenave-Gassiot A, Ong CB, Wenk
Leukoc Biol 88, 33–39. MR et al. (2020) Remodeling of whole-body lipid
147 Sharma RB & Alonso LC (2014) Lipotoxicity in the metabolism and a diabetic-like phenotype caused by
pancreatic beta cell: not just survival and function, but loss of CDK1 and hepatocyte division. eLife 9,
proliferation as well? Curr Diab Rep 14, 492. e63835.
148 Newsholme P, Keane D, Welters HJ & Morgan NG 161 Morita I, Tanimoto K, Akiyama N, Naya N, Fujieda
(2007) Life and death decisions of the pancreatic K, Iwasaki T & Yukioka H (2017) Chronic

642 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

hyperinsulinemia contributes to insulin resistance inhibiting glycogen synthesis. Diabetes 45,


under dietary restriction in association with altered 1396–1404.
lipid metabolism in Zucker diabetic fatty rats. Am J 175 Considine RV, Nyce MR, Allen LE, Morales LM,
Physiol Endocrinol Metab 312, E264–E272. Triester S, Serrano J, Colberg J, Lanza-Jacoby S &
162 Erion K & Corkey BE (2018) b-Cell failure or b-cell Caro JF (1995) Protein kinase C is increased in the
abuse? Front Endocrinol (Lausanne) 9, 532. liver of humans and rats with non-insulin-dependent
163 Goodpaster BH, He J, Watkins S & Kelley DE (2001) diabetes mellitus: an alteration not due to
Skeletal muscle lipid content and insulin resistance: hyperglycemia. J Clin Invest 95, 2938–2944.
evidence for a paradox in endurance-trained athletes. 176 Kishimoto A, Takai Y, Mori T, Kikkawa U &
J Clin Endocrinol Metab 86, 5755–5761. Nishizuka Y (1980) Activation of calcium and
164 Stefan N, Kantartzis K & H€aring HU (2008) Causes phospholipid-dependent protein kinase by
and metabolic consequences of Fatty liver. Endocr Rev diacylglycerol, its possible relation to
29, 939–960. phosphatidylinositol turnover. J Biol Chem 255,
165 Powell DJ, Turban S, Gray A, Hajduch E & Hundal 2273–2276.
HS (2004) Intracellular ceramide synthesis and protein 177 Gray S, Idris I, Davis KR & Donnelly R (2003)
kinase Czeta activation play an essential role in Increased skeletal muscle expression of PKC-theta but
palmitate-induced insulin resistance in rat L6 skeletal not PKC-alpha mRNA in type 2 diabetes: inverse
muscle cells. Biochem J 382, 619–629. relationship with in-vivo insulin sensitivity. Eur J Clin
166 Itani SI, Ruderman NB, Schmieder F & Boden G Invest 33, 983–987.
(2002) Lipid-induced insulin resistance in human 178 Park JW, Park WJ & Futerman AH (2014) Ceramide
muscle is associated with changes in diacylglycerol, synthases as potential targets for therapeutic
protein kinase C, and IkappaB-alpha. Diabetes 51, intervention in human diseases. Biochem Biophys Acta
2005–2011. 1841, 671–681.
167 Szendroedi J, Yoshimura T, Phielix E, Koliaki C, 179 Holland WL, Knotts TA, Chavez JA, Wang LP,
Marcucci M, Zhang D, Jelenik T, M€ uller J, Herder C, Hoehn KL & Summers SA (2007) Lipid mediators of
Nowotny P et al. (2014) Role of diacylglycerol insulin resistance. Nutr Rev 65, S39–S46.
activation of PKCh in lipid-induced muscle insulin 180 Pickersgill L, Litherland GJ, Greenberg AS, Walker M
resistance in humans. Proc Natl Acad Sci USA 111, & Yeaman SJ (2007) Key role for ceramides in
9597–9602. mediating insulin resistance in human muscle cells.
168 Erion DM & Shulman GI (2010) Diacylglycerol- J Biol Chem 282, 12583–12589.
mediated insulin resistance. Nat Med 16, 400–402. 181 JeBailey L, Wanono O, Niu W, Roessler J, Rudich A
169 Samuel VT, Petersen KF & Shulman GI (2010) Lipid- & Klip A (2007) Ceramide- and oxidant-induced
induced insulin resistance: unravelling the mechanism. insulin resistance involve loss of insulin-dependent
Lancet 375, 2267–2277. Rac-activation and actin remodeling in muscle cells.
170 Shulman GI (2014) Ectopic fat in insulin resistance, Diabetes 56, 394–403.
dyslipidemia, and cardiometabolic disease. New Engl J 182 Straczkowski M, Kowalska I, Baranowski M,
Med 371, 2237–2238. Nikolajuk A, Otziomek E, Zabielski P, Adamska A,
171 Boden G, Lebed B, Schatz M, Homko C & Lemieux S Blachnio A, Gorski J & Gorska M (2007) Increased
(2001) Effects of acute changes of plasma free fatty skeletal muscle ceramide level in men at risk of
acids on intramyocellular fat content and insulin developing type 2 diabetes. Diabetologia 50,
resistance in healthy subjects. Diabetes 50, 1612–1617. 2366–2373.
172 Jacobs S, Sahyoun NE, Saltiel AR & Cuatrecasas P 183 Summers SA, Garza LA, Zhou H & Birnbaum MJ
(1983) Phorbol esters stimulate the phosphorylation of (1998) Regulation of insulin-stimulated glucose
receptors for insulin and somatomedin C. Proc Natl transporter GLUT4 translocation and Akt kinase
Acad Sci USA 80, 6211–6213. activity by ceramide. Mol Cell Biol 18, 5457–5464.
173 Karasik A, Rothenberg PL, Yamada K, White MF & 184 Wang CN, O’Brien L & Brindley DN (1998) Effects of
Kahn CR (1990) Increased protein kinase C activity is cell-permeable ceramides and tumor necrosis factor-
linked to reduced insulin receptor autophosphorylation alpha on insulin signaling and glucose uptake in
in liver of starved rats. J Biol Chem 265, 10226–10231. 3T3-L1 adipocytes. Diabetes 47, 24–31.
174 Avignon A, Yamada K, Zhou X, Spencer B, Cardona 185 Holland WL, Miller RA, Wang ZV, Sun K, Barth
O, Saba-Siddique S, Galloway L, Standaert ML & BM, Bui HH, Davis KE, Bikman BT, Halberg N,
Farese RV (1996) Chronic activation of protein kinase Rutkowski JM et al. (2011) Receptor-mediated
C in soleus muscles and other tissues of insulin- activation of ceramidase activity initiates the
resistant type II diabetic Goto-Kakizaki (GK), obese/ pleiotropic actions of adiponectin. Nat Med 17,
aged, and obese/Zucker rats. A mechanism for 55–63.

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 643
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

186 Chavez JA & Summers SA (2012) A ceramide-centric Carmona F, Sanchez-Pla A, Salas-Salvad o J et al.
view of insulin resistance. Cell Metab 15, 585–594. (2019) Non-targeted metabolomic biomarkers and
187 Chavez JA, Knotts TA, Wang L-P, Li G, Dobrowsky metabotypes of type 2 diabetes: a cross-sectional study
RT, Florant GL & Summers SA (2003) A role for of PREDIMED trial participants. Diabetes Metab 45,
ceramide, but not diacylglycerol, in the antagonism of 167–174.
insulin signal transduction by saturated fatty acids. 200 Wu G, Fang YZ, Yang S, Lupton JR & Turner ND
J Biol Chem 278, 10297–10303. (2004) Glutathione metabolism and its implications for
188 Holland WL & Summers SA (2008) Sphingolipids, health. J Nutr 134, 489–492.
insulin resistance, and metabolic disease: new insights 201 Deykin D, Balko C & Kreisberg RA (1972) Glucose-
from in vivo manipulation of sphingolipid metabolism. lactate inter-relations in man. New Engl J Med 287,
Endocr Rev 29, 381–402. 132–137.
189 Teruel T, Hernandez R & Lorenzo M (2001) Ceramide 202 Avogaro A, Toffolo G, Miola M, Valerio A, Tiengo
mediates insulin resistance by tumor necrosis factor- A, Cobelli C & Del Prato S (1996) Intracellular
alpha in brown adipocytes by maintaining Akt in an lactate- and pyruvate-interconversion rates are
inactive dephosphorylated state. Diabetes 50, 2563–2571. increased in muscle tissue of non-insulin-dependent
190 Chaurasia B & Summers SA (2015) Ceramides – diabetic individuals. J Clin Invest 98, 108–115.
lipotoxic inducers of metabolic disorders. Trends 203 Pagel-Langenickel I, Bao J, Pang L & Sack MN
Endocrinol Metab 26, 538–550. (2010) The role of mitochondria in the
191 Stratford S, DeWald DB & Summers SA (2001) pathophysiology of skeletal muscle insulin resistance.
Ceramide dissociates 3’-phosphoinositide production Endocr Rev 31, 25–51.
from pleckstrin homology domain translocation. 204 Lovejoy J, Newby FD, Gebhart SS & DiGirolamo M
Biochem J 354, 359–368. (1992) Insulin resistance in obesity is associated with
192 Powell DJ, Hajduch E, Kular G & Hundal HS (2003) elevated basal lactate levels and diminished lactate
Ceramide disables 3-phosphoinositide binding to the appearance following intravenous glucose and insulin.
pleckstrin homology domain of protein kinase B Metabolism 41, 22–27.
(PKB)/Akt by a PKCzeta-dependent mechanism. Mol 205 Chen YD, Varasteh BB & Reaven GM (1993) Plasma
Cell Biol 23, 7794–7808. lactate concentration in obesity and type 2 diabetes.
193 Salinas M, L opez-Valdaliso R, Martın D, Alvarez A Diabete Metab 19, 348–354.
& Cuadrado A (2000) Inhibition of PKB/Akt1 by C2- 206 Robert JJ, Beaufrere B, Koziet J, Desjeux JF, Bier
ceramide involves activation of ceramide-activated DM, Young VR & Lestradet H (1985) Whole body de
protein phosphatase in PC12 cells. Mol Cell Neurosci novo amino acid synthesis in type I (insulin-
15, 156–169. dependent) diabetes studied with stable isotope-labeled
194 Hajduch E, Balendran A, Batty IH, Litherland GJ, leucine, alanine, and glycine. Diabetes 34, 67–73.
Blair AS, Downes CP & Hundal HS (2001) Ceramide 207 Talasniemi JP, Pennanen S, Savolainen H, Niskanen L
impairs the insulin-dependent membrane recruitment & Liesivuori J (2008) Analytical investigation: assay of
of protein kinase B leading to a loss in downstream D-lactate in diabetic plasma and urine. Clin Biochem
signalling in L6 skeletal muscle cells. Diabetologia 44, 41, 1099–1103.
173–183. 208 Connor H, Woods HF & Ledingham JG (1983)
195 Zhang Y, Li X, Becker KA & Gulbins E (2009) Comparison of the kinetics and utilisation of D(-)-and
Ceramide-enriched membrane domains structure and L(+)-sodium lactate in normal man. Ann Nutr Metab
function. Biochem Biophys Acta 1788, 178–183. 27, 481–487.
196 Cremesti A, Paris F, Grassme H, Holler N, Tschopp 209 Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A,
J, Fuks Z, Gulbins E & Kolesnick R (2001) Ceramide Deck LM, Royer RE, Vander Jagt DL, Semenza GL
enables fas to cap and kill. J Biol Chem 276, & Dang CV (2010) Inhibition of lactate dehydrogenase
23954–23961. A induces oxidative stress and inhibits tumor
197 Vandanmagsar B, Youm Y-H, Ravussin A, Galgani progression. Proc Natl Acad Sci USA 107, 2037–2042.
JE, Stadler K, Mynatt RL, Ravussin E, Stephens JM 210 Higuchi I, Kimura Y, Kobayashi M, Narumi K,
& Dixit VD (2011) The NLRP3 inflammasome Furugen A, Miyoshi H, Nakamura A, Yamada T,
instigates obesity-induced inflammation and insulin Atsumi T & Iseki K (2020) Relationships between
resistance. Nat Med 17, 179–188. plasma lactate, plasma alanine, genetic variations in
198 Albeituni S & Stiban J (2019) Roles of ceramides and lactate transporters and type 2 diabetes in the
other sphingolipids in immune cell function and Japanese population. Drug Metab Pharmacokinet 35,
inflammation. Adv Exp Med Biol 1161, 169–191. 131–138.
199 Urpi-Sarda M, Almanza-Aguilera E, Llorach R, 211 Snell K (1980) Muscle alanine synthesis and hepatic
Vazquez-Fresno R, Estruch R, Corella D, Sorli JV, gluconeogenesis. Biochem Soc Trans 8, 205–213.

644 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

212 Haymond MW, Karl IE, Clarke WL, Pagliara AS & functions. Proc Jpn Acad Ser B Phys Biol Sci 95,
Santiago JV (1982) Differences in circulating 246–260.
gluconeogenic substrates during short-term fasting in 224 Sener A, Conget I, Rasschaert J, Leclercq-Meyer V,
men, women, and children. Metabolism 31, 33–42. Villanueva-Penacarrillo ML, Valverde I & Malaisse
213 Caldez MJ, Van Hul N, Koh HWL, Teo XQ, Fan JJ, WJ (1994) Insulinotropic action of glutamic acid
Tan PY, Dewhurst MR, Too PG, Talib SZA, Chiang dimethyl ester. Am J Physio 267 (4 Pt 1), E573–E584.
BE et al. (2018) Metabolic remodeling during liver 225 Soni A, Amisten S, Rorsman P & Salehi A (2013)
regeneration. Dev Cell 47, 425–438. GPRC5B a putative glutamate-receptor candidate is
214 Gar C, Rottenkolber M, Sacco V, Moschko S, negative modulator of insulin secretion. Biochem
Banning F, Hesse N, Popp D, H€ ubener C, Seissler J Biophys Res Commun 441, 643–648.
& Lechner A (2018) Patterns of plasma glucagon 226 Feldmann N, del Rio RM, Gjinovci A, Tamarit-
dynamics do not match metabolic phenotypes in Rodriguez J, Wollheim CB & Wiederkehr A (2011)
young women. J Clin Endocrinol Metab 103, Reduction of plasma membrane glutamate transport
972–982. potentiates insulin but not glucagon secretion in
215 Locasale JW & Cantley LC (2011) Metabolic flux and pancreatic islet cells. Mol Cell Endocrinol 338, 46–57.
the regulation of mammalian cell growth. Cell Metab 227 Maechler P & Wollheim CB (1999) Mitochondrial
14, 443–451. glutamate acts as a messenger in glucose-induced
216 Just P-A, Charawi S, Denis RGP, Savall M, Traore insulin exocytosis. Nature 402, 685–689.
M, Foretz M, Bastu S, Magassa S, Senni N, Sohier P 228 Gheni G, Ogura M, Iwasaki M, Yokoi N, Minami K,
et al. (2020) Lkb1 suppresses amino acid-driven Nakayama Y, Harada K, Hastoy B, Wu X, Takahashi
gluconeogenesis in the liver. Nat Commun 11, 6127. H et al. (2014) Glutamate acts as a key signal linking
217 Okun JG, Rusu PM, Chan AY, Wu Y, Yap YW, glucose metabolism to incretin/cAMP action to
Sharkie T, Schumacher J, Schmidt KV, Roberts- amplify insulin secretion. Cell Rep 9, 661–673.
Thomson KM, Russell RD et al. (2021) Liver alanine 229 Casimir M, Lasorsa FM, Rubi B, Caille D, Palmieri
catabolism promotes skeletal muscle atrophy and F, Meda P & Maechler P (2009) Mitochondrial
hyperglycaemia in type 2 diabetes. Nat Metab 3, glutamate carrier GC1 as a newly identified player in
394–409. the control of glucose-stimulated insulin secretion.
218 Alecu I, Othman A, Penno A, Saied EM, Arenz C, J Biol Chem 284, 25004–25014.
von Eckardstein A & Hornemann T (2017) Cytotoxic 230 Siegel EG & Creutzfeldt W (1985) Stimulation of
1-deoxysphingolipids are metabolized by a cytochrome insulin release in isolated rat islets by GIP in
P450-dependent pathway. J Lipid Res 58, 60–71. physiological concentrations and its relation to islet
219 Galsgaard KD, Jepsen SL, Kjeldsen SAS, Pedersen J, cyclic AMP content. Diabetologia 28, 857–861.
Wewer Albrechtsen NJ & Holst JJ (2020) Alanine, 231 Holst JJ, Knop FK, Vilsbøll T, Krarup T & Madsbad
arginine, cysteine, and proline, but not glutamine, are S (2011) Loss of incretin effect is a specific, important,
substrates for, and acute mediators of, the liver-a-cell and early characteristic of type 2 diabetes. Diabetes
axis in female mice. Am J Physiol Endocrinol Metab Care 34 (Suppl 2), S251–S257.
318, E920–E929. 232 Huang X-T, Li C, Peng X-P, Guo J, Yue S-J, Liu W
220 Dean ED, Li M, Prasad N, Wisniewski SN, Von et al. (2017) An excessive increase in glutamate
Deylen A, Spaeth J, Maddison L, Botros A, Sedgeman contributes to glucose-toxicity in b-cells via activation
LR, Bozadjieva N et al. (2017) Interrupted glucagon of pancreatic NMDA receptors in rodent diabetes. Sci
signaling reveals hepatic a cell axis and role for Rep 7, 44120.
L-glutamine in a cell proliferation. Cell Metab 25, 233 Marquard J, Otter S, Welters A, Stirban A, Fischer A,
1362–1373. Eglinger J, Herebian D, Kletke O, Klemen MS, Stozer
221 Kim J, Okamoto H, Huang Z, Anguiano G, Chen S, A et al. (2015) Characterization of pancreatic NMDA
Liu Q, Cavino K, Xin Y, Na E, Hamid R et al. (2017) receptors as possible drug targets for diabetes
Amino acid transporter Slc38a5 controls glucagon treatment. Nat Med 21, 363–372.
receptor inhibition-induced pancreatic a cell 234 Welters A, Kl€ uppel C, Mrugala J, W€ ormeyer L,
hyperplasia in mice. Cell Metab 25, 1348–1361.e8. Meissner T, Mayatepek E et al. (2017) NMDAR
222 Longuet C, Robledo AM, Dean ED, Dai C, Ali S, antagonists for the treatment of diabetes mellitus-
McGuinness I et al. (2013) Liver-specific disruption of current status and future directions. Diabetes Obes
the murine glucagon receptor produces a-cell Metab 19 (Suppl 1), 95–106.
hyperplasia: evidence for a circulating a-cell growth 235 Chevalier S, Marliss EB, Morais JA, Lamarche M &
factor. Diabetes 62, 1196–1205. Gougeon R (2005) Whole-body protein anabolic
223 Takahashi H, Yokoi N & Seino S (2019) Glutamate as response is resistant to the action of insulin in obese
intracellular and extracellular signals in pancreatic islet women. Am J Clin Nutr 82, 3553–3565.

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 645
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

236 Hack V, St€ utz O, Kinscherf R, Schykowski M, 250 Lutchmansingh FK, Hsu JW, Bennett FI, Badaloo
Kellerer M, Holm E & Dr€ oge W (1996) Elevated AV, McFarlane-Anderson N, Gordon-Strachan GM,
venous glutamate levels in (pre)catabolic conditions Wright-Pascoe RA, Jahoor F & Boyne MS (2018)
result at least partly from a decreased glutamate Glutathione metabolism in type 2 diabetes and its
transport activity. J Mol Med 74, 337–343. relationship with microvascular complications and
237 Bao Y, Zhao T, Wang X, Qiu Y, Su M, Jia W & Jia glycemia. PLoS One 13, e0198626.
W (2009) Metabonomic variations in the drug-treated 251 Meidute Abaraviciene S, Lundquist I, Galvanovskis J,
type 2 diabetes mellitus patients and healthy Flodgren E, Olde B & Salehi A (2008) Palmitate-
volunteers. J Proteome Res 8, 1623–1630. induced beta-cell dysfunction is associated with
238 Nieuwdorp M, Stroes ES, Meijers JC & B€ uller H excessive NO production and is reversed by
(2005) Hypercoagulability in the metabolic syndrome. thiazolidinedione-mediated inhibition of GPR40
Curr Opin Pharmacol 5, 155–159. transduction mechanisms. PLoS One 3, e2182.
239 Davı G, Chiarelli F, Santilli F, Pomilio M, Vigneri S, 252 Furukawa S, Fujita T, Shimabukuro M, Iwaki M,
Falco A, Basili S, Ciabattoni G & Patrono C (2003) Yamada Y, Nakajima Y, Nakayama O, Makishima
Enhanced lipid peroxidation and platelet activation in M, Matsuda M & Shimomura I (2004) Increased
the early phase of type 1 diabetes mellitus: role of oxidative stress in obesity and its impact on metabolic
interleukin-6 and disease duration. Circulation 107, syndrome. J Clin Invest 114, 1752–1761.
3199–3203. 253 Halim M & Halim A (2019) The effects of
240 Hu H, Johansson BL, Hjemdahl P & Li N (2004) inflammation, aging and oxidative stress on the
Exercise-induced platelet and leucocyte activation is pathogenesis of diabetes mellitus (type 2 diabetes).
not enhanced in well-controlled type 1 diabetes, Diabetes Metab Syndr 13, 1165–1172.
despite increased activity at rest. Diabetologia 47, 853– 254 Gao Z, Hwang D, Bataille F, Lefevre M, York D,
859. Quon MJ et al. (2002) Serine phosphorylation of
241 Tolosa L, Caraballo-Miralles V, Olmos G & Llad oJ insulin receptor substrate 1 by inhibitor kappa B
(2011) TNF-a potentiates glutamate-induced spinal kinase complex. J Biol Chem 277, 48115–48121.
cord motoneuron death via NF-jB. Mol Cell Neurosci 255 Aguirre V, Uchida T, Yenush L, Davis R & White
46, 176–186. MF (2000) The c-Jun NH(2)-terminal kinase promotes
242 Lu SC (2000) Regulation of glutathione synthesis. insulin resistance during association with insulin
Curr Top Cell Regul 36, 95–116. receptor substrate-1 and phosphorylation of Ser(307).
243 Oppenheimer L, Wellner VP, Griffith OW & Meister J Biol Chem 275, 9047–9054.
A (1979) Glutathione synthetase. Purification from rat 256 Rui L, Aguirre V, Kim JK, Shulman GI, Lee A,
kidney and mapping of the substrate binding sites. J Corbould A et al. (2001) Insulin/IGF-1 and TNF-
Biol Chem 254, 5184–5190. alpha stimulate phosphorylation of IRS-1 at inhibitory
244 Yan N & Meister A (1990) Amino acid sequence of Ser307 via distinct pathways. J Clin Invest 107, 181–
rat kidney gamma-glutamylcysteine synthetase. J Biol 189.
Chem 265, 1588–1593. 257 Rollins BJ, Walz A & Baggiolini M (1991)
245 Ballatori N, Krance SM, Notenboom S, Shi S, Tieu K Recombinant human MCP-1/JE induces chemotaxis,
& Hammond CL (2009) Glutathione dysregulation calcium flux, and the respiratory burst in human
and the etiology and progression of human diseases. monocytes. Blood 78, 1112–1116.
Biol Chem 390, 191–214. 258 Al-Amily IM, Duner P, Groop L & Salehi A (2019)
246 Liu RM & Gaston Pravia KA (2010) Oxidative stress The functional impact of G protein-coupled receptor
and glutathione in TGF-beta-mediated fibrogenesis. 142 (Gpr142) on pancreatic b-cell in rodent. Pflugers
Free Radic Biol Med 48, 1–15. Archiv 471, 633–645.
247 Nguyen D, Hsu JW, Jahoor F & Sekhar RV (2014) 259 Boden G & Chen X (1995) Effects of fat on glucose
Effect of increasing glutathione with cysteine and uptake and utilization in patients with non-insulin-
glycine supplementation on mitochondrial fuel dependent diabetes. J Clin Invest 96, 1261–1268.
oxidation, insulin sensitivity, and body composition in 260 Dresner A, Laurent D, Marcucci M, Griffin ME,
older HIV-infected patients. J Clin Endocrinol Metab Dufour S, Cline GW, Slezak LA, Andersen DK,
99, 169–177. Hundal RS, Rothman DL et al. (1999) Effects of free
248 Caldez MJ, Bjorklund M & Kaldis P (2020) Cell cycle fatty acids on glucose transport and IRS-1-associated
regulation in NAFLD: when imbalanced metabolism phosphatidylinositol 3-kinase activity. J Clin Invest
limits cell division. Hepatol Int 14, 463–474. 103, 253–259.
249 Brownlee M (2001) Biochemistry and molecular cell 261 Guo W, Wong S, Xie W, Lei T & Luo Z (2007)
biology of diabetic complications. Nature 414, 813– Palmitate modulates intracellular signaling, induces
820. endoplasmic reticulum stress, and causes apoptosis in

646 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. M. Sanches et al. Metabolic interorgan crosstalk driving T2D

mouse 3T3-L1 and rat primary preadipocytes. Am J 273 Davis BK, Wen H & Ting JP (2011) The
Physiol Endocrinol Metab 293, E576–E586. inflammasome NLRs in immunity, inflammation, and
262 Wei Y, Wang D, Topczewski F & Pagliassotti MJ associated diseases. Annu Rev Immunol 29, 707–735.
(2006) Saturated fatty acids induce endoplasmic 274 Platnich JM & Muruve DA (2019) NOD-like receptors
reticulum stress and apoptosis independently of and inflammasomes: a review of their canonical and
ceramide in liver cells. Am J Physiol Endocrinol Metab non-canonical signaling pathways. Arch Biochem
291, E275–E281. Biophys 670, 4–14.
263 Karaskov E, Scott C, Zhang L, Teodoro T, Ravazzola 275 Kim YK, Shin JS & Nahm MH (2016) NOD-like
M & Volchuk A (2006) Chronic palmitate but not receptors in infection, immunity, and diseases. Yonsei
oleate exposure induces endoplasmic reticulum stress, Med J 57, 5–14.
which may contribute to INS-1 pancreatic beta-cell 276 Zasłona Z, Palsson-McDermott EM, Menon D,
apoptosis. Endocrinology 147, 3398–3407. Haneklaus M, Flis E, Prendeville H et al. (2017) The
264 Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, induction of pro-IL-1b by lipopolysaccharide requires
Harding HP & Ron D (2000) Coupling of stress in the endogenous prostaglandin E. J Immunol 198, 3558–3564.
ER to activation of JNK protein kinases by 277 Serhan CN & Savill J (2005) Resolution of
transmembrane protein kinase IRE1. Science 287, 664– inflammation: the beginning programs the end. Nat
666. Immunol 6, 1191–1197.
265 Arkan MC, Hevener AL, Greten FR, Maeda S, Li 278 von Moltke J, Trinidad NJ, Moayeri M, Kintzer AF,
ZW, Long JM et al. (2005) IKK-beta links Wang SB, van Rooijen N, Brown CR, Krantz BA,
inflammation to obesity-induced insulin resistance. Nat Leppla SH, Gronert K et al. (2012) Rapid induction
Med 11, 191–198. of inflammatory lipid mediators by the inflammasome
266 Cai D, Yuan M, Frantz DF, Melendez PA, Hansen in vivo. Nature 490, 107–111.
L, Lee J et al. (2005) Local and systemic insulin 279 Brash AR (2001) Arachidonic acid as a bioactive
resistance resulting from hepatic activation of molecule. J Clin Invest 107, 1339–1345.
IKK-beta and NF-kappaB. Nat Med 11, 280 Tallima H & El Ridi R (2018) Arachidonic acid:
183–190. physiological roles and potential health benefits – a
267 Sabio G, Das M, Mora A, Zhang Z, Jun JY, Ko HJ, review. J Adv Res 11, 33–41.
Barrett T, Kim JK & Davis RJ (2008) A stress 281 Dennis EA, Cao J, Hsu YH, Magrioti V & Kokotos
signaling pathway in adipose tissue regulates hepatic G (2011) Phospholipase A2 enzymes: physical
insulin resistance. Science 322, 1539–1543. structure, biological function, disease implication,
268 Tuncman G, Hirosumi J, Solinas G, Chang L, Karin chemical inhibition, and therapeutic intervention.
M & Hotamisligil GS (2006) Functional in vivo Chem Rev 111, 6130–6185.
interactions between JNK1 and JNK2 isoforms in 282 Ishimoto T, Akiba S, Sato T & Fujii T (1994)
obesity and insulin resistance. Proc Natl Acad Sci Contribution of phospholipases A2 and D to
USA 103, 10741–10746. arachidonic acid liberation and prostaglandin D2
269 Solinas G, Vilcu C, Neels JG, Bandyopadhyay GK, formation with increase in intracellular Ca2+
Luo J-L, Naugler W, Grivennikov S, Wynshaw-Boris concentration in rat peritoneal mast cells. Europ J
A, Scadeng M, Olefsky JM et al. (2007) JNK1 in Biochem 219, 401–406.
hematopoietically derived cells contributes to diet- 283 Ueda N, Tsuboi K & Uyama T (2013) Metabolism of
induced inflammation and insulin resistance without endocannabinoids and related N-acylethanolamines:
affecting obesity. Cell Metab 6, 386–397. canonical and alternative pathways. FEBS J 280,
270 Maedler K, Spinas GA, Lehmann R, Sergeev P, 1874–1894.
Weber M, Fontana A et al. (2001) Glucose induces 284 Kano M (2014) Control of synaptic function by
beta-cell apoptosis via upregulation of the Fas endocannabinoid-mediated retrograde signaling. Proc
receptor in human islets. Diabetes 50, 1683–1690. Jpn Acad Ser B Phys Biol Sci 90, 235–250.
271 Donath MY, Gross DJ, Cerasi E & Kaiser N (1999) 285 Brock TG & Peters-Golden M (2007) Activation and
Hyperglycemia-induced beta-cell apoptosis in regulation of cellular eicosanoid biosynthesis.
pancreatic islets of Psammomys obesus during ScientificWorldJournal 7, 1273–1284.
development of diabetes. Diabetes 48, 738–744. 286 Esser-von BJ (2019) Eicosanoids in tissue repair.
272 Schumann DM, Maedler K, Franklin I, Konrad D, Immunol Cell Biol 97, 279–288.
Storling J, Boni-Schnetzler M, Gjinovci A, Kurrer 287 Harizi H, Corcuff JB & Gualde N (2008) Arachidonic-
MO, Gauthier BR, Bosco D et al. (2007) The acid-derived eicosanoids: roles in biology and
Fas pathway is involved in pancreatic beta cell immunopathology. Trends Mol Med 14, 461–469.
secretory function. Proc Natl Acad Sci USA 104, 288 Sala A, Folco G & Murphy RC (2010) Transcellular
2861–2866. biosynthesis of eicosanoids. Pharmacol Rep 62, 503–510.

The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of 647
Federation of European Biochemical Societies
17424658, 2023, 3, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.16306 by Cochrane Colombia, Wiley Online Library on [23/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Metabolic interorgan crosstalk driving T2D J. M. Sanches et al.

289 Murphy JF, Steele C, Belton O & Fitzgerald DJ Western diet. Am J Physiol Endocrinol Metab 295,
(2003) Induction of cyclooxygenase-1 and -2 E1065–E1075.
modulates angiogenic responses to engagement of 301 Sun L, Liang L, Gao X, Zhang H, Yao P, Hu Y, Ma
alphavbeta3. Br J Haematol 121, 157–164. Y, Wang F, Jin Q, Li H et al. (2016) Early prediction
290 Robertson RP (1998) Dominance of cyclooxygenase-2 of developing type 2 diabetes by plasma acylcarnitines:
in the regulation of pancreatic islet prostaglandin a population-based study. Diabetes Care 39, 1563–
synthesis. Diabetes 47, 1379–1383. 1570.
291 Persaud SJ, Burns CJ, Belin VD & Jones PM (2004) 302 Choi CS, Savage DB, Abu-Elheiga L, Liu Z-X, Kim
Glucose-induced regulation of COX-2 expression in S, Kulkarni A, Distefano A, Hwang Y-J, Reznick
human islets of Langerhans. Diabetes 53, S190–S192. RM, Codella R et al. (2007) Continuous fat oxidation
292 Shanmugam N, Todorov IT, Nair I, Omori K, in acetyl-CoA carboxylase 2 knockout mice increases
Reddy MA & Natarajan R (2006) Increased total energy expenditure, reduces fat mass, and
expression of cyclooxygenase-2 in human pancreatic improves insulin sensitivity. Proc Natl Acad Sci USA
islets treated with high glucose or ligands of the 104, 16480–16485.
advanced glycation endproduct-specific receptor 303 Grapov D, Adams SH, Pedersen TL, Garvey WT &
(AGER), and in islets from diabetic mice. Newman JW (2012) Type 2 diabetes associated
Diabetologia 49, 100–107. changes in the plasma non-esterified fatty acids,
293 Amior L, Srivastava R, Nano R, Bertuzzi F & oxylipins and endocannabinoids. PLoS One 7, e48852.
Melloul D (2019) The role of Cox-2 and prostaglandin 304 Yi L, He J, Liang Y, Yuan D, Gao H & Zhou H
E. FASEB J 33, 4975–4986. (2007) Simultaneously quantitative measurement of
294 Mochizuki N & Kwon YG (2008) 15-lipoxygenase-1 in comprehensive profiles of esterified and non-esterified
the vasculature: expanding roles in angiogenesis. Circ fatty acid in plasma of type 2 diabetic patients. Chem
Res 102, 143–145. Phys Lipid 150, 204–216.
295 Decleves A-E, Mathew AV, Armando AM, Han X, 305 Liu L, Li Y, Guan C, Li K, Wang C, Feng R & Sun
Dennis EA, Quehenberger O & Sharma K (2019) C (2010) Free fatty acid metabolic profile and
AMP-activated protein kinase activation ameliorates biomarkers of isolated post-challenge diabetes and
eicosanoid dysregulation in high-fat-induced kidney type 2 diabetes mellitus based on GC-MS and
disease in mice. J Lipid Res 60, 937–952. multivariate statistical analysis. J Chromatogr B Analyt
296 Dobrian AD, Lieb DC, Cole BK, Taylor-Fishwick Technol Biomed Life Sci 878, 2817–2825.
DA, Chakrabarti SK & Nadler JL (2011) Functional 306 Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP,
and pathological roles of the 12- and 15-lipoxygenases. McCabe E, Lewis GD, Fox CS, Jacques PF,
Prog Lipid Res 50, 115–131. Fernandez C et al. (2011) Metabolite profiles and the
297 Natarajan R & Nadler JL (2004) Lipid inflammatory risk of developing diabetes. Nat Med 17, 448–453.
mediators in diabetic vascular disease. Arterioscler 307 Wang-Sattler R, Yu Z, Herder C, Messias AC, Floegel
Thromb Vasc Biol 24, 1542–1548. A, He Y, Heim K, Campillos M, Holzapfel C,
298 Patricia MK, Natarajan R, Dooley AN, Hernandez F, Thorand B et al. (2012) Novel biomarkers for pre-
Gu J-L, Berliner JA, Rossi JJ, Nadler JL, Meidell RS diabetes identified by metabolomics. Mol Syst Biol 8,
& Hedrick CC (2001) Adenoviral delivery of a 615.
leukocyte-type 12 lipoxygenase ribozyme inhibits 308 Oresic M, Simell S, Sysi-Aho M, N€ant€ o-Salonen K,
effects of glucose and platelet-derived growth factor in Sepp€anen-Laakso T, Parikka V, Katajamaa M,
vascular endothelial and smooth muscle cells. Circ Res Hekkala A, Mattila I, Keskinen P et al. (2008)
88, 659–665. Dysregulation of lipid and amino acid metabolism
299 Sears DD, Miles PD, Chapman J, Ofrecio JM, precedes islet autoimmunity in children who later
Almazan F, Thapar D & Miller YI (2009) 12/15- progress to type 1 diabetes. J Exp Med 205, 2975–
lipoxygenase is required for the early onset of high fat 2984.
diet-induced adipose tissue inflammation and insulin 309 Drogan D, Dunn WB, Lin W, Buijsse B, Schulze MB,
resistance in mice. PLoS One 4, e7250. Langenberg C, Brown M, Floegel A, Dietrich S,
300 Nunemaker CS, Chen M, Pei H, Kimble SD, Keller Rolandsson O et al. (2015) Untargeted metabolic
SR, Carter JD, Yang Z, Smith KM, Wu R, Bevard profiling identifies altered serum metabolites of type 2
MH et al. (2008) 12-Lipoxygenase-knockout mice are diabetes mellitus in a prospective, nested case control
resistant to inflammatory effects of obesity induced by study. Clin Chem 61, 487–497.

648 The FEBS Journal 290 (2023) 620–648 ª 2021 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of
Federation of European Biochemical Societies

You might also like