Diabetes Type 1 and Type 2 are widely occurring diseases. In spite of a vast amount of biomedical literature about diabetic processes in general, links to certain biological processes are only becoming evident these days. One such area of biology is the sulfation of small molecules, such as steroid hormones or metabolites from the gastrointestinal tract, as well as larger biomolecules, such as proteins and proteoglycans. Thus, modulating the physicochemical propensities of the different sulfate acceptors, resulting in enhanced solubility, expedited circulatory transit, or enhanced macromolecular interaction. This review lists evidence for the involvement of sulfation pathways in the maintenance of functional pancreatic beta-cell mass and the implications for diabetes, grouped into various classes of sulfated biomolecule. Complex heparan sulfates might play a role in the development and maintenance of beta-cells. The sulfolipids sulfatide and sulfo-cholesterol might contribute to beta-cell health. In beta-cells, there are only very few proteins with confirmed sulfation on some tyrosine residues, with the IRS4 molecule being one of them. Sulfated steroid hormones, such as estradiol-sulfate and vitamin-D-sulfate, may facilitate downstream steroid signaling in beta-cells, following de-sulfation. Indoxyl sulfate is a metabolite from the intestine, that causes kidney damage, contributing to diabetic kidney disease. Finally, from a technological perspective, there is heparan sulfate, heparin, and chondroitin sulfate, that all might be involved in next-generation beta-cell transplantation. Sulfation pathways may play a role in pancreatic beta-cells through multiple mechanisms. A more coherent understanding of sulfation pathways in diabetes will facilitate discussion and guide future research.

Type 1 and type 2 diabetes are complex disease entities, causing the morbidity and mortality of millions of people on our planet. These forms of diabetes are linked to deficient insulin production by the endocrine pancreas, leading to subsequent deregulation of glucose metabolism, and also lipid and protein metabolic pathways, manifesting as pathologies that affect multiple organs (extensively covered in [1]). The deficiency in insulin in diabetes could be caused by the lack of insulin-producing pancreatic β-cells – either by autoimmune destruction of said cells in type 1 diabetes or defective development of such cells, or by a decline in functional pancreatic β-cell mass caused by cell stress followed by cell death (as in Type 2 diabetes), where loss of function manifests as aberrant production and/or release of the hormone insulin from the β-cells. Thus, strategies for treatment of diabetes have generally been focused on increasing the sensitivity of target tissues to insulin, on increasing the function of existing/remaining pancreatic β-cells, or on replacement of β-cells.

A survey of the literature on publicly available databases showed that aberrations in sulfation pathways [2] have been variously linked to liver diseases (e.g. hepatocellular carcinoma, liver fibrosis, drug induced liver injury) [3], skeletal dysplasia, chondrodysplasias, and growth disorders [4], colitis and colonic carcinogenesis [5], migraines, autism [6]. The link between sulfation pathways and diabetes is a relatively recent one. Sulfation pathways are about making and breaking esters of sulfuric acid and various biomolecules [7]. They are the non-reducing branch of overall sulfur metabolism [8]; until recently an underexploited area of life-science research [9]. In all living organisms, sulfation pathways need sulfate in an activated form, and 3′-phospho-adenosine-5′-phospho-sulfate (PAPS) is the near-universal sulfuryl donor for cells to accomplish this task [10,11].

There are more than 50 sulfotransferase genes encoded in the human genome [7]. They all bind the PAPS cofactor in a similar manner, but they employ strikingly different molecular ways to recognize and bind a large variety of substrates [12,13]. Sulfation targets can be low-molecular weight compounds, such as steroids, other hormones, metabolites, or drugs. Interestingly, also lipids can be sulfated – a prominent sulfo-lipid is the galacto-cerebroside sulfatide [14], where the modification with sulfate has become eponymous. In proteins, tyrosine side chains of peptides and proteins can be sulfated, post-translationally. Recently, sulfation has been added to the list of possible modifications of histone proteins [15]; and PAPS synthases are known to shuttle into the cellular nucleus [16]. What remains is the sulfation of complex proteoglycans and glycosaminoglycans; high-molecular-weight protein-bound sugar chains, found predominantly in the extracellular matrix, or attached to extracellular domains of transmembrane proteins. In humans, chondroitin-sulfate, heparan-sulfate and heparin, to some extent, belong to this category.

The biosynthesis of PAPS is energy-demanding, and, hence, upregulated expression of the actual enzymes for PAPS synthesis – PAPS synthases PAPSS1 and 2 – may indicate active sulfation processes, where PAPS-dependent sulfotransferases add sulfate moieties to various biomolecules. Publicly available bulk RNA-sequencing datasets [17,18] suggest that the gene for 3′-phosphoadenosine 5′-phosphosulfate synthase 2, PAPSS2, in humans, is notably more expressed in pancreatic islets compared with hepatocytes.

Type 2 diabetes mellitus is one of several established risk factors for the development of pancreatic ductal adenocarcinoma [19]. Carbohydrate sulfotransferase CHST15 was first reported as a predictive marker for pancreatic ductal adenocarcinoma [20], linked with worse prognosis [21]. Reciprocally, silencing of CHST15 gene expression blocks tumor growth in a T-cell-dependent manner in mice [22,23]. Other carbohydrate sulfotransferases might be linked to different malignancies [24]. These findings show that targeting sulfation pathways may improve prognosis for these tumors by inducing an immunological effect.

This review will link different aspects of diabetes and glucose tolerance to a number of sulfated biomolecules. Some intersectional aspects of this review are summarized in Table 1. There is scope for sulfation pathways to play a role in pancreatic β-cell function and therefore to be linked with the pathogenesis of diabetes or regulation of insulin secretion. We hope that a more coherent understanding of sulfation pathways in diabetes will facilitate discussion, enabling us to ask the right questions for future research.

Table 1
Cross-sectional involvement of sulfation pathways in pancreatic β-cells
Research findingReference (s)
Pancreatic β-cell function and survival 
Heparin and heparan sulfate proteoglycans are involved in pancreatic β-cell function and survival [25,34,39
Cholesterol sulfate protects β-cells against apoptosis under stressful conditions, thus preserving β-cell mass [96
Estrogens regulate β-cell survival, function, and proliferation [98
Signaling 
High-affinity binding to heparan sulfates differentiates paracrine signaling from systemic signaling, in the case of FGF signaling [32,33
α-cells and β-cells have specific heparan-sulfate sulfation patterns impacting paracrine signaling [38
Insulin receptor substrate 4 (IRS4) is tyrosine-sulfated on Tyr921 IRS4 plays a role in insulin receptor signaling, in addition to IRS1 and IRS2, and may be A putative substrate for IGFR1, with anti-apoptotic effects [65,66,71
Insulin secretion 
Heparan sulfate 3-O-sulfotransferase isoform-1 involved in insulin secretion [37
Sulfate-removing enzyme iduronate 2-sulfatase IDS linked to insulin content in mouse islets, and lysosomal degradation of secretory peptides [46
Murine Tpst2 over-expression and impaired insulin secretion were linked in the pancreatic islets of hypothyroid mice [55
Microtubules impact on the glucose-regulated release of insulin containing vesicles in pancreatic β-cells, mediated by vesicular transport and exocytosis [72–76
β-cell development 
Heparan sulfate synthesis involved in β-cell development and maintenance of β-cell mass, with impact on glucose tolerance and insulin secretory capacity [40
Sulfation during pancreas development balances differentiation and proliferation [135
Heparan sulfate supports postnatal mouse islet proliferation and function [38,136
Microtubules contribute to pancreatic β-cell heterogeneity [77,78
β-cell pathophysiology 
Sulfated glycosaminoglycan, a substrate for the IDS enzyme, promotes the formation of amyloid from pro-islet amyloid polypeptide processing Intermediates in vitro, thus leading to loss of β-cell mass [48
Increased expression of vitamin-D-binding protein has been associated with dedifferentiation of β-cells and with β-cell dysfunction [120,121
Testosterone exposure during pregnancy caused defective β-cell programming in the offspring of various mammals [107–111
Metabolism 
Heparan sulfate glucosamine-6-O-endosulfatase-2 (Sulf2) in hepatocytes removes sulfate groups from syndecan-1-type heparan sulfate proteoglycans, thus reducing triglyceride-rich lipoprotein uptakes in cultured hepatocytes [49
Cholesterol sulfate regulates glucose metabolism, by inhibiting hepatic gluconeogenesis and lipogenesis, thus contributing to Type 2 diabetes [94,95
Autoimmune diabetes 
Heparan sulfates protect against autoimmune diabetes and immune cell infiltration [34,51
Heparan sulfates and other sulfated carbohydrates protect β-cells against an autoimmune response and help maintain a functional β-cell mass [52
Anti-inflammatory properties of the sulfatide C24:0 isoform protect β-cells from autoimmune attacks [90
Research findingReference (s)
Pancreatic β-cell function and survival 
Heparin and heparan sulfate proteoglycans are involved in pancreatic β-cell function and survival [25,34,39
Cholesterol sulfate protects β-cells against apoptosis under stressful conditions, thus preserving β-cell mass [96
Estrogens regulate β-cell survival, function, and proliferation [98
Signaling 
High-affinity binding to heparan sulfates differentiates paracrine signaling from systemic signaling, in the case of FGF signaling [32,33
α-cells and β-cells have specific heparan-sulfate sulfation patterns impacting paracrine signaling [38
Insulin receptor substrate 4 (IRS4) is tyrosine-sulfated on Tyr921 IRS4 plays a role in insulin receptor signaling, in addition to IRS1 and IRS2, and may be A putative substrate for IGFR1, with anti-apoptotic effects [65,66,71
Insulin secretion 
Heparan sulfate 3-O-sulfotransferase isoform-1 involved in insulin secretion [37
Sulfate-removing enzyme iduronate 2-sulfatase IDS linked to insulin content in mouse islets, and lysosomal degradation of secretory peptides [46
Murine Tpst2 over-expression and impaired insulin secretion were linked in the pancreatic islets of hypothyroid mice [55
Microtubules impact on the glucose-regulated release of insulin containing vesicles in pancreatic β-cells, mediated by vesicular transport and exocytosis [72–76
β-cell development 
Heparan sulfate synthesis involved in β-cell development and maintenance of β-cell mass, with impact on glucose tolerance and insulin secretory capacity [40
Sulfation during pancreas development balances differentiation and proliferation [135
Heparan sulfate supports postnatal mouse islet proliferation and function [38,136
Microtubules contribute to pancreatic β-cell heterogeneity [77,78
β-cell pathophysiology 
Sulfated glycosaminoglycan, a substrate for the IDS enzyme, promotes the formation of amyloid from pro-islet amyloid polypeptide processing Intermediates in vitro, thus leading to loss of β-cell mass [48
Increased expression of vitamin-D-binding protein has been associated with dedifferentiation of β-cells and with β-cell dysfunction [120,121
Testosterone exposure during pregnancy caused defective β-cell programming in the offspring of various mammals [107–111
Metabolism 
Heparan sulfate glucosamine-6-O-endosulfatase-2 (Sulf2) in hepatocytes removes sulfate groups from syndecan-1-type heparan sulfate proteoglycans, thus reducing triglyceride-rich lipoprotein uptakes in cultured hepatocytes [49
Cholesterol sulfate regulates glucose metabolism, by inhibiting hepatic gluconeogenesis and lipogenesis, thus contributing to Type 2 diabetes [94,95
Autoimmune diabetes 
Heparan sulfates protect against autoimmune diabetes and immune cell infiltration [34,51
Heparan sulfates and other sulfated carbohydrates protect β-cells against an autoimmune response and help maintain a functional β-cell mass [52
Anti-inflammatory properties of the sulfatide C24:0 isoform protect β-cells from autoimmune attacks [90

Heparan sulfates are structurally extremely diverse carbohydrate chains, that are bound to an extracellular or transmembrane protein. The main classes of heparan sulfate are syndecans and the membrane-anchored glypicans. Heparin shares many similarities in the composition of carbohydrates and in the sulfation patterns; heparins however are not protein-bound. Heparin and heparan sulfate proteoglycans, as well as enzymes that degrade heparan sulfates, have been shown to be important in the regulation of pancreatic β-cell function and survival (reviewed in [25]). Heparan sulfate proteoglycans consist of heterogeneous, highly negatively charged polysaccharides, tethered to proteins on the cell surface or in the extracellular matrix [26]. The structural diversity of heparan sulfates is massive – rivaling that of proteins [27]. Heparan sulfate diversity is created from different heparan modifying enzymes acting in additive or mutually exclusive ways [12], such as the heparan sulfate sulfotransferases targeting the 2-OH, 3-OH, or 6-OH hydroxyl groups. Thus, patterns are created in heparan sulfate that are referred to as sulfation code [28,29].

Sulfated proteoglycans are important for cell development anywhere in the human body. These molecules act like inter-cellular lubricants; they also modulate how cytokines and growth factors distribute within tissue [30]. A striking example of what heparan sulfates can do to a signaling pathway are fibroblast growth factors (FGFs) [31]. More than 20 different FGF molecules target four different FGF receptors (FGFRs), but different binding affinities toward heparan sulfates make some of them endocrine or systemic signaling compounds [32]. The others cause paracrine signaling [33], spatially confined by heparan sulfates.

In the context of the Islets of Langerhans, heparan sulfate was found to be expressed at high levels in mouse pancreatic islets [34], with heparan sulfate proteoglycans concentrated in the intracellular space of mouse β-cells [35,36]. The sulfation pathway gene heparan sulfate 3-O-sulfotransferase isoform-1 has been shown to be involved in insulin secretion [37]. Differential sulfation patterns have been observed between α- and β-cells [38]. These specific heparan-sulfate-sulfation-patterns are thought to be important for paracrine signaling between islets cell types [38], potentially important for appropriate control of blood glucose levels.

Loss of heparan sulfate during culture of mouse islets was associated with increased β-cell death, which was reversed by culture with heparin (a highly sulfated, soluble variant of heparan sulfate) [34,39]. Disruption of heparan sulfate synthesis in mice led to reduced β-cell mass, and reduced insulin secretory capacity [40], potentially through an impact on β-cell development and maturation, culminating in impaired glucose tolerance. In humans, genetic variation in the EXT2 gene, which encodes for exostosin glycosyltransferase 2, an enzyme involved in the heparan sulfate biosynthesis, is associated with increased risk for Type 2 diabetes [41,42] and impaired glucose clearance [43]. Additionally, loss-of-function mutations in the EXT2 gene, and the related EXT1 gene, was shown to be associated with lowered insulin secretion and impaired β-cell reserve in humans, as assessed by hyper-glycemic clamp and arginine challenge [44]. Thus, these genetic data indicate that heparan sulfates are required for the development and maintenance of functional β-cell mass in humans.

Iduronate 2-sulfatase (IDS) is a lysosomal enzyme involved in the degradation of proteoglycans. Interrogation of the single-cell RNA-seq dataset generated by Camunas-Soler and colleagues [45] indicated that expression of the IDS gene is lowered by approximately 30% (pAdjusted, 1.62 × 10−9) in β-cells from people living with Type 2 diabetes versus β-cells from individuals not living with Type 2 diabetes. Reduction of murine Ids expression was previously associated with reduced insulin content in mouse islets, whilst the secretory response to glucose was maintained [46]. Due to its localization in the lysosomes in pancreatic islets, it was hypothesized that Ids may be involved in the lysosomal degradation of secretory peptides. Ids expression in mouse pancreatic islets is regulated by glucose [46] and over-expression of Ids in INS1E cells, a rat insulinoma cell line, enhanced glucose-stimulated insulin secretion via a pathway dependent on protein kinase C alpha [47]. Increased levels of sulfated glycosaminoglycan, a substrate for IDS, increase the formation of amyloid from pro-islet amyloid polypeptide processing intermediates in vitro [48]. Amyloid deposition in pancreatic β-cells leads to loss of β-cell mass.

Genetically diabetic (db/db) mice overexpress the enzyme, heparan sulfate glucosamine-6-O-endosulfatase-2 (Sulf2) in hepatocytes, which may remove sulfate groups from syndecan-1-type heparan sulfate proteoglycans [49]. This in turn leads to suppression of the uptake of atherogenic postprandial triglyceride-rich lipoproteins in cultured hepatocytes [49]. Inhibition of Sulf2 in db/db mice using antisense oligonucleotides was shown to normalize the capacity of hepatocytes to bind very-low density lipoprotein and abolish postprandial hypertriglyceridemia [50]. Sulf2 protein content in mouse liver and in cultured hepatocytes is suppressed by adiponectin and insulin [49]. Thus, appropriate sulfation of cell surface proteo-glycans is linked to mechanisms that regulate energy flux.

Finally, heparan sulfates may play a role in protection against autoimmune diabetes, as autoimmune destruction of islets of non-obese diabetic mice and in streptozotocin-induced diabetic mice was prevented by inhibition of heparinase – stopping heparan sulfates to be degraded thereby reduced immune cell infiltration [34,51]. Thus, the evidence points to the involvement of heparan sulfate in protection of β-cells against an autoimmune response and maintenance of a functional β-cell mass [52], indicating a potential role in the pathophysiology of both Type 1 and 2 diabetes.

Tyrosine sulfation, a post-translational modification of proteins, is catalyzed by the tyrosyl-protein sulfotransferases TPST1 and TPST2 [7]. Tyrosine sulfation is a common modification found on secreted proteins as well as extracellular parts of transmembrane proteins [53]. In addition, intracellular protein sulfation was reported very recently for the very first time [15]. Tyrosine sulfation is important for protein-protein interactions and has been shown to be involved in the regulation of hemostasis, the inflammatory response and chemokine recognition [54].

TPST1 and 2 are expressed in pancreatic islets; in fact, both genes are among the highest-expressed sulfotransferases in β-cells, in publicly available datasets [17,18]. What is the evidence for a role for protein sulfation in the regulation of islet hormone production and action? Hypothyroid growth-retarded mice show impaired insulin secretion, and at the same time, murine Tpst2 is over-expressed in the pancreatic islets [55], suggesting a possible link between Tpst2 and insulin secretion. Indeed, increased sulfation was reported to contribute to atherosclerosis related to chronic kidney disease [56], a prevalent chronic complication in Type 2 diabetes.

Could it be that insulin itself is sulfated? There are a total of four tyrosine residues in human pre-pro-insulin, all of them end up in the 51-amino-acids-comprising mature insulin, highlighted in Figure 1. PubMed lists the search phrase ‘sulfated insulin’ in 16 publications, all before 1998; reaching back to a study in 1947 [57]. In the past, human insulin was an extreme rarity; instead, insulin from the pancreases of various animals, mainly pork and beef, was available for use in humans. Synthetically sulfated insulin was a type of formulation, believed to reduce antigenic responses in diabetic patients with immune insulin resistance [58]. Although there were some promising reports around sulfated insulin [59,60], the use of sulfated insulin ceased toward the late 1990s [61], as recombinant human insulin became readily available.

Short and long lasting insulin variants.

Figure 1
Short and long lasting insulin variants.

With regard to their molecular composition, established rapid-acting and long-acting insulins are compared with the newly introduced once-weekly insulin icodec. TyrA14Glu, TyrB16His, and PheB25His are mutations not contained in any of the commonly used insulins so far. The C14 acylation of detemir is myristic acid; the C16 acylation in degludec is hexadecanedioic acid; only the C20 acylation in icodec is more complex, two oligoethylene glycol units connect LysB29 to an oligoethylene icosanedioic diacid, via a gamma-glutamate moiety. Please note that detemir, degludec, and icodec all are ΔThrB30 insulins. Rapid and long insulins are presented according to [145], the insulin icodec molecule is described in [64].

Figure 1
Short and long lasting insulin variants.

With regard to their molecular composition, established rapid-acting and long-acting insulins are compared with the newly introduced once-weekly insulin icodec. TyrA14Glu, TyrB16His, and PheB25His are mutations not contained in any of the commonly used insulins so far. The C14 acylation of detemir is myristic acid; the C16 acylation in degludec is hexadecanedioic acid; only the C20 acylation in icodec is more complex, two oligoethylene glycol units connect LysB29 to an oligoethylene icosanedioic diacid, via a gamma-glutamate moiety. Please note that detemir, degludec, and icodec all are ΔThrB30 insulins. Rapid and long insulins are presented according to [145], the insulin icodec molecule is described in [64].

Close modal

Chemical sulfation of insulin mimics biological sulfation to a certain extent. Tyrosyl-protein sulfotransferase TPST1 and TPST2 enzymes modify proteins on selected tyrosine side chains [62], during the proteins transit through the Golgi network [53,63]. The TPST1 and TPST2 sulfating enzymes tend to recognize tyrosine residues surrounded by negatively charged residues, located in accessible and somewhat flexible parts of the protein, thus creating similar, but not overlapping tyrosine sulfation patterns.

Although no direct evidence for sulfation of native insulin can be found in the literature, a recently approved ultra-long-lasting version of insulin, weekly ‘icodec’, is noteworthy here. This insulin carries a long acyl chain at LysB29; additionally, icodec features three amino acid changes, TyrA14Glu, TyrB16His, and PheB25His, featured in Figure 1 [64]. These are unusual replacements, as they all affect aromatic residues. One in particular, TyrA14Glu, actually mimics the negatively charged side chain of tyrosine sulfate, enhancing solubility and slowing down pharmacokinetics.

Sulfation of other proteins may play a role in the regulation of β-cell function, supported by recent literature on mass-spectrometry surveys. Tyr921 of insulin receptor substrate 4 – IRS4 – was recently identified as a target for protein sulfation [65]; and IRS4 plays a role in insulin receptor signaling, in addition to IRS1 and IRS2. An alignment of this motif is shown in Figure 2. Alternatively, IRS4 is a putative substrate for IGFR1 [66]. Resistance to insulin and IGF1 in pancreatic β-cells has been shown to lead to compromised β-cell function [67–69]. Although the IRS1 and IRS2 are the major adaptor proteins involved in the transmission of the insulin signal (reviewed in [70]), over-expression of IRS4 has been shown to be protective against apoptosis in pancreatic β-cell lines lacking IRS2 [71].

Alignment of different insulin receptor substrates.

Figure 2
Alignment of different insulin receptor substrates.

Protein sequences NP_005535 for IRS1, NP_003740 for IRS2, and NP_003595 for IRS4 were aligned using the multiple sequence alignment tool Muscle [146]. Sulfated IRS4 tyrosine Y921 corresponds to Y896 in IRS1 and Y919 in IRS2. Negatively charged amino acids depicted in red, positively charged ones, in blue.

Figure 2
Alignment of different insulin receptor substrates.

Protein sequences NP_005535 for IRS1, NP_003740 for IRS2, and NP_003595 for IRS4 were aligned using the multiple sequence alignment tool Muscle [146]. Sulfated IRS4 tyrosine Y921 corresponds to Y896 in IRS1 and Y919 in IRS2. Negatively charged amino acids depicted in red, positively charged ones, in blue.

Close modal

Amongst the sulfated proteins identified by Daly and colleagues are also α- and β-tubulin, which are building blocks for microtubules. Microtubules are involved in vesicular transport and the regulation of exocytosis (reviewed in [72]); and this includes glucose-regulated release of insulin containing vesicles in pancreatic β-cells [73–76]. Microtubule also contribute to pancreatic β-cell heterogeneity [77,78].

Coming back to the link between protein sulfation and insulin secretion, the following sulfation pathway proteins have been identified in insulin-secretory-granules, in close proximity to pro-insulin and mature insulin [79]: Sulfated glycoprotein 1 precursor; arylsulfatase B; galactosamine (N-acetyl)-6-sulfate sulfatase; glucosamine (N-acetyl)-6-sulfatase. Arylsulfatase B and galactosamine (N-acetyl)-6-sulfate sulfatase are lysosomal enzymes involved in the breakdown of glycosaminoglycans. The expression of arylsulfatase B has been shown to be elevated in the liver of streptozotocin-induced diabetic rats [80]. This protein’s function, and likewise that of galactosamine (N-acetyl)-6-sulfate sulfatase, within the islet secretory vesicles is as yet unknown. However, they may, like the iduronate 2-sulfatase IDS, discussed above, be involved in the degradation of secretory peptidoglycan, thereby modulating β-cell function. Glucosamine (N-acetyl)-6-sulfatase is a lysosomal enzyme that is involved in the metabolism of heparin, heparan sulfate and keratan sulfate, and their involvement in the regulation of β-cell function and survival was discussed above. The sulfatide lipid is also present in exocytotic granules, as described further below.

Taken together, a number of sulfated proteins from within β-cells have been reported. With improved analytics, as recently shown [65], it is likely that more sulfated proteins will be reported from within islets.

Sulfation is important for the regulation of lipid metabolism and dysregulation of sulfation pathways have been shown to lead to dyslipidemia. Sulfated glycoprotein-1 is a saposin precursor (prosaposin); it is involved in the regulation of glycolipid metabolism, leading to ceramide production (reviewed in [81]. Ceramides are known to antagonize insulin action, thereby contributing to insulin resistance (reviewed in [82]). In the context of the pancreatic β-cell, elevated ceramide content is associated with increased β-cell apoptosis and defective insulin production and secretion (reviewed in [83]). Sulfation of ceramide heads is widespread in a special class of sulfated galacto-cerebroside, the sulfatides. Sulfatides are made by the cerebroside sulfotransferase GAL3ST1 and specifically degraded by arylsulfatase A [84]. Interestingly, arylsulfatase A was recently identified as regulator of sulfatide content and glycemic control [85], alongside the sphingolipid synthesizing enzyme hexosaminidase A [86].

Sulfatides can strongly influence the properties of the membranes they are part of [87]. Roeske-Nielsen and colleagues have identified a variation in the cerebroside sulfotransferase gene GAL3ST1 that is linked to exercise-modified insulin resistance and to Type 2 diabetes [88]. The same group also showed that sulfatide protects insulin-producing cells against cytokine-induced apoptosis [89]. A specific version of sulfatide, the C24:0 isoform, may have anti-inflammatory properties in the context of Type 1 diabetes, as it may protect β-cells from autoimmune attacks [90]. Finally, sulfatide might be a good addition to injectable commercial insulin formulations, because it strongly inhibits fibroblast growth [14], thus potentially reducing adverse fibroblast growth and improve well-being in patients with diabetes.

Cholesterol is a central component of nearly all eukaryotic membranes. This sterol mainly gets sulfated by the cytoplasmic sulfotransferase SULT2B1b [91] and de-sulfated by the steroid sulfatase STS [7,92]. Rare genetic mutations in the human X-linked STS steroid sulfatase gene lead to a scaling of the skin, due to accumulation of cholesterol sulfate [5]. Cholesterol sulfate more and more is recognized as an emerging regulator of lipid metabolism, inflammation, and cell fate [93].

SULT2B1b and cholesterol sulfate regulate glucose metabolism, as cholesterol sulfate inhibits hepatic gluconeogenesis [94] and lipogenesis [95], thus contributing to Type 2 diabetes [94]. Mechanistic studies in rodents suggest, however, that cholesterol sulfate protects β-cells against apoptosis under stressful conditions, thus preserving β-cell mass and protecting against the development of diabetes mellitus [96]. The same authors reported cholesterol sulfate levels to be elevated in bodily samples from patients with Type 1 or Type 2 diabetes [96]. The knockout of the mouse Slc10a6 transporter surprisingly resulted in elevated serum levels of cholesterol sulfate [97]. Studying the role of sodium-dependent organic anion transporters (SOATs) might bring interesting new insights for cholesterol sulfate in the future. Because of downstream steroidogenesis, where cholesterol is converted to highly potent steroid hormones, causing their very own effects, studying cholesterol sulfate in molecular detail may be associated with unique challenges.

The action of steroid hormones is regulated by their sulfation status [3]. There is evidence in the literature to indicate that sex steroids are involved in the regulation of pancreatic β-cell function and potential paracrine regulation (reviewed in [98]). The estrogen sulfotransferase SULT1E1 (also known as EST) catalyzes the sulfation of estrogens, temporarily inactivating these steroid hormones. SULT1E1 expression may be induced in inflammation-linked disease states, such as Type 2 diabetes, and some of its effects appear to be independent of estrogens [99], suggesting that other aromatic metabolites or steroids to be involved. The effects of the sulfation pathway gene SULT1E1 in energy homeostasis and insulin sensitivity may be sex- and tissue-specific [100,101].

Cells in the human islets of Langerhans express dedicated enzymes for the downstream conversion of steroid hormones [102,103], using sulfated steroid precursors from the circulation, such as DHEAS [3]. Estrogens in pancreatic β-cells have been shown to be involved in the regulation of β-cell survival, function, and proliferation in the context of both Type 1 and Type 2 diabetes, in rodent systems (reviewed in [98]). Activation of the androgen receptor [104] has been shown to augment insulin secretion in male [105], but to be deleterious to β-cell function in female rodent islets [106]. Offspring that had been exposed to high levels of testosterone during pregnancy showed defective β-cell programming in mice [107], sheep [108,109], and non-human primates [110,111]; findings that resemble what occurs in women with polycystic ovarian syndrome and their offsprings. It remains to be seen whether steroids currently under study, such as 11-oxo-androgens [5] or bis-sulfated steroid diols [112,113], play a role in the pancreatic β-cell as well.

The literature linking vitamin D and diabetes is vast (reviewed in [114,115]), with evidence that vitamin D is important for the development [116], survival [117], and function [118,119] of pancreatic β-cells. We have previously shown [120] that loss of expression of vitamin D-binding protein, which carries vitamin-D-metabolites in the circulation, in mice, leads to elevated basal glucagon secretion but a decrease in glucagon secretion in response to glucose in pancreatic α-cells, which is paralleled by increased insulin secretion in response to glucose challenge, and that this is due to dysregulation of the actin-network [120]. In the islet, vitamin D-binding protein is primarily expressed in pancreatic α-cells [120], but its expression in pancreatic β-cells is induced by glucose and increased expression has been associated with dedifferentiation of β-cells and with β-cell dysfunction [121].

Sulfated vitamin D has only recently come into the focus of clinical analytics [122,123], with sulfated conjugates of vitamin D representing a high proportion of the circulating vitamin D metabolome. Several vitamin D metabolites are sulfated primarily by the cytoplasmic sulfotransferase SULT2A1 enzyme [124] in liver [125]. We now know that vitamin D-sulfates are a significant part of the vitamin D-metabolome both in serum [122,126] and breast milk [123]. However, it remains to be seen whether the sulfation status of vitamin D would impact β-cell function and/or glucose tolerance.

The gut microbiota converts tryptophan from dietary protein to various metabolites. Indoxyl sulfate is one of the metabolites of tryptophan metabolism, produced by the intestinal bacterial flora and subsequent liver sulfation. Indoxyl sulfate has been mentioned many times in various metabolomics papers in recent years. Harlacher et al. linked indoxyl sulfate to an increase in inflammation, oxidative stress, leukocyte migration and adhesion, cell death and a thrombotic phenotype via the activation of the aryl-hydrocarbon-receptor [127]. Studies in human umbilical vein endothelial cells suggest that indoxyl sulfate may accelerate chronic kidney disease and drive vascular disease, by inducing oxidative stress and endothelial senescence [128]. In an animal model, indoxyl sulfate is taken up by proximal tubular cells through the organic anion transporters OAT1 and OAT3, and it induces reactive oxygen species (ROS) with impairment of cellular antioxidative system [128]. Via a number of signaling pathways, indoxyl sulfate induces nephro-vascular senescence [129].

In patients with Type 2 diabetes, indoxyl sulfate was linked to cardiovascular risk [130], and diabetic nephropathy [131]. In contrast with p-cresyl sulfate, indoxyl sulfate levels specifically correlate with glycated protein products in hemodialysis patients [132]; putatively increasing glucose toxicity and enhancing atherosclerosis (Figure 3). In patients with diabetes, urine indoxyl sulfate levels correlate with urinary markers of oxidative stress, suggesting a role as inducer of oxidative stress for indoxyl sulfate [133]. Studying sulfated metabolites, such as indoxyl sulfate, may reveal novel biomarkers to monitor disease progression. Furthermore, the elucidation of the signaling pathways around indoxyl sulfate toxicity may reveal new strategies for therapeutic intervention. Finally, even such simple biotechnological approaches as indole-absorbing matrices [134] for indoxyl sulfate removal might represent interesting avenues for future research and application.

Indoxyl-sulfate and related metabolites.

Figure 3
Indoxyl-sulfate and related metabolites.

Molecular structures of tryptophan (showing the indole side chain only), of indoxyl-sulfate, and of para-cresyl-sulfate. Although indoxyl-sulfate and para-cresyl-sulfate are both produced in the intestine, they have different downstream effects.

Figure 3
Indoxyl-sulfate and related metabolites.

Molecular structures of tryptophan (showing the indole side chain only), of indoxyl-sulfate, and of para-cresyl-sulfate. Although indoxyl-sulfate and para-cresyl-sulfate are both produced in the intestine, they have different downstream effects.

Close modal

The potential of sulfation pathways might be harnessed in enabling technologies which could lead to better treatment for diabetes, particularly Type 1 diabetes. For example, understanding the regulation of sulfation during the development of pancreatic islets may be important in our efforts to generate pancreatic β-cells from embryonic stem cells or pluripotent stem cells for transplantation. Regulation of sulfation has been shown to be important during the development of the endocrine pancreas: inhibition of sulfation was reported to increase the balance toward differentiation versus proliferation in rodent embryonic pancreas [135]. Heparan sulfate has also been reported to be important for regulation of postnatal mouse islet proliferation and function [38,136]. Harnessing such pathways could be beneficial for the development of protocols that lead to efficient production of pancreatic β-cells in cell culture.

One of the big issues with islet transplantation is hypoxia-induced cell death in islets of Langerhans whilst in culture prior to transplantation. It has previously been shown that the culture of human and mouse islets in heparin leads to decreased islet cell death whilst in culture [34,137,138]. Indeed, heparin is administered during the transplant process for graft protection and to prevent blood coagulation at the graft site (UK Guidelines on Pancreas and Islet Transplantation 2019 https://bts.org.uk/wp-content/uploads/2019/09/FINAL-Pancreas-guidelines-FINAL-version-following-consultation.-Sept-2019.pdf). Coating of the islets with heparin has been shown to prevent instant blood-mediated inflammatory reaction following transplant in pigs [139]. However, culture of human islets in heparin has been shown to lead to an increased deposition of human islet amyloid polypeptide, which would impair β-cell function, despite also preventing β-cell apoptosis [137].

Applications of sulfation pathways research might result in enabling technologies. This may encompass preservation of (intracellular) islet levels of heparan sulfate during the islet isolation process to optimize islet survival after transplantation [36]. Coating the pancreatic islet surface with a chondroitin sulfate-branched-PEG nanocoating may improve transplant success [140], a strategy also applicable to other types of cell therapies. 3D-bioprinted double-crosslinked alginate/chondroitin sulfate patches might improve diabetic wound healing [141,142]. Finally, chondroitin sulfate-coated gold nanoparticles might enable oral delivery of insulin [143,144], with the potential of revolutionizing diabetes treatment.

There is much evidence to suggest that sulfation pathways could be important in the maintenance of functional β-cell mass and regulation of β-cell function. This paper looks from the side of sulfation pathways on diabetes, discussing implications of heparan-sulfate and heparin, sulfatide and sulfo-cholesterol, various sulfated proteins, sulfo-steroid hormones and vitamin D-sulfate, as well as sulfated metabolites, on different types of diabetes and various aspects of β-cell physiology. A biotechnological section concludes this review, showing exciting new developments where sulfation pathways may be implemented in drug manufacturing and delivery, coated nanoparticles and advanced xeno-grafting and β-cell transplantation.

What else might be relevant for sulfation pathways in diabetes? More and more insulin variants become known, to put the evolution of insulin as a hormone into new perspectives, including viral insulin-like peptides. Certainly, it will be interesting to identify more and other sulfated proteins in β-cells and around them. Another route of research could involve tracing co-expression patterns of sulfation pathways genes in diabetes-related datasets. Finally, there are several molecules in sulfation pathways that await their biotechnical application, such as sulfatides for improving insulin injections.

Hopefully, a better understanding of the role of sulfation pathways in β-cells could lead to better understanding of the regulatory mechanisms involved, opening up new avenues to better treatment for diabetes.

  • More and more evidence suggest that sulfation pathways play a role in the maintenance of β-cell mass and regulation of β-cell function.

  • Sulfation pathways including heparan-sulfate and heparin, sulfatide and sulfo-cholesterol, various sulfated proteins, sulfo-steroid hormones and vitamin D-sulfate, as well as sulfated metabolites, may be implicated in different types of diabetes.

  • New developments in sulfation pathways research may be implemented in drug manufacturing and delivery, coated nanoparticles and advanced xeno-grafting and β-cell transplantation.

  • A better understanding of the role of sulfation pathways in β-cells could lead to insights into the regulatory mechanisms involved, opening up new avenues to better treatments of diabetes.

The authors declare that there are no competing interests associated with the manuscript.

FGF

fibroblast growth factor

IDS

iduronate 2-sulfatase

PAPS

3′-phospho-adenosine-5′-phospho-sulfate

ROS

reactive oxygen species

1.
Krentz
A.J.
(
2024
)
Insulin: deficiency, excess and resistance in human disease
.
Academic Press
London
2.
Brylski
O.
,
Shrestha
P.
,
House
P.J.
,
Gnutt
P.
,
Mueller
J.W.
and
Ebbinghaus
S.
(
2022
)
Disease-related protein variants of the highly conserved enzyme PAPSS2 show marginal stability and aggregation in cells
.
Front Mol. Biosci.
9
,
860387
[PubMed]
3.
Mueller
J.W.
,
Gilligan
L.C.
,
Idkowiak
J.
,
Arlt
W.
and
Foster
P.A.
(
2015
)
The regulation of steroid action by sulfation and desulfation
.
Endocr. Rev.
36
,
526
563
[PubMed]
4.
Oostdijk
W.
,
Idkowiak
J.
,
Mueller
J.W.
,
House
P.J.
,
Taylor
A.E.
,
O'Reilly
M.W.
et al.
(
2015
)
PAPSS2 deficiency causes androgen excess via impaired DHEA sulfation–in vitro and in vivo studies in a family harboring two novel PAPSS2 mutations
.
J. Clin. Endocrinol. Metab.
100
,
E672
E680
[PubMed]
5.
Foster
P.A.
and
Mueller
J.W.
(
2018
)
Sulfation pathways: insights into steroid sulfation and desulfation pathways
.
J. Mol. Endocrinol.
61
,
T271
T283
[PubMed]
6.
Davies
W.
(
2018
)
Sulfation pathways: the steroid sulfate axis and its relationship to maternal behaviour and mental health
.
J. Mol. Endocrinol.
61
,
T199
T210
[PubMed]
7.
Gunal
S.
,
Hardman
R.
,
Kopriva
S.
and
Mueller
J.W.
(
2019
)
Sulfation pathways from red to green
.
J. Biol. Chem.
294
,
12293
12312
[PubMed]
8.
McCully
K.S.
(
1972
)
Macromolecular basis for homocystein-induced changes in proteoglycan structure in growth and arteriosclerosis
.
Am. J. Pathol.
66
,
83
96
[PubMed]
9.
Leung
A.W.
,
Backstrom
I.
and
Bally
M.B.
(
2016
)
Sulfonation, an underexploited area: from skeletal development to infectious diseases and cancer
.
Oncotarget
7
,
55811
55827
[PubMed]
10.
Mueller
J.W.
and
Shafqat
N.
(
2013
)
Adenosine-5′-phosphosulfate–a multifaceted modulator of bifunctional 3′-phospho-adenosine-5′-phosphosulfate synthases and related enzymes
.
FEBS J.
280
,
3050
3057
[PubMed]
11.
Brylski
O.
,
Shrestha
P.
,
Gnutt
P.
,
Gnutt
D.
,
Mueller
J.W.
and
Ebbinghaus
S.
(
2021
)
Cellular ATP levels determine the stability of a nucleotide kinase
.
Front Mol. Biosci.
8
,
790304
[PubMed]
12.
Gesteira
T.F.
,
Marforio
T.D.
,
Mueller
J.W.
,
Calvaresi
M.
and
Coulson-Thomas
V.J.
(
2021
)
Structural determinants of substrate recognition and catalysis by heparan sulfate sulfotransferases
.
ACS Catal.
11
,
10974
10987
[PubMed]
13.
Hirschmann
F.
,
Krause
F.
,
Baruch
P.
,
Chizhov
I.
,
Mueller
J.W.
,
Manstein
D.J.
et al.
(
2017
)
Structural and biochemical studies of sulphotransferase 18 from Arabidopsis thaliana explain its substrate specificity and reaction mechanism
.
Sci. Rep.
7
,
4160
[PubMed]
14.
Roeske-Nielsen
A.
,
Mansson
J.E.
,
Tekin
H.
,
Rieneck
K.
,
Bendtzen
K.
and
Buschard
K.
(
2023
)
Sulfatide inhibits fibroblast growth, activation and oxidative stress induced by ectopic insulin
.
Diabetes Obes. Metab.
25
,
2514
2525
[PubMed]
15.
Yu
W.
,
Zhou
R.
,
Li
N.
,
Lei
Z.C.
,
Guo
D.
,
Peng
F.
et al.
(
2023
)
Histone tyrosine sulfation by SULT1B1 regulates H4R3me2a and gene transcription
.
Nat. Chem. Biol.
19
,
855
864
[PubMed]
16.
Schroder
E.
,
Gebel
L.
,
Eremeev
A.A.
,
Morgner
J.
,
Grum
D.
,
Knauer
S.K.
et al.
(
2012
)
Human PAPS synthase isoforms are dynamically regulated enzymes with access to nucleus and cytoplasm
.
PloS ONE
7
,
e29559
[PubMed]
17.
Diedisheim
M.
,
Oshima
M.
,
Albagli
O.
,
Huldt
C.W.
,
Ahlstedt
I.
,
Clausen
M.
et al.
(
2018
)
Modeling human pancreatic beta cell dedifferentiation
.
Mol. Metab.
10
,
74
86
[PubMed]
18.
Colli
M.L.
,
Ramos-Rodriguez
M.
,
Nakayasu
E.S.
,
Alvelos
M.I.
,
Lopes
M.
,
Hill
J.L.E.
et al.
(
2020
)
An integrated multi-omics approach identifies the landscape of interferon-alpha-mediated responses of human pancreatic beta cells
.
Nat. Commun.
11
,
2584
[PubMed]
19.
Dunne
R.F.
and
Roeland
E.J.
(
2022
)
The interplay among pancreatic cancer, cachexia, body composition, and diabetes
.
Hematol. Oncol. Clin. North Am.
36
,
897
910
[PubMed]
20.
Ito
Z.
,
Takakura
K.
,
Suka
M.
,
Kanai
T.
,
Saito
R.
,
Fujioka
S.
et al.
(
2017
)
Prognostic impact of carbohydrate sulfotransferase 15 in patients with pancreatic ductal adenocarcinoma
.
Oncol. Lett.
13
,
4799
4805
[PubMed]
21.
Matsuda
Y.
,
Fujii
Y.
,
Matsukawa
M.
,
Ishiwata
T.
,
Nishimura
M.
and
Arai
T.
(
2019
)
Overexpression of carbohydrate sulfotransferase 15 in pancreatic cancer stroma is associated with worse prognosis
.
Oncol. Lett.
18
,
4100
4105
[PubMed]
22.
Ye
J.
,
Suizu
F.
,
Yamakawa
K.
,
Mukai
Y.
,
Kato
M.
,
Yoneyama
H.
et al.
(
2023
)
Silencing of tumoral carbohydrate sulfotransferase 15 reactivates lymph node pancreatic cancer T cells in mice
.
Eur. J. Immunol.
53
,
e2250160
[PubMed]
23.
Ye
J.
,
Suizu
F.
,
Yamakawa
K.
,
Mukai
Y.
,
Yoneyama
H.
,
Kondo
J.
et al.
(
2024
)
Intra-tumoral administration of CHST15 siRNA remodels tumor microenvironment and augments tumor-infiltrating T cells in pancreatic cancer
.
Mol. Ther. Oncol.
32
,
200812
[PubMed]
24.
Begolli
G.
,
Markovic
I.
,
Knezevic
J.
and
Debeljak
Z.
(
2023
)
Carbohydrate sulfotransferases: a review of emerging diagnostic and prognostic applications
.
Biochem. Med. (Zagreb)
33
,
030503
[PubMed]
25.
Takahashi
I.
(
2022
)
Importance of heparan sulfate proteoglycans in pancreatic islets and beta-cells
.
Int. J. Mol. Sci.
23
,
[PubMed]
26.
Hiebert
L.M.
(
2021
)
Heparan sulfate proteoglycans in diabetes
.
Semin. Thromb. Hemost.
47
,
261
273
[PubMed]
27.
Alotaibi
F.S.
,
Alsadun
M.M.R.
,
Alsaiari
S.A.
,
Ramakrishnan
K.
,
Yates
E.A.
and
Fernig
D.G.
(
2024
)
Interactions of proteins with heparan sulfate
.
Essays Biochem.
68
,
[PubMed]
28.
Holmes
S.G.
,
Nagarajan
B.
and
Desai
U.R.
(
2022
)
3-O-Sulfation induces sequence-specific compact topologies in heparan sulfate that encode a dynamic sulfation code
.
Comput. Struct. Biotechnol. J.
20
,
3884
3898
[PubMed]
29.
Shanthamurthy
C.D.
,
Gimeno
A.
,
Leviatan Ben-Arye
S.
,
Kumar
N.V.
,
Jain
P.
,
Padler-Karavani
V.
et al.
(
2021
)
Sulfation code and conformational plasticity of l-iduronic acid homo-oligosaccharides mimic the biological functions of heparan sulfate
.
ACS Chem. Biol.
16
,
2481
2489
[PubMed]
30.
Bandari
S.
,
Exner
S.
,
Ortmann
C.
,
Bachvarova
V.
,
Vortkamp
A.
and
Grobe
K.
(
2015
)
Sweet on Hedgehogs: regulatory roles of heparan sulfate proteoglycans in Hedgehog-dependent cell proliferation and differentiation
.
Curr. Protein Pept. Sci.
16
,
66
76
[PubMed]
31.
Kyrou
I.
,
Weickert
M.O.
,
Gharanei
S.
,
Randeva
H.S.
and
Tan
B.K.
(
2017
)
Fibroblast growth factors: new insights, new targets in the management of diabetes
.
Minerva Endocrinol.
42
,
248
270
[PubMed]
32.
Ghanem
M.
,
Archer
G.
,
Crestani
B.
and
Mailleux
A.A.
(
2024
)
The endocrine FGFs axis: a systemic anti-fibrotic response that could prevent pulmonary fibrogenesis?
Pharmacol. Ther.
259
,
108669
[PubMed]
33.
Nagai
N.
,
Habuchi
H.
,
Sugaya
N.
,
Nakamura
M.
,
Imamura
T.
,
Watanabe
H.
et al.
(
2013
)
Involvement of heparan sulfate 6-O-sulfation in the regulation of energy metabolism and the alteration of thyroid hormone levels in male mice
.
Glycobiology
23
,
980
992
[PubMed]
34.
Ziolkowski
A.F.
,
Popp
S.K.
,
Freeman
C.
,
Parish
C.R.
and
Simeonovic
C.J.
(
2012
)
Heparan sulfate and heparanase play key roles in mouse beta cell survival and autoimmune diabetes
.
J. Clin. Invest.
122
,
132
141
[PubMed]
35.
Bogdani
M.
,
Korpos
E.
,
Simeonovic
C.J.
,
Parish
C.R.
,
Sorokin
L.
and
Wight
T.N.
(
2014
)
Extracellular matrix components in the pathogenesis of type 1 diabetes
.
Curr. Diab. Rep.
14
,
552
[PubMed]
36.
Choong
F.J.
,
Freeman
C.
,
Parish
C.R.
and
Simeonovic
C.J.
(
2015
)
Islet heparan sulfate but not heparan sulfate proteoglycan core protein is lost during islet isolation and undergoes recovery post-islet transplantation
.
Am. J. Transplant.
15
,
2851
2864
[PubMed]
37.
Takahashi
I.
,
Ohashi
K.
and
Nata
K.
(
2012
)
Involvement of heparan sulfate 3-O-sulfotransferase isoform-1 in the insulin secretion pathway
.
J. Diab. Investig.
3
,
362
370
38.
Theodoraki
A.
,
Hu
Y.
,
Poopalasundaram
S.
,
Oosterhof
A.
,
Guimond
S.E.
,
Disterer
P.
et al.
(
2015
)
Distinct patterns of heparan sulphate in pancreatic islets suggest novel roles in paracrine islet regulation
.
Mol. Cell. Endocrinol.
399
,
296
310
[PubMed]
39.
Dhounchak
S.
,
Popp
S.K.
,
Brown
D.J.
,
Laybutt
D.R.
,
Biden
T.J.
,
Bornstein
S.R.
et al.
(
2021
)
Heparan sulfate proteoglycans in beta cells provide a critical link between endoplasmic reticulum stress, oxidative stress and type 2 diabetes
.
PloS ONE
16
,
e0252607
[PubMed]
40.
Matsuzawa
T.
,
Yoshikawa
T.
,
Iida
T.
,
Karpati
A.
,
Kitano
H.
,
Harada
R.
et al.
(
2018
)
Heparan sulfate in pancreatic beta-cells contributes to normal glucose homeostasis by regulating insulin secretion
.
Biochem. Biophys. Res. Commun.
499
,
688
695
[PubMed]
41.
Sladek
R.
,
Rocheleau
G.
,
Rung
J.
,
Dina
C.
,
Shen
L.
,
Serre
D.
et al.
(
2007
)
A genome-wide association study identifies novel risk loci for type 2 diabetes
.
Nature
445
,
881
885
[PubMed]
42.
Liu
L.
,
Yang
X.
,
Wang
H.
,
Cui
G.
,
Xu
Y.
,
Wang
P.
et al.
(
2013
)
Association between variants of EXT2 and type 2 diabetes: a replication and meta-analysis
.
Hum. Genet.
132
,
139
145
[PubMed]
43.
Rong
R.
,
Hanson
R.L.
,
Ortiz
D.
,
Wiedrich
C.
,
Kobes
S.
,
Knowler
W.C.
et al.
(
2009
)
Association analysis of variation in/near FTO, CDKAL1, SLC30A8, HHEX, EXT2, IGF2BP2, LOC387761, and CDKN2B with type 2 diabetes and related quantitative traits in Pima Indians
.
Diabetes
58
,
478
488
[PubMed]
44.
Bernelot Moens
S.J.
,
Mooij
H.L.
,
Hassing
H.C.
,
Kruit
J.K.
,
Witjes
J.J.
,
van de Sande
M.A.J.
et al.
(
2014
)
Carriers of loss-of-function mutations in EXT display impaired pancreatic beta-cell reserve due to smaller pancreas volume
.
PloS ONE
9
,
e115662
[PubMed]
45.
Camunas-Soler
J.
,
Dai
X.Q.
,
Hang
Y.
,
Bautista
A.
,
Lyon
J.
,
Suzuki
K.
et al.
(
2020
)
Patch-Seq links single-cell transcriptomes to human islet dysfunction in diabetes
.
Cell Metab.
31
,
1017e1014
1031e1014
46.
Coronado-Pons
I.
,
Novials
A.
,
Casas
S.
,
Clark
A.
and
Gomis
R.
(
2004
)
Identification of iduronate-2-sulfatase in mouse pancreatic islets
.
Am. J. Physiol. Endocrinol. Metab.
287
,
E983
E990
[PubMed]
47.
Piquer
S.
,
Casas
S.
,
Quesada
I.
,
Nadal
A.
,
Julia
M.
,
Novials
A.
et al.
(
2009
)
Role of iduronate-2-sulfatase in glucose-stimulated insulin secretion by activation of exocytosis
.
Am. J. Physiol. Endocrinol. Metab.
297
,
E793
E801
[PubMed]
48.
Wang
H.
and
Raleigh
D.P.
(
2014
)
The ability of insulin to inhibit the formation of amyloid by pro-islet amyloid polypeptide processing intermediates is significantly reduced in the presence of sulfated glycosaminoglycans
.
Biochemistry
53
,
2605
2614
[PubMed]
49.
Chen
K.
,
Liu
M.L.
,
Schaffer
L.
,
Li
M.
,
Boden
G.
,
Wu
X.
et al.
(
2010
)
Type 2 diabetes in mice induces hepatic overexpression of sulfatase 2, a novel factor that suppresses uptake of remnant lipoproteins
.
Hepatology
52
,
1957
1967
[PubMed]
50.
Hassing
H.C.
,
Mooij
H.
,
Guo
S.
,
Monia
B.P.
,
Chen
K.
,
Kulik
W.
et al.
(
2012
)
Inhibition of hepatic sulfatase-2 in vivo: a novel strategy to correct diabetic dyslipidemia
.
Hepatology
55
,
1746
1753
[PubMed]
51.
Song
W.Y.
,
Jiang
X.H.
,
Ding
Y.
,
Wang
Y.
,
Zhou
M.X.
,
Xia
Y.
et al.
(
2020
)
Inhibition of heparanase protects against pancreatic beta cell death in streptozotocin-induced diabetic mice via reducing intra-islet inflammatory cell infiltration
.
Br. J. Pharmacol.
177
,
4433
4447
[PubMed]
52.
Bogdani
M.
,
Speake
C.
,
Dufort
M.J.
,
Johnson
P.Y.
,
Larmore
M.J.
,
Day
A.J.
et al.
(
2020
)
Hyaluronan deposition in islets may precede and direct the location of islet immune-cell infiltrates
.
Diabetologia
63
,
549
560
[PubMed]
53.
Stewart
V.
and
Ronald
P.C.
(
2022
)
Sulfotyrosine residues: Interaction specificity determinants for extracellular protein-protein interactions
.
J. Biol. Chem.
298
,
102232
[PubMed]
54.
Sasaki
N.
(
2012
)
Current status and future prospects for research on tyrosine sulfation
.
Curr. Pharm. Biotechnol.
13
,
2632
2641
[PubMed]
55.
Taguchi
Y.
,
Tasaki
Y.
,
Terakado
K.
,
Kobayashi
K.
,
Machida
T.
and
Kobayashi
T.
(
2010
)
Impaired insulin secretion from the pancreatic islets of hypothyroidal growth-retarded mice
.
J. Endocrinol.
206
,
195
204
[PubMed]
56.
Dai
D.
,
Zhu
Z.
,
Han
H.
,
Xu
T.
,
Feng
S.
,
Zhang
W.
et al.
(
2023
)
Enhanced tyrosine sulfation is associated with chronic kidney disease-related atherosclerosis
.
BMC Biol.
21
,
151
[PubMed]
57.
Glendening
M.B.
,
Greenberg
D.M.
and
Fraenkel-Conrat
H.
(
1947
)
Biologically active insulin sulfate
.
J. Biol. Chem.
167
,
125
128
[PubMed]
58.
Davidson
J.K.
,
Fineberg
S.E.
,
De Meyts
P.
,
Fineberg
N.S.
and
Galloway
J.A.
(
1992
)
Immunological and metabolic responses of patients with history of antibody-induced beef insulin resistance to treatment with beef, pork, human, and sulfated beef insulin
.
Diabetes Care
15
,
702
704
[PubMed]
59.
Naquet
P.
,
Ellis
J.
,
Kenshole
A.
,
Semple
J.W.
and
Delovitch
T.L.
(
1989
)
Sulfated beef insulin treatment elicits CD8+ T cells that may abrogate immunologic insulin resistance in type I diabetes
.
J. Clin. Invest.
84
,
1479
1487
[PubMed]
60.
Davidson
M.B.
,
Kumar
D.
and
Smith
W.
(
1991
)
Successful treatment of unusual case of brittle diabetes with sulfated beef insulin
.
Diabetes Care
14
,
1109
1110
[PubMed]
61.
Carpentier
A.
,
Wither
J.
,
Vukusic
B.
,
Lawday
K.
,
Boss
A.H.
and
Lewis
G.F.
(
1998
)
An epitaph for sulfated insulin: immunologic profile of the last patients as they are switched from sulfated beef to human insulin
.
Diabetes Care
21
,
1571
1572
[PubMed]
62.
Liu
C.C.
(
2020
)
A new path to tyrosine sulfation
.
Nat. Chem. Biol.
16
,
365
366
[PubMed]
63.
Goettsch
S.
,
Badea
R.A.
,
Mueller
J.W.
,
Wotzlaw
C.
,
Schoelermann
B.
,
Schulz
L.
et al.
(
2006
)
Human TPST1 transmembrane domain triggers enzyme dimerisation and localisation to the Golgi compartment
.
J. Mol. Biol.
361
,
436
449
[PubMed]
64.
Nishimura
E.
,
Pridal
L.
,
Glendorf
T.
,
Hansen
B.F.
,
Hubalek
F.
,
Kjeldsen
T.
et al.
(
2021
)
Molecular and pharmacological characterization of insulin icodec: a new basal insulin analog designed for once-weekly dosing
.
BMJ Open Diab. Res. Care
9
,
e002301
[PubMed]
65.
Daly
L.A.
,
Byrne
D.P.
,
Perkins
S.
,
Brownridge
P.J.
,
McDonnell
E.
,
Jones
A.R.
et al.
(
2023
)
Custom workflow for the confident identification of sulfotyrosine-containing peptides and their discrimination from phosphopeptides
.
J. Proteome Res.
22
,
3754
3772
[PubMed]
66.
Wauman
J.
,
De Smet
A.S.
,
Catteeuw
D.
,
Belsham
D.
and
Tavernier
J.
(
2008
)
Insulin receptor substrate 4 couples the leptin receptor to multiple signaling pathways
.
Mol. Endocrinol.
22
,
965
977
[PubMed]
67.
Kulkarni
R.N.
,
Bruning
J.C.
,
Winnay
J.N.
,
Postic
C.
,
Magnuson
M.A.
and
Kahn
C.R.
(
1999
)
Tissue-specific knockout of the insulin receptor in pancreatic beta cells creates an insulin secretory defect similar to that in type 2 diabetes
.
Cell
96
,
329
339
[PubMed]
68.
Ueki
K.
,
Okada
T.
,
Hu
J.
,
Liew
C.W.
,
Assmann
A.
,
Dahlgren
G.M.
et al.
(
2006
)
Total insulin and IGF-I resistance in pancreatic beta cells causes overt diabetes
.
Nat. Genet.
38
,
583
588
[PubMed]
69.
Goldfine
A.B.
and
Kulkarni
R.N.
(
2012
)
Modulation of beta-cell function: a translational journey from the bench to the bedside
.
Diabetes Obes. Metab.
14
,
152
160
[PubMed]
70.
Kulkarni
R.N.
(
2007
)
Insulin Action in the Islet β-Cell
. In
Mechanisms of Insulin Action: Medical Intelligence Unit
(
Saltiel
A.R.
and
Pessin
J.E.
, eds), pp.
133
151
,
Springer New York
,
New York, NY
71.
Lingohr
M.K.
,
Dickson
L.M.
,
Wrede
C.E.
,
Briaud
I.
,
McCuaig
J.F.
,
Myers
M.G.
Jr.
et al.
(
2003
)
Decreasing IRS-2 expression in pancreatic beta-cells (INS-1) promotes apoptosis, which can be compensated for by introduction of IRS-4 expression
.
Mol. Cell. Endocrinol.
209
,
17
31
[PubMed]
72.
Fourriere
L.
,
Jimenez
A.J.
,
Perez
F.
and
Boncompain
G.
(
2020
)
The role of microtubules in secretory protein transport
.
J. Cell Sci.
133
,
jcs237016
[PubMed]
73.
Bracey
K.M.
,
Gu
G.
and
Kaverina
I.
(
2022
)
Microtubules in pancreatic beta cells: convoluted roadways toward precision
.
Front Cell Dev. Biol.
10
,
915206
[PubMed]
74.
Ho
K.H.
,
Yang
X.
,
Osipovich
A.B.
,
Cabrera
O.
,
Hayashi
M.L.
,
Magnuson
M.A.
et al.
(
2020
)
Glucose regulates microtubule disassembly and the dose of insulin secretion via tau phosphorylation
.
Diabetes
69
,
1936
1947
[PubMed]
75.
Trogden
K.P.
,
Zhu
X.
,
Lee
J.S.
,
Wright
C.V.E.
,
Gu
G.
and
Kaverina
I.
(
2019
)
Regulation of glucose-dependent golgi-derived microtubules by cAMP/EPAC2 promotes secretory vesicle biogenesis in pancreatic beta cells
.
Curr. Biol.
29
,
2339e2335
2350e2335
76.
Smith
R.
,
Bacos
K.
,
Fedele
V.
,
Soulet
D.
,
Walz
H.A.
,
Obermuller
S.
et al.
(
2009
)
Mutant huntingtin interacts with beta-tubulin and disrupts vesicular transport and insulin secretion
.
Hum. Mol. Genet.
18
,
3942
3954
[PubMed]
77.
Trogden
K.P.
,
Lee
J.
,
Bracey
K.M.
,
Ho
K.H.
,
McKinney
H.
,
Zhu
X.
et al.
(
2021
)
Microtubules regulate pancreatic beta-cell heterogeneity via spatiotemporal control of insulin secretion hot spots
.
Elife
10
,
e59912
[PubMed]
78.
Da Silva Xavier
G.
and
Rutter
G.A.
(
2020
)
Metabolic and functional heterogeneity in pancreatic beta cells
.
J. Mol. Biol.
432
,
1395
1406
[PubMed]
79.
Brunner
Y.
,
Coute
Y.
,
Iezzi
M.
,
Foti
M.
,
Fukuda
M.
,
Hochstrasser
D.F.
et al.
(
2007
)
Proteomics analysis of insulin secretory granules
.
Mol. Cell. Proteomics
6
,
1007
1017
[PubMed]
80.
Samarji
R.
and
Balbaa
M.
(
2014
)
Anti-diabetic activity of different oils through their effect on arylsulfatases
.
J. Diab. Metab. Disord.
13
,
116
[PubMed]
81.
Darmoise
A.
,
Maschmeyer
P.
and
Winau
F.
(
2010
)
The immunological functions of saposins
.
Adv. Immunol.
105
,
25
62
[PubMed]
82.
Sokolowska
E.
and
Blachnio-Zabielska
A.
(
2019
)
The role of ceramides in insulin resistance
.
Front Endocrinol. (Lausanne)
10
,
577
[PubMed]
83.
Boslem
E.
,
Meikle
P.J.
and
Biden
T.J.
(
2012
)
Roles of ceramide and sphingolipids in pancreatic beta-cell function and dysfunction
.
Islets
4
,
177
187
[PubMed]
84.
Takahashi
T.
and
Suzuki
T.
(
2012
)
Role of sulfatide in normal and pathological cells and tissues
.
J. Lipid Res.
53
,
1437
1450
[PubMed]
85.
Montgomery
M.K.
,
Bayliss
J.
,
Nie
S.
,
De Nardo
W.
,
Keenan
S.N.
,
Miotto
P.M.
et al.
(
2022
)
Deep proteomic profiling unveils arylsulfatase A as a non-alcoholic steatohepatitis inducible hepatokine and regulator of glycemic control
.
Nat. Commun.
13
,
1259
[PubMed]
86.
Montgomery
M.K.
,
Bayliss
J.
,
Nie
S.
,
de Nardo
W.
,
Keenan
S.N.
,
Anari
M.
et al.
(
2023
)
Liver-secreted hexosaminidase a regulates insulin-like growth factor signaling and glucose transport in skeletal muscle
.
Diabetes
72
,
715
727
[PubMed]
87.
Capelluto
D.G.S.
(
2022
)
The repertoire of protein-sulfatide interactions reveal distinct modes of sulfatide recognition
.
Front Mol. Biosci.
9
,
1080161
[PubMed]
88.
Roeske-Nielsen
A.
,
Buschard
K.
,
Manson
J.E.
,
Rastam
L.
and
Lindblad
U.
(
2009
)
A variation in the cerebroside sulfotransferase gene is linked to exercise-modified insulin resistance and to type 2 diabetes
.
Exp. Diab. Res.
2009
,
429593
89.
Roeske-Nielsen
A.
,
Dalgaard
L.T.
,
Mansson
J.E.
and
Buschard
K.
(
2010
)
The glycolipid sulfatide protects insulin-producing cells against cytokine-induced apoptosis, a possible role in diabetes
.
Diabetes Metab. Res. Rev.
26
,
631
638
[PubMed]
90.
Buschard
K.
and
Antvorskov
J.C.
(
2022
)
The C24:0 sulfatide isoform as an important molecule in type 1 diabetes
.
Front Biosci. (Landmark Ed)
27
,
331
[PubMed]
91.
Cook
I.
and
Leyh
T.S.
(
2023
)
Sulfotransferase 2B1b, sterol sulfonation, and disease
.
Pharmacol. Rev.
75
,
521
531
[PubMed]
92.
Foster
P.A.
and
Mueller
J.W.
(
2023
)
New structural insights provide a different angle on steroid sulfatase action
.
J. Steroid Biochem. Mol. Biol.
232
,
106353
[PubMed]
93.
Sanchez
L.D.
,
Pontini
L.
,
Marinozzi
M.
,
Sanchez-Aranguren
L.C.
,
Reis
A.
and
Dias
I.H.K.
(
2021
)
Cholesterol and oxysterol sulfates: pathophysiological roles and analytical challenges
.
Br. J. Pharmacol.
178
,
3327
3341
[PubMed]
94.
Bi
Y.
,
Shi
X.
,
Zhu
J.
,
Guan
X.
,
Garbacz
W.G.
,
Huang
Y.
et al.
(
2018
)
Regulation of cholesterol sulfotransferase SULT2B1b by hepatocyte nuclear factor 4alpha constitutes a negative feedback control of hepatic gluconeogenesis
.
Mol. Cell. Biol.
38
,
e00654
17
[PubMed]
95.
Shi
X.
,
Cheng
Q.
,
Xu
L.
,
Yan
J.
,
Jiang
M.
,
He
J.
et al.
(
2014
)
Cholesterol sulfate and cholesterol sulfotransferase inhibit gluconeogenesis by targeting hepatocyte nuclear factor 4alpha
.
Mol. Cell. Biol.
34
,
485
497
[PubMed]
96.
Zhang
X.
,
Deng
D.
,
Cui
D.
,
Liu
Y.
,
He
S.
,
Zhang
H.
et al.
(
2022
)
Cholesterol sulfate exerts protective effect on pancreatic beta-cells by regulating beta-cell mass and insulin secretion
.
Front Pharmacol.
13
,
840406
[PubMed]
97.
Bakhaus
K.
,
Bennien
J.
,
Fietz
D.
,
Sanchez-Guijo
A.
,
Hartmann
M.
,
Serafini
R.
et al.
(
2018
)
Sodium-dependent organic anion transporter (Slc10a6(-/-)) knockout mice show normal spermatogenesis and reproduction, but elevated serum levels for cholesterol sulfate
.
J. Steroid Biochem. Mol. Biol.
179
,
45
54
[PubMed]
98.
Mauvais-Jarvis
F.
(
2016
)
Role of sex steroids in beta cell function, growth, and survival
.
Trends Endocrinol. Metab.
27
,
844
855
[PubMed]
99.
Barbosa
A.C.S.
,
Feng
Y.
,
Yu
C.
,
Huang
M.
and
Xie
W.
(
2019
)
Estrogen sulfotransferase in the metabolism of estrogenic drugs and in the pathogenesis of diseases
.
Expert Opin. Drug Metab. Toxicol.
15
,
329
339
[PubMed]
100.
Gao
J.
,
He
J.
,
Shi
X.
,
Stefanovic-Racic
M.
,
Xu
M.
,
O'Doherty
R.M.
et al.
(
2012
)
Sex-specific effect of estrogen sulfotransferase on mouse models of type 2 diabetes
.
Diabetes
61
,
1543
1551
[PubMed]
101.
Garbacz
W.G.
,
Jiang
M.
,
Xu
M.
,
Yamauchi
J.
,
Dong
H.H.
and
Xie
W.
(
2017
)
Sex- and tissue-specific role of estrogen sulfotransferase in energy homeostasis and insulin sensitivity
.
Endocrinology
158
,
4093
4104
[PubMed]
102.
Iqbal
M.J.
,
Greenway
B.
,
Wilkinson
M.L.
,
Johnson
P.J.
and
Williams
R.
(
1983
)
Sex-steroid enzymes, aromatase and 5 alpha-reductase in the pancreas: a comparison of normal adult, foetal and malignant tissue
.
Clin. Sci. (Lond.)
65
,
71
75
[PubMed]
103.
Sakurai
N.
,
Miki
Y.
,
Suzuki
T.
,
Watanabe
K.
,
Narita
T.
,
Ando
K.
et al.
(
2006
)
Systemic distribution and tissue localizations of human 17beta-hydroxysteroid dehydrogenase type 12
.
J. Steroid Biochem. Mol. Biol.
99
,
174
181
[PubMed]
104.
Xu
W.
,
Qadir
M.M.F.
,
Nasteska
D.
,
Mota de Sa
P.
,
Gorvin
C.M.
,
Blandino-Rosano
M.
et al.
(
2023
)
Architecture of androgen receptor pathways amplifying glucagon-like peptide-1 insulinotropic action in male pancreatic beta cells
.
Cell Rep.
42
,
112529
[PubMed]
105.
Navarro
G.
,
Xu
W.
,
Jacobson
D.A.
,
Wicksteed
B.
,
Allard
C.
,
Zhang
G.
et al.
(
2016
)
Extranuclear actions of the androgen receptor enhance glucose-stimulated insulin secretion in the male
.
Cell Metab.
23
,
837
851
[PubMed]
106.
Navarro
G.
,
Allard
C.
,
Morford
J.J.
,
Xu
W.
,
Liu
S.
,
Molinas
A.J.
et al.
(
2018
)
Androgen excess in pancreatic beta cells and neurons predisposes female mice to type 2 diabetes
.
JCI Insight
3
,
107.
Roland
A.V.
,
Nunemaker
C.S.
,
Keller
S.R.
and
Moenter
S.M.
(
2010
)
Prenatal androgen exposure programs metabolic dysfunction in female mice
.
J. Endocrinol.
207
,
213
223
[PubMed]
108.
Hogg
K.
,
Wood
C.
,
McNeilly
A.S.
and
Duncan
W.C.
(
2011
)
The in utero programming effect of increased maternal androgens and a direct fetal intervention on liver and metabolic function in adult sheep
.
PloS ONE
6
,
e24877
[PubMed]
109.
Rae
M.
,
Grace
C.
,
Hogg
K.
,
Wilson
L.M.
,
McHaffie
S.L.
,
Ramaswamy
S.
et al.
(
2013
)
The pancreas is altered by in utero androgen exposure: implications for clinical conditions such as polycystic ovary syndrome (PCOS)
.
PloS ONE
8
,
e56263
[PubMed]
110.
Abbott
D.H.
,
Bruns
C.R.
,
Barnett
D.K.
,
Dunaif
A.
,
Goodfriend
T.L.
,
Dumesic
D.A.
et al.
(
2010
)
Experimentally induced gestational androgen excess disrupts glucoregulation in rhesus monkey dams and their female offspring
.
Am. J. Physiol. Endocrinol. Metab.
299
,
E741
E751
[PubMed]
111.
Nicol
L.E.
,
O'Brien
T.D.
,
Dumesic
D.A.
,
Grogan
T.
,
Tarantal
A.F.
and
Abbott
D.H.
(
2014
)
Abnormal infant islet morphology precedes insulin resistance in PCOS-like monkeys
.
PloS ONE
9
,
e106527
[PubMed]
112.
Lightning
T.A.
,
Gesteira
T.F.
and
Mueller
J.W.
(
2021
)
Steroid disulfates - Sulfation double trouble
.
Mol. Cell. Endocrinol.
524
,
111161
[PubMed]
113.
Mueller
J.W.
,
Vogg
N.
,
Lightning
T.A.
,
Weigand
I.
,
Ronchi
C.L.
,
Foster
P.A.
et al.
(
2021
)
Steroid sulfation in adrenal tumors
.
J. Clin. Endocrinol. Metab.
106
,
3385
3397
[PubMed]
114.
MacGirlley
R.
,
Phoswa
W.N.
and
Mokgalaboni
K.
(
2023
)
Modulatory properties of vitamin D in Type 2 diabetic patients: a focus on inflammation and dyslipidemia
.
Nutrients
15
,
4575
[PubMed]
115.
Li
C.
,
Fu
J.
,
Ye
Y.
,
Li
J.
,
He
Y.
and
Fang
T.
(
2024
)
The impact of vitamin D on the etiopathogenesis and the progression of type 1 and type 2 diabetes in children and adults
.
Front Endocrinol. (Lausanne)
15
,
1360525
[PubMed]
116.
Schavinski
A.Z.
,
Reis
N.G.
,
Morgan
H.J.N.
,
Assis
A.P.
,
Moro
M.L.
,
Valentim
R.R.
et al.
(
2024
)
Maternal vitamin D deficiency impairs the development of beta cells in offspring rats in a sex-dependent manner
.
Int. J. Mol. Sci.
25
,
[PubMed]
117.
Morro
M.
,
Vila
L.
,
Franckhauser
S.
,
Mallol
C.
,
Elias
G.
,
Ferre
T.
et al.
(
2020
)
Vitamin D receptor overexpression in beta-cells ameliorates diabetes in mice
.
Diabetes
69
,
927
939
[PubMed]
118.
Lau
S.L.
,
Stokes
R.A.
,
Ng
B.
,
Cheng
K.
,
Clifton-Bligh
R.
and
Gunton
J.E.
(
2022
)
Metabolic changes in vitamin D receptor knockout mice
.
PloS ONE
17
,
e0267573
[PubMed]
119.
Neelankal John
A.
,
Iqbal
Z.
,
Colley
S.
,
Morahan
G.
,
Makishima
M.
and
Jiang
F.X.
(
2018
)
Vitamin D receptor-targeted treatment to prevent pathological dedifferentiation of pancreatic beta cells under hyperglycaemic stress
.
Diabetes Metab.
44
,
269
280
[PubMed]
120.
Viloria
K.
,
Nasteska
D.
,
Briant
L.J.B.
,
Heising
S.
,
Larner
D.P.
,
Fine
N.H.F.
et al.
(
2020
)
Vitamin-D-binding protein contributes to the maintenance of alpha cell function and glucagon secretion
.
Cell Rep.
31
,
107761
[PubMed]
121.
Kuo
T.
,
Damle
M.
,
Gonzalez
B.J.
,
Egli
D.
,
Lazar
M.A.
and
Accili
D.
(
2019
)
Induction of alpha cell-restricted Gc in dedifferentiating beta cells contributes to stress-induced beta-cell dysfunction
.
JCI Insight
5
,
e128351
[PubMed]
122.
Huynh
K.
,
Kempegowda
P.
,
Tamblyn
J.
,
MW
O.R.
,
Mueller
J.W.
,
Hewison
M.
et al.
(
2021
)
Development of a LC-MS/MS method to measure serum 3-sulfate and 3-glucuronide 25-hydroxyvitamin D3 metabolites; comparisons to unconjugated 25OHD in pregnancy and polycystic ovary syndrome
.
Steroids
169
,
108812
[PubMed]
123.
Reynolds
C.J.
,
Dyer
R.B.
,
Vizenor
B.A.
,
Koszewski
N.J.
,
Singh
R.J.
and
Thacher
T.D.
(
2024
)
Analysis of vitamin D(3)-sulfate and 25-hydroxyvitamin D(3)-sulfate in breastmilk by LC-MS/MS
.
J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.
1232
,
123954
[PubMed]
124.
Kurogi
K.
,
Sakakibara
Y.
,
Suiko
M.
and
Liu
M.C.
(
2017
)
Sulfation of vitamin D(3) -related compounds-identification and characterization of the responsible human cytosolic sulfotransferases
.
FEBS Lett.
591
,
2417
2425
[PubMed]
125.
Wong
T.
,
Wang
Z.
,
Chapron
B.D.
,
Suzuki
M.
,
Claw
K.G.
,
Gao
C.
et al.
(
2018
)
Polymorphic human sulfotransferase 2A1 mediates the formation of 25-hydroxyvitamin D(3)-3-O-sulfate, a major circulating vitamin D metabolite in humans
.
Drug Metab. Dispos.
46
,
367
379
[PubMed]
126.
Jenkinson
C.
,
Desai
R.
,
McLeod
M.D.
,
Mueller
J.W.
,
Hewison
M.
and
Handelsman
D.J.
(
2022
)
Circulating conjugated and unconjugated vitamin D metabolite measurements by liquid chromatography mass spectrometry
.
J. Clin. Endocrinol. Metab.
107
,
435
449
[PubMed]
127.
Harlacher
E.
,
Wollenhaupt
J.
,
Baaten
C.
and
Noels
H.
(
2022
)
Impact of uremic toxins on endothelial dysfunction in chronic kidney disease: a systematic review
.
Int. J. Mol. Sci.
23
,
531
[PubMed]
128.
Yu
M.
,
Kim
Y.J.
and
Kang
D.H.
(
2011
)
Indoxyl sulfate-induced endothelial dysfunction in patients with chronic kidney disease via an induction of oxidative stress
.
Clin. J. Am. Soc. Nephrol.
6
,
30
39
[PubMed]
129.
Niwa
T.
and
Shimizu
H.
(
2012
)
Indoxyl sulfate induces nephrovascular senescence
.
J. Ren. Nutr.
22
,
102
106
[PubMed]
130.
Omori
K.
,
Katakami
N.
,
Arakawa
S.
,
Yamamoto
Y.
,
Ninomiya
H.
,
Takahara
M.
et al.
(
2020
)
Identification of plasma inositol and indoxyl sulfate as novel biomarker candidates for atherosclerosis in patients with type 2 diabetes. -findings from metabolome analysis using GC/MS
.
J. Atheroscler. Thromb.
27
,
1053
1067
[PubMed]
131.
Oladi-Ghadikolaei
R.
,
Aliasgharzadeh
A.
,
Shayanfar
A.
,
Soleymani
J.
,
Moradi
M.
,
Jouyban
A.
et al.
(
2023
)
Serum levels of indoxyl sulfate and P-cresol in Type II diabetic patients with and without nephropathy
.
Iran J. Kidney Dis.
17
,
126
134
[PubMed]
132.
Lin
C.J.
,
Lin
J.
,
Pan
C.F.
,
Chuang
C.K.
,
Liu
H.L.
,
Sun
F.J.
et al.
(
2015
)
Indoxyl sulfate, not P-cresyl sulfate, is associated with advanced glycation end products in patients on long-term hemodialysis
.
Kidney Blood Press. Res.
40
,
121
129
[PubMed]
133.
Wakabayashi
I.
and
Marumo
M.
(
2022
)
Evidence for indoxyl sulfate as an inducer of oxidative stress in patients with diabetes
.
In Vivo
36
,
1790
1794
[PubMed]
134.
Aoyama
I.
and
Niwa
T.
(
2001
)
An oral adsorbent ameliorates renal overload of indoxyl sulfate and progression of renal failure in diabetic rats
.
Am. J. Kidney Dis.
37
,
S7
S12
[PubMed]
135.
Zertal-Zidani
S.
,
Bounacer
A.
and
Scharfmann
R.
(
2007
)
Regulation of pancreatic endocrine cell differentiation by sulphated proteoglycans
.
Diabetologia
50
,
585
595
[PubMed]
136.
Takahashi
I.
,
Noguchi
N.
,
Nata
K.
,
Yamada
S.
,
Kaneiwa
T.
,
Mizumoto
S.
et al.
(
2009
)
Important role of heparan sulfate in postnatal islet growth and insulin secretion
.
Biochem. Biophys. Res. Commun.
383
,
113
118
[PubMed]
137.
Potter
K.J.
,
Werner
I.
,
Denroche
H.C.
,
Montane
J.
,
Plesner
A.
,
Chen
Y.
et al.
(
2015
)
Amyloid formation in human islets is enhanced by heparin and inhibited by heparinase
.
Am. J. Transplant.
15
,
1519
1530
[PubMed]
138.
Zhang
S.
,
Yan
H.
,
Ma
X.
,
Zheng
W.
and
Wang
W.
(
2022
)
Effects of different routes of heparin on instant blood-mediated inflammatory reaction after portal vein islet transplantation
.
Zhong Nan Da Xue Xue Bao Yi Xue Ban
47
,
1
7
[PubMed]
139.
Cabric
S.
,
Sanchez
J.
,
Lundgren
T.
,
Foss
A.
,
Felldin
M.
,
Kallen
R.
et al.
(
2007
)
Islet surface heparinization prevents the instant blood-mediated inflammatory reaction in islet transplantation
.
Diabetes
56
,
2008
2015
[PubMed]
140.
Yang
J.
,
Jiang
S.
,
Guan
Y.
,
Deng
J.
,
Lou
S.
,
Feng
D.
et al.
(
2019
)
Pancreatic islet surface engineering with a starPEG-chondroitin sulfate nanocoating
.
Biomater. Sci.
7
,
2308
2316
[PubMed]
141.
Liao
W.
,
Duan
X.
,
Xie
F.
,
Zheng
D.
,
Yang
P.
,
Wang
X.
et al.
(
2023
)
3D-bioprinted double-crosslinked angiogenic alginate/chondroitin sulfate patch for diabetic wound healing
.
Int. J. Biol. Macromol.
236
,
123952
[PubMed]
142.
Shah
S.A.
,
Sohail
M.
,
Khan
S.A.
and
Kousar
M.
(
2021
)
Improved drug delivery and accelerated diabetic wound healing by chondroitin sulfate grafted alginate-based thermoreversible hydrogels
.
Mater. Sci. Eng. C. Mater. Biol. Appl.
126
,
112169
[PubMed]
143.
Cho
H.J.
,
Oh
J.
,
Choo
M.K.
,
Ha
J.I.
,
Park
Y.
and
Maeng
H.J.
(
2014
)
Chondroitin sulfate-capped gold nanoparticles for the oral delivery of insulin
.
Int. J. Biol. Macromol.
63
,
15
20
[PubMed]
144.
Wang
S.
,
Meng
S.
,
Zhou
X.
,
Gao
Z.
and
Piao
M.G.
(
2023
)
pH-responsive and mucoadhesive nanoparticles for enhanced oral insulin delivery: the effect of hyaluronic acid with different molecular weights
.
Pharmaceutics
15
,
820
[PubMed]
145.
Borgono
C.A.
and
Zinman
B.
(
2012
)
Insulins: past, present, and future
.
Endocrinol. Metab. Clin. North Am.
41
,
1
24
[PubMed]
146.
Madeira
F.
,
Madhusoodanan
N.
,
Lee
J.
,
Eusebi
A.
,
Niewielska
A.
,
Tivey
A.R.N.
et al.
(
2024
)
The EMBL-EBI Job Dispatcher sequence analysis tools framework in 2024
.
Nucleic Acids Res.
52
,
W521
W525
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).