It is the cache of ${baseHref}. It is a snapshot of the page. The current page could have changed in the meantime.
Tip: To quickly find your search term on this page, press Ctrl+F or ⌘-F (Mac) and use the find bar.

World Journal of Diabetes-Baishideng Publishing
World J Diabetes. 2011 August 15; 2 (8) : 119-126.
Published online 2011 August 15. doi: 10.4239/wjd.v2.i8.119.
Nutritional programming of pancreatic β-cell plasticity
David J Hill.
David J Hill, Department of Medicine, Physiology and Pharmacology, and Paediatrics, University of Western Ontario, London, Ontario N6A 5B8, Canada
David J Hill, Lawson Health Research Institute, St. Joseph’s Health Care, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
Author contributions: Hill DJ contributed solely to this paper.
Correspondence to: David J Hill, DPhil, Lawson Health Research Institute, St. Joseph’s Health Care, 268 Grosvenor Street, London, Ontario, N6A 4V2, Canada. david.hill@lhrionhealth.ca
Telephone: +1-519-6466100-64716 Fax: +1-519-6466110
Received March 1, 2011; Revised August 7, 2011; Accepted August 14, 2011;
Abstract
Nutritional insufficiency during pregnancy has been shown to alter the metabolism of the offspring and can increase the risk of type 2 diabetes. The phenotype in the offspring involves changes to the morphology and functional capacity of the endocrine pancreas, and in the supporting islet microvasculature. Pancreatic β-cells possess a plastic potential and can partially recover from catastrophic loss. This is partly due to the existence of progenitors within the islets and the ability to generate new islets by neogenesis from the pancreatic ducts. This regenerative capacity is induced by bone marrow-derived stem cells, including endothelial cell progenitors and is associated with increased angiogenesis within the islets. Nutritional insults in early life, such as feeding a low protein diet to the mother, impair the regenerative capacity of the β-cells. The mechanisms underlying this include a reduced ability of β-cells to differentiate from the progenitor population, changes in the inductive signals from the microvasculature and an altered presence of endothelial progenitors. Statin treatment within animal models was associated with angiogenesis in the islet microvasculature, improved vascular function and an increase in β-cell mass. This demonstrates that reversal of the impaired β-cell phenotype observed following nutritional insult in early life is potentially possible.
Keywords: Islet, β-cell, Plasticity, Diabetes, Nutrition, Statin
INTRODUCTION
Epidemiological studies have demonstrated that dietary restriction during pregnancy results in a reduced birth weight[ 1], leading to permanent changes in organ development, including the endocrine pancreas[ 2] and contributing to an adult predisposition to several chronic disease conditions, including type 2 diabetes and cardiovascular disease. Using an established model of dietary protein restriction during pregnancy and lactation, it has been extensively reported that dietary insufficiency in early life alters normal pancreatic development, which ultimately contributes to impaired glucose homeostasis in adulthood[ 3 - 7]. A low protein (LP) diet given to rats during pregnancy results in reduced β-cell mass (islet size) due to altered cell cycle kinetics and a lower proliferation rate but a greater incidence of apoptosis. The endocrine pancreas demonstrates impaired glucose-stimulated insulin release and greater cytokine-induced cell death. Offspring of LP-fed dams are glucose intolerant by 130 d of age.
However, the nutritional insult early in life not only changes the β-cell phenotype but also has a profound effect on the micro-vasculature of the pancreas. Intra-islet vascular volume and endothelial cell vascular endothelial growth factor (VEGF) receptor abundance were reduced in LP-fed offspring and both were reversed by supplementation of the LP diet with taurine, an amino acid which normally is present at high concentrations within islets but which is depleted in animals exposed to a LP diet[ 7]. Islet vasculature is also diminished in other models of intrauterine growth retardation such as uterine artery ligation of the mother[ 8] and in that study, it was reversible by administration of the glucagon-like polypeptide-1 analog, exendin 4, to the offspring, presumably by increasing β-cell-derived VEGF which promoted local angiogenesis. We recently applied the LP diet model to the mouse and found that the β-cell regeneration that normally occurs in juveniles following depletion of β-cell mass with streptozotocin (STZ) treatment was prevented if the offspring had been previously exposed to a LP diet[ 9]. It is therefore possible that the phenotypic changes seen in the endocrine pancreas as a result of nutritional insult in early life may represent impaired mechanisms of β-cell plasticity and that these relate to deficiencies in islet vasculogenesis. This review explores the capacity for β-cell plasticity, the relationship to the islet microvasculature and how such deficiencies might be reversed to prevent the risk of future diabetes.
DEVELOPMENT OF PANCREATIC β-CELLS
Both islet endocrine cells and acinar tissue develop from pancreatic ductal epithelium during fetal and neonatal development in the rat and human fetus[ 10 , 11]. The initial development of both lineages depends on the expression of key transcription factors such as Pdx1 and Ptf1 within the ductal cells[ 10]. Pdx1 is also required in the mature β-cell where it trans-activates the insulin and GLUT2 gene promoters. Other transcription factors, including neurogenin-3 (Ngn3), β2/NeuroD, Pax-4 and -6, and Nkx2.2, are necessary to complete the differentiation of individual endocrine cell lineages[ 12]. Pancreatic ductal cells or multi-potential stem cells can be manipulated in vitro to yield islet-like structures with multiple endocrine cell types[ 13 - 15]. The number of these structures can be potentiated by introducing extracellular matrix (ECM)[ 16], or specific combinations of growth factors such as activin, exendin-4, hepatocyte growth factor (HGF)[ 17], fibroblast growth factor-1 or leukemia inhibitory factor[ 18]. Regardless, the yield of new β-cells is generally low, most likely because the optimal environment for β-cell generation requires other supporting cell types.
EVIDENCE OF ENDOGENOUS β-CELL REGENERATION
Plasticity in β-cell mass is a physiological response and is seen during pregnancy[ 19 , 20] and with obesity[ 21]. A delicate balance of proliferation and apoptotic loss maintains β-cell mass in vivo. The human fetus and neonatal rodent undergo significant remodeling of their endocrine pancreas involving β-cell proliferation, neogenesis and apoptosis[ 22]. In humans there is histological evidence of β-cell neogenesis and a regenerative response in children and adolescents with type 1 diabetes[ 23 , 24]. Recently, proliferation of remaining β-cells was shown in deceased patients with new onset type 1 diabetes but not in those with long-standing disease or type 2 diabetes[ 25].
The origins of new β-cells in animal models of regeneration are various. Partial pancreatectomy induced the expansion of both endocrine and exocrine pancreatic mass[ 26 , 27], while injection of STZ into young rodents was shown to induce islet neogenesis from the ducts, similar to that occurring in embryogenesis[ 28]. Pancreatic ductal ligation has been shown to stimulate a doubling of β-cell mass in adult rats[ 29] by both islet neogenesis and hypertrophy of existing β-cells[ 30]. Surviving β-cells have been shown to spontaneously proliferate after cessation of their selective doxycycline-induced apoptosis by diphtheria toxin[ 31], supporting the concept that during regeneration β-cells are released from a tight control of cell replication. However, hormone-negative cells expressing Thy1.1 and CD133 have been identified in adult rat pancreatic ducts that subsequently expressed Pdx1 and both insulin and glucagon[ 32]. Similar duel insulin and glucagon-expressing cells have been identified in neonatal rat islets during β-cell regeneration following STZ treatment[ 33] and could represent resident endocrine cell progenitors. Seaberg et al[ 34] showed that multi-potential pancreatic stem cells existed within mouse islets and pancreatic ducts but were extremely rare. Conversely, Dor et al[ 35] and Nir et al[ 36] showed that following partial pancreatectomy, repopulation of β-cells within mouse islets occurred solely by replication of existing β-cells. A number of reports now show this conclusion to be misinterpreted.
Liu et al[ 37] used the same mouse model as Dor et al[ 35], where β-cells were lineage-tagged with human placental alkaline phosphatase (HPAP) to show that β-cell progenitors existed within the islets with little or no insulin expression and that these proliferated following β-cell depletion with STZ. Such cells were in the periphery of the islets and expressed the transcription factor MafB, a marker of immature β-cells. Similarly, Szabat et al[ 38] identified cells in mouse islets that were Pdx1-positive but insulin-negative, and which co-expressed MafB and Nkx2.2. These could mature into insulin-expressing β-cells in vitro that expressed MafA and Glut2, or could remain as progenitors. Finally, Thorel et al[ 39] showed that after near-total induced β-cell loss, new β-cells could be generated by trans-differentiation from α-cells. We have utilized the transgenic mouse model of Melton[ 35], in which approximately 30%-40% of β-cells and their subsequent progeny are genetically tagged with HPAP, to show that neonatal islets can be de-differentiated to a progenitor cell population in vitro and subsequently re-differentiated into pseudo-islet structures that express many of the transcription factor signatures of functional β-cells[ 40]. HPAP-tagged β-cells contribute both to the de-differentiated and re-differentiated cell populations. In summary, in postnatal life β-cell regeneration seems to predominantly occur within existing islets but may proceed both from a differentiation of resident progenitors and by the proliferation of mature β-cells. Additionally, substantial plasticity exists within existing β-cells, at least in vitro, to de-differentiate to a more primitive progenitor phenotype and subsequently to re-differentiate back into endocrine cells.
CONTRIBUTION OF THE MICROVASCULATURE TO β-CELL REGENERATION
Pancreatic islet vascular endothelium can induce β-cell growth, differentiation and function through the actions of paracrine growth factors and through integrin signals across the shared basement membrane[ 41 , 42]. Paracrine actions within the islet allow a synthesis of VEGF from the β-cells which contributes to endothelial cell proliferation, while a reciprocal production of HGF by the endothelial cells promotes β-cell growth[ 43]. We found that administration of STZ in the young rat not only caused a loss of β-cells, but an associated decrease in islet vasculature and that recovery of β-cell mass only occurred subsequent to recovery of the microvasculature[ 44]. However, the β-cell regenerative environment is likely to include not only vascular endothelium, but also the endothelial precursor cells (EPC)[ 45], mesenchymal stromal cells and bone marrow-derived hematopoietic lineage stem cells. Understanding how these components contribute to the regenerative environment and their communication with β-cells or their progenitors is key to understanding the control of β-cell regeneration.
BONE MARROW STEM CELLS AND β-CELL REGENERATION
Transplantation of bone marrow progenitor cells was shown by us and others to cause a reversal of hyperglycemia in animal models of diabetes and in newly diagnosed individuals with type 1 diabetes[ 46 - 49]. The ability of such cells to selectively home to damaged tissues has been variously linked to their expression of L-selectin[ 50], β2-integrins[ 51] and stromal cell-derived factor-1[ 52]. In some studies, a direct trans-differentiation of bone marrow-derived stem cells into insulin-positive β-cells was demonstrated, either in vivo or following in vitro lineage manipulation[ 53 - 55] but the direct contribution of bone marrow stem cells to new β-cells has generally been found to be low and inconsistent with the resulting increase in insulin secretion and/or normalization of blood glucose[ 46 , 56 - 58]. However, following bone marrow stem cell transfer, islet neovascularization was seen[ 46 , 59], accompanied by an increase in endogenous β-cells by replication or neogenesis of new islets from the pancreatic ducts[ 46 , 56]. There is debate as to which bone marrow-derived cells ‘induce’ β-cell regeneration. Yoder et al[ 60] concluded that bone marrow contained both pro-angiogenic hematopoietic progenitors of myeloid/monocyte lineage and true EPC that were not of hematopoietic lineage. Pro-angiogenic hematopoietic progenitors were hypothesized to function as paracrine supportive cells that induced vasculogenesis and tissue regeneration but the majority did not form functional endothelial cells. In the context of β-cell regeneration, these cells would be synergistic to the direct interactions known to occur between vascular endothelium and β-cells. In most papers, pro-angiogenic hematopoietic progenitors and true EPC are not distinguished between and are collectively described as EPC.
An alternate mechanism whereby hematopoietic lineage stem cell progeny could contribute to β-cell replication is by the generation of macrophages. In the macrophage-deficient colony stimulating factor 1 knock-out mouse ( op/op), animals develop osteopetrosis as adults but young animals demonstrated abnormal islet morphogenesis, a much reduced β-cell mass and deficiencies in β-cell replication[ 61]. Islet neogenesis at the pancreatic ducts was enhanced, suggesting that islets could form but the β-cell population could not expand appropriately. There is also evidence that macrophages have a key role in islet angiogenesis through the expression of matrix metalloproteinase-9[ 62].
Most studies on the contribution of bone marrow-derived stem cells to β-cell survival or regeneration have transplanted cells with a genetic tag, such as green fluorescent protein, into irradiated recipient animals made diabetic with STZ or into diabetes-prone animals such as the NOD mouse[ 63 , 64]. As little as 1% allogeneic chimerism of repopulated marrow was able to reverse diabetes in the latter[ 64]. However, in human pancreata from individuals who had previously received hematopoietic stem cell transplants from the opposite gender, there was no evidence of colonization within the islets[ 65]. The mobilization of bone marrow stem cells to colonize the pancreas appears to be linked to the presence of pancreatic tissue damage in either the endocrine or exocrine compartments[ 62]. It cannot be assumed that the bone marrow-derived cells will be of equivalent lineage phenotype when they colonize the pancreas vs their subsequent ability to induce β-cell renewal. The entire environment of the pancreas following β-cell loss, including bone marrow-derived cells, the remodeled ECM and the cytokine/growth factor milieu, is likely to represent the combined elements necessary to optimize β-cell regeneration.
We utilized mice expressing Cre recombinase under control of the Vav promoter, which were crossed with ROSA26 yellow fluorescent protein (YFP) transgenic mice, such that hematopoietic lineage cells and their progeny could be tracked[ 66]. The Vav gene is ubiquitously but specifically expressed by all hematopoietic lineage cells where it functions as a signal transduction molecule and it remains active on differentiated cell progeny including T cells, B cells and macrophages[ 67]. YFP-tagged cells were located within the pancreas at all ages, lining the ductal epithelium and around and within the islets. Following STZ treatment, the presence of these cells was significantly increased, temporally corresponding with a recovery of β-cell mass. No co-localization of insulin or other pancreatic endocrine hormones was found in the hematopoietic lineage cells but approximately 30% of such cells co-stained with CD31, a marker of macrophages, EPC and endothelial cells, which significantly increased after STZ. A sub-population of hematopoietic lineage cells around the islets demonstrated the macrophage markers F4-80 and Mac-1 and some large YFP-positive cells within the islets showed a nuclear presence of Pdx1 and could be endocrine progenitors. This strongly suggests that endogenous bone-marrow-derived stem cells are involved in β-cell recovery after induced diabetes.
EVIDENCE THAT EARLY NUTRITIONAL INSULTS IMPAIR β-CELL REGENERATION
Exposure to a LP diet during gestation affected pancreatic endocrine plasticity postnatally as mice were unable to recover β-cell mass following exposure to STZ[ 9]. In female animals, this was associated with a reduced number of islets relative to STZ treatment alone but this was not seen in males. Other studies have also shown that nutrient deficiency early in life affects tissue plasticity. A LP diet during gestation significantly impaired recovery of male adult rats following ischemia-reperfusion[ 68] while maternal calorie restriction during gestation and lactation impaired β-cell replacement after STZ treatment[ 69 , 70]. However, changes in islet morphometry resulting from prior exposure to dietary insult are not specific to β-cells, as the α-cell mass was also increased[ 9]. Thus, the change in islet tissue plasticity is likely to represent a fundamental change in phenotype in islet cell progenitors that contribute to multiple cell types. One type of progenitor that has been characterized in islet cells expresses the transcription factor Pdx1 but not insulin[ 38]. Such cells are rapidly able to differentiate into insulin-expressing β-cells in vitro and in vivo and may represent a strategic reserve of latent β-cells that could be mobilized in situations of extreme metabolic demand. We found that 4%-8% of islet cells in neonatal mice were Pdx1-positive but insulin-negative by immune-histochemistry[ 9]. Following exposure of mice to a LP diet during gestation, the offspring showed no change in the percentage of such cells that were present within islets. However, after exposure to STZ, the number of Pdx1-positive/insulin-negative cells was increased, which may indicate that normal maturational pathways that allow such cells to differentiate into functional β-cells are impaired following dietary insult.
Further evidence that dietary restriction in early life alters β-cell progenitor phenotype comes from manipulation of islets in vitro. We isolated islets from neonatal mouse islets previously exposed to a control diet or a LP diet fed to the mothers during gestation. Islets were de-differentiated by culture for 4 wk on a type 1 collagen matrix in the presence of epidermal growth factor to yield ductal epithelial cell-like monolayers[ 71]. The doubling time of cells from LP-fed offspring was significantly prolonged. Cell monolayers were then re-differentiated to form pseudo-islets over 4 wk by culture on Matrigel in the presence of insulin-like growth factor-II (IGF-II) and fibroblast growth factor-7. Cells derived from LP-fed mice demonstrated a relative impairment of pseudo-islet formation and insulin content and release. The relative gene expression of transcription factors involved in β-cell generation from precursors and endocrine hormones was determined by DNA microarray analysis. De-differentiated islet cultures showed a reduction in the expression of Pdx1, Pax6 and Ngn3, and of insulin and somatostatin mRNAs (Table 1). After pseudo-islets were subsequently generated, the expression of each of these genes returned to values close to those seen in fresh islets if the donor animals had been exposed to the control diet. However, in animals exposed to the LP diet, pseudo-islets did recover expression of transcription factors, insulin or somatostatin to a similar extent. These results suggest that a maternal LP diet alters pancreatic endocrine stem cell presence and phenotype in the offspring, leading to reduced islet plasticity in postnatal life, and that this can be demonstrated in vitro.
Table 1
Table 1
Changes in relative gene expression assessed by DNA microarray for monolayer cells cultures
Maternal malnutrition can also alter the development of tissue vasculogenesis in the offspring, which may also limit β-cell plasticity through a disruption of endothelium-β-cell signaling. Offspring of pregnant rats given a LP diet during gestation exhibit a reduction in capillary density in a variety of tissues, including skeletal muscle, endometrium, ovaries and pancreatic islets[ 5 , 72 - 74]. Endothelial dilatation was similarly impaired[ 75]. Within the pancreas, the number of EPC, characterized as being nestin and CD34-positive, was significantly reduced in offspring of LP-fed mothers[ 6], which in other tissues has been associated with a reduced expression of IGF-II[ 76].
REVERSAL STRATEGIES: ABILITY OF STATINS TO INCREASE β-CELL MASS
Statins are potent and safe drugs widely used to treat familial dyslipidemia[ 77 , 78] and to lower cholesterol levels in patients with or at risk of cardiovascular disease[ 79]. However, statins also exert pleiotropic actions unrelated to their cholesterol-lowering effect. These include improvements in endothelial cell function such as increased NO synthesis and anti-oxidant effects[ 80], stabilization and reduction of atherosclerotic plaque[ 80 - 82] and inhibition of inflammatory responses as measured by circulating C-reactive protein and cytokines. In offspring of rats given a LP diet during gestation, blood vessel dilation was impaired but this was corrected by postnatal treatment with atorvastatin[ 83]. Diabetes is associated with a reduction in circulating EPC and their abundance in bone marrow[ 84]. This is likely be related to the increased presence of oxidized LDL-cholesterol which has been shown to decrease EPC migration, differentiation into endothelial cells and survival[ 85]. Statin treatment increased the mobilization of EPC from bone marrow in a diabetic pig model[ 86], increased proliferation of EPC in vitro[ 87] and delayed diabetes onset in two different mouse models of T1D, including STZ treatment[ 88]. This was independent of inhibition of HMG-CoA reductase activity[ 89].
Treatment of neonatal rats with atorvastatin significantly increased β-cell mass in both STZ-treated and control animals and improved glucose tolerance[ 90]. Atorvastatin treatment was associated with an increased number of intra-islet endothelial cells, suggesting that vasculogenesis had preceded the increase in β-cell mass. This was supported by an increase in the proportion of intra-islet endothelial cells undergoing DNA synthesis and a parallel increase in the proliferation rate of adjacent β-cells. Hyperglycemia induced apoptosis in isolated human pancreatic islet endothelial cells but this was prevented by exposure to statin via the Akt intra-cellular survival pathway[ 91]. It is not clear if the trophic effects of atorvastatin are exerted directly on the β-cells or if they are mediated by secondary trophic effects that enhance migration of bone marrow-derived stem cells into the pancreas and/or islet vasculogenesis.
What is the potential for using statins to increase the islet microvasculature and enhance β-cell mass in humans? In individuals with type 1 diabetes of duration greater than 10 years, treatment with atorvastatin for 6 mo in a placebo-controlled study resulted in a significant improvement in blood vessel flow-mediated dilation and C-reactive protein, a marker of inflammation[ 92]. Similarly, in young adults with type 1 diabetes with a mean age of 34 years and normal blood cholesterol, just 6 wk of treatment with atorvastatin resulted in improved flow-mediated dilation and reduced LDL-cholesterol[ 93]. Diabetic subjects with microalbuminurea similarly benefited from 6 wk of atorvastatin therapy with a significant decrease in apolipoprotein B, LDL-cholesterol and oxidized LDL[ 94]. Young, normo-cholesterolemic males with type 1 diabetes had endothelial dysfunction assessed by flow-mediated dilation which improved significantly after only 4 wk of treatment with pravastatin and reached control patient values[ 95]. These beneficial effects of statins on the vasculature of individuals with diabetes were demonstrated in the absence of hypercholesterolemia. Recently, a randomized, placebo-controlled clinical trial tested the effect of atorvastatin therapy over 18 mo on residual β-cell function in young adults with new onset type 1 diabetes[ 96]. C-peptide levels were measured after a mixed meal test as an indicator of endogenous insulin release. This gradually declined with time in both placebo and atorvastatin-treated subjects but was significantly better preserved with atorvastatin. This strongly suggests that statin treatment could help retain or enhance residual β-cell mass.
CONCLUSION
In summary, nutritional insults in early life result in decreased β-cell mass and function in the offspring that persists into adult life and provides increased risk of glucose intolerance and type 2 diabetes. This is likely to involve a restriction on β-cell plasticity that can be directly mediated through effects on β-cell progenitors and the function of mature β-cells, but may also be indirect through a decreased islet vasculogenesis and availability of EPC, resulting in an impaired trophic signaling between the vascular endothelium and the β-cell. Likely reversal strategies include the use of statins to improve microvascular volume and function.
Footnotes
Supported by the Canadian Institutes of Health Research, the Canadian Diabetes Association and the Juvenile Diabetes Research Foundation
Peer reviewers: Cong-Yi Wang, MD, PhD, Professor, The Center for Biomedical Research, 1095 Jiefang Ave, Wuhan 430030, China; CBGM, Georgia’s Health Sciences University, 1120 15th Street, CA4098, Augusta, GA 30912, United States
S- Editor Wu X L- Editor Roemmele A E- Editor Zheng XM
References
1.
Barker DJP. 1994 Programming the baby. In:Mothers , Babies , Disease in later life.,editors.London: BMJ Publishing Group, London; 1994.pp.14-36.
2.
Desai M, Crowther NJ, Lucas A, Hales CN. Organ-selective growth in the offspring of protein-restricted mothers. Br J Nutr. 1996; 76:591-603. [PubMed]  [DOI]
3.
Petrik J, Reusens B, Arany E, Remacle C, Coelho C, Hoet JJ, Hill DJ. A low protein diet alters the balance of islet cell replication and apoptosis in the fetal and neonatal rat and is associated with a reduced pancreatic expression of insulin-like growth factor-II. Endocrinology. 1999; 140:4861-4873. [PubMed]  [DOI]
4.
Boujendar S, Reusens B, Merezak S, Ahn MT, Arany E, Hill D, Remacle C. Taurine supplementation to a low protein diet during foetal and early postnatal life restores a normal proliferation and apoptosis of rat pancreatic islets. Diabetologia. 2002; 45:856-866. [PubMed]  [DOI]
5.
Boujendar S, Arany E, Hill D, Remacle C, Reusens B. Taurine supplementation of a low protein diet fed to rat dams normalizes the vascularization of the fetal endocrine pancreas. J Nutr. 2003; 133:2820-2825. [PubMed]
6.
Joanette EA, Reusens B, Arany E, Thyssen S, Remacle RC, Hill DJ. Low-protein diet during early life causes a reduction in the frequency of cells immunopositive for nestin and CD34 in both pancreatic ducts and islets in the rat. Endocrinology. 2004; 145:3004-3013. [PubMed]  [DOI]
7.
Chamson-Reig A, Thyssen SM, Hill DJ, Arany E. Exposure of the pregnant rat to low protein diet causes impaired glucose homeostasis in the young adult offspring by different mechanisms in males and females. Exp Biol Med (Maywood). 2009; 234:1425-1436. [PubMed]  [DOI]
8.
Ham JN, Crutchlow MF, Desai BM, Simmons RA, Stoffers DA. Exendin-4 normalizes islet vascularity in intrauterine growth restricted rats: potential role of VEGF. Pediatr Res. 2009; 66:42-46. [PubMed]  [DOI]
9.
Cox AR, Gottheil SK, Arany EJ, Hill DJ. The effects of low protein during gestation on mouse pancreatic development and beta cell regeneration. Pediatr Res. 2010; 68:16-22. [PubMed]
10.
Murtaugh LC. Pancreas and beta-cell development: from the actual to the possible. Development. 2007; 134:427-438. [PubMed]  [DOI]
11.
Cerf ME. Transcription factors regulating beta-cell function. Eur J Endocrinol. 2006; 155:671-679. [PubMed]  [DOI]
12.
Habener JF, Kemp DM, Thomas MK. Minireview: transcriptional regulation in pancreatic development. Endocrinology. 2005; 146:1025-1034. [PubMed]  [DOI]
13.
Soria B. In-vitro differentiation of pancreatic beta-cells. Differentiation. 2001; 68:205-219. [PubMed]  [DOI]
14.
Ramiya VK, Maraist M, Arfors KE, Schatz DA, Peck AB, Cornelius JG. Reversal of insulin-dependent diabetes using islets generated in vitro from pancreatic stem cells. Nat Med. 2000; 6:278-282. [PubMed]  [DOI]
15.
Bonner-Weir S, Toschi E, Inada A, Reitz P, Fonseca SY, Aye T, Sharma A. The pancreatic ductal epithelium serves as a potential pool of progenitor cells. Pediatr Diabetes. 2004; 5 Suppl 2:16-22. [PubMed]  [DOI]
16.
Kerr-Conte J, Pattou F, Lecomte-Houcke M, Xia Y, Boilly B, Proye C, Lefebvre J. Ductal cyst formation in collagen-embedded adult human islet preparations. A means to the reproduction of nesidioblastosis in vitro. Diabetes. 1996; 45:1108-1114. [PubMed]  [DOI]
17.
Chase LG, Ulloa-Montoya F, Kidder BL, Verfaillie CM. Islet-derived fibroblast-like cells are not derived via epithelial-mesenchymal transition from Pdx-1 or insulin-positive cells. Diabetes. 2007; 56:3-7. [PubMed]  [DOI]
18.
Atouf F, Park CH, Pechhold K, Ta M, Choi Y, Lumelsky NL. No evidence for mouse pancreatic beta-cell epithelial-mesenchymal transition in vitro. Diabetes. 2007; 56:699-702. [PubMed]  [DOI]
19.
Sorenson RL, Brelje TC. Adaptation of islets of Langerhans to pregnancy: beta-cell growth, enhanced insulin secretion and the role of lactogenic hormones. Horm Metab Res. 1997; 29:301-307. [PubMed]  [DOI]
20.
Parsons JA, Brelje TC, Sorenson RL. Adaptation of islets of Langerhans to pregnancy: increased islet cell proliferation and insulin secretion correlates with the onset of placental lactogen secretion. Endocrinology. 1992; 130:1459-1466. [PubMed]  [DOI]
21.
Klöppel G, Löhr M, Habich K, Oberholzer M, Heitz PU. Islet pathology and the pathogenesis of type 1 and type 2 diabetes mellitus revisited. Surv Synth Pathol Res. 1985; 4:110-125. [PubMed]
22.
Scaglia L, Cahill CJ, Finegood DT, Bonner-Weir S. Apoptosis participates in the remodeling of the endocrine pancreas in the neonatal rat. Endocrinology. 1997; 138:1736-1741. [PubMed]  [DOI]
23.
Gepts W, De Mey J. Islet cell survival determined by morphology. An immunocytochemical study of the islets of Langerhans in juvenile diabetes mellitus. Diabetes. 1978; 27 Suppl 1:251-261. [PubMed]
24.
Volk BW, Wellman KF. The pancreas in idiopathic diabetes. In:Volk BW, Arquilla ER, eds The Diabetic Pancreas.,editors. 2nd ed. New York: Plenum Medical; 1985.pp.353.
25.
Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG. Evidence of increased islet cell proliferation in patients with recent-onset type 1 diabetes. Diabetologia. 2010; 53:2020-2028. [PubMed]  [DOI]
26.
Bonner-Weir S, Trent DF, Weir GC. Partial pancreatectomy in the rat and subsequent defect in glucose-induced insulin release. J Clin Invest. 1983; 71:1544-1553. [PubMed]  [DOI]
27.
Sétáló G, Blatniczky L, Vigh S. Development and growth of the islets of Langerhans through acino-insular transformation in regenerating rat pancreas. Acta Biol Acad Sci Hung. 1972; 23:309-325. [PubMed]
28.
Dutrillaux MC, Portha B, Rozé C, Hollande E. Ultrastructural study of pancreatic B cell regeneration in newborn rats after destruction by streptozotocin. Virchows Arch B Cell Pathol Incl Mol Pathol. 1982; 39:173-185. [PubMed]  [DOI]
29.
Wang RN, Klöppel G, Bouwens L. Duct- to islet-cell differentiation and islet growth in the pancreas of duct-ligated adult rats. Diabetologia. 1995; 38:1405-1411. [PubMed]  [DOI]
30.
Xu X, D'Hoker J, Stangé G, Bonné S, De Leu N, Xiao X, Van de Casteele M, Mellitzer G, Ling Z, Pipeleers D. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell. 2008; 132:197-207. [PubMed]  [DOI]
31.
Herrera PL, Huarte J, Zufferey R, Nichols A, Mermillod B, Philippe J, Muniesa P, Sanvito F, Orci L, Vassalli JD. Ablation of islet endocrine cells by targeted expression of hormone-promoter-driven toxigenes. Proc Natl Acad Sci U S A. 1994; 91:12999-13003. [PubMed]  [DOI]
32.
Stevenson KS, McGlynn L, Hodge M, McLinden H, George WD, Davies RW, Shiels PG. Isolation, characterization, and differentiation of thy1.1-sorted pancreatic adult progenitor cell populations. Stem Cells Dev. 2009; 18:1389-1398. [PubMed]  [DOI]
33.
Thyssen S, Arany E, Hill DJ. Ontogeny of regeneration of beta-cells in the neonatal rat after treatment with streptozotocin. Endocrinology. 2006; 147:2346-2356. [PubMed]  [DOI]
34.
Seaberg RM, Smukler SR, Kieffer TJ, Enikolopov G, Asghar Z, Wheeler MB, Korbutt G, van der Kooy D. Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat Biotechnol. 2004; 22:1115-1124. [PubMed]  [DOI]
35.
Dor Y, Brown J, Martinez OI, Melton DA. Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature. 2004; 429:41-46. [PubMed]  [DOI]
36.
Nir T, Melton DA, Dor Y. Recovery from diabetes in mice by beta cell regeneration. J Clin Invest. 2007; 117:2553-2561. [PubMed]  [DOI]
37.
Liu H, Guz Y, Kedees MH, Winkler J, Teitelman G. Precursor cells in mouse islets generate new beta-cells in vivo during aging and after islet injury. Endocrinology. 2010; 151:520-528. [PubMed]  [DOI]
38.
Szabat M, Luciani DS, Piret JM, Johnson JD. Maturation of adult beta-cells revealed using a Pdx1/insulin dual-reporter lentivirus. Endocrinology. 2009; 150:1627-1635. [PubMed]  [DOI]
39.
Thorel F, Népote V, Avril I, Kohno K, Desgraz R, Chera S, Herrera PL. Conversion of adult pancreatic alpha-cells to beta-cells after extreme beta-cell loss. Nature. 2010; 464:1149-1154. [PubMed]  [DOI]
40.
Beamish CA, Strutt BJ, Arany EJ, Hill DJ. Neonatal mouse β-cells can be differentiated in vitro. Montreal: Int. Diabetes Fed; 2009.pp.D-0851.
41.
Lammert E, Cleaver O, Melton D. Role of endothelial cells in early pancreas and liver development. Mech Dev. 2003; 120:59-64. [PubMed]  [DOI]
42.
Nikolova G, Jabs N, Konstantinova I, Domogatskaya A, Tryggvason K, Sorokin L, Fässler R, Gu G, Gerber HP, Ferrara N. The vascular basement membrane: a niche for insulin gene expression and Beta cell proliferation. Dev Cell. 2006; 10:397-405. [PubMed]  [DOI]
43.
Johansson M, Mattsson G, Andersson A, Jansson L, Carlsson PO. Islet endothelial cells and pancreatic beta-cell proliferation: studies in vitro and during pregnancy in adult rats. Endocrinology. 2006; 147:2315-2324. [PubMed]
44.
Nicholson JM, Arany EJ, Hill DJ. Changes in islet microvasculature following streptozotocin-induced beta-cell loss and subsequent replacement in the neonatal rat. Exp Biol Med (Maywood). 2010; 235:189-198. [PubMed]  [DOI]
45.
Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997; 275:964-967. [PubMed]  [DOI]
46.
Hess D, Li L, Martin M, Sakano S, Hill D, Strutt B, Thyssen S, Gray DA, Bhatia M. Bone marrow-derived stem cells initiate pancreatic regeneration. Nat Biotechnol. 2003; 21:763-770. [PubMed]  [DOI]
47.
Xie QP, Huang H, Xu B, Dong X, Gao SL, Zhang B, Wu YL. Human bone marrow mesenchymal stem cells differentiate into insulin-producing cells upon microenvironmental manipulation in vitro. Differentiation. 2009; 77:483-491. [PubMed]  [DOI]
48.
Sordi V, Piemonti L. The contribution of hematopoietic stem cells to beta-cell replacement. Curr Diab Rep. 2009; 9:119-124. [PubMed]  [DOI]
49.
Couri CE, Oliveira MC, Stracieri AB, Moraes DA, Pieroni F, Barros GM, Madeira MI, Malmegrim KC, Foss-Freitas MC, Simões BP. C-peptide levels and insulin independence following autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA. 2009; 301:1573-1579. [PubMed]  [DOI]
50.
Biancone L, Cantaluppi V, Duò D, Deregibus MC, Torre C, Camussi G. Role of L-selectin in the vascular homing of peripheral blood-derived endothelial progenitor cells. J Immunol. 2004; 173:5268-5274. [PubMed]
51.
Chavakis E, Aicher A, Heeschen C, Sasaki K, Kaiser R, El Makhfi N, Urbich C, Peters T, Scharffetter-Kochanek K, Zeiher AM. Role of beta2-integrins for homing and neovascularization capacity of endothelial progenitor cells. J Exp Med. 2005; 201:63-72. [PubMed]  [DOI]
52.
Huang Y, Kucia M, Hussain LR, Wen Y, Xu H, Yan J, Ratajczak MZ, Ildstad ST. Bone marrow transplantation temporarily improves pancreatic function in streptozotocin-induced diabetes: potential involvement of very small embryonic-like cells. Transplantation. 2010; 89:677-685. [PubMed]  [DOI]
53.
Ianus A, Holz GG, Theise ND, Hussain MA. In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J Clin Invest. 2003; 111:843-850. [PubMed]  [DOI]
54.
Zhao M, Amiel SA, Ajami S, Jiang J, Rela M, Heaton N, Huang GC. Amelioration of streptozotocin-induced diabetes in mice with cells derived from human marrow stromal cells. PLoS One. 2008; 3:e2666. [PubMed]  [DOI]
55.
Luo L, Luo JZ, Xiong F, Abedi M, Greer D. Cytokines inducing bone marrow SCA+ cells migration into pancreatic islet and conversion into insulin-positive cells in vivo. PLoS One. 2009; 4:e4504. [PubMed]  [DOI]
56.
Hasegawa Y, Ogihara T, Yamada T, Ishigaki Y, Imai J, Uno K, Gao J, Kaneko K, Ishihara H, Sasano H. Bone marrow (BM) transplantation promotes beta-cell regeneration after acute injury through BM cell mobilization. Endocrinology. 2007; 148:2006-2015. [PubMed]  [DOI]
57.
Lechner A, Yang YG, Blacken RA, Wang L, Nolan AL, Habener JF. No evidence for significant transdifferentiation of bone marrow into pancreatic beta-cells in vivo. Diabetes. 2004; 53:616-623. [PubMed]  [DOI]
58.
Taneera J, Rosengren A, Renstrom E, Nygren JM, Serup P, Rorsman P, Jacobsen SE. Failure of transplanted bone marrow cells to adopt a pancreatic beta-cell fate. Diabetes. 2006; 55:290-296. [PubMed]  [DOI]
59.
Mathews V, Hanson PT, Ford E, Fujita J, Polonsky KS, Graubert TA. Recruitment of bone marrow-derived endothelial cells to sites of pancreatic beta-cell injury. Diabetes. 2004; 53:91-98. [PubMed]  [DOI]
60.
Yoder MC, Mead LE, Prater D, Krier TR, Mroueh KN, Li F, Krasich R, Temm CJ, Prchal JT, Ingram DA. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood. 2007; 109:1801-1809. [PubMed]  [DOI]
61.
Banaei-Bouchareb L, Gouon-Evans V, Samara-Boustani D, Castellotti MC, Czernichow P, Pollard JW, Polak M. Insulin cell mass is altered in Csf1op/Csf1op macrophage-deficient mice. J Leukoc Biol. 2004; 76:359-367. [PubMed]  [DOI]
62.
Tessem JS, Jensen JN, Pelli H, Dai XM, Zong XH, Stanley ER, Jensen J, DeGregori J. Critical roles for macrophages in islet angiogenesis and maintenance during pancreatic degeneration. Diabetes. 2008; 57:1605-1617. [PubMed]  [DOI]
63.
Choi JB, Uchino H, Azuma K, Iwashita N, Tanaka Y, Mochizuki H, Migita M, Shimada T, Kawamori R, Watada H. Little evidence of transdifferentiation of bone marrow-derived cells into pancreatic beta cells. Diabetologia. 2003; 46:1366-1374. [PubMed]  [DOI]
64.
Zorina TD, Subbotin VM, Bertera S, Alexander AM, Haluszczak C, Gambrell B, Bottino R, Styche AJ, Trucco M. Recovery of the endogenous beta cell function in the NOD model of autoimmune diabetes. Stem Cells. 2003; 21:377-388. [PubMed]  [DOI]
65.
Butler AE, Huang A, Rao PN, Bhushan A, Hogan WJ, Rizza RA, Butler PC. Hematopoietic stem cells derived from adult donors are not a source of pancreatic beta-cells in adult nondiabetic humans. Diabetes. 2007; 56:1810-1816. [PubMed]  [DOI]
66.
Chamson-Reig A, Arany EJ, Hill DJ. Lineage tracing and resulting phenotype of haemopoietic-derived cells in the pancreas during beta cell regeneration. Diabetologia. 2010; 53:2188-2197. [PubMed]  [DOI]
67.
de Boer J, Williams A, Skavdis G, Harker N, Coles M, Tolaini M, Norton T, Williams K, Roderick K, Potocnik AJ. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur J Immunol. 2003; 33:314-325. [PubMed]  [DOI]
68.
Ravelli AC, van der Meulen JH, Michels RP, Osmond C, Barker DJ, Hales CN, Bleker OP. Glucose tolerance in adults after prenatal exposure to famine. Lancet. 1998; 351:173-177. [PubMed]  [DOI]
69.
Elmes MJ, Gardner DS, Langley-Evans SC. Fetal exposure to a maternal low-protein diet is associated with altered left ventricular pressure response to ischaemia-reperfusion injury. Br J Nutr. 2007; 98:93-100. [PubMed]  [DOI]
70.
Garofano A, Czernichow P, Bréant B. Impaired beta-cell regeneration in perinatally malnourished rats: a study with STZ. FASEB J. 2000; 14:2611-2617. [PubMed]  [DOI]
71.
Beamish CA, Strutt B, Arany E, Hill , DJ . Imaging of re-differentiated islet like clusters using a novel immunocytochemical method. Diabetes. 2007; 56:(Suppl 1) 1678.
72.
Pladys P, Sennlaub F, Brault S, Checchin D, Lahaie I, Lê NL, Bibeau K, Cambonie G, Abran D, Brochu M. Microvascular rarefaction and decreased angiogenesis in rats with fetal programming of hypertension associated with exposure to a low-protein diet in utero. Am J Physiol Regul Integr Comp Physiol. 2005; 289:R1580-R1588. [PubMed]  [DOI]
73.
Bittencourt Brasil F, Silva Faria T, Barcellos Sampaio FJ, da Fonte Ramos C. The effect of maternal malnutrition during lactation on the endometrial ERalpha expression, collagen type, and blood vessels in the rats offspring at puberty. Anat Rec (Hoboken). 2010; 293:162-170. [PubMed]  [DOI]
74.
Ferreira RV, Gombar FM, da Silva Faria T, Costa WS, Sampaio FJ, da Fonte Ramos C. Metabolic programming of ovarian angiogenesis and folliculogenesis by maternal malnutrition during lactation. Fertil Steril. 2010; 93:2572-2580. [PubMed]  [DOI]
75.
Sathishkumar K, Elkins R, Yallampalli U, Yallampalli C. Protein restriction during pregnancy induces hypertension and impairs endothelium-dependent vascular function in adult female offspring. J Vasc Res. 2009; 46:229-239. [PubMed]  [DOI]
76.
Maeng YS, Choi HJ, Kwon JY, Park YW, Choi KS, Min JK, Kim YH, Suh PG, Kang KS, Won MH. Endothelial progenitor cell homing: prominent role of the IGF2-IGF2R-PLCbeta2 axis. Blood. 2009; 113:233-243. [PubMed]  [DOI]
77.
Shepherd J, Cobbe SM, Ford I, Isles CG, Lorimer AR, MacFarlane PW, McKillop JH, Packard CJ. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. West of Scotland Coronary Prevention Study Group. N Engl J Med. 1995; 333:1301-1307. [PubMed]  [DOI]
78.
Daniels SR, Greer FR. Lipid screening and cardiovascular health in childhood. Pediatrics. 2008; 122:198-208. [PubMed]  [DOI]
79.
Sacks FM, Pfeffer MA, Moye LA, Rouleau JL, Rutherford JD, Cole TG, Brown L, Warnica JW, Arnold JM, Wun CC. The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events Trial investigators. N Engl J Med. 1996; 335:1001-1009. [PubMed]  [DOI]
80.
Wang CY, Liu PY, Liao JK. Pleiotropic effects of statin therapy: molecular mechanisms and clinical results. Trends Mol Med. 2008; 14:37-44. [PubMed]  [DOI]
81.
Ikeda U, Shimpo M, Ohki R, Inaba H, Takahashi M, Yamamoto K, Shimada K. Fluvastatin inhibits matrix metalloproteinase-1 expression in human vascular endothelial cells. Hypertension. 2000; 36:325-329. [PubMed]
82.
Dulak J, Loboda A, Jazwa A, Zagorska A, Dörler J, Alber H, Dichtl W, Weidinger F, Frick M, Jozkowicz A. Atorvastatin affects several angiogenic mediators in human endothelial cells. Endothelium. 2006; 12:233-241. [PubMed]
83.
Torrens C, Kelsall CJ, Hopkins LA, Anthony FW, Curzen NP, Hanson MA. Atorvastatin restores endothelial function in offspring of protein-restricted rats in a cholesterol-independent manner. Hypertension. 2009; 53:661-667. [PubMed]  [DOI]
84.
Fadini GP, Sartore S, Albiero M, Baesso I, Murphy E, Menegolo M, Grego F, Vigili de Kreutzenberg S, Tiengo A, Agostini C. Number and function of endothelial progenitor cells as a marker of severity for diabetic vasculopathy. Arterioscler Thromb Vasc Biol. 2006; 26:2140-2146. [PubMed]
85.
Ma FX, Zhou B, Chen Z, Ren Q, Lu SH, Sawamura T, Han ZC. Oxidized low density lipoprotein impairs endothelial progenitor cells by regulation of endothelial nitric oxide synthase. J Lipid Res. 2006; 47:1227-1237. [PubMed]  [DOI]
86.
Mohler ER, Shi Y, Moore J, Bantly A, Hamamdzic D, Yoder M, Rader DJ, Putt M, Zhang L, Parmacek M. Diabetes reduces bone marrow and circulating porcine endothelial progenitor cells, an effect ameliorated by atorvastatin and independent of cholesterol. Cytometry A. 2009; 75:75-82. [PubMed]  [DOI]
87.
Assmus B, Urbich C, Aicher A, Hofmann WK, Haendeler J, Rössig L, Spyridopoulos I, Zeiher AM, Dimmeler S. HMG-CoA reductase inhibitors reduce senescence and increase proliferation of endothelial progenitor cells via regulation of cell cycle regulatory genes. Circ Res. 2003; 92:1049-1055. [PubMed]  [DOI]
88.
Rydgren T, Vaarala O, Sandler S. Simvastatin protects against multiple low-dose streptozotocin-induced type 1 diabetes in CD-1 mice and recurrence of disease in nonobese diabetic mice. J Pharmacol Exp Ther. 2007; 323:180-185. [PubMed]  [DOI]
89.
Rydgren T, Sandler S. The protective effect of simvastatin against low dose streptozotocin induced type 1 diabetes in mice is independent of inhibition of HMG-CoA reductase. Biochem Biophys Res Commun. 2009; 379:1076-1079. [PubMed]  [DOI]
90.
Marchand KC, Arany EJ, Hill DJ. Effects of atorvastatin on the regeneration of pancreatic {beta}-cells after streptozotocin treatment in the neonatal rodent. Am J Physiol Endocrinol Metab. 2010; 299:E92-E100. [PubMed]  [DOI]
91.
Favaro E, Miceli I, Bussolati B, Schmitt-Ney M, Cavallo Perin P, Camussi G, Zanone MM. Hyperglycemia induces apoptosis of human pancreatic islet endothelial cells: effects of pravastatin on the Akt survival pathway. Am J Pathol. 2008; 173:442-450. [PubMed]  [DOI]
92.
Konduracka E, Galicka-Latala D, Cieslik G, Rostoff P, Fedak D, Sieradzki J, Naskalski J, Piwowarska W. Effect of atorvastatin on endothelial function and inflammation in long-duration type 1 diabetic patients without coronary heart disease and arterial hypertension. Diabetes Obes Metab. 2008; 10:719-725. [PubMed]  [DOI]
93.
Mullen MJ, Wright D, Donald AE, Thorne S, Thomson H, Deanfield JE. Atorvastatin but not L-arginine improves endothelial function in type I diabetes mellitus: a double-blind study. J Am Coll Cardiol. 2000; 36:410-416. [PubMed]  [DOI]
94.
Dogra GK, Watts GF, Chan DC, Stanton K. Statin therapy improves brachial artery vasodilator function in patients with Type 1 diabetes and microalbuminuria. Diabet Med. 2005; 22:239-242. [PubMed]  [DOI]
95.
Joyce M, Moore K, Thompson C, Fitzgerald P, Fennessy F, Kelly CJ, Bouchier-Hayes DJ. Hydroxy-methylglutaryl-coenzyme A reductase inhibition improves endothelial dysfunction in type-1 diabetes. Eur J Vasc Endovasc Surg. 2004; 27:432-437. [PubMed]  [DOI]
96.
Kolb H, Herder C, Schloot NC, Koenig W, Heise T, Heinemann L, Martin S. Impact of atorvastatin treatment on residual beta cell function in recent-onset type 1 diabetes. Diabetes. 2010; 59:396.