Revisão Acesso aberto Revisado por pares

Progenitor Cell Dysfunctions Underlie Some Diabetic Complications

2015; Elsevier BV; Volume: 185; Issue: 10 Linguagem: Inglês

10.1016/j.ajpath.2015.05.003

ISSN

1525-2191

Autores

Mélanie Rodrigues, Victor W. Wong, Robert C. Rennert, Christopher R. Davis, Michael T. Longaker, Geoffrey C. Gurtner,

Tópico(s)

Metabolism, Diabetes, and Cancer

Resumo

Stem cells and progenitor cells are integral to tissue homeostasis and repair. They contribute to health through their ability to self-renew and commit to specialized effector cells. Recently, defects in a variety of progenitor cell populations have been described in both preclinical and human diabetes. These deficits affect multiple aspects of stem cell biology, including quiescence, renewal, and differentiation, as well as homing, cytokine production, and neovascularization, through mechanisms that are still unclear. More important, stem cell aberrations resulting from diabetes have direct implications on tissue function and seem to persist even after return to normoglycemia. Understanding how diabetes alters stem cell signaling and homeostasis is critical for understanding the complex pathophysiology of many diabetic complications. Moreover, the success of cell-based therapies will depend on a more comprehensive understanding of these deficiencies. This review has three goals: to analyze stem cell pathways dysregulated during diabetes, to highlight the effects of hyperglycemic memory on stem cells, and to define ways of using stem cell therapy to overcome diabetic complications. Stem cells and progenitor cells are integral to tissue homeostasis and repair. They contribute to health through their ability to self-renew and commit to specialized effector cells. Recently, defects in a variety of progenitor cell populations have been described in both preclinical and human diabetes. These deficits affect multiple aspects of stem cell biology, including quiescence, renewal, and differentiation, as well as homing, cytokine production, and neovascularization, through mechanisms that are still unclear. More important, stem cell aberrations resulting from diabetes have direct implications on tissue function and seem to persist even after return to normoglycemia. Understanding how diabetes alters stem cell signaling and homeostasis is critical for understanding the complex pathophysiology of many diabetic complications. Moreover, the success of cell-based therapies will depend on a more comprehensive understanding of these deficiencies. This review has three goals: to analyze stem cell pathways dysregulated during diabetes, to highlight the effects of hyperglycemic memory on stem cells, and to define ways of using stem cell therapy to overcome diabetic complications. Diabetes is characterized by insulin resistance and hyperglycemia, and affects a diverse array of cells, leading to a myriad of tissue complications. These include, but are not limited to, cardiac arrest, stroke, nephropathy, retinopathy, and non-traumatic lower limb amputations.1Centers for Disease Control and PreventionNational diabetes statistics report: estimates of diabetes and its burden in the United States, 2014. U.S. Department of Health and Human Services, Atlanta, GA2014http://www.cdc.gov/diabetes/pubs/statsreport14/national-diabetes-report-web.pdfGoogle Scholar Results from randomized clinical trials indicate that adequate glycemic control in diabetic patients reduces the risk of developing one or several of these complications.2UK Prospective Diabetes Study (UKPDS) Group: Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34).Lancet. 1998; 352: 854-865Abstract Full Text Full Text PDF PubMed Scopus (4830) Google Scholar, 3UK Prospective Diabetes Study (UKPDS) Group: Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33).Lancet. 1998; 352: 837-853Abstract Full Text Full Text PDF PubMed Scopus (13228) Google Scholar, 4Skyler J.S. Bergenstal R. Bonow R.O. Buse J. Deedwania P. Gale E.A. Howard B.V. Kirkman M.S. Kosiborod M. Reaven P. Sherwin R.S. American Diabetes Association, American College of Cardiology Foundation, American Heart AssociationIntensive glycemic control and the prevention of cardiovascular events: implications of the ACCORD, ADVANCE, and VA Diabetes Trials: a position statement of the American Diabetes Association and a Scientific Statement of the American College of Cardiology Foundation and the American Heart Association.J Am Coll Cardiol. 2009; 53: 298-304Abstract Full Text Full Text PDF PubMed Scopus (178) Google Scholar The Diabetes Control and Complications Trial reports a reduction in the development or progression of diabetic nephropathy (50% reduction), neuropathy (60% reduction), and retinopathy (76% reduction) after intensive glycemic control.5The Diabetes Control and Complications Trial Research Group: The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus.N Engl J Med. 1993; 329: 977-986Crossref PubMed Scopus (0) Google Scholar However, 33% of Americans with diabetes remain undiagnosed, approximately 12% of US adults with diabetes exhibit poor glycemic control, and different medical organizations recommend different glycemic targets, increasing the occurrence of diabetic complications.1Centers for Disease Control and PreventionNational diabetes statistics report: estimates of diabetes and its burden in the United States, 2014. U.S. Department of Health and Human Services, Atlanta, GA2014http://www.cdc.gov/diabetes/pubs/statsreport14/national-diabetes-report-web.pdfGoogle Scholar, 6Huang E.S. Liu J.Y. Moffet H.H. John P.M. Karter A.J. Glycemic control, complications, and death in older diabetic patients: the diabetes and aging study.Diabetes Care. 2011; 34: 1329-1336Crossref PubMed Scopus (85) Google Scholar Furthermore, a substantial fraction of patients develop progressive disease despite lowering glycemia, making it critical to study the cellular and molecular modifications that lead to diabetic complications.7Hsu W.C. Lau K.H. Matsumoto M. Moghazy D. Keenan H. King G.L. Improvement of insulin sensitivity by isoenergy high carbohydrate traditional Asian diet: a randomized controlled pilot feasibility study.PLoS One. 2014; 9: e106851Crossref PubMed Google Scholar, 8Tonna S. El-Osta A. Cooper M.E. Tikellis C. Metabolic memory and diabetic nephropathy: potential role for epigenetic mechanisms.Nat Rev Nephrol. 2010; 6: 332-341Crossref PubMed Scopus (40) Google Scholar, 9Yong T.Y. Phillipov G. Phillips P.J. Management outcomes of patients with type 2 diabetes: targeting the 10-year absolute risk of coronary heart disease.Med J Aust. 2007; 186: 622-624PubMed Google Scholar At the tissue level, vascular complications are the most serious manifestations of diabetes.10Tanimura S. Tadokoro Y. Inomata K. Binh N.T. Nishie W. Yamazaki S. Nakauchi H. Tanaka Y. McMillan J.R. Sawamura D. Yancey K. Shimizu H. Nishimura E.K. Hair follicle stem cells provide a functional niche for melanocyte stem cells.Cell Stem Cell. 2011; 8: 177-187Abstract Full Text Full Text PDF PubMed Scopus (60) Google Scholar Closer analysis at the cellular and molecular levels reveals that diabetic complications emerge from alterations in the intracellular signaling of a wider range of cell types. These cellular changes, in turn, arise from variations in the oxidation reduction and glycation state after exposure to hyperglycemia.11Brownlee M. Biochemistry and molecular cell biology of diabetic complications.Nature. 2001; 414: 813-820Crossref PubMed Scopus (4291) Google Scholar, 12Brem H. Tomic-Canic M. Cellular and molecular basis of wound healing in diabetes.J Clin Invest. 2007; 117: 1219-1222Crossref PubMed Scopus (328) Google Scholar Stem cells and progenitor cells are one of the more critical cell types to be affected by the glycemic modulations.13Ferraro F. Lymperi S. Mendez-Ferrer S. Saez B. Spencer J.A. Yeap B.Y. Masselli E. Graiani G. Prezioso L. Rizzini E.L. Mangoni M. Rizzoli V. Sykes S.M. Lin C.P. Frenette P.S. Quaini F. Scadden D.T. Diabetes impairs hematopoietic stem cell mobilization by altering niche function.Sci Transl Med. 2011; 3: 104ra1Crossref Scopus (33) Google Scholar, 14Rennert R.C. Sorkin M. Januszyk M. Duscher D. Kosaraju R. Chung M.T. Lennon J. Radiya-Dixit A. Raghvendra S. Maan Z.N. Hu M.S. Rajadas J. Rodrigues M. Gurtner G.C. Diabetes impairs the angiogenic potential of adipose-derived stem cells by selectively depleting cellular subpopulations.Stem Cell Res Ther. 2014; 5: 79Crossref PubMed Scopus (3) Google Scholar, 15Hernandez S.L. Gong J.H. Chen L. Wu I.H. Sun J.K. Keenan H.A. King G.L. Characterization of circulating and endothelial progenitor cells in patients with extreme-duration type 1 diabetes.Diabetes Care. 2014; 37: 2193-2201Crossref PubMed Google Scholar This review addresses the emerging role of hyperglycemia on stem and progenitor cells, and the subsequent consequence of these changes on specific tissues. Stem cells are the fundamental building blocks of tissue and defined by the ability to self-renew and the capacity to differentiate into progenitor cells that perform specific functions. True stem cells exhibit both these criteria, whereas progenitor cells or transit amplifying cells cannot self-renew in perpetuity. In the adult tissue, hematopoietic stem cells (HSCs) are the only cells known to repopulate the hematopoietic system, making them indispensable for repair and regeneration.16Seita J. Weissman I.L. Hematopoietic stem cell: self-renewal versus differentiation.Wiley Interdiscip Rev Syst Biol Med. 2010; 2: 640-653Crossref PubMed Scopus (92) Google Scholar, 17Oguro H. Ding L. Morrison S.J. SLAM family markers resolve functionally distinct subpopulations of hematopoietic stem cells and multipotent progenitors.Cell Stem Cell. 2013; 13: 102-116Abstract Full Text Full Text PDF PubMed Scopus (50) Google Scholar HSCs reside in the bone marrow, harbored by mesenchymal stromal cells (MSCs) with multipotent differentiation capacity.18Pittenger M.F. Mackay A.M. Beck S.C. Jaiswal R.K. Douglas R. Mosca J.D. Moorman M.A. Simonetti D.W. Craig S. Marshak D.R. Multilineage potential of adult human mesenchymal stem cells.Science. 1999; 284: 143-147Crossref PubMed Scopus (11830) Google Scholar Herein, MSCs are believed to regulate the quiescence, proliferative potential, differentiation fate, and trafficking of HSCs through release of growth factors and chemokines.19Mendez-Ferrer S. Michurina T.V. Ferraro F. Mazloom A.R. Macarthur B.D. Lira S.A. Scadden D.T. Ma'ayan A. Enikolopov G.N. Frenette P.S. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche.Nature. 2010; 466: 829-834Crossref PubMed Scopus (867) Google Scholar, 20Morrison S.J. Scadden D.T. The bone marrow niche for haematopoietic stem cells.Nature. 2014; 505: 327-334Crossref PubMed Scopus (137) Google Scholar MSCs have also been isolated from fetal bone marrow, umbilical cord, placenta, and adipose tissue. They are provasculogenic, and facilitate angiogenesis after injury by functioning as pericytes.21Crisan M. Yap S. Casteilla L. Chen C.W. Corselli M. Park T.S. Andriolo G. Sun B. Zheng B. Zhang L. Norotte C. Teng P.N. Traas J. Schugar R. Deasy B.M. Badylak S. Buhring H.J. Giacobino J.P. Lazzari L. Huard J. Peault B. A perivascular origin for mesenchymal stem cells in multiple human organs.Cell Stem Cell. 2008; 3: 301-313Abstract Full Text Full Text PDF PubMed Scopus (1345) Google Scholar There is consensus on the basis of correlative tissue engineering, in vitro observations, and stem cell niche studies that MSCs contribute to local healing.22Crisan M. Chen C.W. Corselli M. Andriolo G. Lazzari L. Peault B. Perivascular multipotent progenitor cells in human organs.Ann N Y Acad Sci. 2009; 1176: 118-123Crossref PubMed Scopus (93) Google Scholar, 23Chan C.K. Lindau P. Jiang W. Chen J.Y. Zhang L.F. Chen C.C. Seita J. Sahoo D. Kim J.B. Lee A. Park S. Nag D. Gong Y. Kulkarni S. Luppen C.A. Theologis A.A. Wan D.C. DeBoer A. Seo E.Y. Vincent-Tompkins J.D. Loh K. Walmsley G.G. Kraft D.L. Wu J.C. Longaker M.T. Weissman I.L. Clonal precursor of bone, cartilage, and hematopoietic niche stromal cells.Proc Natl Acad Sci U S A. 2013; 110: 12643-12648Crossref PubMed Scopus (23) Google Scholar, 24Chan C.K. Seo E.Y. Chen J.Y. Lo D. McArdle A. Sinha R. Tevlin R. Seita J. Vincent-Tompkins J. Wearda T. Lu W.J. Senarath-Yapa K. Chung M.T. Marecic O. Tran M. Yan K.S. Upton R. Walmsley G.G. Lee A.S. Sahoo D. Kuo C.J. Weissman I.L. Longaker M.T. Identification and specification of the mouse skeletal stem cell.Cell. 2015; 160: 285-298Abstract Full Text Full Text PDF PubMed Scopus (14) Google Scholar However, the presence of MSCs in circulation is disputed.25Tondreau T. Meuleman N. Delforge A. Dejeneffe M. Leroy R. Massy M. Mortier C. Bron D. Lagneaux L. Mesenchymal stem cells derived from CD133-positive cells in mobilized peripheral blood and cord blood: proliferation, Oct4 expression, and plasticity.Stem Cells. 2005; 23: 1105-1112Crossref PubMed Scopus (264) Google Scholar, 26Hoogduijn M.J. Verstegen M.M. Engela A.U. Korevaar S.S. Roemeling-van Rhijn M. Merino A. Franquesa M. de Jonge J. Ijzermans J.N. Weimar W. Betjes M.G. Baan C.C. van der Laan L.J. No evidence for circulating mesenchymal stem cells in patients with organ injury.Stem Cells Dev. 2014; 23: 2328-2335Crossref PubMed Scopus (5) Google Scholar, 27Li S. Huang K.J. Wu J.C. Hu M.S. Sanyal M. Hu M. Longaker M.T. Lorenz H.P. Peripheral blood-derived mesenchymal stem cells: candidate cells responsible for healing critical-sized calvarial bone defects.Stem Cells Transl Med. 2015; 4: 359-368Crossref PubMed Google Scholar Because of the rare distribution of MSCs and the lack of definite markers for their identification in vivo, it remains difficult to determine whether these cells are indispensable for physiological regeneration at distant sites. In addition to HSCs and MSCs, the bone marrow is also reported to contain endothelial precursor cells that circulate and aid in physiological and pathological neovascularization.28Asahara T. Murohara T. Sullivan A. Silver M. van der Zee R. Li T. Witzenbichler B. Schatteman G. Isner J.M. Isolation of putative progenitor endothelial cells for angiogenesis.Science. 1997; 275: 964-967Crossref PubMed Scopus (5569) Google Scholar, 29Asahara T. Masuda H. Takahashi T. Kalka C. Pastore C. Silver M. Kearne M. Magner M. Isner J.M. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization.Circ Res. 1999; 85: 221-228Crossref PubMed Google Scholar Early endothelial precursor cell studies reported migration of these cells toward ischemia after injury, where they initiated vasculogenesis, the process of de novo vessel formation.30Ceradini D.J. Kulkarni A.R. Callaghan M.J. Tepper O.M. Bastidas N. Kleinman M.E. Capla J.M. Galiano R.D. Levine J.P. Gurtner G.C. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1.Nat Med. 2004; 10: 858-864Crossref PubMed Scopus (1458) Google Scholar, 31Schmidt A. Brixius K. Bloch W. Endothelial precursor cell migration during vasculogenesis.Circ Res. 2007; 101: 125-136Crossref PubMed Scopus (64) Google Scholar However, the identity and existence of endothelial precursor cells have since been highly controversial, especially because the cells are derived through inconsistent protocols of expanding peripheral blood mononuclear cells. It is suggested, on the basis of discrepancies in surface marker identity, that these cells either do not circulate or are most likely monocytes or macrophages.32Rehman J. Li J. Orschell C.M. March K.L. Peripheral blood "endothelial progenitor cells" are derived from monocyte/macrophages and secrete angiogenic growth factors.Circulation. 2003; 107: 1164-1169Crossref PubMed Scopus (1174) Google Scholar, 33Purhonen S. Palm J. Rossi D. Kaskenpaa N. Rajantie I. Yla-Herttuala S. Alitalo K. Weissman I.L. Salven P. Bone marrow-derived circulating endothelial precursors do not contribute to vascular endothelium and are not needed for tumor growth.Proc Natl Acad Sci U S A. 2008; 105: 6620-6625Crossref PubMed Scopus (258) Google Scholar Stem cells have the unique ability to reside in a quiescent G0 phase. Injury and tissue loss triggers their activation, and the cells enter the G1 phase of the cell cycle, where they commit to either self-renewal or differentiation.34Cheung T.H. Rando T.A. Molecular regulation of stem cell quiescence.Nat Rev Mol Cell Biol. 2013; 14: 329-340Crossref PubMed Scopus (85) Google Scholar Between the G0 and G1 phases of the cell cycle exists a newly described reversible phase of quiescence called the Galert phase that is proposed to prime stem cells for either renewal or differentiation.35Rodgers J.T. King K.Y. Brett J.O. Cromie M.J. Charville G.W. Maguire K.K. Brunson C. Mastey N. Liu L. Tsai C.R. Goodell M.A. Rando T.A. mTORC1 controls the adaptive transition of quiescent stem cells from G0 to G(Alert).Nature. 2014; 510: 393-396PubMed Google Scholar Imbalance within these states can have pathological consequences on the body's ability to repair injured tissues.36Rossi L. Lin K.K. Boles N.C. Yang L. King K.Y. Jeong M. Mayle A. Goodell M.A. Less is more: unveiling the functional core of hematopoietic stem cells through knockout mice.Cell Stem Cell. 2012; 11: 302-317Abstract Full Text Full Text PDF PubMed Scopus (47) Google Scholar, 37Beerman I. Seita J. Inlay M.A. Weissman I.L. Rossi D.J. Quiescent hematopoietic stem cells accumulate DNA damage during aging that is repaired upon entry into cell cycle.Cell Stem Cell. 2014; 15: 37-50Abstract Full Text Full Text PDF PubMed Scopus (35) Google Scholar, 38Malam Z. Cohn R.D. Stem cells on alert: priming quiescent stem cells after remote injury.Cell Stem Cell. 2014; 15: 7-8Abstract Full Text Full Text PDF PubMed Google Scholar There is an increasing emphasis on glucose metabolism in determining stem cell fate.39Wang Y.H. Israelsen W.J. Lee D. Yu V.W. Jeanson N.T. Clish C.B. Cantley L.C. Vander Heiden M.G. Scadden D.T. Cell-state-specific metabolic dependency in hematopoiesis and leukemogenesis.Cell. 2014; 158: 1309-1323Abstract Full Text Full Text PDF PubMed Scopus (9) Google Scholar A stem cell in an undifferentiated state, a progenitor committed to differentiation, and a terminally differentiated cell are expected to possess varying metabolic demands. Thus, stem cells would benefit from flexibility in metabolic pathways, with a balance of anabolic processes for building and catabolic processes to ensure supply of bioenergetics resources. Complete consumption of substrates, including glucose, that occur during oxidative phosphorylation may be insufficient to support the energy requirements of the stem cell. Instead, stem cells are postulated to prefer glycolysis, which allows for partial breakdown of glucose and shunting of intermediates through the pentose phosphate pathway. This allows for both the catabolic process of ATP generation and the production of substrates for anabolic processes.40Folmes C.D. Dzeja P.P. Nelson T.J. Terzic A. Metabolic plasticity in stem cell homeostasis and differentiation.Cell Stem Cell. 2012; 11: 596-606Abstract Full Text Full Text PDF PubMed Scopus (97) Google Scholar, 41Folmes C.D. Nelson T.J. Martinez-Fernandez A. Arrell D.K. Lindor J.Z. Dzeja P.P. Ikeda Y. Perez-Terzic C. Terzic A. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming.Cell Metab. 2011; 14: 264-271Abstract Full Text Full Text PDF PubMed Scopus (189) Google Scholar When stem cells switch the bulk of their ATP generation from glycolysis to oxidative phosphorylation, they are found to undergo differentiation.41Folmes C.D. Nelson T.J. Martinez-Fernandez A. Arrell D.K. Lindor J.Z. Dzeja P.P. Ikeda Y. Perez-Terzic C. Terzic A. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming.Cell Metab. 2011; 14: 264-271Abstract Full Text Full Text PDF PubMed Scopus (189) Google Scholar, 42Zhang J. Khvorostov I. Hong J.S. Oktay Y. Vergnes L. Nuebel E. Wahjudi P.N. Setoguchi K. Wang G. Do A. Jung H.J. McCaffery J.M. Kurland I.J. Reue K. Lee W.N. Koehler C.M. Teitell M.A. UCP2 regulates energy metabolism and differentiation potential of human pluripotent stem cells.EMBO J. 2011; 30: 4860-4873Crossref PubMed Scopus (102) Google Scholar Similarly, the balance between ATP/AMP ratios has been found to control the transition between quiescence and proliferation.43Gurumurthy S. Xie S.Z. Alagesan B. Kim J. Yusuf R.Z. Saez B. Tzatsos A. Ozsolak F. Milos P. Ferrari F. Park P.J. Shirihai O.S. Scadden D.T. Bardeesy N. The Lkb1 metabolic sensor maintains haematopoietic stem cell survival.Nature. 2010; 468: 659-663Crossref PubMed Scopus (151) Google Scholar Therefore, maintaining a balance between glycolysis and oxidative phosphorylation offers a potential strategy to deterministically alter the fate of downstream proliferation versus differentiation and improve homeostasis and regeneration. Regulations of these pathways have implications during hyperglycemia and diabetes. Other nutrient- and glucose-sensing mechanisms detailing the exit from quiescence and the activation of stem cells are being actively investigated. Induction of autophagy, the process of self-eating of cellular components, is emerging as one of the alternate mechanisms by which stem cells rapidly meet their bioenergetics demands and get activated. This mode of activation has been studied in embryonic stem cells, HSCs, MSCs, and neural stem cells (NSCs).44Tang A.H. Rando T.A. Induction of autophagy supports the bioenergetic demands of quiescent muscle stem cell activation.EMBO J. 2014; 33: 2782-2797Crossref PubMed Scopus (10) Google Scholar, 45Nuschke A. Rodrigues M. Stolz D.B. Chu C.T. Griffith L. Wells A. Human mesenchymal stem cells/multipotential stromal cells consume accumulated autophagosomes early in differentiation.Stem Cell Res Ther. 2014; 5: 140Crossref PubMed Google Scholar, 46Wang C. Liang C.C. Bian Z.C. Zhu Y. Guan J.L. FIP200 is required for maintenance and differentiation of postnatal neural stem cells.Nat Neurosci. 2013; 16: 532-542Crossref PubMed Scopus (5) Google Scholar Correlative studies demonstrate that aging and diabetes perturb and inhibit the autophagic machinery of cells.47Rubinsztein D.C. Marino G. Kroemer G. Autophagy and aging.Cell. 2011; 146: 682-695Abstract Full Text Full Text PDF PubMed Scopus (419) Google Scholar, 48Yang L. Li P. Fu S. Calay E.S. Hotamisligil G.S. Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance.Cell Metab. 2010; 11: 467-478Abstract Full Text Full Text PDF PubMed Scopus (262) Google Scholar This would suggest that a specific disruption of the autophagic signaling in stem cells in response to the diabetic state might contribute to the dysfunctional stem cell phenotype. Diabetes is associated with a myriad of tissue-specific complications. These include, but are not limited to, renal failure, blindness, neuropathy, atherosclerosis, and cardiac failure. Diabetes also results in generic complications, such as impaired neovascularization and microvascular complications, resulting in ulcers and chronic nonhealing wounds. There is increasing evidence that suggests that stem cell dysfunction underlies some of these complications. Exposure to hyperglycemia is found to produce both stem cell deficits and alterations in the tissue microenvironment surrounding the stem cell.49Ceradini D.J. Gurtner G.C. Homing to hypoxia: HIF-1 as a mediator of progenitor cell recruitment to injured tissue.Trends Cardiovasc Med. 2005; 15: 57-63Abstract Full Text Full Text PDF PubMed Scopus (218) Google Scholar, 50Thangarajah H. Vial I.N. Grogan R.H. Yao D. Shi Y. Januszyk M. Galiano R.D. Chang E.I. Galvez M.G. Glotzbach J.P. Wong V.W. Brownlee M. Gurtner G.C. HIF-1alpha dysfunction in diabetes.Cell Cycle. 2010; 9: 75-79Crossref PubMed Google Scholar, 51Capla J.M. Grogan R.H. Callaghan M.J. Galiano R.D. Tepper O.M. Ceradini D.J. Gurtner G.C. Diabetes impairs endothelial progenitor cell-mediated blood vessel formation in response to hypoxia.Plast Reconstr Surg. 2007; 119: 59-70Crossref PubMed Scopus (61) Google Scholar, 52El-Ftesi S. Chang E.I. Longaker M.T. Gurtner G.C. Aging and diabetes impair the neovascular potential of adipose-derived stromal cells.Plast Reconstr Surg. 2009; 123: 475-485Crossref PubMed Scopus (0) Google Scholar, 53Lerman O.Z. Galiano R.D. Armour M. Levine J.P. Gurtner G.C. Cellular dysfunction in the diabetic fibroblast: impairment in migration, vascular endothelial growth factor production, and response to hypoxia.Am J Pathol. 2003; 162: 303-312Abstract Full Text Full Text PDF PubMed Google Scholar These impairments include, but are not limited to, changes in migration, recruitment, survival, self-renewal, and differentiation capacity (Figure 1). This section details the changes produced by hyperglycemia in stem cells of the bone marrow, nervous system, bone, and heart. Diabetes produces a global remodeling of the bone marrow. From a stem cell standpoint, there is decreased efflux of diabetic progenitors from the marrow and reduced vasculogenic potential of these cells.54Fadini G.P. Albiero M. Vigili de Kreutzenberg S. Boscaro E. Cappellari R. Marescotti M. Poncina N. Agostini C. Avogaro A. Diabetes impairs stem cell and proangiogenic cell mobilization in humans.Diabetes Care. 2013; 36: 943-949Crossref PubMed Scopus (38) Google Scholar Murine models of both type 1 and type 2 diabetes confirm this impaired egress of bone marrow progenitors after injury.13Ferraro F. Lymperi S. Mendez-Ferrer S. Saez B. Spencer J.A. Yeap B.Y. Masselli E. Graiani G. Prezioso L. Rizzini E.L. Mangoni M. Rizzoli V. Sykes S.M. Lin C.P. Frenette P.S. Quaini F. Scadden D.T. Diabetes impairs hematopoietic stem cell mobilization by altering niche function.Sci Transl Med. 2011; 3: 104ra1Crossref Scopus (33) Google Scholar, 55Januszyk M. Sorkin M. Glotzbach J.P. Vial I.N. Maan Z.N. Rennert R.C. Duscher D. Thangarajah H. Longaker M.T. Butte A.J. Gurtner G.C. Diabetes irreversibly depletes bone marrow-derived mesenchymal progenitor cell subpopulations.Diabetes. 2014; 63: 3047-3056Crossref PubMed Scopus (3) Google Scholar Furthermore, clinical studies demonstrate that diabetes causes a reduction of hematopoietic tissue, increased fat deposition, and microvascular rarefaction in the bone marrow.56Spinetti G. Cordella D. Fortunato O. Sangalli E. Losa S. Gotti A. Carnelli F. Rosa F. Riboldi S. Sessa F. Avolio E. Beltrami A.P. Emanueli C. Madeddu P. Global remodeling of the vascular stem cell niche in bone marrow of diabetic patients: implication of the microRNA-155/FOXO3a signaling pathway.Circ Res. 2013; 112: 510-522Crossref PubMed Scopus (31) Google Scholar These changes have a cumulative negative outcome on progenitor cells. More important, diabetic patients with satisfactory glycemic control display more circulatory progenitors than patients with poor glycemic control, but fewer progenitors compared with nondiabetics.57Yue W.S. Lau K.K. Siu C.W. Wang M. Yan G.H. Yiu K.H. Tse H.F. Impact of glycemic control on circulating endothelial progenitor cells and arterial stiffness in patients with type 2 diabetes mellitus.Cardiovasc Diabetol. 2011; 10: 113Crossref PubMed Scopus (29) Google Scholar This suggests that normalizing glucose levels after hyperglycemia may not be sufficient to normalize tissue homeostasis. The effect of diabetes on stem cells is not limited to the adult tissue alone. Maternal diabetes produces a hyperglycemic environment for the developing embryo and has been linked to congenital malformations in various tissues, including the nervous system. NSCs are self-renewing multipotent cells that produce neurons and glia (astrocytes and oligodendrocytes) of the central nervous system. Exposure of the fetus to high glucose has been found to impair NSCs in the developing brain, altering cell fate and producing neural tube defects. These include spina bifida, anencephaly, craniorachischisis, and encephalocele.58Zabihi S. Loeken M.R. Understanding diabetic teratogenesis: where are we now and where are we going?.Birth Defects Res A Clin Mol Teratol. 2010; 88: 779-790Crossref PubMed Google Scholar Specifically, exposure to high glucose produces epigenetic changes in NSCs: histone H3K9 trimethylation, DNA methylation, and decreased histone H3K9 acetylation. These changes alter genes, such as Dcx and Pafah1b1, which regulate neural migration and microtubule formation in the cortex. Neurogenesis and neural migration are affected by these changes.59Shyamasundar S. Jadhav S.P. Bay B.H. Tay S.S. Kumar S.D. Rangasamy D. Dheen S.T. Analysis of epigenetic factors in mouse embryonic neural stem cells exposed to hyperglycemia.PLoS One. 2013; 8: e65945Crossref PubMed Scopus (6) Google Scholar Hyperglycemia also induces reactive oxygen species (ROS) production and intracellular oxidative stress in NSCs, altering the balance between their proliferation and apoptosis.60Fu J. Tay S.S. Ling E.A. Dheen S.T. Aldose reductase is implicated in high glucose-induced oxidative stress in mouse embryonic neural stem cells.J Neurochem. 2007; 103: 1654-1665Crossref PubMed Scopus (9) Google Scholar Reduction of ROS is, therefore, one strategy to overcome NSC dysfunction during fetal development in the presence of high glucose. In the bone microenvironment, after injury, diabetes has been found to impair blood supply to the fracture site undermining bony union. Diabetes also leads to increased bone resorption and osteopenia, significantly increasing the risk of fractures.61Vestergaard P. Rejnmark L. Mosekilde L. Relative fracture risk in patients with diabetes mellitus, and the impact of insulin and oral antidiabetic medication on relative fracture risk.Diabetologia. 2005; 48: 1292-1299Crossref PubMed Scopus (171) Google Scholar Increased reactive oxygen

Referência(s)