1
|
Menger MM, Nalbach L, Roma LP, Laschke MW, Menger MD, Ampofo E. Erythropoietin exposure of isolated pancreatic islets accelerates their revascularization after transplantation. Acta Diabetol 2021; 58:1637-1647. [PMID: 34254190 PMCID: PMC8542558 DOI: 10.1007/s00592-021-01760-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/06/2021] [Indexed: 12/15/2022]
Abstract
AIMS The exposure of isolated pancreatic islets to pro-angiogenic factors prior to their transplantation represents a promising strategy to accelerate the revascularization of the grafts. It has been shown that erythropoietin (EPO), a glycoprotein regulating erythropoiesis, also induces angiogenesis. Therefore, we hypothesized that EPO exposure of isolated islets improves their posttransplant revascularization. METHODS Flow cytometric, immunohistochemical and quantitative real-time (qRT)-PCR analyses were performed to study the effect of EPO on the viability, cellular composition and gene expression of isolated islets. Moreover, islets expressing a mitochondrial or cytosolic H2O2 sensor were used to determine reactive oxygen species (ROS) levels. The dorsal skinfold chamber model in combination with intravital fluorescence microscopy was used to analyze the revascularization of transplanted islets. RESULTS We found that the exposure of isolated islets to EPO (3 units/mL) for 24 h does not affect the viability and the production of ROS when compared to vehicle-treated and freshly isolated islets. However, the exposure of islets to EPO increased the number of CD31-positive cells and enhanced the gene expression of insulin and vascular endothelial growth factor (VEGF)-A. The revascularization of the EPO-cultivated islets was accelerated within the initial phase after transplantation when compared to both controls. CONCLUSION These findings indicate that the exposure of isolated islets to EPO may be a promising approach to improve clinical islet transplantation.
Collapse
Affiliation(s)
- Maximilian M Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
- Department of Trauma and Reconstructive Surgery, Faculty of Medicine, BG Hospital Tuebingen, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Leticia P Roma
- Biophysics Department, Center for Human and Molecular Biology, Saarland University, 66421, Homburg, Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| |
Collapse
|
2
|
Kazmierak W, Korolczuk A, Kurzepa J, Czechowska G, Boguszewska-Czubara A, Madro A. The influence of erythropoietin on apoptosis and fibrosis in the early phase of chronic pancreatitis in rats. Arch Med Sci 2021; 17:1100-1108. [PMID: 34336038 PMCID: PMC8314426 DOI: 10.5114/aoms.2020.99800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 08/01/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Chronic pancreatitis (CP) is a continuing, inflammatory process of the pancreas, characterised by irreversible morphological changes. The identification of pancreatic stellate cells resulted in the development of research on the pathogenesis of CP. Erythropoietin (Epo) regulates the interaction between apoptosis and inflammation of the brain, kidney, and heart muscle. Erythropoietin receptors were also found in the pancreas, in particular on the islet cells. Our objective was to evaluate the influence of Epo on fibrosis and apoptosis in experimental CP. MATERIAL AND METHODS The experiments were performed on 48 male Wistar rats (250-350 g). The animals were divided into six equal groups (I - control, II - chronic cerulein - induced pancreatitis, III - 1 ml of Epo sc, IV - 0.5 ml of Epo sc, V - CP treated with 1 ml Epo, VI - CP treated with 0.5 ml Epo). The blood for gelatinases and pancreata for the morphological examinations and immunohistochemistry were collected. RESULTS A slight reduction of interstitial oedema and less severe fibrosis were noticed in the groups treated with Epo. Reduced expression of caspase-3 and α-actin, and a lack of Bcl-2 expression were observed in areas with inflammation. There was no expression of caspase-9 observed in all groups. There were no statistically significant differences between the groups in the activity of gelatinases. CONCLUSIONS Erythropoietin seems to have the effect of reducing fibrosis and apoptosis in an experimental model of CP.
Collapse
Affiliation(s)
- Weronika Kazmierak
- Department of Gastroenterology with Endoscopic Unit, Medical University of Lublin, Lublin, Poland
| | - Agnieszka Korolczuk
- Department of Clinical Pathomorphology, Medical University of Lublin, Lublin, Poland
| | - Jacek Kurzepa
- Department of Medicinal Chemistry, Medical University of Lublin, Lublin, Poland
| | - Grażyna Czechowska
- Department of Gastroenterology with Endoscopic Unit, Medical University of Lublin, Lublin, Poland
| | | | - Agnieszka Madro
- Department of Gastroenterology with Endoscopic Unit, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
3
|
Golmohammadi MG, Ajam R, Shahbazi A, Chinifroush-Asl MM, Banaei S. Protective effect of vitamin D3 and erythropoietin on renal ischemia/reperfusion-induced liver and kidney damage in rats. JOURNAL OF HERBMED PHARMACOLOGY 2020. [DOI: 10.34172/jhp.2020.37] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: Renal ischemia reperfusion (IR) contributes to the development of acute renal failure (ARF). Free radicals are considered to be principal components involved in the pathophysiological alterations observed during IR. In this study, we evaluated the effects of vitamin D and erythropoietin (EPO) in IR–induced renal and liver damage. Methods: Wistar rats were divided into five groups of 6 each. 1) The control, 2) IR, 3) VD3 (1,25-dihydroxyvitamin D3) + IR, 4) EPO+ IR, and 5) VD3+EPO+ IR groups. The rats were unilaterally nephrectomized and subjected to 45 minutes of renal pedicle occlusion followed by 24 h reperfusion. Vitamin D (10 mg/kg, IP) and EPO (1000 U/kg, IP) were administered prior to ischemia. After 24 hours reperfusion, the blood samples were collected for the determination of biochemical parameters and kidney and liver samples were taken for histological studies. Results: Renal ischemia significantly decreased kidney and liver functions. IR significantly increased blood urea nitrogen-creatinine (BUN-Cr), glucose, total protein and liver enzyme levels and significantly decreased hemoglobin (Hb) and hematocrit (Hct) values. Histopathological findings of the IR group confirmed that there were glomerular atrophy and acute tubular necrosis in the renal tissues and lymphocyte infiltration in the liver samples. Treatment with vitamin D and EPO boosted liver and kidney functions and improved the morphological changes. Conclusion: It seems that vitamin D or EPO administration could protect the kidney and liver damage induced by IR. Also, the combination of vitamin D and EPO may exert more beneficial effects than either agent used alone.
Collapse
Affiliation(s)
| | - Reza Ajam
- General Practitioner, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Shahbazi
- General Practitioner, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Shokofeh Banaei
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
4
|
Menger MM, Nalbach L, Roma LP, Körbel C, Wrublewsky S, Glanemann M, Laschke MW, Menger MD, Ampofo E. Erythropoietin accelerates the revascularization of transplanted pancreatic islets. Br J Pharmacol 2020; 177:1651-1665. [PMID: 31721150 DOI: 10.1111/bph.14925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 10/30/2019] [Accepted: 11/03/2019] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Pancreatic islet transplantation is a promising therapeutic approach for Type 1 diabetes. A major prerequisite for the survival of grafted islets is a rapid revascularization after transplantation. Erythropoietin (EPO), the primary regulator of erythropoiesis, has been shown to promote angiogenesis. Therefore, we investigated in this study whether EPO improves the revascularization of transplanted islets. EXPERIMENTAL APPROACH Islets from FVB/N mice were transplanted into dorsal skinfold chambers of recipient animals, which were daily treated with an intraperitoneal injection of EPO (500 IU·kg-1 ) or vehicle (control) throughout an observation period of 14 days. In a second set of experiments, animals were only pretreated with EPO over a 6-day period prior to islet transplantation. The revascularization of the grafts was assessed by repetitive intravital fluorescence microscopy and immunohistochemistry. In addition, a streptozotocin-induced diabetic mouse model was used to study the effect of EPO-pretreatment on the endocrine function of the grafts. KEY RESULTS EPO treatment slightly accelerated the revascularization of the islet grafts. This effect was markedly more pronounced in EPO-pretreated animals, resulting in significantly higher numbers of engrafted islets and an improved perfusion of endocrine tissue without affecting systemic haematocrit levels when compared with controls. Moreover, EPO-pretreatment significantly accelerated the recovery of normoglycaemia in diabetic mice after islet transplantation. CONCLUSION AND IMPLICATIONS These findings demonstrate that, particularly, short-term EPO-pretreatment represents a promising therapeutic approach to improve the outcome of islet transplantation, without an increased risk of thromboembolic events.
Collapse
Affiliation(s)
- Maximilian M Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Leticia P Roma
- Biophysics Department, Center for Human and Molecular Biology, Saarland University, Homburg/Saar, Germany
| | - Christina Körbel
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias Glanemann
- Department for General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
5
|
Montesanto A, Bonfigli AR, De Luca M, Crocco P, Garagnani P, Marasco E, Pirazzini C, Giuliani C, Romagnoli F, Franceschi C, Passarino G, Testa R, Olivieri F, Rose G. Erythropoietin (EPO) haplotype associated with all-cause mortality in a cohort of Italian patients with Type-2 Diabetes. Sci Rep 2019; 9:10395. [PMID: 31316151 PMCID: PMC6637129 DOI: 10.1038/s41598-019-46894-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 06/27/2019] [Indexed: 01/04/2023] Open
Abstract
Type-2 Diabetes (T2D), diabetic complications, and their clinical risk factors harbor a substantial genetic component but the genetic factors contributing to overall diabetes mortality remain unknown. Here, we examined the association between genetic variants at 21 T2D-susceptibility loci and all-cause mortality in an elderly cohort of 542 Italian diabetic patients who were followed for an average of 12.08 years. Univariate Cox regression analyses detected age, waist-to-hip ratio (WHR), glycosylated haemoglobin (HbA1c), diabetes duration, retinopathy, nephropathy, chronic kidney disease (CKD), and anaemia as predictors of all-cause mortality. When Cox proportional hazards multivariate models adjusted for these factors were run, three erythropoietin (EPO) genetic variants in linkage disequilibrium (LD) with each other (rs1617640-T/G, rs507392-T/C and rs551238-A/C) were significantly (False Discovery Rate < 0.1) associated with mortality. Haplotype multivariate analysis revealed that patients carrying the G-C-C haplotype have an increased probability of survival, while an opposite effect was observed among subjects carrying the T-T-A haplotype. Our findings provide evidence that the EPO gene is an independent predictor of mortality in patients with T2D. Thus, understanding the mechanisms by which the genetic variability of EPO affects the mortality of T2D patients may provide potential targets for therapeutic interventions to improve the survival of these patients.
Collapse
Affiliation(s)
- Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, Italy
| | | | - Maria De Luca
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Paolina Crocco
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, Italy.,Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden
| | - Elena Marasco
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Chiara Pirazzini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Cristina Giuliani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Fabio Romagnoli
- Diabetology Unit, IRCCS INRCA, National Institute, Ancona, Italy
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, Italy
| | - Roberto Testa
- Clinical Laboratory and Molecular Diagnostics, IRCCS INRCA, Ancona, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, National Institute IRCCS INRCA, Ancona, Italy
| | - Giuseppina Rose
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Rende, Italy.
| |
Collapse
|
6
|
Erythropoietin-induced cytoprotection in intestinal epithelial cells is linked to system Xc<sup/>. Exp Cell Res 2017; 352:202-206. [PMID: 28167131 DOI: 10.1016/j.yexcr.2017.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 01/28/2017] [Accepted: 02/03/2017] [Indexed: 02/06/2023]
Abstract
Necrotizing enterocolitis is a common but serious complication among premature babies. Currently, there are limited treatment options. These include intensive supportive care and surgical intervention. In this study, we hypothesize that erythropoietin (Epo) could be protective against cell necrosis by increasing the levels of glutathione. This can be regulated by increasing the activity of system xC-. This was demonstrated using intestinal epithelial cells (IEC-6) as a model system. S4-CPG and sulfasalazine pharmacologically inhibit xCT, which induced cell death. Our data showed a dose dependent decrease in cell viability when treated with both inhibitors. In addition, the IEC-6 cells displayed a dose dependent increase when treated with Epo. In conclusion, Epo can be protective against cell death and ultimately be considered as a treatment option for intestinal epithelial cell death.
Collapse
|
7
|
A Nonhematopoietic Erythropoietin Analogue, ARA 290, Inhibits Macrophage Activation and Prevents Damage to Transplanted Islets. Transplantation 2016; 100:554-62. [PMID: 26683514 DOI: 10.1097/tp.0000000000001026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Erythropoietin exerts anti-inflammatory, antiapoptotic, and cytoprotective effects in addition to its hematopoietic action. A nonhematopoietic erythropoietin analogue, ARA 290, has similar properties. The efficacy of pancreatic islet transplantation (PITx) is reduced due to islet damage that occurs during isolation and from the severe inflammatory reactions caused by the transplantation procedure. We investigated whether ARA 290 protects islets and ameliorates inflammatory responses following PITx thus improving engraftment. METHODS The effects of ARA 290 on pancreatic islets of C57BL/6J (H-2) mice and on murine macrophages were investigated using an in vitro culture model. As a marginal PITx, 185 islets were transplanted into the liver of streptozotocin-induced diabetic mice (H-2) via the portal vein. Recipients were given ARA 290 (120 μg/kg) intraperitoneally just before and at 0, 6, and 24 hours after PITx. Liver samples were obtained at 12 hours after PITx, and expression levels of proinflammatory cytokines were assessed. RESULTS ARA 290 protected islets from cytokine-induced damage and apoptosis. Secretion of pro-inflammatory cytokines (IL-6, IL-12, and TNF-α) from macrophages was significantly inhibited by ARA 290. After the marginal PITx, ARA 290 treatment significantly improved the blood glucose levels when compared to those of control animals (P < 0.001). Upregulation of monocyte chemoattractant protein-1, macrophage inflammatory protein-1β, IL-1β, and IL-6 messenger RNA expression within the liver was suppressed by ARA 290 treatment. CONCLUSIONS ARA 290 protected pancreatic islets from cytokine-induced damage and apoptosis and ameliorated the inflammatory response after PITx. ARA 290 appears to be a promising candidate for improvement of PITx.
Collapse
|
8
|
Chen LN, Sun Q, Liu SQ, Hu H, Lv J, Ji WJ, Wang M, Chen MX, Zhou J. Erythropoietin improves glucose metabolism and pancreatic β-cell damage in experimental diabetic rats. Mol Med Rep 2015; 12:5391-8. [PMID: 26126591 DOI: 10.3892/mmr.2015.4006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 06/03/2015] [Indexed: 11/05/2022] Open
Abstract
Previous studies have implicated erythropoietin (EPO) signaling in the regulation of glucose metabolism. Whether EPO can be used treat diabetes and the underlying mechanism remain to be elucidated. The present study aimed to investigate whether EPO affects glucose metabolism, and the underlying mechanisms, in experimental diabetic rats. The effects of EPO (300 U/kg three times a week for 4 weeks) on glucose metabolism, hematopoietic function, blood selenium content and the ultrastructure of pancreatic β‑cells were investigated in low dose (25 mg/kg body weight) streptozotocin‑induced experimental diabetic rats provided with a high‑fat diet. The results demonstrated that EPO significantly decreased the fasting blood glucose, the area under the curve of the oral glucose tolerance and insulin tolerance tests and L‑alanine gluconeogenesis. Ultrastructural examination of the pancreatic islets revealed that EPO prevented the dysfunction of pancreatic β‑cells in experimental diabetic rats, ameliorated cytoplasmic vacuolation and fragmentation of mitochondria, and increased the number of secretory granules. EPO administration increased the activities of superoxide dismutase and glutathione peroxidase, and decreased the level of malondialdehyde. Additionally, EPO increased blood selenium in the diabetic rats and produced a hematopoietic effect. These results indicated that EPO modulated glucose metabolism and improved pancreatic β‑cells damage by increasing anti‑oxidation. The detailed mechanisms underlying these effects require further investigation.
Collapse
Affiliation(s)
- Li-Na Chen
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| | - Qiang Sun
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| | - Shu-Qing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| | - Juan Lv
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| | - Wen-Jun Ji
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| | - Meng Wang
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| | - Ming-Xia Chen
- Department of Electron Microscopy Room, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| | - Jun Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xian, Shaanxi 710061, P.R. China
| |
Collapse
|
9
|
Wang L, Di L, Noguchi CT. Erythropoietin, a novel versatile player regulating energy metabolism beyond the erythroid system. Int J Biol Sci 2014; 10:921-39. [PMID: 25170305 PMCID: PMC4147225 DOI: 10.7150/ijbs.9518] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 06/04/2014] [Indexed: 12/12/2022] Open
Abstract
Erythropoietin (EPO), the required cytokine for promoting the proliferation and differentiation of erythroid cells to stimulate erythropoiesis, has been reported to act as a pleiotropic cytokine beyond hematopoietic system. The various activities of EPO are determined by the widespread distribution of its cell surface EPO receptor (EpoR) in multiple tissues including endothelial, neural, myoblasts, adipocytes and other cell types. EPO activity has been linked to angiogenesis, neuroprotection, cardioprotection, stress protection, anti-inflammation and especially the energy metabolism regulation that is recently revealed. The investigations of EPO activity in animals and the expression analysis of EpoR provide more insights on the potential of EPO in regulating energy metabolism and homeostasis. The findings of crosstalk between EPO and some important energy sensors and the regulation of EPO in the cellular respiration and mitochondrial function further provide molecular mechanisms for EPO activity in metabolic activity regulation. In this review, we will summarize the roles of EPO in energy metabolism regulation and the activity of EPO in tissues that are tightly associated with energy metabolism. We will also discuss the effects of EPO in regulating oxidative metabolism and mitochondrial function, the interactions between EPO and important energy regulation factors, and the protective role of EPO from stresses that are related to metabolism, providing a brief overview of previously less appreciated EPO biological function in energy metabolism and homeostasis.
Collapse
Affiliation(s)
- Li Wang
- 1. Faculty of Health Sciences, University of Macau, SAR of People's Republic of China
| | - Lijun Di
- 1. Faculty of Health Sciences, University of Macau, SAR of People's Republic of China
| | - Constance Tom Noguchi
- 2. Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, U.S.A
| |
Collapse
|
10
|
Abstract
The hematopoietic growth factor erythropoietin (Epo) circulates in plasma and controls the oxygen carrying capacity of the blood (Fisher. Exp Biol Med (Maywood) 228:1-14, 2003). Epo is produced primarily in the adult kidney and fetal liver and was originally believed to play a role restricted to stimulation of early erythroid precursor proliferation, inhibition of apoptosis, and differentiation of the erythroid lineage. Early studies showed that mice with targeted deletion of Epo or the Epo receptor (EpoR) show impaired erythropoiesis, lack mature erythrocytes, and die in utero around embryonic day 13.5 (Wu et al. Cell 83:59-67, 1995; Lin et al. Genes Dev. 10:154-164, 1996). These animals also exhibited heart defects, abnormal vascular development as well as increased apoptosis in the brain suggesting additional functions for Epo signaling in normal development of the central nervous system and heart. Now, in addition to its well-known role in erythropoiesis, a diverse array of cells have been identified that produce Epo and/or express the Epo-R including endothelial cells, smooth muscle cells, and cells of the central nervous system (Masuda et al. J Biol Chem. 269:19488-19493, 1994; Marti et al. Eur J Neurosci. 8:666-676, 1996; Bernaudin et al. J Cereb Blood Flow Metab. 19:643-651, 1999; Li et al. Neurochem Res. 32:2132-2141, 2007). Endogenously produced Epo and/or expression of the EpoR gives rise to autocrine and paracrine signaling in different organs particularly during hypoxia, toxicity, and injury conditions. Epo has been shown to regulate a variety of cell functions such as calcium flux (Korbel et al. J Comp Physiol B. 174:121-128, 2004) neurotransmitter synthesis and cell survival (Velly et al. Pharmacol Ther. 128:445-459, 2010; Vogel et al. Blood. 102:2278-2284, 2003). Furthermore Epo has neurotrophic effects (Grimm et al. Nat Med. 8:718-724, 2002; Junk et al. Proc Natl Acad Sci U S A. 99:10659-10664, 2002), can induce an angiogenic phenotype in cultured endothelial cells and is a potent angiogenic factor in vivo (Ribatti et al. Eur J Clin Invest. 33:891-896, 2003) and might enhance ventilation in hypoxic conditions (Soliz et al. J Physiol. 568:559-571, 2005; Soliz et al. J Physiol. 583, 329-336, 2007). Thus multiple functions have been identified breathing new life and exciting possibilities into what is really an old growth factor.This review will address the function of Epo in non-hematopoietic tissues with significant emphasis on the brain and heart.
Collapse
Affiliation(s)
- Omolara O Ogunshola
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
11
|
Murua A, Orive G, Hernández RM, Pedraz JL. Emerging technologies in the delivery of erythropoietin for therapeutics. Med Res Rev 2011; 31:284-309. [PMID: 19967731 DOI: 10.1002/med.20184] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Deciphering the function of proteins and their roles in signaling pathways is one of the main goals of biomedical research, especially from the perspective of uncovering pathways that may ultimately be exploited for therapeutic benefit. Over the last half century, a greatly expanded understanding of the biology of the glycoprotein hormone erythropoietin (Epo) has emerged from regulator of the circulating erythrocyte mass to a widely used therapeutic agent. Originally viewed as the renal hormone responsible for erythropoiesis, recent in vivo studies in animal models and clinical trials demonstrate that many other tissues locally produce Epo independent of its effects on red blood cell mass. Thus, not only its hematopoietic activity but also the recently discovered nonerythropoietic actions in addition to new drug delivery systems are being thoroughly investigated in order to fulfill the specific Epo release requirements for each therapeutic approach. The present review focuses on updating the information previously provided by similar reviews and recent experimental approaches are presented to describe the advances in Epo drug delivery achieved in the last few years and future perspectives.
Collapse
Affiliation(s)
- Ainhoa Murua
- Laboratory of Pharmacy and Pharmaceutical Technology, Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, SLFPB-EHU, Faculty of Pharmacy, University of the Basque Country, 01006, Vitoria-Gasteiz, Spain
| | | | | | | |
Collapse
|
12
|
Choi D, Cai EP, Schroer SA, Wang L, Woo M. Vhl is required for normal pancreatic β cell function and the maintenance of β cell mass with age in mice. J Transl Med 2011; 91:527-38. [PMID: 21242957 DOI: 10.1038/labinvest.2010.207] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Type 2 diabetes is hallmarked by insulin resistance and insufficient β-cell function. Islets of type 2 diabetes patients have been shown to have decreased hypoxia-inducible factor (HIF)-1α/β expression. Target genes of the HIF pathway are involved in angiogenesis, survival, proliferation, and energy metabolism, and von Hippel-Lindau protein (VHL) is a negative regulator of this pathway. We hypothesized that increased HIF-mediated gene transcription by VHL deletion in the β-cells would increase β-cell mass and function. We generated β-cell-specific VHL-knockout mice using the Cre-loxP recombination system driven by the rat insulin promoter to assess the role of VHL in glucose homeostasis and β-cell function. VHL deletion in the pancreatic β-cells led to impaired glucose tolerance due to defects in glucose-stimulated insulin secretion and β-cell mass with age. VHL-knockout islets had decreased GLUT2, but increased glucose transporter 1 and vascular endothelial growth factor expression. Furthermore, there were significant aberrations in islet morphology in the VHL-knockout mice, likely due to increased islet vasculature. Given that erythropoietin (EPO) is a target gene of the HIF pathway, which is not expressed in islets, we tested whether activating EPO signaling by systemic administration with recombinant human EPO (rHuEPO) can overcome the β-cell defects that occurred with VHL loss. We observed improved glucose tolerance and restoration of GLUT2 expression in VHL-deficient β-cells in response to rHuEPO. Contrary to our hypothesis, loss of VHL and increased transcription of HIF-target genes resulted in impaired β-cell function and mass, which can be overcome with exogenous EPO. Our results indicate a critical role for VHL in β-cell function and mass, and that EPO administration improved β-cell function making it a potential strategy for diabetes treatment.
Collapse
Affiliation(s)
- Diana Choi
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
13
|
Shuai H, Zhang J, Zhang J, Xie J, Zhang M, Yu Y, Zhang L. Erythropoietin protects pancreatic β-cell line NIT-1 cells against cytokine-induced apoptosis via phosphatidylinositol 3-kinase/Akt signaling. Endocr Res 2011; 36:25-34. [PMID: 21226565 DOI: 10.3109/07435800.2010.534753] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Erythropoietin (EPO) is a cytokine that regulates the proliferation, differentiation, and survival of erythroid progenitor cells. EPO has recently been demonstrated to have a tissue-protective role by mediating anti-apoptotic signals through the erythropoietin receptor (EPOR) in various tissues, including brain, liver, and heart. We have previously examined pancreatic β-cell line NIT-1 cells for the expression of EPOR by real-time PCR and determined that these cells were protected by EPO against cytokine-induced apoptosis. The precise underlying anti-apoptotic mechanisms in pancreatic β-cells are poorly understood. The purpose of this study is to examine erythropoietin receptor expression in the NIT-1 pancreatic beta-cell line and the intracellular pathway related with its anti-apoptosis effect in NIT-1 cells. METHODS we examined the expression of EPOR by western blot. We investigate the role of erythropoietin in the survival of these cells, and whether the PI3K/AKT pathway is involved in this protective process. RESULTS NIT-1 cells expressed EPOR and, in the presence of certain cytokines, exposure of NIT-1 cells to recombinant human erythropoietin (rhEPO) significantly improved the impaired insulin secretion and inhibited cytokine-induced apoptosis. RhEPO caused a rapid activation of Akt and increased expression of Bcl-2. The protective anti-apoptotic effect of rhEPO was significantly abolished by a specific phosphatidylinositol 3-kiniase (PI3K) inhibitor, LY294002. CONCLUSIONS Our findings indicate that EPOR is expressed in pancreatic β-cell line NIT-1 cells and suggest that EPO may act as a survival factor requiring the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Hongxia Shuai
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | | | | | | | | | | | | |
Collapse
|
14
|
Choi D, Schroer SA, Lu SY, Wang L, Wu X, Liu Y, Zhang Y, Gaisano HY, Wagner KU, Wu H, Retnakaran R, Woo M. Erythropoietin protects against diabetes through direct effects on pancreatic beta cells. ACTA ACUST UNITED AC 2010; 207:2831-42. [PMID: 21149549 PMCID: PMC3005231 DOI: 10.1084/jem.20100665] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In mouse models of type 1 and type 2 diabetes, administration of human erythropoietin protects against disease by acting directly on pancreatic β cells. A common feature among all forms of diabetes mellitus is a functional β-cell mass insufficient to maintain euglycemia; therefore, the promotion of β-cell growth and survival is a fundamental goal for diabetes prevention and treatment. Evidence has suggested that erythropoietin (EPO) exerts cytoprotective effects on nonerythroid cells. However, the influence of EPO on pancreatic β cells and diabetes has not been evaluated to date. In this study, we report that recombinant human EPO treatment can protect against diabetes development in streptozotocin-induced and db/db mouse models of type 1 and type 2 diabetes, respectively. EPO exerts antiapoptotic, proliferative, antiinflammatory, and angiogenic effects within the islets. Using β-cell–specific EPO receptor and JAK2 knockout mice, we show that these effects of EPO result from direct biological effects on β cells and that JAK2 is an essential intracellular mediator. Thus, promotion of EPO signaling in β cells may be a novel therapeutic strategy for diabetes prevention and treatment.
Collapse
Affiliation(s)
- Diana Choi
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Sims B, Clarke M, Njah W, Hopkins ES, Sontheimer H. Erythropoietin-induced neuroprotection requires cystine glutamate exchanger activity. Brain Res 2010; 1321:88-95. [PMID: 20102705 DOI: 10.1016/j.brainres.2010.01.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 01/12/2010] [Accepted: 01/13/2010] [Indexed: 12/25/2022]
Abstract
Erythropoietin (Epo) has been used for many years in neonates for the treatment of anemia of prematurity. Epo has also been proposed for treatment of neonatal brain injury, as mounting evidence suggests neuroprotective properties for Epo. However, Epo's neuroprotective mechanism of action is poorly understood. In this study we hypothesized that Epo may confer neuroprotection by enhancing cellular redox defense brought about by cellular glutathione (GSH). This was examined in cultures of differentiated cortical neural stem cells and using the B104 cell line as model systems. Our data shows that Epo causes a time- and dose-dependent increase in expression and activity of system Xc(-), the transporter responsible for uptake of cystine for the production of glutathione. Cystine uptake increases 3-5 fold in differentiated neural stem cells and B104 cells treated with Epo. Exposure of cells to 100 microM kainate suppressed cellular GSH and caused excitotoxicity, but GSH levels and cell viability were completely restored by Epo in the continued presence of kainate. This rescue effect of Epo vanished if system Xc(-) was inhibited pharmacologically using S4-CPG in the presence of Epo leading to marked cell death of B104 cells and cultured mouse cortical neural stem cells. This could also be achieved using xCT siRNA to decrease xCT expression. This data suggests that system Xc(-) activity and protein expression are positively regulated by Epo directly explaining its neuroprotective effect.
Collapse
Affiliation(s)
- Brian Sims
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | |
Collapse
|
16
|
Epo delivery by genetically engineered C2C12 myoblasts immobilized in microcapsules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 670:54-67. [PMID: 20384218 DOI: 10.1007/978-1-4419-5786-3_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ver the last half century, the use of erythropoietin (Epo) in the management of malignancies has been extensively studied. Originally viewed as the renal hormone responsible for red blood cell production, many recent in vivo and clinical approaches demonstrate that various tissues locally produce Epo in response to physical or metabolic stress. Thus, not only its circulating erythrocyte mass regulator activity but also the recently discovered nonhematological actions are being thoroughly investigated in order to fulfill the specific Epo delivery requirements for each therapeutic approach.
Collapse
|
17
|
Ucan BH, Irkorucu O, Cakmak GK, Tascilar O, Tekin IO, Acikgoz S, Emre AU, Bahadir B, Ankarali H, Comert M. Erythropoietin: a possible cytoprotective cytokine in acute necrotizing pancreatitis. ACTA ACUST UNITED AC 2009; 16:530-7. [PMID: 19333535 DOI: 10.1007/s00534-009-0082-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Accepted: 12/09/2008] [Indexed: 01/30/2023]
Abstract
BACKGROUND/PURPOSE Despite decades of research and clinical trials, a specific therapeutic treatment for acute pancreatitis (AP) has yet to be developed. The aim of the present study was to investigate the effects of erythropoietin on the severity of taurocolic acid-induced acute necrotizing pancreatitis. METHODS Forty-seven male Wistar albino rats were randomized into seven experimental groups. In group I, animals were sham-operated (n = 5). In groups II, III, IV, IIepo, IIIepo, and IVepo, AP was induced by sodium taurodeoxycholate treatment (n = 7). In groups II, III, and IV, 1 ml normal saline and in groups IIepo, IIIepo, and IVepo, 1000 U/kg body weight erythropoietin (EPO) was administered intramuscularly immediately after the induction of AP. Animals were killed at 24, 48, and 72 h postoperatively. Histopathological and biochemical evaluations were performed. RESULTS The serum levels of interleukin-6 (IL-6) and tissue levels of malondialdehyde were found to be significantly lower in EPO-administered groups when compared with the levels in groups without EPO treatment. The severity of pancreatic edema, acinar necrosis, inflammation, and perivascular infiltrate were reduced in all the EPO groups compared with the no-treatment groups. CONCLUSIONS Our findings may reflect the possible cytoprotective effect of EPO in acute necrotizing pancreatitis.
Collapse
Affiliation(s)
- Bulent Hamdi Ucan
- Zonguldak Karaelmas University, School of Medicine, Zonguldak, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Adenovirus Infection Activates Akt1 and Induces Cell Proliferation in Pancreatic Islets1. Transplantation 2009; 87:821-4. [DOI: 10.1097/tp.0b013e318199c686] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
19
|
Mohammed JS, Wang Y, Harvat TA, Oberholzer J, Eddington DT. Microfluidic device for multimodal characterization of pancreatic islets. LAB ON A CHIP 2009; 9:97-106. [PMID: 19209341 PMCID: PMC3759253 DOI: 10.1039/b809590f] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
A microfluidic device to perfuse pancreatic islets while simultaneously characterizing their functionality through fluorescence imaging of the mitochondrial membrane potential and intracellular calcium ([Ca(2+)](i)) in addition to enzyme linked immunosorbent assay (ELISA) quantification of secreted insulin was developed and characterized. This multimodal characterization of islet function will facilitate rapid assessment of tissue quality immediately following isolation from donor pancreas and allow more informed transplantation decisions to be made which may improve transplantation outcomes. The microfluidic perfusion chamber allows flow rates of up to 1 mL min(-1), without any noticeable perturbation or shear of islets. This multimodal quantification was done on both mouse and human islets. The ability of this simple microfluidic device to detect subtle variations in islet responses in different functional assays performed in short time-periods demonstrates that the microfluidic perfusion chamber device can be used as a new gold standard to perform comprehensive islet analysis and obtain a more meaningful predictive value for islet functionality prior to transplantation into recipients, which is currently difficult to predict using a single functional assay.
Collapse
|
20
|
Brines M, Cerami A. Erythropoietin-mediated tissue protection: reducing collateral damage from the primary injury response. J Intern Med 2008; 264:405-32. [PMID: 19017170 DOI: 10.1111/j.1365-2796.2008.02024.x] [Citation(s) in RCA: 247] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In its classic hormonal role, erythropoietin (EPO) is produced by the kidney and regulates the number of erythrocytes within the circulation to provide adequate tissue oxygenation. EPO also mediates other effects directed towards optimizing oxygen delivery to tissues, e.g. modulating regional blood flow and reducing blood loss by promoting thrombosis within damaged vessels. Over the past 15 years, many unexpected nonhaematopoietic functions of EPO have been identified. In these more recently appreciated nonhormonal roles, locally-produced EPO signals through a different receptor isoform and is a major molecular component of the injury response, in which it counteracts the effects of pro-inflammatory cytokines. Acutely, EPO prevents programmed cell death and reduces the development of secondary, pro-inflammatory cytokine-induced injury. Within a longer time frame, EPO provides trophic support to enable regeneration and healing. As the region immediately surrounding damage is typically relatively deficient in endogenous EPO, administration of recombinant EPO can provide increased tissue protection. However, effective use of EPO as therapy for tissue injury requires higher doses than for haematopoiesis, potentially triggering serious adverse effects. The identification of a tissue-protective receptor isoform has facilitated the engineering of nonhaematopoietic, tissue-protective EPO derivatives, e.g. carbamyl EPO, that avoid these complications. Recently, regions within the EPO molecule mediating tissue protection have been identified and this has enabled the development of potent tissue-protective peptides, including some mimicking EPO's tertiary structure but unrelated in primary sequence.
Collapse
Affiliation(s)
- M Brines
- Warren Pharmaceuticals, Ossining, NY 10562, USA.
| | | |
Collapse
|
21
|
Bosio E, Seveso M, Dedja A, Luca G, Calvitti M, Calafiore R, Rigotti P, Busetto R, Ancona E, Cozzi E. Cobalt Protoporpyhrin Reduces Caspase-3,-7 Enzyme Activity in Neonatal Porcine Islets, But Does Not Inhibit Cell Death Induced by TNF-α. Cell Transplant 2008; 17:587-98. [DOI: 10.3727/096368908786092784] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Apoptotic phenomena observed in vitro following isolation and following transplantation contribute significantly to islet graft loss. Strategies to reduce apoptosis of islet tissue prior to and posttransplantation may improve graft survival and function and reduce the amount of tissue necessary to achieve insulin independence. The expression of cytoprotective proteins is one such strategy that may prolong islet survival. In this light, heme-oxygenase 1 (HO-1) upregulation has been studied in both allo- and xenotransplantation models. In this study, the effect of HO-1 on apoptosis in neonatal porcine islet-like cell clusters (NPICC) was assessed. In in vitro assessments of NPICC apoptosis, NPICC showed a high sensitivity to apoptotic stimulation using a combination of TNF-α and cycloheximide. Stimulation with TNF-α alone was sufficient to induce reproducible apoptotic responses as demonstrated by caspase-3,-7 activation and subdiploid DNA analysis. Dose-dependent, high-level HO-1 protein expression was achieved following culture of NPICC in medium containing either cobalt protoporphyrin (CoPP) or cobalt mesoporphyrin (CoMP). CoPP treatment resulted in the reduction of caspase-3,-7 enzyme activity following TNF-α stimulation. However, such an effect was not associated with a reduction in the levels of cell death. Indeed, the inhibition of caspase enzyme activity resulted in decreased PARP-1 cleavage, which may lead to heightened levels of necrosis in treated NPICC cultures, possibly explaining the observed commitment of NPICC to the death pathway.
Collapse
Affiliation(s)
- Erika Bosio
- CORIT (Consorzio per la Ricerca sul Trapianto d'Organi), Padua, Italy
- Department of Medical and Surgical Sciences, University of Padua, Padua, Italy
| | - Michela Seveso
- CORIT (Consorzio per la Ricerca sul Trapianto d'Organi), Padua, Italy
| | - Arben Dedja
- CORIT (Consorzio per la Ricerca sul Trapianto d'Organi), Padua, Italy
- Department of Medical and Surgical Sciences, University of Padua, Padua, Italy
| | - Giovanni Luca
- Department of Internal Medicine, Section of Internal Medicine and Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy
| | - Mario Calvitti
- Department of Internal Medicine, Section of Internal Medicine and Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy
| | - Riccardo Calafiore
- Department of Internal Medicine, Section of Internal Medicine and Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy
| | - Paolo Rigotti
- CORIT (Consorzio per la Ricerca sul Trapianto d'Organi), Padua, Italy
- Department of Medical and Surgical Sciences, University of Padua, Padua, Italy
| | - Roberto Busetto
- Department of Clinical Veterinary Science, University of Padua, Legnaro, Italy
| | - Ermanno Ancona
- CORIT (Consorzio per la Ricerca sul Trapianto d'Organi), Padua, Italy
- Department of Medical and Surgical Sciences, University of Padua, Padua, Italy
- Direzione Sanitaria, Padua General Hospital, Padua, Italy
- Clinica Chirurgica III, Padua General Hospital, Padua, Italy
| | - Emanuele Cozzi
- CORIT (Consorzio per la Ricerca sul Trapianto d'Organi), Padua, Italy
- Department of Medical and Surgical Sciences, University of Padua, Padua, Italy
- Direzione Sanitaria, Padua General Hospital, Padua, Italy
- Clinica Chirurgica III, Padua General Hospital, Padua, Italy
| |
Collapse
|
22
|
Sweet IR, Yanay O, Waldron L, Gilbert M, Fuller JM, Tupling T, Lernmark A, Osborne WRA. Treatment of diabetic rats with encapsulated islets. J Cell Mol Med 2008; 12:2644-50. [PMID: 18373735 PMCID: PMC3828880 DOI: 10.1111/j.1582-4934.2008.00322.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Immunoprotection of islets using bioisolator systems permits introduction of allogeneic cells to diabetic patients without the need for immunosuppression. Using TheraCyte™ immunoisolation devices, we investigated two rat models of type 1 diabetes mellitus (T1DM), BB rats and rats made diabetic by streptozotocin (STZ) treatment. We chose to implant islets after the onset of diabetes to mimic the probable treatment of children with T1DM as they are usually diagnosed after disease onset. We encapsulated 1000 rat islets and implanted them subcutaneously (SQ) into diabetic biobreeding (BB) rats and STZ-induced diabetic rats, defined as two or more consecutive days of blood glucose >350 mg/dl. Rats were monitored for weight and blood glucose. Untreated BB rats rapidly lost weight and were euthanized at >20% weight loss that occurred between 4 and 10 days from implantation. For period of 30–40 days following islet implantation weights of treated rats remained steady or increased. Rapid weight loss occurred after surgical removal of devices that contained insulin positive islets. STZ-treated rats that received encapsulated islets showed steady weight gain for up to 130 days, whereas untreated control rats showed steady weight loss that achieved >20% at around 55 days. Although islet implants did not normalize blood glucose, treated rats were apparently healthy and groomed normally. Autologous or allogeneic islets were equally effective in providing treatment. TheraCyte™ devices can sustain islets, protect allogeneic cells from immune attack and provide treatment for diabetic-mediated weight loss in both BB rats and STZ-induced diabetic rats.
Collapse
Affiliation(s)
- Ian R Sweet
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Shuai H, Zhang J, Yu Y, Zhang M. Expression of EPO receptor in pancreatic cells and its effect on cell apoptosis. ACTA ACUST UNITED AC 2008; 28:49-51. [DOI: 10.1007/s11596-008-0112-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Indexed: 11/29/2022]
|
24
|
Lai Y, Drobinskaya I, Kolossov E, Chen C, Linn T. Genetic modification of cells for transplantation. Adv Drug Deliv Rev 2008; 60:146-59. [PMID: 18037530 DOI: 10.1016/j.addr.2007.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 08/02/2007] [Indexed: 01/16/2023]
Abstract
Progress in gene therapy has produced promising results that translate experimental research into clinical treatment. Gene modification has been extensively employed in cell transplantation. The main barrier is an effective gene delivery system. Several viral vectors were utilized in end-stage differentiated cells. Recently, successful applications were described with adenovirus-associated vectors. As an alternative, embryonic stem cell- and stem cell-like systems were established for generation of tissue-specified gene-modified cells. Owing to the feasibility for genetic manipulations and the self-renewing potency of these cells they can be used in a way enabling large-scale in vitro production. This approach offers the establishment of in vitro cell culture systems that will deliver sufficient amounts of highly purified, immunoautologous cells suitable for application in regenerative medicine. In this review, the current technology of gene delivery systems to cells is recapitulated and the latest developments for cell transplantation are discussed.
Collapse
|
25
|
Abstract
Significant progress has been made in the field of beta-cell replacement therapies by islet transplantation in patients with unstable Type 1 diabetes mellitus (T1DM). Recent clinical trials have shown that islet transplantation can reproducibly lead to insulin independence when adequate islet numbers are implanted. Benefits include improvement of glycemic control, prevention of severe hypoglycemia and amelioration of quality of life. Numerous challenges still limit this therapeutic option from becoming the treatment of choice for T1DM. The limitations are primarily associated with the low islet yield of human pancreas isolations and the need for chronic immunosuppressive therapies. Herein the authors present an overview of the historical progress of islet transplantation and outline the recent advances of the field. Cellular therapies offer the potential for a cure for patients with T1DM. The progress in beta-cell replacement treatment by islet transplantation as well as those of emerging immune interventions for the restoration of self tolerance justify great optimism for years to come.
Collapse
Affiliation(s)
- Simona Marzorati
- University of Miami Miller School of Medicine, Cell Transplant Center and Clinical Islet Transplant Program, Diabetes Research Institute, 1450 NW, 10th Avenue (R-134), Miami, FL 33136, USA
| | | | | |
Collapse
|
26
|
Pileggi A, Cobianchi L, Inverardi L, Ricordi C. Overcoming the Challenges Now Limiting Islet Transplantation: A Sequential, Integrated Approach. Ann N Y Acad Sci 2006; 1079:383-98. [PMID: 17130583 DOI: 10.1196/annals.1375.059] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Steady improvements in islet cell processing technology and immunosuppressive protocols have made pancreatic islet transplantation a clinical reality for the treatment of patients with Type 1 diabetes mellitus (T1DM). Recent trials are showing that improved glycemic metabolic control, prevention of severe hypoglycemia, and better quality of life can be reproducibly achieved after transplantation of allogeneic islets in patients with unstable T1DM. Despite these encouraging results, challenges ahead comprise obtaining adequate islet cells for transplant, enhancing islets engraftment, sustaining beta cell mass and function over time, and defining effective immune interventions, among others. In order to overcome the current hurdles to the widespread application of islet transplantation there is a need for implementation of integrated, sequential therapeutic approaches.
Collapse
Affiliation(s)
- Antonello Pileggi
- Cell Transplant Center, Diabetes Research Institute, Miller School of Medicine, University of Miami, 1450 NW 10th Avenue (R-134), Miami, FL 33136, USA
| | | | | | | |
Collapse
|
27
|
McGill JB, Bell DSH. Anemia and the role of erythropoietin in diabetes. J Diabetes Complications 2006; 20:262-72. [PMID: 16798479 DOI: 10.1016/j.jdiacomp.2005.08.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2004] [Revised: 07/29/2005] [Accepted: 08/01/2005] [Indexed: 11/18/2022]
Abstract
Anemia is more common in patients with diabetes than without diabetes, and the problem is magnified in patients with renal impairment. Diabetic patients with anemia may be at increased risk of adverse outcomes from diabetic retinopathy, nephropathy, neuropathy, and cardiovascular disease. The etiology of anemia in diabetes is multifactorial and includes inflammation, nutritional deficiencies, concomitant autoimmune diseases, drugs, and hormonal changes in addition to kidney disease. Anemia that is associated with erythropoietin deficiency may have prognostic significance for persons with nephropathy or heart failure. In early diabetic nephropathy, damage to the peritubular fibroblasts can occur and lead to erythropoietin deficiency and anemia prior to the loss of filtration. Correction of the anemia not only leads to less fatigue, greater exercise tolerance, and an improved quality of life but also to a reduction in mortality and hospital admissions for congestive heart failure (CHF). Data are accumulating that suggest that treatment of anemia will slow the progression of microvascular and macrovascular complications, including postural hypotension from autonomic neuropathy, retinopathy, and loss of renal function from diabetic nephropathy. Promptly diagnosing and treating anemia in patients with diabetes may result in an improved quality of life and decreased morbidity and mortality.
Collapse
Affiliation(s)
- Janet B McGill
- Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
28
|
Matsumoto S, Noguchi H, Yonekawa Y, Okitsu T, Iwanaga Y, Liu X, Nagata H, Kobayashi N, Ricordi C. Pancreatic islet transplantation for treating diabetes. Expert Opin Biol Ther 2006; 6:23-37. [PMID: 16370912 DOI: 10.1517/14712598.6.1.23] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic islet transplantation is one of the options for treating diabetes and has been shown to improve the quality of life of severe diabetic patients. Since the Edmonton protocol was announced, islet transplantation have advanced considerably, including islet after kidney transplantation, utilisation of non-heart-beating donors, single-donor islet transplantation and living-donor islet transplantation. These advances were based on revised immunosuppression protocols, improved pancreas procurement and islet isolation methods, and enhanced islet engraftment. Further improvements are necessary to make islet transplantation a routine clinical treatment. To synergise efforts towards a cure for type 1 diabetes, a Diabetes Research Institute (DRI) Federation is currently being established to include leading diabetes research centres worldwide, including DRIs in Miami, Edmonton and Kyoto among others.
Collapse
Affiliation(s)
- Shinichi Matsumoto
- Transplantation Unit, Kyoto University Hospital, Diabetes Research Institute Kyoto, Shogoin, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Emamaullee JA, Rajotte RV, Liston P, Korneluk RG, Lakey JRT, Shapiro AMJ, Elliott JF. XIAP overexpression in human islets prevents early posttransplant apoptosis and reduces the islet mass needed to treat diabetes. Diabetes 2005; 54:2541-8. [PMID: 16123341 DOI: 10.2337/diabetes.54.9.2541] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Edmonton Protocol for treatment of type 1 diabetes requires islets from two or more donors to achieve euglycemia in a single recipient, primarily because soon after portal infusion, the majority of the transplanted cells undergo apoptosis due to hypoxia and hypoxia reperfusion injury. X-linked inhibitor of apoptosis protein (XIAP) is a potent endogenous inhibitor of apoptosis that is capable of blocking the activation of multiple downstream caspases, and XIAP overexpression has previously been shown to enhance engraftment of a murine beta-cell line. In this study, human islets transduced with a XIAP-expressing recombinant adenovirus were resistant to apoptosis and functionally recovered following in vitro stresses of hypoxia and hypoxia with reoxygenation (models reperfusion injury). Furthermore Ad-XIAP transduction dramatically reduced the number of human islets required to reverse hyperglycemia in chemically diabetic immunodeficient mice. These results suggest that by transiently overexpressing XIAP in the immediate posttransplant period, human islets from a single donor might be used to effectively treat two diabetic recipients.
Collapse
Affiliation(s)
- Juliet A Emamaullee
- 1-21 Medical Sciences Building, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada.
| | | | | | | | | | | | | |
Collapse
|