1
|
Geng A, Yuan S, Yu QC, Zeng YA. The role of endothelial cells in pancreatic islet development, transplantation and culture. Front Cell Dev Biol 2025; 13:1558137. [PMID: 40330424 PMCID: PMC12052768 DOI: 10.3389/fcell.2025.1558137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/03/2025] [Indexed: 05/08/2025] Open
Abstract
Endothelial cells (ECs) play pivotal roles in the development and maintenance of tissue homeostasis. During development, vasculature actively involves in organ morphogenesis and functional maturation, through the secretion of angiocrine factors and extracellular matrix components. Islets of Langerhans, essential functional units of glucose homeostasis, are embedded in a dense endothelial capillary network. Islet vasculature not only supplies nutrients and oxygen to endocrine cells but also facilitate the rapid delivery of pancreatic hormones to target tissues, thereby ensuring precise glucose regulation. Diabetes mellitus is a major disease burden and is caused by islet dysfunction or depletion, often accompanied by vessel loss and dysregulation. Therefore, elucidating the regulatory mechanisms of ECs within islets hold profound implications for diabetes therapy. This review provides an overview of recent research advancements on the functional roles of ECs in islet biology, transplantation, and in vitro islet organoid culture.
Collapse
Affiliation(s)
- Ajun Geng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shubo Yuan
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing Cissy Yu
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Arial Zeng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
2
|
Gao X, Gao J, Sun Y, Zhao J, Geng L, Wang C, Qiao M, Wang J. The common pathogenesis of nodular goiter in both sexes: An exploration into gene expression and signaling pathways. Heliyon 2024; 10:e33411. [PMID: 39035545 PMCID: PMC11259846 DOI: 10.1016/j.heliyon.2024.e33411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024] Open
Abstract
The past few years have witnessed an increasing incidence of nodular goiter (NG), with a well-documented higher prevalence in females than males. This gender disparity has led research to focus primarily on female subjects, potentially overlooking common pathogenic mechanisms in both sexes. In this study, we investigated the shared pathogenesis of NG in males and females. Utilizing a rat model and RNA sequencing, we identified differentially expressed genes associated with the disease. We further validated these findings in normal human thyroid cells and human papillary thyroid cancer cells. A randomized experiment was conducted with equal numbers of male and female rats divided into control and NG model groups. The NG model was established using propylthiouracil and various assessments such as thyroid ultrasonography, thyroid index, thyroid function, and thyroid histology were performed. Transcriptome analysis revealed numerous upregulated and downregulated genes in both male and female model groups. Key genes like KDR, FLT1, PDGFB, and CAV1, and pathways including PI3K-Akt, MAPK, Ras, fluid shear stress and atherosclerosis, calcium signaling, and Rap1 signaling pathways were linked with the disease. Western blot and immunofluorescence analysis confirmed these findings, which were further supported by cell-based experiments. In conclusion, our findings suggest that abnormal expression of specific genes and pathways leading to irregular cell growth, blood vessel formation, and inflammation may be common factors in the pathogenesis of NG in both males and females.
Collapse
Affiliation(s)
- Xiangju Gao
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jie Gao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ya Sun
- Institute of Traditional Chinese Medicine Innovation, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jing Zhao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Li Geng
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Changlin Wang
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Mingqi Qiao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jieqiong Wang
- Emotional Disease Syndrome Liver Storage Pharmacological Young Scientific Research Innovation Team in Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| |
Collapse
|
3
|
Doherty DT, Khambalia HA, van Dellen D, Jennings RE, Piper Hanley K. Unlocking the post-transplant microenvironment for successful islet function and survival. Front Endocrinol (Lausanne) 2023; 14:1250126. [PMID: 37711891 PMCID: PMC10497759 DOI: 10.3389/fendo.2023.1250126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023] Open
Abstract
Islet transplantation (IT) offers the potential to restore euglycemia for patients with type 1 diabetes mellitus (T1DM). Despite improvements in islet isolation techniques and immunosuppressive regimes, outcomes remain suboptimal with UK five-year graft survivals (5YGS) of 55% and most patients still requiring exogenous insulin after multiple islet infusions. Native islets have a significant non-endocrine component with dense extra-cellular matrix (ECM), important for islet development, cell survival and function. Collagenase isolation necessarily disrupts this complex islet microenvironment, leaving islets devoid of a supporting framework and increasing vulnerability of transplanted islets. Following portal venous transplantation, a liver injury response is potentially induced, which typically results in inflammation and ECM deposition from liver specific myofibroblasts. The impact of this response may have important impact on islet survival and function. A fibroblast response and ECM deposition at the kidney capsule and eye chamber alongside other implantation sites have been shown to be beneficial for survival and function. Investigating the implantation site microenvironment and the interactions of transplanted islets with ECM proteins may reveal therapeutic interventions to improve IT and stem-cell derived beta-cell therapy.
Collapse
Affiliation(s)
- Daniel T. Doherty
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- Department of Renal & Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Hussein A. Khambalia
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- Department of Renal & Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - David van Dellen
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- Department of Renal & Pancreatic Transplantation, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Rachel E. Jennings
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- Department of Endocrinology, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Karen Piper Hanley
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
4
|
Nemati M, Karbalaei N, Mokarram P, Dehghani F, Dastghaib S, Aghaei Z. Cotransplant With Pancreatic Islet Homogenate Improved Survival and Long-Term Efficacy of Islet Transplant in Streptozotocin-Diabetic Rats. EXP CLIN TRANSPLANT 2022; 20:164-172. [PMID: 35282811 DOI: 10.6002/ect.2021.0385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Pancreatic islet transplant is suggested as a promising treatment option in diabetes, but the number of viable and functional islets and the long-term efficacy of transplanted islets have not been satisfactory. Islet isolation leads to destruction of the extracellular matrix and loss of trophic support of islets, which reduces their survival and function. Reconstruction of islet microenvironment with biomaterials may preserve islet survival and graft efficacy. Accordingly, we investigated the effects of pancreatic islet homogenate on islet quality and graft outcomes in diabetic rats. MATERIALS AND METHODS Islets were isolated from the pancreas of Sprague Dawley rats and were cultured with or without pancreatic islet homogenate. Before transplant, viability, insulin content, and insulin released from cultured islets were assessed. Islets were then transplanted into subcapsular space of diabetic rat kidney. Transplant outcomes were evaluated by plasma glucose and insulin levels, glucose tolerance tests, and stress oxidative markers. RESULTS Viability and insulin release in the pancreatic islet homogenate-treated islets were significantly higher than that in the control islets. After transplant of islets, recipient rats with pancreatic islet homogenate showed significant decreases in blood glucose and malondialdehyde levels and increases in superoxide dismutase activity and plasma insulin levels. CONCLUSIONS Islet treatment with pancreatic islet homogenate could improve islet survival and transplant function and outcomes. Oxidative stress reduction might be a secondary beneficial effect of improved quality of treated islets.
Collapse
Affiliation(s)
- Marzieh Nemati
- From the Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,From the Department of Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | | | |
Collapse
|
5
|
Chung WY, Pollard CA, Kumar R, Drogemuller CJ, Naziruddin B, Stover C, Issa E, Isherwood J, Cooke J, Levy MF, Coates PTH, Garcea G, Dennison AR. A comparison of the inflammatory response following autologous compared with allogenic islet cell transplantation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:98. [PMID: 33569400 PMCID: PMC7867892 DOI: 10.21037/atm-20-3519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background The initial response to islet transplantation and the subsequent acute inflammation is responsible for significant attrition of islets following both autologous and allogenic procedures. This multicentre study compares this inflammatory response using cytokine profiles and complement activation. Methods Inflammatory cytokine and complement pathway activity were examined in two cohorts of patients undergoing total pancreatectomy followed either by autologous (n=11) or allogenic (n=6) islet transplantation. Two patients who underwent total pancreatectomy alone (n=2) served as controls. Results The peak of cytokine production occurred immediately following induction of anaesthesia and during surgery. There was found to be a greater elevation of the following cytokines: TNF-alpha (P<0.01), MCP-1 (P=0.0013), MIP-1α (P=0.001), MIP-1β (P=0.00020), IP-10 (P=0.001), IL-8 (P=0.004), IL-1α (P=0.001), IL-1ra (0.0018), IL-10 (P=0.001), GM-CSF (P=0.001), G-CSF (P=0.0198), and Eotaxin (P=0.01) in the allogenic group compared to autografts and controls. Complement activation and consumption was observed in all three pathways, and there were no significant differences in between the groups although following allogenic transplantation ∆IL-10 and ∆VEGF levels were significantly elevated those patients who became insulin-independent compared with those who were insulin-dependent. Conclusions The cytokine profiles following islet transplantation suggests a significantly greater acute inflammatory response following allogenic islet transplantation compared with auto-transplantation although a significant, non-specific inflammatory response occurs following both forms of islet transplantation.
Collapse
Affiliation(s)
- Wen Yuan Chung
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Cristina A Pollard
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Rohan Kumar
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | | | | | - Cordula Stover
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Eyad Issa
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - John Isherwood
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Jill Cooke
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Marlon F Levy
- Baylor Research Institute, Dallas & Fort Worth, TX, USA
| | - P Toby H Coates
- Australian Islet Consortium, Royal Adelaide Hospital, South Australia, Australia
| | - Giuseppe Garcea
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Ashley R Dennison
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| |
Collapse
|
6
|
Figueiredo H, Figueroa ALC, Garcia A, Fernandez-Ruiz R, Broca C, Wojtusciszyn A, Malpique R, Gasa R, Gomis R. Targeting pancreatic islet PTP1B improves islet graft revascularization and transplant outcomes. Sci Transl Med 2020; 11:11/497/eaar6294. [PMID: 31217339 DOI: 10.1126/scitranslmed.aar6294] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/16/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Deficient vascularization is a major driver of early islet graft loss and one of the primary reasons for the failure of islet transplantation as a viable treatment for type 1 diabetes. This study identifies the protein tyrosine phosphatase 1B (PTP1B) as a potential modulator of islet graft revascularization. We demonstrate that grafts of pancreatic islets lacking PTP1B exhibit increased revascularization, which is accompanied by improved graft survival and function, and recovery of normoglycemia and glucose tolerance in diabetic mice transplanted with PTP1B-deficient islets. Mechanistically, we show that the absence of PTP1B leads to activation of hypoxia-inducible factor 1α-independent peroxisome proliferator-activated receptor γ coactivator 1α/estrogen-related receptor α signaling and enhanced expression and production of vascular endothelial growth factor A (VEGF-A) by β cells. These observations were reproduced in human islets. Together, these findings reveal that PTP1B regulates islet VEGF-A production and suggest that this phosphatase could be targeted to improve islet transplantation outcomes.
Collapse
Affiliation(s)
- Hugo Figueiredo
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,University of Barcelona, 08036 Barcelona, Spain.,Escuela de Medicina y Ciencias de la Salud, Dept. Medicina Cardiovascular y Metabolómica, Tecnológico de Monterrey, 66278 San Pedro Garza García, Nuevo León, Mexico
| | - Ana Lucia C Figueroa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,University of Barcelona, 08036 Barcelona, Spain
| | - Ainhoa Garcia
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Rebeca Fernandez-Ruiz
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Christophe Broca
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, 34295 Montpellier, France
| | - Anne Wojtusciszyn
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, 34295 Montpellier, France.,Department of Endocrinology, Diabetes and Nutrition, University Hospital of Montpellier, Lapeyronie Hospital, 34295 Montpellier, France.,Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Rita Malpique
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Rosa Gasa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Ramon Gomis
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain. .,University of Barcelona, 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.,Universitat Oberta de Catalunya (UOC), 08018 Barcelona, Spain.,Department of Endocrinology and Nutrition, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
7
|
Nemati M, Karbalaei N, Mokarram P, Dehghani F. Effects of platelet-rich plasma on the pancreatic islet survival and function, islet transplantation outcome and pancreatic pdx 1 and insulin gene expression in streptozotocin-induced diabetic rats. Growth Factors 2020; 38:137-151. [PMID: 33569978 DOI: 10.1080/08977194.2021.1881502] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Platelet-rich plasma (PRP) is a therapeutic option in different fields based on its growth factors. We investigated influence of PRP on islet survival, function, transplantation outcomes, and pancreatic genes expression in diabetic rats. In vitro: pancreatic isolated islets were incubated with/without PRP then viability, insulin secretion, and content were assessed. In vivo: Series 1 were designed to determine whether islet treatment with PRP improves transplantation outcome in diabetic rats by evaluating plasma glucose and insulin concentrations and oxidative parameters. Series 2, effects of PRP subcutaneous injection were evaluated on pancreatic genes expression and glucose tolerance test in diabetic rats. PRP enhanced viability and secretary function of islet. Reduced glucose and malondialdehyde levels as well as increased insulin levels, superoxide dismutase activity, and expressions of pdx1 and insulin were observed in diabetic rats. PRP treatment has positive effects on islet viability, function, transplantation outcome, and pancreatic genes expression in diabetic rats.
Collapse
Affiliation(s)
- Marzieh Nemati
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Karbalaei
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Mokarram
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Dehghani
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Is the renal subcapsular space the preferred site for clinical porcine islet xenotransplantation? Review article. Int J Surg 2019; 69:100-107. [PMID: 31369877 DOI: 10.1016/j.ijsu.2019.07.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 07/27/2019] [Indexed: 12/29/2022]
Abstract
It can reasonably be anticipated that, within 5-10 years, islet allotransplantation or pig islet xenotransplantation may be the preferred options for β-cell replacement therapy. The portal vein/liver is currently the preferred clinical site for free islet transplantation, constituting 90% of clinical islet transplants. Despite being the site of choice for rodent and some large animal studies, the renal subcapsular space is rarely used clinically, even though the introduction of islets intraportally is not entirely satisfactory (particularly for pig islet xenotransplantation). We questioned why this might be so. Is it perhaps based on prior clinical evidence, or from experience in nonhuman primates? When we have questioned experts in the field, no definitive answers have been forthcoming. We have therefore reviewed the relevant literature, and still cannot find a convincing reason why the renal subcapsular space has been so relatively abandoned as a site for clinical islet transplantation. Owing to its sequestered environment, subcapsular transplantation might avoid some of the remaining challenges of intraportal transplantation. This may be particularly true when using pig islets for xenotransplantation, which are exceptionally pure in comparison to human islets used in auto- or allo-transplantation. With evidence from the literature, we question the notion that the subcapsular space is inhospitable to islet transplantation and suggest that, when porcine islet transplantation is introduced, this site should perhaps be reconsidered.
Collapse
|
9
|
Jansson L, Carlsson PO. Pancreatic Blood Flow with Special Emphasis on Blood Perfusion of the Islets of Langerhans. Compr Physiol 2019; 9:799-837. [PMID: 30892693 DOI: 10.1002/cphy.c160050] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pancreatic islets are more richly vascularized than the exocrine pancreas, and possess a 5- to 10-fold higher basal and stimulated blood flow, which is separately regulated. This is reflected in the vascular anatomy of the pancreas where islets have separate arterioles. There is also an insulo-acinar portal system, where numerous venules connect each islet to the acinar capillaries. Both islets and acini possess strong metabolic regulation of their blood perfusion. Of particular importance, especially in the islets, is adenosine and ATP/ADP. Basal and stimulated blood flow is modified by local endothelial mediators, the nervous system as well as gastrointestinal hormones. Normally the responses to the nervous system, especially the parasympathetic and sympathetic nerves, are fairly similar in endocrine and exocrine parts. The islets seem to be more sensitive to the effects of endothelial mediators, especially nitric oxide, which is a permissive factor to maintain the high basal islet blood flow. The gastrointestinal hormones with pancreatic effects mainly influence the exocrine pancreatic blood flow, whereas islets are less affected. A notable exception is incretin hormones and adipokines, which preferentially affect islet vasculature. Islet hormones can influence both exocrine and endocrine blood vessels, and these complex effects are discussed. Secondary changes in pancreatic and islet blood flow occur during several conditions. To what extent changes in blood perfusion may affect the pathogenesis of pancreatic diseases is discussed. Both type 2 diabetes mellitus and acute pancreatitis are conditions where we think there is evidence that blood flow may contribute to disease manifestations. © 2019 American Physiological Society. Compr Physiol 9:799-837, 2019.
Collapse
Affiliation(s)
- Leif Jansson
- Uppsala University, Department of Medical Cell Biology, Uppsala, Sweden
| | - Per-Ola Carlsson
- Uppsala University, Department of Medical Cell Biology, Uppsala, Sweden.,Uppsala University, Department of Medical Sciences, Uppsala, Sweden
| |
Collapse
|
10
|
Haque MR, Jeong JH, Lee KW, Shin DY, Kim GS, Kim SJ, Byun Y. Effects of Transplanted Islets Nano-Encapsulated with Hyperbranched Polyethylene Glycol and Heparin on Microenvironment Reconstruction and Glucose Control. Bioconjug Chem 2018; 29:2945-2953. [PMID: 29985588 DOI: 10.1021/acs.bioconjchem.8b00364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The microenvironment of pancreatic islets gets disrupted during enzyme digestion and causes islets to remain in a vulnerable state, leading to poor outcome in the initial days of transplantation. To avoid immune invasion while allowing the reconstruction of the microenvironment of the transplanted site, we propose immunoisolation polymers, which can nanoencapsulate islets quickly without cytotoxicity. Here, nonhuman primate (NHP) islets were nanoencapsulated with hyperbranched polyethylene glycol (hb-PEG) and heparin by layer-by-layer technology and transplanted into the kidney subcapsular space of diabetic C57BL/6 mice. An immunosuppressive drug protocol was applied to increase the survival time until the animals were sacrificed. The recipients of NHP islets exhibited high nonfasting blood glucose level (BGL) for 2-3 weeks, which was normalized afterward. Immunohistochemical (IHC) analysis revealed an immature vascular basement membrane and cell surface integrins directly associated with poor initial insulin production. The transplanted grafts regained their own microenvironment within a month without any outside stimuli. No lymphocyte infiltration was observed in the grafts at any time. Humoral and cell-mediated immune responses were prominently diminished by the hb-PEG/Heparin nanoencapsulated islets. Immunoisolation accompanied by an immunosuppressive drug protocol protects islets by helping them avoid immunogenesis while at the same time allowing them to reconstruct their microenvironment.
Collapse
Affiliation(s)
| | - Jee-Heon Jeong
- College of Pharmacy , Yeungnam University , Gyeongsan , Gyeongbuk 712-749 , Republic of Korea
| | - Kyo Won Lee
- Transplantation Research Center , Samsung Biomedical Research Institute , 81 Ilwon-ro , Gangnam-gu, Seoul 06351 , Republic of Korea.,Department of Surgery, Samsung Medical Center , Sungkyunkwan University School of Medicine , Seoul 06351 , Republic of Korea
| | - Du Yeon Shin
- Transplantation Research Center , Samsung Biomedical Research Institute , 81 Ilwon-ro , Gangnam-gu, Seoul 06351 , Republic of Korea.,Department of Surgery, Samsung Medical Center , Sungkyunkwan University School of Medicine , Seoul 06351 , Republic of Korea.,Stem Cell & Regenerative Medicine Institute , Samsung Medical Center , Seoul 06351 , Republic of Korea.,Department of Health Sciences & Technology, Samsung Advanced Institute for Health Sciences & Technology, Graduate School , Sungkyunkwan University , Seoul 06351 , Republic of Korea
| | - Geun-Soo Kim
- Transplantation Research Center , Samsung Biomedical Research Institute , 81 Ilwon-ro , Gangnam-gu, Seoul 06351 , Republic of Korea.,Department of Surgery, Samsung Medical Center , Sungkyunkwan University School of Medicine , Seoul 06351 , Republic of Korea.,Stem Cell & Regenerative Medicine Institute , Samsung Medical Center , Seoul 06351 , Republic of Korea.,Department of Health Sciences & Technology, Samsung Advanced Institute for Health Sciences & Technology, Graduate School , Sungkyunkwan University , Seoul 06351 , Republic of Korea
| | - Sung Joo Kim
- Transplantation Research Center , Samsung Biomedical Research Institute , 81 Ilwon-ro , Gangnam-gu, Seoul 06351 , Republic of Korea.,Department of Surgery, Samsung Medical Center , Sungkyunkwan University School of Medicine , Seoul 06351 , Republic of Korea.,Stem Cell & Regenerative Medicine Institute , Samsung Medical Center , Seoul 06351 , Republic of Korea.,Department of Health Sciences & Technology, Samsung Advanced Institute for Health Sciences & Technology, Graduate School , Sungkyunkwan University , Seoul 06351 , Republic of Korea
| | | |
Collapse
|
11
|
Current advanced therapy cell-based medicinal products for type-1-diabetes treatment. Int J Pharm 2018; 543:107-120. [PMID: 29597032 DOI: 10.1016/j.ijpharm.2018.03.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/19/2018] [Accepted: 03/24/2018] [Indexed: 12/11/2022]
Abstract
In the XXI century diabetes mellitus has become one of the main threats to human health with higher incidence in regions such as Europe and North America. Type 1 diabetes mellitus (T1DM) occurs as a consequence of the immune-mediated destruction of insulin producing β-cells located in the endocrine part of the pancreas, the islets of Langerhans. The administration of exogenous insulin through daily injections is the most prominent treatment for T1DM but its administration is frequently associated to failure in glucose metabolism control, finally leading to hyperglycemia episodes. Other approaches have been developed in the past decades, such as whole pancreas and islet allotransplantation, but they are restricted to patients who exhibit frequent episodes of hypoglycemia or renal failure because the lack of donors and islet survival. Moreover, patients transplanted with either whole pancreas or islets require of immune suppression to avoid the rejection of the transplant. Currently, advanced therapy medicinal products (ATMP), such as implantable devices, have been developed in order to reduce immune rejection response while increasing cell survival. To overcome these issues, ATMPs must promote vascularization, guaranteeing the nutritional contribution, while providing O2 until vasculature can surround the device. Moreover, it should help in the immune-protection to avoid acute and chronic rejection. The transplanted cells or islets should be embedded within biomaterials with tunable properties like injectability, stiffness and porosity mimicking natural ECM structural characteristics. And finally, an infinitive cell source that solves the donor scarcity should be found such as insulin producing cells derived from mesenchymal stem cells (MSCs), embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Several companies have registered their ATMPs and future studies envision new prototypes. In this review, we will discuss the mechanisms and etiology of diabetes, comparing the clinical trials in the last decades in order to define the main characteristics for future ATMPs.
Collapse
|
12
|
Jones GL, Juszczak MT, Hughes SJ, Kooner P, Powis SH, Press M. Time Course and Quantification of Pancreatic Islet Revasculariztion following Intraportal Transplantation. Cell Transplant 2017; 16:505-16. [PMID: 17708340 DOI: 10.3727/000000007783464993] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A large proportion of islets are lost after transplantation partly due to a lack of functional vasculature. Islets revascularize from host tissue but the process takes up to 2 weeks and has been suggested to result in reduced vascular density in engrafted islets. We describe a method for observing and quantifying the revascularization of intraportally transplanted islets that includes number, density, and branching of islet capillaries. Syngeneic islets were transplanted selectively into the two right posterior lobes of the liver of adult Lewis rats. Sections of the livers were dual stained for insulin and Bandeiraea simplicifolia and analyzed for islet morphology, area, and vascular density from day 0 to day 14 posttransplant and compared to native islets. Vascular density was 1431 ± 75.7 vessels/mm2 in native islets and fell to 325.3 ± 30.8 vessels/mm2 (p < 0.001) by day 1 posttransplant and subsequently increased until day 14 when it was significantly higher than in native islets (2612.5 ± 107.8 vessels/mm2, p < 0.001). The percentage of islet area occupied by vascular space was 9.1 ± 0.9% in native islets. After falling to 2.3 ± 0.3% (p < 0.001) 1 day posttransplant this rose to supranormal levels (21.5 ± 0.8%, p < 0.001) by day 14. The index of capillary branching was 0.771 ± 0.017 in native islets and fell to 0.465 ± 0.02 (p = 0.001) by day 3 but returned to native values by day 7 posttransplantation (0.726 ± 0.03). This technique provides a robust method for tracking and quantifying the revascularization of intraportally transplanted islets, which should enable the comparison of different strategies aimed at accelerating islet revascularization.
Collapse
Affiliation(s)
- Gareth L Jones
- Centre for Nephrology, Royal Free Campus, Royal Free and University College Medical School, London, NW3 2PF, UK
| | | | | | | | | | | |
Collapse
|
13
|
Sigrist S, Mechine-Neuville A, Mandes K, Calenda V, Braun S, Legeay G, Bellocq JP, Pinget M, Kessler L. Influence of VEGF on the Viability of Encapsulated Pancreatic Rat Islets after Transplantation in Diabetic Mice. Cell Transplant 2017; 12:627-35. [PMID: 14579931 DOI: 10.3727/000000003108747109] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
After pancreatic islet transplantation, insufficient blood supply is responsible for the loss of islet viability. The aim of our study was: 1) to determine the influence of vascular endothelial growth factor (VEGF) on the survival of encapsulated rat islets transplanted into healthy and diabetic mice and 2) to evaluate the metabolic efficiency of the VEGF-supplemented grafts. Twenty-four hours after culture, 50 rat islets immobilized into collagen in the presence of VEGF (100 ng/ml) and encapsulated (AN69 membrane, HOSPAL) were grafted in the peritoneal cavity of healthy or streptozotocin-induced diabetic mice (n = 6). Seven, 14, and 28 days after implantation, the encapsulation device and tissue surrounding the device were removed and the following parameters were analyzed: the number and the diameter of buds, the distance between devices and buds, the amount of cellular adhesion on the capsule surface, and the level of insulin secreted by encapsulated islet. For reversal of diabetes, 1000 rat islets encapsulated in the presence of VEGF were implanted in the peritoneal cavity of diabetic mice and fasting glycemia was analyzed. After 7 days of islet implantation in the absence of VEGF, the bud diameter was 16.1 ± 6.9 μm in diabetic mice and 34.4 ± 3.9 μm in healthy mice. However, the number of buds increased by a factor 2.5 in the presence of VEGF in both types of mice. Furthermore, when islets were transplanted in the presence of VEGF, the distance between the device and the buds was significantly decreased in both types of mice (p < 0.001) after 7, 14, and 28 days of islet implantation. Capsule analysis showed a decrease in cellular adhesion when the islets were encapsulated in the presence of VEGF. Insulin secretion of the islets was higher in the presence of VEGF compared with islets alone at all steps of the study. When 1000 rat islets were transplanted in the presence of VEGF, the glycemia level decreased to 6.2 ± 0.8 mmol/L after 3 days and remained stable until at least 28 days. In contrast, in the absence of VEGF, the initial decrease in the glucose level was rapidly followed by a relapse in hyperglycemia. In summary, VEGF increased the viability of engrafted encapsulated islets, increasing the duration of a normalized glycemia in diabetic mice following transplantation. Local adjunction of VEGF may therefore improve the clinical outcome of islet transplantation.
Collapse
Affiliation(s)
- S Sigrist
- Centre européen d'étude du Diabète, Faculté de Médecine, 11, rue Humann, 67 000, Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Johansson A, Sandvik D, Carlsson PO. Inhibition of p38 MAP Kinase in the Early Posttransplantation Phase Redistributes Blood Vessels from the Surrounding Stroma into the Transplanted Endocrine Tissue. Cell Transplant 2017; 15:483-8. [PMID: 17121159 DOI: 10.3727/000000006783981729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transplanted pancreatic islets attain a chronically decreased vascular density following transplantation, despite the increased concentrations of vascular endothelial growth factor (VEGF) secreted from beta-cells in response to hypoxia during culture and in the immediate posttransplantation phase. VEGF, however, exerts dual effects on endothelial cells, and in islet endothelial cells of the adult, the vascular permeability-inducing effects of VEGF seem normally more pronounced than those to induce angiogenesis. p38 MAP kinase activity has recently been shown to serve as a switch to separate these properties of VEGF; inhibition of p38 MAP kinase activity enhances VEGF-induced angiogenesis and, at the same time, abrogates VEGF-induced vascular permeability. We hypothesized that the revascularization of transplanted islets may be hampered by a predisposition of adult islet endothelial cells to react to VEGF by forming fenestrae rather than migrating and proliferating. We therefore administered the p38 MAP kinase inhibitor SB203580 by daily IP injections for the first 14 days following transplantation, and then studied the influence of this treatment on the oxygen tension, blood perfusion, and vascular density of the islet grafts 1 month posttransplantation. SB203580 treatment redistributed islet graft blood vessels from the stroma into the endocrine tissue, and this redistribution of blood vessels into the endocrine tissue was accompanied by an increased oxygenation of the islet cells. However, the total number of blood vessels in the tissue was not affected. The blood perfusion of the islet grafts was also similar in control and SB203580-treated animals. Our results suggest that effects of VEGF to preferentially induce vascular permeability may partially contribute to, but is not the main cause of, low revascularization of transplanted islets.
Collapse
Affiliation(s)
- Asa Johansson
- Department of Medical Cell Biology, Uppsala University, SE-751 23 Uppsala, Sweden
| | | | | |
Collapse
|
15
|
Kidszun A, Schneider D, Erb D, Hertl G, Schmidt V, Eckhard M, Preissner KT, Breier G, Bretzel RG, Linn T. Isolated Pancreatic Islets in Three-Dimensional Matrices are Responsive to Stimulators and Inhibitors of Angiogenesis. Cell Transplant 2017; 15:489-97. [PMID: 17121160 DOI: 10.3727/000000006783981774] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The formation of a new microvasculature is essential for the long-term survival and function of the islet graft. In this study we examined endothelium of isolated pancreatic islets by stimulation with growth factors, different culture conditions, and genetic modification. We also inspected the effect of immunosuppressives used in human transplantation on angiogenesis. Isolated islets were embedded in a three-dimensional fibrin or Matrigel matrix. The effect of hyperglycemia, hypoxia, and the addition of VEGF and bFGF was investigated. We exposed islets from transgenic mice expressing the VEGF gene (RIP1VEGF-A) to high glucose (16.7 mmol/L) medium and tested the immunosuppressive agents rapamycin (100 ng/ml) and FK506 (100 ng/ml). To quantify angiogenesis the percentage of sprouting islets was determined. New endothelial capillary-like structures protruded from isolated pancreatic islets. Addition of VEGF to the islets and transgenic RIP-VEGF islets showed a two- to threefold increase of sprouting islets compared to control. Hypoxic culture conditions stimulated angiogenesis, resulting in a twofold increase of capillary sprouting. Rapamycin and FK506 proved to be potent inhibitors of angiogenesis in this system, because a decrease of sprouting islets of more than 20% by both agents was observed. Isolated pancreatic islets are capable of forming new capillary structures and are susceptible to pro- and antiangiogenic stimuli.
Collapse
Affiliation(s)
- André Kidszun
- Medical Clinic and Policlinic 3, Justus Liebig University, Rodthohl 6, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Jansson L, Barbu A, Bodin B, Drott CJ, Espes D, Gao X, Grapensparr L, Källskog Ö, Lau J, Liljebäck H, Palm F, Quach M, Sandberg M, Strömberg V, Ullsten S, Carlsson PO. Pancreatic islet blood flow and its measurement. Ups J Med Sci 2016; 121:81-95. [PMID: 27124642 PMCID: PMC4900068 DOI: 10.3109/03009734.2016.1164769] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pancreatic islets are richly vascularized, and islet blood vessels are uniquely adapted to maintain and support the internal milieu of the islets favoring normal endocrine function. Islet blood flow is normally very high compared with that to the exocrine pancreas and is autonomously regulated through complex interactions between the nervous system, metabolites from insulin secreting β-cells, endothelium-derived mediators, and hormones. The islet blood flow is normally coupled to the needs for insulin release and is usually disturbed during glucose intolerance and overt diabetes. The present review provides a brief background on islet vascular function and especially focuses on available techniques to measure islet blood perfusion. The gold standard for islet blood flow measurements in experimental animals is the microsphere technique, and its advantages and disadvantages will be discussed. In humans there are still no methods to measure islet blood flow selectively, but new developments in radiological techniques hold great hopes for the future.
Collapse
Affiliation(s)
- Leif Jansson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- CONTACT Leif Jansson, Department of Medical Cell Biology, Biomedical Centre, Box 571, Husargatan 3, SE-75123 Uppsala, Sweden
| | - Andreea Barbu
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Birgitta Bodin
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Carl Johan Drott
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Daniel Espes
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Xiang Gao
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Liza Grapensparr
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Örjan Källskog
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Hanna Liljebäck
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - My Quach
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Monica Sandberg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Sara Ullsten
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Shalaly ND, Ria M, Johansson U, Åvall K, Berggren PO, Hedhammar M. Silk matrices promote formation of insulin-secreting islet-like clusters. Biomaterials 2016; 90:50-61. [PMID: 26986856 DOI: 10.1016/j.biomaterials.2016.03.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 11/29/2022]
Abstract
Ex vivo expansion of endocrine cells constitutes an interesting alternative to be able to match the unmet need of transplantable pancreatic islets. However, endocrine cells become fragile once removed from their extracellular matrix (ECM) and typically become senescent and loose insulin expression during conventional 2D culture. Herein we develop a protocol where 3D silk matrices functionalized with ECM-derived motifs are used for generation of insulin-secreting islet-like clusters from mouse and human primary cells. The obtained clusters were shown to attain an islet-like spheroid shape and to maintain functional insulin release upon glucose stimulation in vitro. Furthermore, in vivo imaging of transplanted murine clusters showed engraftment with increasing vessel formation during time. There was no sign of cell death and the clusters maintained or increased in size throughout the period, thus suggesting a suitable cluster size for transplantation.
Collapse
Affiliation(s)
- Nancy Dekki Shalaly
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden
| | - Massimiliano Ria
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital, S-17176 Stockholm, Sweden
| | - Ulrika Johansson
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden
| | - Karin Åvall
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital, S-17176 Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital, S-17176 Stockholm, Sweden
| | - My Hedhammar
- Division of Protein Technology, School of Biotechnology, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden; Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, S-750 07 Uppsala, Sweden.
| |
Collapse
|
18
|
Cotransplantation of Polymerized Hemoglobin Reduces β-Cell Hypoxia and Improves β-Cell Function in Intramuscular Islet Grafts. Transplantation 2015; 99:2077-82. [DOI: 10.1097/tp.0000000000000815] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
19
|
Nerve Growth Factor Improves Survival and Function of Transplanted Islets Via TrkA-mediated β Cell Proliferation and Revascularization. Transplantation 2015; 99:1132-43. [PMID: 25806408 DOI: 10.1097/tp.0000000000000655] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Nerve growth factor (NGF), which plays important roles in promoting growth and differentiation of nerve cells, has recently been reported as a regulator in pancreatic β cells in terms of insulin releasing function. In this study, we examined whether NGF stimulation would promote islet graft survival and function in islet transplantation. METHODS We found that supplementation of cultured islets with NGF improved the viability of islet cells and induced the production of insulin, vascular endothelial growth factor, and cellular proliferative markers. Because a specific inhibitor of TrkA, K252a, blocked all these effects, we propose that the TrkA receptor is the mediator of NGF stimulation. RESULTS After transplantation to the kidney subcapsule and liver of syngenic diabetic mice, a higher rate of normoglycemic achievement, increased serum insulin, and improved glucose tolerance were observed in the mice transplanted with NGF-pretreated islet grafts. Histological analysis revealed higher expression of insulin and vascular endothelial growth factor, an increase in proliferative β cells, and revascularization in NGF-pretreated islet grafts without activation of any inflammatory cells. CONCLUSIONS The NGF treatment can therefore serve as a new and promising therapeutic tool for improving islet graft viability and function in islet transplantation.
Collapse
|
20
|
Najjar M, Manzoli V, Abreu M, Villa C, Martino MM, Molano RD, Torrente Y, Pileggi A, Inverardi L, Ricordi C, Hubbell JA, Tomei AA. Fibrin gels engineered with pro-angiogenic growth factors promote engraftment of pancreatic islets in extrahepatic sites in mice. Biotechnol Bioeng 2015; 112:1916-26. [PMID: 25786390 DOI: 10.1002/bit.25589] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/21/2015] [Accepted: 03/09/2015] [Indexed: 01/08/2023]
Abstract
With a view toward reduction of graft loss, we explored pancreatic islet transplantation within fibrin matrices rendered pro-angiogenic by incorporation of minimal doses of vascular endothelial growth factor-A165 and platelet-derived growth factor-BB presented complexed to a fibrin-bound integrin-binding fibronectin domain. Engineered matrices allowed for extended release of pro-angiogenic factors and for their synergistic signaling with extracellular matrix-binding domains in the post-transplant period. Aprotinin addition delayed matrix degradation and prolonged pro-angiogenic factor availability within the graft. Both subcutaneous (SC) and epididymal fat pad (EFP) sites were evaluated. We show that in the SC site, diabetes reversal in mice transplanted with 1,000 IEQ of syngeneic islets was not observed for islets transplanted alone, while engineered matrices resulted in a diabetes median reversal time (MDRT) of 38 days. In the EFP site, the MDRT with 250 IEQ of syngeneic islets within the engineered matrices was 24 days versus 86 days for islets transplanted alone. Improved function of engineered grafts was associated with enhanced and earlier (by day 7) angiogenesis. Our findings show that by engineering the transplant site to promote prompt re-vascularization, engraftment and long-term function of islet grafts can be improved in relevant extrahepatic sites.
Collapse
Affiliation(s)
- Mejdi Najjar
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida
| | - Vita Manzoli
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida.,Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy
| | - Maria Abreu
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida
| | - Chiara Villa
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida.,Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Italy
| | - Mikaël M Martino
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - R Damaris Molano
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida
| | - Yvan Torrente
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida.,Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Italy
| | - Antonello Pileggi
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida.,Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Biomedical Engineering, University of Miami, Miami, Florida
| | - Luca Inverardi
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida.,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida.,Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Biomedical Engineering, University of Miami, Miami, Florida.,Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Jeffrey A Hubbell
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida.,Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Institute for Molecular Engineering, University of Chicago, Illinois
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Avenue, Miami, Florida. .,Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida. .,Department of Biomedical Engineering, University of Miami, Miami, Florida.
| |
Collapse
|
21
|
Abstract
The ultimate goal of diabetes therapy is the restoration of physiologic metabolic control. For type 1 diabetes, research efforts are focused on the prevention or early intervention to halt the autoimmune process and preserve β cell function. Replacement of pancreatic β cells via islet transplantation reestablishes physiologic β cell function in patients with diabetes. Emerging research shows that microRNAs (miRNAs), noncoding small RNA molecules produced by a newly discovered class of genes, negatively regulate gene expression. MiRNAs recognize and bind to partially complementary sequences of target messenger RNA (mRNA), regulating mRNA translation and affecting gene expression. Correlation between miRNA signatures and genome-wide RNA expression allows identification of multiple miRNA-mRNA pairs in biological processes. Because miRNAs target functionally related genes, they represent an exciting and indispensable approach for biomarkers and drug discovery. We are studying the role of miRNA in the context of islet immunobiology. Our research aims at understanding the mechanisms underlying pancreatic β cell loss and developing clinically relevant approaches for preservation and restoration of β cell function to treat insulin-dependent diabetes. Herein, we discuss some of our recent efforts related to the study of miRNA in islet inflammation and islet engraftment. Our working hypothesis is that modulation of the expression of specific microRNAs in the transplant microenvironment will be of assistance in enhancing islet engraftment and promoting long-term function.
Collapse
|
22
|
Figliolini F, Cantaluppi V, De Lena M, Beltramo S, Romagnoli R, Salizzoni M, Melzi R, Nano R, Piemonti L, Tetta C, Biancone L, Camussi G. Isolation, characterization and potential role in beta cell-endothelium cross-talk of extracellular vesicles released from human pancreatic islets. PLoS One 2014; 9:e102521. [PMID: 25028931 PMCID: PMC4100900 DOI: 10.1371/journal.pone.0102521] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/19/2014] [Indexed: 12/13/2022] Open
Abstract
The cross-talk between beta cells and endothelium plays a key role in islet physiopathology and in the revascularization process after islet transplantation. However, the molecular mechanisms involved in this cross-talk are not fully elucidated. Extracellular vesicles (EVs) are secreted membrane nanoparticles involved in inter-cellular communication through the transfer of proteins and nucleic acids. The aims of this study were: 1) isolation and characterization of EVs from human islets; 2) evaluation of the pro-angiogenic effect of islet-derived EVs on human islet endothelial cells (IECs). EVs were isolated by ultracentrifugation from conditioned medium of human islets and characterized by nanotrack analysis (Nanosight), FACS, western blot, bioanalyzer, mRNA/microRNA RT-PCR array. On IECs, we evaluated EV-induced insulin mRNA transfer, proliferation, resistance to apoptosis, in vitro angiogenesis, migration, gene and protein profiling. EVs sized 236±54 nm, expressed different surface molecules and islet-specific proteins (insulin, C-peptide, GLP1R) and carried several mRNAs (VEGFa, eNOS) and microRNAs (miR-27b, miR-126, miR-130 and miR-296) involved in beta cell function, insulin secretion and angiogenesis. Purified EVs were internalized into IECs inducing insulin mRNA expression, protection from apoptosis and enhancement of angiogenesis. Human islets release biologically active EVs able to shuttle specific mRNAs and microRNAs (miRNAs) into target endothelial cells. These results suggest a putative role for islet-derived EVs in beta cell-endothelium cross-talk and in the neoangiogenesis process which is critical for engraftment of transplanted islets.
Collapse
Affiliation(s)
| | | | - Michela De Lena
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Silvia Beltramo
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Renato Romagnoli
- Liver Transplantation Center, University of Torino, Torino, Italy
| | - Mauro Salizzoni
- Liver Transplantation Center, University of Torino, Torino, Italy
| | - Raffaella Melzi
- Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milano, Italy
| | - Rita Nano
- Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milano, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milano, Italy
| | - Ciro Tetta
- Fresenius Medical Care, Bad Homburg, Germany
| | - Luigi Biancone
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
23
|
Figliuzzi M, Bonandrini B, Silvani S, Remuzzi A. Mesenchymal stem cells help pancreatic islet transplantation to control type 1 diabetes. World J Stem Cells 2014; 6:163-172. [PMID: 24772243 PMCID: PMC3999774 DOI: 10.4252/wjsc.v6.i2.163] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/20/2013] [Accepted: 03/04/2014] [Indexed: 02/06/2023] Open
Abstract
Islet cell transplantation has therapeutic potential to treat type 1 diabetes, which is characterized by autoimmune destruction of insulin-producing pancreatic islet β cells. It represents a minimal invasive approach for β cell replacement, but long-term blood control is still largely unachievable. This phenomenon can be attributed to the lack of islet vasculature and hypoxic environment in the immediate post-transplantation period that contributes to the acute loss of islets by ischemia. Moreover, graft failures continue to occur because of immunological rejection, despite the use of potent immunosuppressive agents. Mesenchymal stem cells (MSCs) have the potential to enhance islet transplantation by suppressing inflammatory damage and immune mediated rejection. In this review we discuss the impact of MSCs on islet transplantation and focus on the potential role of MSCs in protecting islet grafts from early graft failure and from autoimmune attack.
Collapse
|
24
|
Quaranta P, Antonini S, Spiga S, Mazzanti B, Curcio M, Mulas G, Diana M, Marzola P, Mosca F, Longoni B. Co-transplantation of endothelial progenitor cells and pancreatic islets to induce long-lasting normoglycemia in streptozotocin-treated diabetic rats. PLoS One 2014; 9:e94783. [PMID: 24733186 PMCID: PMC3986409 DOI: 10.1371/journal.pone.0094783] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/20/2014] [Indexed: 01/09/2023] Open
Abstract
Graft vascularization is a crucial step to obtain stable normoglycemia in pancreatic islet transplantation. Endothelial progenitor cells (EPCs) contribute to neoangiogenesis and to the revascularization process during ischaemic events and play a key role in the response to pancreatic islet injury. In this work we co-transplanted EPCs and islets in the portal vein of chemically-induced diabetic rats to restore islet vascularization and to improve graft survival. Syngenic islets were transplanted, either alone or with EPCs derived from green fluorescent protein (GFP) transgenic rats, into the portal vein of streptozotocin-induced diabetic rats. Blood glucose levels were monitored and intraperitoneal glucose tolerance tests were performed. Real time-PCR was carried out to evaluate the gene expression of angiogenic factors. Diabetic-induced rats showed long-lasting (6 months) normoglycemia upon co-transplantation of syngenic islets and EPCs. After 3–5 days from transplantation, hyperglycaemic levels dropped to normal values and lasted unmodified as long as they were checked. Further, glucose tolerance tests revealed the animals' ability to produce insulin on-demand as indexed by a prompt response in blood glucose clearance. Graft neovascularization was evaluated by immunohistochemistry: for the first time the measure of endothelial thickness revealed a donor-EPC-related neovascularization supporting viable islets up to six months after transplant. Our results highlight the importance of a newly formed viable vascular network together with pancreatic islets to provide de novo adequate supply in order to obtain enduring normoglycemia and prevent diabetes-related long-term health hazards.
Collapse
Affiliation(s)
- Paola Quaranta
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Sara Antonini
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Benedetta Mazzanti
- Department of Clinical and Experimental Medicine, Haematology Section, University of Florence, Florence, Italy
| | - Michele Curcio
- U.O. Immunohaematology, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Marco Diana
- Department of Chemistry and Pharmacy, “G. Minardi” Laboratory of Cognitive Neuroscience, University of Sassari, Sassari, Italy
| | - Pasquina Marzola
- Department of Computer Science, University of Verona, Verona, Italy
| | - Franco Mosca
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Biancamaria Longoni
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
- * E-mail:
| |
Collapse
|
25
|
Brissova M, Aamodt K, Brahmachary P, Prasad N, Hong JY, Dai C, Mellati M, Shostak A, Poffenberger G, Aramandla R, Levy SE, Powers AC. Islet microenvironment, modulated by vascular endothelial growth factor-A signaling, promotes β cell regeneration. Cell Metab 2014; 19:498-511. [PMID: 24561261 PMCID: PMC4012856 DOI: 10.1016/j.cmet.2014.02.001] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 03/05/2013] [Accepted: 01/27/2014] [Indexed: 12/31/2022]
Abstract
Pancreatic islet endocrine cell and endothelial cell (EC) interactions mediated by vascular endothelial growth factor-A (VEGF-A) signaling are important for islet differentiation and the formation of highly vascularized islets. To dissect how VEGF-A signaling modulates intra-islet vasculature, islet microenvironment, and β cell mass, we transiently increased VEGF-A production by β cells. VEGF-A induction dramatically increased the number of intra-islet ECs but led to β cell loss. After withdrawal of the VEGF-A stimulus, β cell mass, function, and islet structure normalized as a result of a robust, but transient, burst in proliferation of pre-existing β cells. Bone marrow-derived macrophages (MΦs) recruited to the site of β cell injury were crucial for the β cell proliferation, which was independent of pancreatic location and circulating factors such as glucose. Identification of the signals responsible for the proliferation of adult, terminally differentiated β cells will improve strategies aimed at β cell regeneration and expansion.
Collapse
Affiliation(s)
- Marcela Brissova
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Kristie Aamodt
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Priyanka Brahmachary
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nripesh Prasad
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; Department of Biology, University of Alabama, Huntsville, Huntsville, AL 35899, USA
| | - Ji-Young Hong
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Chunhua Dai
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mahnaz Mellati
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alena Shostak
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Greg Poffenberger
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Radhika Aramandla
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Shawn E Levy
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Alvin C Powers
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
| |
Collapse
|
26
|
Samikannu B, Chen C, Lingwal N, Padmasekar M, Engel FB, Linn T. Dipeptidyl peptidase IV inhibition activates CREB and improves islet vascularization through VEGF-A/VEGFR-2 signaling pathway. PLoS One 2013; 8:e82639. [PMID: 24349326 PMCID: PMC3859629 DOI: 10.1371/journal.pone.0082639] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 11/04/2013] [Indexed: 01/09/2023] Open
Abstract
Substitution of pancreatic islets is a potential therapy to treat diabetes and it depends on reconstitution of islet’s capillary network. In this study, we addressed the question whether stabilization of Glucagon-Like-Peptide-1 (GLP-1) by inhibiting Dipeptidyl Peptidase-IV (DPP-IV) increases β-cell mass by modulating vascularization. Mouse or porcine donor islets were implanted under kidney capsule of diabetic mice treated with DPP-IV inhibitor sitagliptin. Grafts were analyzed for insulin production, β-cell proliferation and vascularization. In addition, the effect of sitagliptin on sprouting and Vascular Endothelial Growth Factor (VEGF)-A expression was examined ex vivo. The cAMP response element-binding (CREB) and VEGF-A/ Vascular Endothelial Growth Factor Receptor (VEGFR)-2 signaling pathway leading to islet vascularization was explored. Sitagliptin increased mean insulin content of islet grafts and area of insulin-positive tissue as well as β-cell proliferation. Interestingly, sitagliptin treatment also markedly increased endothelial cell proliferation, microvessel density and blood flow. Finally, GLP-1 (7-36) stimulated sprouting and VEGF expression, which was significantly enhanced by sitagliptin- mediated inhibition of DPP-IV. Our in vivo data demonstrate that sitagliptin treatment phosphorylated CREB and induced islet vascularization through VEGF-A/VEGFR-2 signaling pathway. This study paves a new pathway for improvement of islet transplantation in treating diabetes mellitus.
Collapse
Affiliation(s)
- Balaji Samikannu
- Third Medical Clinic and Policlinic, Justus-Liebig-University, Giessen, Germany
- * E-mail:
| | - Chunguang Chen
- CRTD / DFG- Center for Regenerative Therapies Dresden, Paul Langerhans Institut Dresden, Dresden, Germany
| | - Neelam Lingwal
- Third Medical Clinic and Policlinic, Justus-Liebig-University, Giessen, Germany
| | - Manju Padmasekar
- Third Medical Clinic and Policlinic, Justus-Liebig-University, Giessen, Germany
| | - Felix B. Engel
- University Hospital Erlangen, Experimental Renal and Cardiovascular Research, Nephropathology Division, Department of Pathology, Erlangen, Germany
| | - Thomas Linn
- Third Medical Clinic and Policlinic, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
27
|
Hypoxia as a target for tissue specific gene therapy. J Control Release 2013; 172:484-94. [DOI: 10.1016/j.jconrel.2013.05.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 05/13/2013] [Accepted: 05/24/2013] [Indexed: 12/28/2022]
|
28
|
Cantley J, Walters SN, Jung MH, Weinberg A, Cowley MJ, Whitworth PT, Kaplan W, Hawthorne WJ, O'connell PJ, Weir G, Grey ST. A Preexistent Hypoxic Gene Signature Predicts Impaired Islet Graft Function and Glucose Homeostasis. Cell Transplant 2013; 22:2147-59. [DOI: 10.3727/096368912x658728] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We examined whether hypoxic exposure prior to the event of transplantation would have a positive or negative effect upon later islet graft function. Mouse islets exposed to hypoxic culture were transplanted into syngeneic recipients. Islet graft function, β-cell physiology, as well as molecular changes were examined. Expression of hypoxia-response genes in human islets pre- and posttransplant was examined by microarray. Hypoxia-preexposed murine islet grafts provided poor glycemic control in their syngeneic recipients, marked by persistent hyperglycemia and pronounced glucose intolerance with failed first- and second-phase glucose-stimulated insulin secretion in vivo. Mechanistically, hypoxic preexposure stabilized HIF-1α with a concomitant increase in hypoxic-response genes including LDHA, and a molecular gene set, which would favor glycolysis and lactate production and impair glucose sensing. Indeed, static incubation studies showed that hypoxia-exposed islets exhibited dysregulated glucose responsiveness with elevated basal insulin secretion. Isolated human islets, prior to transplantation, express a characteristic hypoxia-response gene expression signature, including high levels of LDHA, which is maintained posttransplant. Hypoxic preexposure of an islet graft drives a HIF-dependent switch to glycolysis with subsequent poor glycemic control and loss of glucose-stimulated insulin secretion (GSIS). Early intervention to reverse or prevent these hypoxia-induced metabolic gene changes may improve clinical islet transplantation.
Collapse
Affiliation(s)
- James Cantley
- Diabetes and Obesity Research Program, Garvan Institute, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Stacey N. Walters
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Immunology Program, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - Min-Ho Jung
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Anita Weinberg
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Immunology Program, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - Mark J. Cowley
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Cancer Program, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - P. Tess Whitworth
- Diabetes and Obesity Research Program, Garvan Institute, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Warren Kaplan
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Peter Wills Bioinformatics Centre, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - Wayne J. Hawthorne
- The Centre for Transplant and Renal Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Philip J. O'connell
- The Centre for Transplant and Renal Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Gordon Weir
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Shane T. Grey
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, New South Wales, Australia
- Immunology Program, Garvan Institute, Darlinghurst, New South Wales, Australia
| |
Collapse
|
29
|
Revascularization of transplanted pancreatic islets and role of the transplantation site. Clin Dev Immunol 2013; 2013:352315. [PMID: 24106517 PMCID: PMC3782812 DOI: 10.1155/2013/352315] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/09/2013] [Indexed: 12/16/2022]
Abstract
Since the initial reporting of the successful reversal of hyperglycemia through the transplantation of pancreatic islets, significant research efforts have been conducted in elucidating the process of revascularization and the influence of engraftment site on graft function and survival. During the isolation process the intrinsic islet vascular networks are destroyed, leading to impaired revascularization after transplant. As a result, in some cases a significant quantity of the beta cell mass transplanted dies acutely following the infusion into the portal vein, the most clinically used site of engraftment. Subsequently, despite the majority of patients achieving insulin independence after transplant, a proportion of them recommence small, supplemental exogenous insulin over time. Herein, this review considers the process of islet revascularization after transplant, its limiting factors, and potential strategies to improve this critical step. Furthermore, we provide a characterization of alternative transplant sites, analyzing the historical evolution and their role towards advancing transplant outcomes in both the experimental and clinical settings.
Collapse
|
30
|
Liu D, Xiao H, Du C, Luo S, Li D, Pan L. The effect of fibroblast activation on vascularization in transplanted pancreatic islets. J Surg Res 2013; 183:450-6. [PMID: 23369362 DOI: 10.1016/j.jss.2012.12.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 11/22/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Insufficient revascularization of transplanted pancreatic islets is an important reason why the long-term effects of pancreatic islet transplantation on type I diabetes patients have been so limited. The goal of this study was to investigate the role of fibroblasts (FBs) activated by tumor cell supernatants on the vascularization of transplanted pancreatic islets. MATERIALS AND METHODS Pancreatic islets and activated or inactivated FBs were used for subrenal capsule transplantation. Mouse melanoma cell supernatants were used to activate FBs; the tests of the purity of the pancreatic islet cells of the donor, survival rate, and function of insulin secretion were performed to ensure high-quality transplants. Mice receiving the allogeneic transplantation were given tacrolimus and sirolimus to prevent rejection. The diabetic model was induced by streptozotocin. RESULTS Conditioned medium made of tumor cell supernatants was found to stimulate the expression of α-smooth muscle actin and vascular endothelial growth factor A to an extent notably greater than that of pancreatic islet transplantation alone or pancreatic islet transplantation combined with inactivated FBs. FBs from the recipient were associated with capillary density in the transplanted pancreatic islet most closely to that observed in isogenically transplanted pancreatic islets and the original pancreatic islet. In this way, activated FBs derived from the recipient combined with pancreatic transplantation were able to treat diabetes, and long-term survival was achieved. CONCLUSIONS The current research sheds new light on the revascularization of transplanted pancreatic islets: activated FBs derived from the recipients, when transplanted alongside pancreatic tissue, can promote revascularization inside the transplanted pancreatic islet.
Collapse
Affiliation(s)
- Dingzhi Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chong Qing Medical University, Chong Qing, PR China
| | | | | | | | | | | |
Collapse
|
31
|
Cowley MJ, Weinberg A, Zammit NW, Walters SN, Hawthorne WJ, Loudovaris T, Thomas H, Kay T, Gunton JE, Alexander SI, Kaplan W, Chapman J, O'Connell PJ, Grey ST. Human islets express a marked proinflammatory molecular signature prior to transplantation. Cell Transplant 2012; 21:2063-78. [PMID: 22404979 DOI: 10.3727/096368911x627372] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In the context of islet transplantation, experimental models show that induction of islet intrinsic NF-κB-dependent proinflammatory genes can contribute to islet graft rejection. Isolation of human islets triggers activation of the NF-κB and mitogen-activated kinase (MAPK) stress response pathways. However, the downstream NF-κB target genes induced in human islets during the isolation process are poorly described. Therefore, in this study, using microarray, bioinformatic, and RTqPCR approaches, we determined the pattern of genes expressed by a set of 14 human islet preparations. We found that isolated human islets express a panel of genes reminiscent of cells undergoing a marked NF-κB-dependent proinflammatory response. Expressed genes included matrix metallopeptidase 1 (MMP1) and fibronectin 1 (FN1), factors involved in tissue remodeling, adhesion, and cell migration; inflammatory cytokines IL-1β and IL-8; genes regulating cell survival including A20 and ATF3; and notably high expression of a set of chemokines that would favor neutrophil and monocyte recruitment including CXCL2, CCL2, CXCL12, CXCL1, CXCL6, and CCL28. Of note, the inflammatory profile of isolated human islets was maintained after transplantation into RAG(-/-) recipients. Thus, human islets can provide a reservoir of NF-κB-dependent inflammatory factors that have the potential to contribute to the anti-islet-graft immune response. To test this hypothesis, we extracted rodent islets under optimal conditions, forced activation of NF-κB, and transplanted them into allogenic recipients. These NF-κB activated islets not only expressed the same chemokine profile observed in human islets but also struggled to maintain normoglycemia posttransplantation. Further, NF-κB-activated islets were rejected with a faster tempo as compared to non-NF-κB-activated rodent islets. Thus, isolated human islets can make cell autonomous contributions to the ensuing allograft response by elaborating inflammatory factors that contribute to their own demise. These data highlight the potential importance of islet intrinsic proinflammatory responses as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mark J Cowley
- Peter Wills Bioinformatics Centre, Darlinghurst, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
GUO JW, CHEN C, HUANG Y, LI B. Combinatorial effects of Naomai Yihao Capsules () and vascular endothelial growth factor gene-transfected bone marrow mesenchymal stem cells on angiogenesis in cerebral ischemic tissues in rats. J TRADIT CHIN MED 2012; 32:87-92. [DOI: 10.1016/s0254-6272(12)60038-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
33
|
Advances and challenges in islet transplantation: islet procurement rates and lessons learned from suboptimal islet transplantation. J Transplant 2011; 2011:979527. [PMID: 22235361 PMCID: PMC3253477 DOI: 10.1155/2011/979527] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Accepted: 10/04/2011] [Indexed: 01/22/2023] Open
Abstract
The initial step in successful islet transplantation is procurement of healthy donor islets. Given the limited number of donor pancreata selected for islet isolation and that islets from multiple donors are typically required to obtain insulin independence, it is critical to improve pancreas procurement rates and yield of islets for transplantation. Islets are delicate microorgans that are susceptible to apoptosis, hypoxia, and ischemia during isolation, culture, and the peritransplant period. Once the islets are engrafted, both prompt revascularization and protection from beta-cell death and graft rejection are key to secure long-term survival and function. To facilitate the engraftment of more robust islets suitable for combating the challenging isolation period and proinflammatory transplantation milieu, numerous approaches have been employed to prevent beta-cell dysfunction and death including immune modulation, prevention of apoptosis and hypoxia, as well as stimulation of growth factors, angiogenesis, and reinnervation. In addition to briefly discussing islet isolation procedures, procurement rates, and islet transplantation, the relevant literature pertaining to successful suboptimal islet transplantation is reviewed to provide insight into potential approaches to balance the limited supply of available donor islets.
Collapse
|
34
|
Langlois A, Bietiger W, Seyfritz E, Maillard E, Vivot K, Peronet C, Meyer N, Kessler L, Jeandidier N, Pinget M, Sigrist S. Improvement of Rat Islet Viability during Transplantation: Validation of Pharmacological Approach to Induce VEGF Overexpression. Cell Transplant 2011; 20:1333-42. [DOI: 10.3727/096368910x557182] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Delayed and insufficient revascularization during islet transplantation deprives islets of oxygen and nutrients, resulting in graft failure. Vascular endothelial growth factor (VEGF) could play a critical role in islet revascularization. We aimed to develop pharmacological strategies for VEGF overexpression in pancreatic islets using the iron chelator deferoxamine (DFO), thus avoiding obstacles or safety risks associated with gene therapy. Rat pancreatic islets were infected in vivo using an adenovirus (ADE) encoding human VEGF gene (4.108 pfu/pancreas) or were incubated in the presence of DFO (10 μmol/L). In vitro viability, functionality, and the secretion of VEGF were evaluated in islets 1 and 3 days after treatment. Infected islets or islets incubated with DFO were transplanted into the liver of syngenic diabetic rats and the graft efficiency was estimated in vivo by measuring body weight, glycemia, C-peptide secretion, and animal survival over a period of 2 months. DFO induced transient VEGF overexpression over 3 days, whereas infection with ADE resulted in prolonged VEGF overexpression lasting 14 days; however, this was toxic and decreased islet viability and functionality. The in vivo study showed a decrease in rat deaths after the transplantation of islets treated with DFO or ADE compared with the sham and control group. ADE treatment improved body weight and C-peptide levels. Gene therapy and DFO improved metabolic control in diabetic rats after transplantation, but this effect was limited in the presence of DFO. The pharmacological approach is an interesting strategy for improving graft efficiency during transplantation, but this approach needs to be improved with drugs that are more specific.
Collapse
Affiliation(s)
- A. Langlois
- Centre européen d'étude du Diabète, Strasbourg, France
| | - W. Bietiger
- Centre européen d'étude du Diabète, Strasbourg, France
| | - E. Seyfritz
- Centre européen d'étude du Diabète, Strasbourg, France
| | - E. Maillard
- Centre européen d'étude du Diabète, Strasbourg, France
| | - K. Vivot
- Centre européen d'étude du Diabète, Strasbourg, France
| | - C. Peronet
- Centre européen d'étude du Diabète, Strasbourg, France
| | - N. Meyer
- Faculté de Médecine de Strasbourg, Laboratoire de Biostatistique, Strasbourg, France
| | - L. Kessler
- Service d'endocrinologie, de diabète et des maladies métaboliques, Médicale B Hopital civil, Strasbourg cedex, France
- Université de Strasbourg (UdS), Strasbourg cedex, France
| | - N. Jeandidier
- Service d'endocrinologie, de diabète et des maladies métaboliques, Médicale B Hopital civil, Strasbourg cedex, France
- Université de Strasbourg (UdS), Strasbourg cedex, France
| | - M. Pinget
- Centre européen d'étude du Diabète, Strasbourg, France
- Service d'endocrinologie, de diabète et des maladies métaboliques, Médicale B Hopital civil, Strasbourg cedex, France
- Université de Strasbourg (UdS), Strasbourg cedex, France
| | - S. Sigrist
- Centre européen d'étude du Diabète, Strasbourg, France
| |
Collapse
|
35
|
Källskog Ö, Jansson L. Autoregulation of islet graft blood flow follows the implantation. J Surg Res 2010; 171:865-70. [PMID: 20888593 DOI: 10.1016/j.jss.2010.07.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 07/01/2010] [Accepted: 07/30/2010] [Indexed: 11/15/2022]
Abstract
BACKGROUND Transplantation of pancreatic islets necessitates a revascularization, which is associated with a generalized graft vascular dysfunction, manifested, e.g., as a capillary hypertension, a decreased graft blood perfusion and graft hypoxia. Some of these changes can be due to impaired autoregulation of the newly formed vasculature in the islet grafts, and the aim of the present study was to further examine if this was the case. MATERIALS AND METHODS We implanted 250 syngeneic islets under the renal capsule of rats and studied them 1 or 12-13 mo later. The blood perfusion of the whole kidney, renal cortex, and islet grafts were recorded in anesthetized animals with an ultrasound probe or laser-Doppler probes, respectively. The blood pressure in the kidneys was then gradually decreased by an adjustable clamp, during simultaneous measurement of blood flow values. RESULTS The whole kidney, renal cortex, and islet grafts regulated their blood flow in concert with one another down to pressures of approximately 60 mmHg both 1 and 12-13 mo after implantation. However, the variability was greater at 1 mo. CONCLUSION Islets transplanted under the renal capsule show similar autoregulatory properties with the kidney. It may be that the autoregulatory capacity of the renal interlobular arteries provides the underlying mechanism. This may be of importance for the good long-term survival of transplanted islets at this implantation site in experimental studies.
Collapse
Affiliation(s)
- Örjan Källskog
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
36
|
Johansson A, Olerud J, Johansson M, Carlsson PO. Angiostatic factors normally restrict islet endothelial cell proliferation and migration: implications for islet transplantation. Transpl Int 2010; 22:1182-8. [PMID: 19891047 DOI: 10.1111/j.1432-2277.2009.00939.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
New blood vessel formation in transplanted islets occurs within 7-14 days post-transplantation through both the expansion of donor islet endothelium and ingrowth of blood vessels from the implantation organ. However, several studies indicate that although the islets attract recipient blood vessels, the formed intra-islet vascular network is insufficient, which affects islet post-transplant function. This study aimed to develop an in vitro model to investigate the migration and proliferation properties of isolated liver and islet endothelium.Rat islet or liver endothelium was purified using Bandeiraea simplicifolia(BS-1)-coated Dynabeads. The liver endothelium displayed an increased migration and proliferation to islet-conditioned medium. These effects were fully prevented by adding a neutralizing vascular endothelial growth factor (VEGF)-antibody. In contrast, islet-produced VEGF failed to induce islet endothelial cell migration and only had marginal effects on islet endothelial cell proliferation.These properties could, however, be activated through blocking the effects of either endostatin, thrombospondin-1 or alpha(1)-antitrypsin. In conclusion, VEGF may attract recipient blood vessels towards intrahepatically transplanted islets,but intra-islet vascular expansion is hampered by angiostatic factors present within the islets and the islet endothelium. Inhibition of angiostatic factors early after transplantation may provide a strategy to restore the islet vascular network and improve islet graft function.
Collapse
Affiliation(s)
- Asa Johansson
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Box 571, SE-751 23 Uppsala, Sweden.
| | | | | | | |
Collapse
|
37
|
Weber LM, Lopez CG, Anseth KS. Effects of PEG hydrogel crosslinking density on protein diffusion and encapsulated islet survival and function. J Biomed Mater Res A 2009; 90:720-9. [PMID: 18570315 DOI: 10.1002/jbm.a.32134] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The rational design of immunoprotective hydrogel barriers for transplanting insulin-producing cells requires an understanding of protein diffusion within the hydrogel network and how alterations to the network structure affect protein diffusion. Hydrogels of varying crosslinking density were formed via the chain polymerization of dimethacrylated PEG macromers of varying molecular weight, and the diffusion of six model proteins with molecular weights ranging from 5700 to 67,000 g/mol was observed in these hydrogel networks. Protein release profiles were used to estimate diffusion coefficients for each protein/gel system that exhibited Fickian diffusion. Diffusion coefficients were on the order of 10(-6)-10(-7) cm(2)/s, such that protein diffusion time scales (t(d) = L(2)/D) from 0.5-mm thick gels vary from 5 min to 24 h. Adult murine islets were encapsulated in PEG hydrogels of varying crosslinking density, and islet survival and insulin release was maintained after two weeks of culture in each gel condition. While the total insulin released during a 1 h glucose stimulation period was the same from islets in each sample, increasing hydrogel crosslinking density contributed to delays in insulin release from hydrogel samples within the 1 h stimulation period.
Collapse
Affiliation(s)
- Laney M Weber
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309-0424, USA
| | | | | |
Collapse
|
38
|
Langlois A, Bietiger W, Sencier MC, Maillard E, Pinget M, Kessler L, Sigrist S. Adenoviral infection or deferoxamine? Two approaches to overexpress VEGF in beta-cell lines. J Drug Target 2009; 17:415-22. [PMID: 19527112 DOI: 10.1080/10611860902929832] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Rapid and adequate revascularization of transplanted islets is important for their survival and function during transplantation. Vascular endothelial growth factor (VEGF) could play a critical role with respect to islet revascularization. The aim of this study was to compare two strategies that are used to overexpress VEGF in beta-cells: (1) gene therapy through adenoviral infection and (2) a pharmacological approach using deferoxamine (DFO). beta-Cell lines from rat insulinoma (RINm5F) were either infected using an adenovirus encoding the gene of human VEGF 165 or incubated with DFO. One day after treatment, the viability of RINm5F cells was preserved with 10 micromol/L of DFO (103.95 +/- 5.66% toward control; n = 4). In addition, adenoviral infection maintained the viability of cells for all the concentrations used. In both treatments, overexpression of VEGF was in a comparable level. Finally, the ratio of Bax/Bcl-2 indicated that the apoptosis increased in infected beta-cells whereas treatment with DFO seems to be antiapoptotic. Our results suggest that the use of DFO could be a realistic approach to improve the vascularization of islets during transplantation.
Collapse
Affiliation(s)
- Allan Langlois
- Centre européen d'étude du Diabète, Boulevard René Leriche, BP 30029, 67033 Strasbourg Cedex 2, France.
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
BACKGROUND Islet transplantation success depends on the number and quality of islets transplanted. This study aimed at exploring the molecular mechanisms associated with cold pancreas preservation and their impact on islet cell survival and function. METHODS Rat pancreata were stored in cold University of Wisconsin preservation solution for short (3 hr; control) or long (18 hr) cold ischemia times (CIT). RESULTS Pancreata exposed to long CIT yielded lower islet numbers and showed reduced cellular viability; isolated islets displayed higher levels of phosphorylated stress-activated protein kinase (c-jun N-terminal Kinase and Mitogen-Activated Protein Kinase-p38), and chemokine (C-C) ligand-3, and lower levels of vascular endothelial growth factor, interleukins (IL)-9 and IL-10. Islets obtained from long-CIT pancreata were functionally impaired after transplantation. Differential proteomic expression in pancreatic tissue after CIT included increased eukaryotic translation elongation factor-1-alpha-1 (apoptosis related) and reduced Clade-B (serine protease inhibitor). CONCLUSIONS Our study indicates that cold ischemia stimulates inflammatory pathways (chemokine (c-c)ligand-3, phosphorylation of c-jun N-terminal Kinase and mitogen-activated protein kinase-p38, and eukaryotic translation elongation factor-1-alpha-1) and decreases repair/cytoprotective pathways (IL-10, vascular endothelial growth factor, and Clade-B), all of which may negatively affect the quality and mass of islets obtained from a donor pancreas.
Collapse
|
40
|
Ham JN, Crutchlow MF, Desai BM, Simmons RA, Stoffers DA. Exendin-4 normalizes islet vascularity in intrauterine growth restricted rats: potential role of VEGF. Pediatr Res 2009; 66:42-6. [PMID: 19287346 PMCID: PMC2735861 DOI: 10.1203/pdr.0b013e3181a282a5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Intrauterine growth restriction (IUGR) induced by uterine artery ligation in pregnant rats leads to low birth weight and early insulin secretory defects followed by the development of insulin resistance, decline in beta-cell mass, and diabetes in adulthood. Neonatal administration of Exendin-4 (Ex-4) prevents the deterioration of beta-cell mass and the onset of adult-onset diabetes. Our aim was to determine whether this effect occurs through preservation of islet vascularization. In 2 wk-old IUGR rats, endothelial-specific lectin staining revealed a 40% reduction in islet vascular density (p = 0.027), which was normalized by neonatal Ex-4. VEGF-A protein expression was reduced in IUGR islets compared with controls at postnatal d 1 (P). Neonatal Ex-4 normalized islet VEGF protein expression at P7. Neither IUGR nor Ex-4 administration to IUGR rats affected relative VEGF splice isoform RNA levels. Together, the reduced vascularity in IUGR islets before the deterioration of beta-cell mass, and the enhancement of VEGF expression and normalization of islet vascularity by neonatal Ex-4, suggest islet vascularity as an early determinant of beta-cell mass and as a potential therapeutic target for diabetes prevention.
Collapse
Affiliation(s)
- J Nina Ham
- Department of Medicine and the Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, 415 Curie Boulevard, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
41
|
Andersson A, Carlsson PO, Carlsson C, Olsson R, Nordin A, Johansson M, Palm F, Tyrberg B, Källskog O, Tillmar L, Welsh N, Mattsson G, Jansson L. Promoting islet cell function after transplantation. Cell Biochem Biophys 2009; 40:55-64. [PMID: 15289643 DOI: 10.1385/cbb:40:3:55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Engraftment (i.e., the adaptation of transplanted pancreatic islets to their new surroundings with regard to revascularization, reinnervation, and reorganization of other stromal compartments) is of crucial importance for the survival and function of the endocrine cells. Previous studies suggest that transplantation induces both vascular and stromal dysfunctions in the implanted islets when compared with endogenous islets. Thus the vascular density and the blood perfusion of islet grafts is decreased and accompanied with a capillary hypertension. This leads to hypoxic conditions, with an associated shift toward anaerobic metabolism in grafted islets. An improved engraftment will prevent or compensate for the vascular/stromal dysfunction seen in transplanted islets and thereby augment survival of the islet implant. By such means the number of islets needed to cure the recipient will be lessened. This will increase the number of patients that can be transplanted with the limited material available.
Collapse
Affiliation(s)
- Arne Andersson
- Department of Medical Cell Biology, Biomedical Centre, Uppsala University, Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Movahedi B, Gysemans C, Jacobs-Tulleneers-Thevissen D, Mathieu C, Pipeleers D. Pancreatic duct cells in human islet cell preparations are a source of angiogenic cytokines interleukin-8 and vascular endothelial growth factor. Diabetes 2008; 57:2128-36. [PMID: 18492788 PMCID: PMC2494672 DOI: 10.2337/db07-1705] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Engraftment and function of human islet cell implants is considered to be dependent on their rapid and adequate revascularization. Studies with rodent islet grafts have shown that vascular endothelial growth factor (VEGF) expression by beta-cells can promote this process. The present work examines whether human islet preparations produce VEGF as well as interleukin (IL)-8, another angiogenic protein, and assesses the role of contaminating duct cells in VEGF and IL-8-mediated angiogenesis. RESEARCH DESIGN AND METHODS Human islet and pancreatic duct cell preparations are compared for their respective expression and production of VEGF and IL-8 during culture as well as following transplantation in nonobese diabetic (NOD)/scid mice. The associated angiogenic effects are measured in an in vitro aortic ring assay and in an in vivo chick embryo chorioallantoic membrane assay. RESULTS Cultured pancreatic duct cells expressed 3- and 10-fold more VEGF and IL-8, respectively, than cultured human islet endocrine cells and released both proteins at angiogenic levels. The angiogenic effect of purified duct cells was higher than that of purified endocrine islet cells and was completely blocked by a combination of IL-8 and VEGF antibodies. Human duct cell implants under the kidney capsule of NOD/scid mice expressed higher levels of IL-8 and VEGF than human islet cell implants and induced circulating IL-8 and VEGF levels during the first day posttransplantation. CONCLUSIONS Human duct cell-released IL-8 and VEGF may help revascularization of currently used human islet cell grafts. Further work should examine whether and when this effect can prevail over other inflammatory and immune influences of this cell type.
Collapse
Affiliation(s)
- Babak Movahedi
- Diabetes Research Center, Brussels Free University, Brussels, Belgium
| | | | | | | | | |
Collapse
|
43
|
Abstract
The major forms of diabetes are characterized by pancreatic islet beta-cell dysfunction and decreased beta-cell numbers, raising hope for cell replacement therapy. Although human islet transplantation is a cell-based therapy under clinical investigation for the treatment of type 1 diabetes, the limited availability of human cadaveric islets for transplantation will preclude its widespread therapeutic application. The result has been an intense focus on the development of alternate sources of beta cells, such as through the guided differentiation of stem or precursor cell populations or the transdifferentiation of more plentiful mature cell populations. Realizing the potential for cell-based therapies, however, requires a thorough understanding of pancreas development and beta-cell formation. Pancreas development is coordinated by a complex interplay of signaling pathways and transcription factors that determine early pancreatic specification as well as the later differentiation of exocrine and endocrine lineages. This review describes the current knowledge of these factors as they relate specifically to the emergence of endocrine beta cells from pancreatic endoderm. Current therapeutic efforts to generate insulin-producing beta-like cells from embryonic stem cells have already capitalized on recent advances in our understanding of the embryonic signals and transcription factors that dictate lineage specification and will most certainly be further enhanced by a continuing emphasis on the identification of novel factors and regulatory relationships.
Collapse
Affiliation(s)
- Jennifer M. Oliver-Krasinski
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Doris A. Stoffers
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
44
|
Olerud J, Johansson M, Lawler J, Welsh N, Carlsson PO. Improved vascular engraftment and graft function after inhibition of the angiostatic factor thrombospondin-1 in mouse pancreatic islets. Diabetes 2008; 57:1870-7. [PMID: 18420490 PMCID: PMC2453615 DOI: 10.2337/db07-0724] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 04/10/2008] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Insufficient development of a new intra-islet capillary network after transplantation may be one contributing factor to the failure of islet grafts in clinical transplantation. The present study tested the hypothesis that the angiostatic factor thrombospondin-1 (TSP-1), which is normally present in islets, restricts intra-islet vascular expansion posttransplantation. RESEARCH DESIGN AND METHODS Pancreatic islets of TSP-1-deficient (TSP-1(-/-)) mice or wild-type islets transfected with siRNA for TSP-1 were transplanted beneath the renal capsule of syngeneic or immunocompromised recipient mice. RESULTS Both genetically TSP-1(-/-) islets and TSP-1 siRNA-transfected islet cells demonstrated an increased vascular density when compared with control islets 1 month after transplantation. This was also reflected in a markedly increased blood perfusion and oxygenation of the grafts. The functional importance of the improved vascular engraftment was analyzed by comparing glucose-stimulated insulin release from islet cells transfected with either TSP-1 siRNA or scramble siRNA before implantation. These experiments showed that the increased revascularization of grafts composed of TSP-1 siRNA-transfected islet cells correlated to increments in both their first and second phase of glucose-stimulated insulin secretion. CONCLUSIONS Our findings demonstrate that inhibition of TSP-1 in islets intended for transplantation may be a feasible strategy to improve islet graft revascularization and function.
Collapse
Affiliation(s)
- Johan Olerud
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Magnus Johansson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Jack Lawler
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Nils Welsh
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
45
|
Nico B, Mangieri D, Crivellato E, Longo V, De Giorgis M, Capobianco C, Corsi P, Benagiano V, Roncali L, Ribatti D. HIF activation and VEGF overexpression are coupled with ZO-1 up-phosphorylation in the brain of dystrophic mdx mouse. Brain Pathol 2007; 17:399-406. [PMID: 17784876 PMCID: PMC8095599 DOI: 10.1111/j.1750-3639.2007.00090.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD) metabolic and structural alterations of the central nervous system are described. Here, we investigated in the brain of 10 mdx mice and in five control ones, the expression of hypoxia inducible factor-1alpha (HIF-1alpha) and we correlated it with the expression of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor-2 (VEGFR-2) and of the endothelial tight junction proteins zonula occludens-1 (ZO-1) and claudin-1. Results showed an activation of mRNA HIF-1alpha by reverse transcription polymerase chain reaction (RT-PCR) and a strong HIF1-alpha labeling of perivascular glial cells and cortical neurons by immunohistochemistry, in mdx mouse. Moreover, overexpression of VEGF and VEGFR-2, respectively, in neurons and in endothelial cells coupled with changes to endothelial ZO-1 and claudin-1 expression in the latter were detected by immunoblotting and immunohistochemistry, in the mdx brain. Furthermore, by immunoprecipitation, an up-phosphorylation of ZO-1 was demonstrated in mdx endothelial cells in parallel with the reduction in ZO-1 protein content. These data suggest that the activation of HIF-1alpha in the brain of dystrophic mice coupled with VEGF and VEGFR-2 up-regulation and ZO-1 and claudin-1 rearrangement might contribute to both blood-brain barrier opening and increased angiogenesis.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/physiopathology
- Brain/metabolism
- Brain/physiopathology
- Brain Diseases, Metabolic, Inborn/genetics
- Brain Diseases, Metabolic, Inborn/metabolism
- Brain Diseases, Metabolic, Inborn/physiopathology
- Claudin-1
- Disease Models, Animal
- Endothelial Cells/metabolism
- Female
- Hypoxia-Inducible Factor 1/genetics
- Hypoxia-Inducible Factor 1/metabolism
- Immunohistochemistry
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/physiopathology
- Neuroglia/metabolism
- Neurons/metabolism
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Phosphorylation
- RNA, Messenger/metabolism
- Up-Regulation/physiology
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/metabolism
- Zonula Occludens-1 Protein
Collapse
Affiliation(s)
- Beatrice Nico
- Department of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cross SE, Richards SK, Clark A, Benest AV, Bates DO, Mathieson PW, Johnson PRV, Harper SJ, Smith RM. Vascular endothelial growth factor as a survival factor for human islets: effect of immunosuppressive drugs. Diabetologia 2007; 50:1423-32. [PMID: 17476476 DOI: 10.1007/s00125-007-0670-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Accepted: 03/07/2007] [Indexed: 12/20/2022]
Abstract
AIMS/HYPOTHESIS Rapamycin, part of the immunosuppressive regimen of the Edmonton protocol, has been shown to inhibit vascular endothelial growth factor (VEGF) production and VEGF-mediated survival signalling in tumour cell lines. This study investigates the survival-promoting activities of VEGF in human islets and the effects of rapamycin on islet viability. MATERIALS AND METHODS Levels of VEGF and its receptors in isolated human islets and whole pancreas was determined by western blotting and immunostaining. Islet viability following VEGF or immunosuppressive drug treatment was determined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Islet VEGF release was measured by ELISA. Mouse islets infected with an adenovirus expressing the gene for VEGF were transplanted syngeneically into streptozotocin-induced diabetic mice, with blood glucose levels measured three times per week. RESULTS Isolated human islets produced multiple isoforms of VEGF and VEGF receptors 1, 2 and 3 and the coreceptor neuropilin 1. Exogenous VEGF (10 ng/ml) prevented human islet death induced by serum starvation, which suggests that VEGF can act as a survival factor for human islets. Transplantation of mouse islets infected with a VEGF-expressing adenovirus in a syngeneic model, improved glycaemic control at day 1 post-transplantation (p < 0.05). Rapamycin at 10 and 100 ng/ml significantly reduced islet VEGF release (by 37 +/- 4% and 43 +/- 6%, respectively; p < 0.05) and at 100 ng/ml reduced islet viability (by 36 +/- 9%) and insulin release (by 47 +/- 7%, all vs vehicle-treated controls; p < 0.05). Tacrolimus had no effect on islet VEGF release or viability. CONCLUSIONS/INTERPRETATION Our data suggest that rapamycin may have deleterious effects on islet survival post-transplantation, both through a direct effect on islet viability and indirectly through blockade of VEGF-mediated revascularisation.
Collapse
Affiliation(s)
- S E Cross
- Academic Renal Unit, Southmead Hospital, University of Bristol, Bristol, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cheng Y, Liu YF, Zhang JL, Li TM, Zhao N. Elevation of vascular endothelial growth factor production and its effect on revascularization and function of graft islets in diabetic rats. World J Gastroenterol 2007; 13:2862-6. [PMID: 17569125 PMCID: PMC4395641 DOI: 10.3748/wjg.v13.i20.2862] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine whether the elevated vascular endothelial growth factor (VEGF) expression produced by the transfected vascular endothelial cells (VECs) could stimulate angiogenesis of the graft islets and exert its effect on the graft function.
METHODS: Thirty diabetic recipient rats were divided into three groups (n = 10 per group). In the control group, 300 IEQ islets were transplanted in each rat under the capsule of the right kidney, which were considered as marginal grafts. In the VEC group, VEC together with the islets were transplanted in each rat. In the VEGF group, VEC transfected by pIRES2-EGFP/VEGF165 plasmid and the islets were transplanted in each rat. Blood glucose and insulin levels were evaluated every other day after operation. Intravenous glucose tolerance test (IVGTT) was performed 10 d after the transplantation. Hematoxylin and eosin (HE) staining was used to evaluate the histological features of the graft islets. Immunohistochemical staining was used to detect insulin-6, VEGF and CD34 (MVD) expression in the graft islets.
RESULTS: Blood glucose and insulin levels in the VEGF group restored to normal 3 d after transplantation. In contrast, diabetic rats receiving the same islets with or without normal VECs displayed moderate hyperglycemia and insulin, without a significant difference between these two groups. IVGTT showed that both the amplitude of blood glucose induction and the kinetics of blood glucose in the VEGF group restored to normal after transplantation. H&E and immunohistochemical staining showed the presence of a large amount of graft islets under the capsule of the kidney, which were positively stained with insulin-6 and VEGF antibodies in the VEGF group. In the cell masses, CD34-stained VECs were observed. The similar masses were also seen in the other two groups, but with a fewer positive cells stained with insulin-6 and CD34 antibodies. No VEGF-positive cells appeared in these groups. Microvessel density (MVD) was significantly higher in the VEGF group compared to the other two groups.
CONCLUSION: Elevated VEGF production by trans-fected vascular endothelial cells in the site of islet transplantation stimulates angiogenesis of the islet grafts. The accelerated islet revascularization in early stage could improve the outcome of islet transplantation, and enhance the graft survival.
Collapse
Affiliation(s)
- Ying Cheng
- Organ Transplant Unit of First Affiliated Hospital of China Medical University, 155 Nanjingbei Street, Heping District, Shenyang 110001, Liaoning Province, China.
| | | | | | | | | |
Collapse
|
48
|
Abstract
The islets of Langerhans consist of endocrine cells embedded in a network of specialized capillaries that regulate islet blood flow. Despite evidence for a critical role of islet perfusion in endocrine pancreas function, there is information to support no fewer than three models of endocrine cell perfusion, emphasizing the lack of a universally accepted physiological theory. Islet blood flow is regulated by signals, such as hormones and nutrients that reach the islet vasculature from distant tissues via the bloodstream. In addition, islet perfusion determines communication between endocrine and exocrine cells and between different types of endocrine cells within islets. Interest in islet microcirculation has increased after improvements in islet transplantation, a therapy for diabetes mellitus that requires revascularization of grafted islets in a new host organ. Abnormal revascularization is thought to be partly responsible for differences in graft and native islet function. Similarly, angiogenesis has been shown to be a critical step in the transformation of islet hyperplasia to neoplasia.
Collapse
Affiliation(s)
- Nikiforos Ballian
- The Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 1709 Dryden, Suite 1500, Houston, Texas 77030, USA
| | | |
Collapse
|
49
|
Mathe Z, Dupraz P, Rinsch C, Thorens B, Bosco D, Zbinden M, Morel P, Berney T, Pepper MS. Tetracycline-regulated expression of VEGF-A in beta cells induces angiogenesis: improvement of engraftment following transplantation. Cell Transplant 2007; 15:621-36. [PMID: 17176614 DOI: 10.3727/000000006783981675] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Early revascularization of pancreatic islet cells after transplantation is crucial for engraftment, and it has been suggested that vascular endothelial growth factor-A (VEGF-A) plays a significant role in this process. Although VEGF gene therapy can improve angiogenesis, uncontrolled VEGF secretion can lead to vascular tumor formation. Here we have explored the role of temporal VEGF expression, controlled by a tetracycline (TC)-regulated promoter, on revascularization and engraftment of genetically modified beta cells following transplantation. To this end, we modified the CDM3D beta cell line using a lentiviral vector to promote secretion of VEGF-A either in a TC-regulated (TET cells) or a constitutive (PGK cells) manner. VEGF secretion, angiogenesis, cell proliferation, and stimulated insulin secretion were assessed in vitro. VEGF secretion was increased in TET and PGK cells, and VEGF delivery resulted in angiogenesis, whereas addition of TC inhibited these processes. Insulin secretion by the three cell types was similar. We used a syngeneic mouse model of transplantation to assess the effects of this controlled VEGF expression in vivo. Time to normoglycemia, intraperitoneal glucose tolerance test, graft vascular density, and cellular mass were evaluated. Increased expression of VEGF resulted in significantly better revascularization and engraftment after transplantation when compared to control cells. In vivo, there was a significant increase in vascular density in grafted TET and PGK cells versus control cells. Moreover, the time for diabetic mice to return to normoglycemia and the stimulated plasma glucose clearance were also significantly accelerated in mice transplanted with TET and PGK cells when compared to control cells. VEGF was only needed during the first 2-3 weeks after transplantation; when removed, normoglycemia and graft vascularization were maintained. TC-treated mice grafted with TC-treated cells failed to restore normoglycemia. This approach allowed us to switch off VEGF secretion when the desired effects had been achieved. TC-regulated temporal expression of VEGF using a gene therapy approach presents a novel way to improve early revascularization and engraftment after islet cell transplantation.
Collapse
Affiliation(s)
- Zoltan Mathe
- Transplantation and Surgical Department, Semmelweis University, Budapest, H-1089, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cantaluppi V, Biancone L, Romanazzi GM, Figliolini F, Beltramo S, Ninniri MS, Galimi F, Romagnoli R, Franchello A, Salizzoni M, Perin PC, Ricordi C, Segoloni GP, Camussi G. Antiangiogenic and immunomodulatory effects of rapamycin on islet endothelium: relevance for islet transplantation. Am J Transplant 2006; 6:2601-11. [PMID: 16981910 DOI: 10.1111/j.1600-6143.2006.01534.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Donor intra-islet endothelial cells contribute to neovascularization after transplantation. Several factors may interfere with this process and ultimately influence islet engraftment. Rapamycin, a central immunosuppressant in islet transplantation, is an mTOR inhibitor that has been shown to inhibit cancer angiogenesis. The aim of this study was to evaluate the effects of rapamycin on islet endothelium. Rapamycin inhibited the outgrowth of endothelial cells from freshly purified human islets and the formation of capillary-like structures in vitro and in vivo after subcutaneous injection within Matrigel plugs into SCID mice. Rapamycin decreased migration, proliferation and angiogenic properties of human and mouse islet-derived endothelial cell lines with appearance of apoptosis. The expression of angiogenesis-related factors VEGF, alphaVbeta3 integrin and thrombospondin-1 on islet endothelium was altered in the presence of rapamycin. On the other hand, rapamycin decreased the surface expression of molecules involved in immune processes such as ICAM-1 and CD40 and reduced the adhesion of T cells to islet endothelium. Our results suggest that rapamycin exerts dual effects on islet endothelium inducing a simultaneous inhibition of angiogenesis and a down-regulation of receptors involved in lymphocyte adhesion and activation.
Collapse
Affiliation(s)
- V Cantaluppi
- Department of Internal Medicine and Research Centre for Experimental Medicine (CeRMS), University of Torino, Corso Dogliotti 14, Torino 10126, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|