1
|
Rodacki M, Silva KR, Araujo DB, Dantas JR, Ramos MEN, Zajdenverg L, Baptista LS. Immunomodulatory agents and cell therapy for patients with type 1 diabetes. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2025; 68:e240233. [PMID: 40215356 PMCID: PMC11967186 DOI: 10.20945/2359-4292-2024-0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/17/2024] [Indexed: 04/15/2025]
Abstract
Type 1 diabetes (TID) is a chronic disease caused by autoimmune destruction of pancreatic β-cells, that progresses in three stages: 1) stage 1: β-cell autoimmunity + normoglycemia; 2) stage 2: β-cell autoimmunity + mild dysglycemia; 3) stage 3: symptomatic disease + hyperglycemia. Interventions to prevent or cure T1D in the various stages of the disease have been pursued and may target the prevention of the destruction of β cells, regression of insulitis, preservation or recovery of β cells residual mass. Some therapies show promising results that might change the natural history and the approach to patients with T1D in the next few years. Teplizumab, a humanized monoclonal antibody that binds to CD3, was recently approved in the USA to delay Stage 3 T1D in individuals ≥ 8 years of age. Other non-cellular immunomodulatory therapies, both antigen-specific and non-specific, have shown interesting results either in patients with stage 2 or recent onset stage 3 T1D. Cell therapies such as non-myeloablative transplantation of autologous hematopoietic stem cells, mesenchymal stem cells, and tolerogenic dendritic cells have been also studied in these individuals, aiming immunomodulation. Stem cell-derived islet replacement therapy is promising for patients with long- standing T1D, especially with asymptomatic hypoglycemia not resolved by technology. This review aimed to provide updated information on the main immunomodulatory agents and cell therapy options for type 1 diabetes.
Collapse
Affiliation(s)
- Melanie Rodacki
- Departamento de Medicina Interna, Universidade Federal do Rio de Janeiro,
Rio de Janeiro, RJ, Brasil
| | - Karina Ribeiro Silva
- Laboratório de Pesquisa com Células-Tronco, Departamento de
Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro,
Rio de Janeiro, RJ, Brasil
| | | | - Joana R. Dantas
- Departamento de Medicina Interna, Universidade Federal do Rio de Janeiro,
Rio de Janeiro, RJ, Brasil
| | | | - Lenita Zajdenverg
- Departamento de Medicina Interna, Universidade Federal do Rio de Janeiro,
Rio de Janeiro, RJ, Brasil
| | - Leandra Santos Baptista
- NUMPEX-BIO, Campus Duque de Caxias Professor Geraldo Cidade, Universidade
Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
2
|
Katano T, Inagaki A, Imura T, Yamana H, Saito R, Endo Kumata Y, Suzuki S, Hagiwara Y, Ohashi K, Watanabe K, Tabata Y, Goto M. A novel approach for hepatocyte transplantation at the liver surface. Cell Transplant 2025; 34:9636897251329308. [PMID: 40208805 PMCID: PMC12032460 DOI: 10.1177/09636897251329308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 04/12/2025] Open
Abstract
Hepatocyte transplantation (HTx) is a promising alternative to liver transplantation; however, poor engraftment remains a major challenge. Although co-transplantation with adipose tissue-derived stromal cells (ADSCs) or islets improves engraftment, exposure of these cells to the portal vein enhances innate immune responses, resulting in a significant loss of hepatocytes. Therefore, we investigated HTx at the liver surface as a novel approach that does not involve the portal vein. Hepatocytes were transplanted onto the liver surface of syngeneic analbuminemic rats with or without ADSCs and/or islets. Serum albumin levels and immunohistochemical staining of the transplanted hepatocytes were evaluated. Hepatocyte engraftment was compared between the liver surface and intraportal groups. To examine the detailed mechanisms behind co-transplantation, co-cultured supernatants were analyzed using multiplex assays, and inhibition tests using neutralizing antibodies were performed. Results showed that islet and ADSC co-transplantation markedly enhanced hepatocyte engraftment at the liver surface (P < 0.01), and its efficiency was comparable to that of intraportal transplantation (P = 0.35). In the co-transplantation group, cells were not necessarily in proximity, suggesting that humoral factors are important. In an in vitro study, hepatocyte function was significantly improved by co-culturing with islets and ADSCs (P < 0.01). Multiplex assays and inhibition tests revealed several important humoral factors, most notably insulin, which promoted hepatocyte engraftment. These findings suggest that HTx at the liver surface, together with crucial factors, may be a novel alternative strategy for intraportal transplantation.
Collapse
Affiliation(s)
- Takumi Katano
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Hiroki Yamana
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Ryusuke Saito
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Yukiko Endo Kumata
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Shoki Suzuki
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Yoshiya Hagiwara
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Kazuo Ohashi
- Graduate School and School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Yasuhiko Tabata
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
- Department of Surgery, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
3
|
Endo Kumata Y, Inagaki A, Nakamura Y, Imura T, Saito R, Katano T, Suzuki S, Tokodai K, Kamei T, Unno M, Watanabe K, Tabata Y, Goto M. A novel method of pancreatic islet transplantation at the liver surface using a gelatin hydrogel nonwoven fabric. Cell Transplant 2025; 34:9636897251328419. [PMID: 40264358 PMCID: PMC12035123 DOI: 10.1177/09636897251328419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 04/24/2025] Open
Abstract
Considering the limitations of intraportal transplantation (Tx), we sought to establish an alternative approach for it-transplanting islets onto the liver surface (LS) by optimizing adipose tissue-derived stem cell (ADSC) co-Tx procedures with a gelatin hydrogel nonwoven fabric (GHNF). In the in vivo study, we examined the use of the GHNF, the effectiveness of islet covering materials, and preferred procedures for ADSC co-Tx using a syngeneic rat model. Immunohistochemical staining was performed to evaluate the extracellular matrix (ECM) expression and angiogenesis. In the in vitro study, we analyzed the culture supernatants to identify crucial factors secreted from ADSCs in different ADSC co-Tx procedures. It was shown that the GHNF should be used to cover the islets but not to embed internally (encapsulate) them. Utilization of the GHNF in LS Tx resulted in significantly better glucose changes (P = 0.0002) and cure rate of diabetic recipients (P = 0.0003) than the use of a common adhesion barrier. Although neovascularization was comparable among groups, ECM reconstitution tended to be higher when the GHNF was used. ADSC co-Tx further enhanced ECM reconstitution only when ADSCs were cultured in the GHNF before islet Tx. Leptin, vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), and several chemokines were identified as candidate factors for enhancing ECM reconstitution (P < 0.001). The inhibition assay using antagonist suggested that leptin might be at least in part responsible for the difference in transplant efficiency in distinct ADSC co-Tx methods. This study showed that the GHNF effectively improved the outcomes of LS islet Tx, mainly due to ECM reconstitution around the islets. Furthermore, we established a novel method of LS islet Tx by combining a GHNF with ADSCs, which is equally effective as intraportal Tx.
Collapse
Affiliation(s)
- Yukiko Endo Kumata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Graduate School of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryusuke Saito
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takumi Katano
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shoki Suzuki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuaki Tokodai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiko Tabata
- Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
4
|
Niknam B, Ayenehdeh JM, Hossein-Khannazer N, Vosough M, Tajik N. Adipose Tissue-Derived Mesenchymal Stromal Cells Modulate Inflammatory Response and Improve Allograft Islet Transplant in Mice Model of Type 1 Diabetes. Endocr Res 2024; 49:223-231. [PMID: 38982737 DOI: 10.1080/07435800.2024.2377286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE Type 1 diabetes mellitus (T1DM) is an autoimmune disease where immune cells attack insulin-producing beta cells. Islet transplantation is a promising treatment for T1DM. This study aims to evaluate the effects of adipose tissue-derived mesenchymal stem cells (AT-MSCs) in combination with pancreatic islet transplantation using hydrogel. METHODS T1DM mouse model was established using streptozotocin (STZ). Islets and AT-MSCs were co-embedded in a hydrogel and transplanted into diabetic mice. Five groups with six animals in each (control, hydrogel alone, AT-MSCs embedded hydrogel, islet embedded in hydrogel, and islet + AT-MSCs co-imbedded into a hydrogel) were evaluated in terms of blood glucose, insulin levels and serum and lavage cytokine production. RESULTS During 32 days, blood glucose levels decreased from over 400 mg/dl to less than 150 mg/dl in the transplanted mice. Analysis showed increased transformation growth factor beta (TGF-β1) and IL-4 levels, while IL-17 and IFN-γ levels significantly decreased in the MSC-treated groups. CONCLUSION These findings suggest that using AT-MSCs with hydrogel could be a beneficial alternative for enhancing pancreatic islet engraftment and function.
Collapse
Affiliation(s)
- Bahare Niknam
- Immunology Research Center (IRC), Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamal Mohammadi Ayenehdeh
- Immunology Research Center (IRC), Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nader Tajik
- Immunology Research Center (IRC), Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Kado T, Tomimaru Y, Kobayashi S, Harada A, Sasaki K, Iwagami Y, Yamada D, Noda T, Takahashi H, Kita S, Shimomura I, Miyagawa S, Doki Y, Eguchi H. Skeletal Myoblast Cells Enhance the Function of Transplanted Islets in Diabetic Mice. J Diabetes Res 2024; 2024:5574968. [PMID: 38800586 PMCID: PMC11126349 DOI: 10.1155/2024/5574968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/05/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Islet transplantation (ITx) is an established and safe alternative to pancreas transplantation for type 1 diabetes mellitus (T1DM) patients. However, most ITx recipients lose insulin independence by 3 years after ITx due to early graft loss, such that multiple donors are required to achieve insulin independence. In the present study, we investigated whether skeletal myoblast cells could be beneficial for promoting angiogenesis and maintaining the differentiated phenotypes of islets. In vitro experiments showed that the myoblast cells secreted angiogenesis-related cytokines (vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and stromal-derived factor-1α (SDF-1α)), contributed to maintenance of differentiated islet phenotypes, and enhanced islet cell insulin secretion capacity. To verify these findings in vivo, we transplanted islets alone or with myoblast cells under the kidney capsule of streptozotocin-induced diabetic mice. Compared with islets alone, the group bearing islets with myoblast cells had a significantly lower average blood glucose level. Histological examination revealed that transplants with islets plus myoblast cells were associated with a significantly larger insulin-positive area and significantly higher number of CD31-positive microvessels compared to islets alone. Furthermore, islets cotransplanted with myoblast cells showed JAK-STAT signaling activation. Our results suggest two possible mechanisms underlying enhancement of islet graft function with myoblast cells cotransplantation: "indirect effects" mediated by angiogenesis and "direct effects" of myoblast cells on islets via the JAK-STAT cascade. Overall, these findings suggest that skeletal myoblast cells enhance the function of transplanted islets, implying clinical potential for a novel ITx procedure involving myoblast cells for patients with diabetes.
Collapse
Affiliation(s)
- Takeshi Kado
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Adipose Management, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
6
|
Mizui T, Inagaki A, Nakamura Y, Imura T, Uematsu SS, Miyagi S, Kamei T, Unno M, Watanabe K, Goto M. A Recombinant Peptide Device Combined with Adipose Tissue-Derived Stem Cells Enhances Subcutaneous Islet Engraftment. Cells 2024; 13:499. [PMID: 38534342 PMCID: PMC10968997 DOI: 10.3390/cells13060499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/28/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024] Open
Abstract
Subcutaneous space has been considered an attractive site for islet graft transplantation; however, the oxygen tension and vascularization are insufficient for islet graft survival. We investigated whether subcutaneous pre-implantation of a recombinant peptide (RCP) device with adipose tissue-derived stem cells (ADSCs) enhanced subcutaneous islet engraftment. RCP devices with/without syngeneic ADSCs were pre-implanted into the subcutaneous space of C57BL/6 mice. Syngeneic islets (300 or 120 islet equivalents (IEQs)) were transplanted into the pre-treated space after diabetes induction using streptozotocin. The cure rates of groups in which RCP devices were implanted four weeks before transplantation were significantly better than the intraportal transplantation group when 300 IEQs of islets were transplanted (p < 0.01). The blood glucose changes in the RCP+ADSCs-4w group was significantly ameliorated in comparison to the RCP-4w group when 120 IEQs of islets were transplanted (p < 0.01). Immunohistochemical analyses showed the collagen III expression in the islet capsule of the RCP+ADSCs-4w group was significantly enhanced in comparison to the RCP-4w and RCP+ADSCs-d10 groups (p < 0.01, p < 0.01). In addition, the number of von Willebrand factor-positive vessels within islets in the RCP+ADSCs-4w group was significantly higher than the RCP-4w group. These results suggest that using ADSCs in combination with an RCP device could enhance the restoration of the extracellular matrices, induce more efficient prevascularization within islets, and improve the graft function.
Collapse
Affiliation(s)
- Takahiro Mizui
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.I.); (T.I.); (K.W.)
| | - Yasuhiro Nakamura
- Division of Pathology, Graduate School of Medicine, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan;
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.I.); (T.I.); (K.W.)
| | - Satomi Suzuki Uematsu
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
| | - Shigehito Miyagi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.I.); (T.I.); (K.W.)
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.I.); (T.I.); (K.W.)
| |
Collapse
|
7
|
Yan D, Song Y, Zhang B, Cao G, Zhou H, Li H, Sun H, Deng M, Qiu Y, Yi W, Sun Y. Progress and application of adipose-derived stem cells in the treatment of diabetes and its complications. Stem Cell Res Ther 2024; 15:3. [PMID: 38167106 PMCID: PMC10763319 DOI: 10.1186/s13287-023-03620-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Diabetes mellitus (DM) is a serious chronic metabolic disease that can lead to many serious complications, such as cardiovascular disease, retinopathy, neuropathy, and kidney disease. Once diagnosed with diabetes, patients need to take oral hypoglycemic drugs or use insulin to control blood sugar and slow down the progression of the disease. This has a significant impact on the daily life of patients, requiring constant monitoring of the side effects of medication. It also imposes a heavy financial burden on individuals, their families, and even society as a whole. Adipose-derived stem cells (ADSCs) have recently become an emerging therapeutic modality for DM and its complications. ADSCs can improve insulin sensitivity and enhance insulin secretion through various pathways, thereby alleviating diabetes and its complications. Additionally, ADSCs can promote tissue regeneration, inhibit inflammatory reactions, and reduce tissue damage and cell apoptosis. The potential mechanisms of ADSC therapy for DM and its complications are numerous, and its extensive regenerative and differentiation ability, as well as its role in regulating the immune system and metabolic function, make it a powerful tool in the treatment of DM. Although this technology is still in the early stages, many studies have already proven its safety and effectiveness, providing new treatment options for patients with DM or its complications. Although based on current research, ADSCs have achieved some results in animal experiments and clinical trials for the treatment of DM, further clinical trials are still needed before they can be applied in a clinical setting.
Collapse
Affiliation(s)
- Dongxu Yan
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Yujie Song
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Bing Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Guojie Cao
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Haitao Zhou
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Hong Li
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Hao Sun
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Meng Deng
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Yufeng Qiu
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China.
| | - Yang Sun
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, 127# Changlexi Road, Xi'an, 710032, China.
| |
Collapse
|
8
|
Saito R, Inagaki A, Nakamura Y, Imura T, Kanai N, Mitsugashira H, Endo Kumata Y, Katano T, Suzuki S, Tokodai K, Kamei T, Unno M, Watanabe K, Tabata Y, Goto M. A Gelatin Hydrogel Nonwoven Fabric Combined With Adipose Tissue-Derived Stem Cells Enhances Subcutaneous Islet Engraftment. Cell Transplant 2024; 33:9636897241251621. [PMID: 38756050 PMCID: PMC11102670 DOI: 10.1177/09636897241251621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/18/2024] Open
Abstract
Subcutaneous islet transplantation is a promising treatment for severe diabetes; however, poor engraftment hinders its prevalence. We previously revealed that a gelatin hydrogel nonwoven fabric (GHNF) markedly improved subcutaneous islet engraftment. We herein investigated whether the addition of adipose tissue-derived stem cells (ADSCs) to GHNF affected the outcome. A silicone spacer sandwiched between two GHNFs with (AG group) or without (GHNF group) ADSCs, or a silicone spacer alone (Silicone group) was implanted into the subcutaneous space of healthy mice at 6 weeks before transplantation, then diabetes was induced 7 days before transplantation. Syngeneic islets were transplanted into the pretreated space. Intraportal transplantation (IPO group) was also performed to compare the transplant efficiency. Blood glucose, intraperitoneal glucose tolerance, immunohistochemistry, and inflammatory mediators were evaluated. The results in the subcutaneous transplantation were compared using the Silicone group as a control. The results of the IPO group were also compared with those of the AG group. The AG group showed significantly better blood glucose changes than the Silicone and the IPO groups. The cure rate of AG group (72.7%) was the highest among the groups (GHNF; 40.0%, IPO; 40.0%, Silicone; 0%). The number of vWF-positive vessels in the subcutaneous space of the AG group was significantly higher than that in other groups before transplantation (P < 0.01). Lectin angiography also showed that the same results (P < 0.05). According to the results of the ADSCs tracing, ADSCs did not exist at the transplant site (6 weeks after implantation). The positive rates for laminin and collagen III constructed around the transplanted islets did not differ among groups. Inflammatory mediators were higher in the Silicone group, followed by the AG and GHNF groups. Pretreatment using bioabsorbable scaffolds combined with ADSCs enhanced neovascularization in subcutaneous space, and subcutaneous islet transplantation using GHNF with ADSCs was superior to intraportal islet transplantation.
Collapse
Affiliation(s)
- Ryusuke Saito
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Norifumi Kanai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Mitsugashira
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukiko Endo Kumata
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takumi Katano
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shoki Suzuki
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuaki Tokodai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiko Tabata
- Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
9
|
Abadpour S, Niemi EM, Orrhult LS, Hermanns C, de Vries R, Nogueira LP, Haugen HJ, Josefsen D, Krauss S, Gatenholm P, van Apeldoorn A, Scholz H. Adipose-Derived Stromal Cells Preserve Pancreatic Islet Function in a Transplantable 3D Bioprinted Scaffold. Adv Healthc Mater 2023; 12:e2300640. [PMID: 37781993 PMCID: PMC11469278 DOI: 10.1002/adhm.202300640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 09/07/2023] [Indexed: 10/03/2023]
Abstract
Intra-portal islet transplantation is currently the only clinically approved beta cell replacement therapy, but its outcome is hindered by limited cell survival due to a multifactorial reaction against the allogeneic tissue in liver. Adipose-derived stromal cells (ASCs) can potentially improve the islet micro-environment by their immunomodulatory action. The challenge is to combine both islets and ASCs in a relatively easy and consistent long-term manner in a deliverable scaffold. Manufacturing the 3D bioprinted double-layered scaffolds with primary islets and ASCs using a mix of alginate/nanofibrillated cellulose (NFC) bioink is reported. The diffusion properties of the bioink and the supportive effect of human ASCs on islet viability, glucose sensing, insulin secretion, and reducing the secretion of pro-inflammatory cytokines are demonstrated. Diabetic mice transplanted with islet-ASC scaffolds reach normoglycemia seven days post-transplantation with no significant difference between this group and the group received islets under the kidney capsules. In addition, animals transplanted with islet-ASC scaffolds stay normoglycemic and show elevated levels of C-peptide compared to mice transplanted with islet-only scaffolds. The data present a functional 3D bioprinted scaffold for islets and ASCs transplanted to the extrahepatic site and suggest a possible role of ASCs on improving the islet micro-environment.
Collapse
Affiliation(s)
- Shadab Abadpour
- Department of Transplant MedicineOslo University HospitalOslo0372Norway
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
| | - Essi M. Niemi
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Department of Vascular SurgeryAker HospitalOslo University HospitalOslo0586Norway
| | - Linnea Strid Orrhult
- 3D Bioprinting CenterWWSCDepartment of Chemistry and Chemical EngineeringChalmers University of TechnologyGothenburg41296Sweden
| | - Carolin Hermanns
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | - Rick de Vries
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | | | | | - Dag Josefsen
- Section for Cellular TherapyRadiumhospitaletOslo University HospitalOslo0379Norway
| | - Stefan Krauss
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Department of Immunology and Transfusion MedicineOslo University HospitalOslo0372Norway
| | - Paul Gatenholm
- 3D Bioprinting CenterWWSCDepartment of Chemistry and Chemical EngineeringChalmers University of TechnologyGothenburg41296Sweden
- CELLHEAL ASSandvika1337Norway
| | - Aart van Apeldoorn
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | - Hanne Scholz
- Department of Transplant MedicineOslo University HospitalOslo0372Norway
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Section for Cellular TherapyRadiumhospitaletOslo University HospitalOslo0379Norway
| |
Collapse
|
10
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
11
|
Kanai N, Inagaki A, Nakamura Y, Imura T, Mitsugashira H, Saito R, Miyagi S, Watanabe K, Kamei T, Unno M, Tabata Y, Goto M. A gelatin hydrogel nonwoven fabric improves outcomes of subcutaneous islet transplantation. Sci Rep 2023; 13:11968. [PMID: 37488155 PMCID: PMC10366205 DOI: 10.1038/s41598-023-39212-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 07/21/2023] [Indexed: 07/26/2023] Open
Abstract
Subcutaneous islet transplantation is a promising treatment for severe diabetes; however, poor engraftment hinders its prevalence. We previously reported that a recombinant peptide (RCP) enhances subcutaneous islet engraftment. However, it is impractical for clinical use because RCP must be removed when transplanting islets. We herein investigated whether a novel bioabsorbable gelatin hydrogel nonwoven fabric (GHNF) could improve subcutaneous islet engraftment. A silicon spacer with or without GHNF was implanted into the subcutaneous space of diabetic mice. Syngeneic islets were transplanted into the pretreated space or intraportally (Ipo group). Blood glucose, intraperitoneal glucose tolerance, immunohistochemistry, CT angiography and gene expression were evaluated. The cure rate and glucose tolerance of the GHNF group were significantly better than in the control and Ipo groups (p < 0.01, p < 0.05, respectively). In the GHNF group, a limited increase of vWF-positive vessels was detected in the islet capsule, whereas laminin (p < 0.05), collagen III and IV were considerably enhanced. TaqMan arrays revealed a significant upregulation of 19 target genes (including insulin-like growth factor-2) in the pretreated space. GHNF markedly improved the subcutaneous islet transplantation outcomes, likely due to ECM compensation and protection of islet function by various growth factors, rather than enhanced neovascularization.
Collapse
Affiliation(s)
- Norifumi Kanai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-0872, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Graduate School of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, 983-8536, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Hiroaki Mitsugashira
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-0872, Japan
| | - Ryusuke Saito
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-0872, Japan
| | - Shigehito Miyagi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-0872, Japan
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-0872, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-0872, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences (LiMe), Kyoto University, Kyoto, 606-8507, Japan
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-0872, Japan.
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| |
Collapse
|
12
|
Mikłosz A, Chabowski A. Adipose-derived Mesenchymal Stem Cells Therapy as a new Treatment Option for Diabetes Mellitus. J Clin Endocrinol Metab 2023; 108:1889-1897. [PMID: 36916961 PMCID: PMC10348459 DOI: 10.1210/clinem/dgad142] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/01/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023]
Abstract
The worldwide increase in the prevalence of diabetes mellitus has raised the demand for new therapeutic strategies targeting diabetic symptoms and its chronic complications. Among different treatment options for diabetes, adipose-derived mesenchymal stem cells (ADMSCs) therapy attract the most attention. The therapeutic effects of ADMSCs are based primarily on their paracrine release of immunomodulatory, anti-inflammatory, and trophic factors. Animal models of diabetes as well as human clinical trials have shown that ADMSCs can effectively facilitate endogenous β cell regeneration, preserve residual β cell mass, reduce islet graft rejection, regulate the immune system, and ultimately improve insulin sensitivity or ameliorate insulin resistance in peripheral tissues. Nevertheless, transplantation of mesenchymal stem cells is associated with certain risks; therefore recently much attention has been devoted to ADMSCs derivatives, such as exosomes or conditioned media, as therapeutic agents for the treatment of diabetes. Compared to ADMSCs, cell-free therapy has even better therapeutic potential. This narrative review summarizes recent outcomes and molecular mechanisms of ADMSCs action in the treatment for both type 1 DM and type 2 DM, as well as shows their feasibility, benefits, and current limitations.
Collapse
Affiliation(s)
- Agnieszka Mikłosz
- Department of Physiology, Medical University of Bialystok, 15-222 Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-222 Bialystok, Poland
| |
Collapse
|
13
|
Meng F, Shen F, Chu X, Ling H, Qiao Y, Liu D. Hsa_circ_0008500 inhibits apoptosis of adipose-derived stem cells under high glucose through hsa-miR-1273h-5p/ELK1 axis. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 37014014 DOI: 10.1002/tox.23801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
Preliminary researches have confirmed that the number of apoptosis of adipose tissue-derived stem cells (ADSCs) in patients with diabetes is significantly increased, leading to a difficult healing wound. Increasing researches revealed that circular RNAs (circRNAs) can control apoptosis. However, it is still unclear whether and how circRNAs are critical for regulating ADSCs apoptosis. In this study, we utilized in vitro model in which ADSCs were cultivated with normal glucose (NG) (5.5 mM) or high glucose (HG) (25 mM) medium, respectively, and found that more apoptotic ADSCs were observed in HG medium comparing to ADSCs in NG medium. Furthermore, we found that hsa_circ_0008500 attenuated HG-mediated ADSCs apoptosis. In addition, Hsa_circ_0008500 could directly interact with hsa-miR-1273h-5p, acting as a miRNA sponge, which subsequently suppressed Ets-like protein-1(ELK1) expression, the downstream target of hsa-miR-1273h-5p. Thus, these results indicated that targeting the hsa_circ_0008500/hsa-miR-1273h-5p/ELK1 signaling pathway in ADSCs may be a potential target for repairing diabetic wounds.
Collapse
Affiliation(s)
- Fandong Meng
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Fengjie Shen
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Xuan Chu
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Hongwei Ling
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Yun Qiao
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Deshan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
14
|
Cheng HY, Anggelia MR, Lin CH, Wei FC. Toward transplantation tolerance with adipose tissue-derived therapeutics. Front Immunol 2023; 14:1111813. [PMID: 37187733 PMCID: PMC10175575 DOI: 10.3389/fimmu.2023.1111813] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Solid organ and composite tissue allotransplanation have been widely applied to treat end-stage organ failure and massive tissue defects, respectively. Currently there are a lot of research endeavors focusing on induction of transplantation tolerance, to relieve the burden derived from long-term immunosuppressant uptake. The mesenchymal stromal cells (MSCs) have been demonstrated with potent immunomodulatory capacities and applied as promising cellular therapeutics to promote allograft survival and induce tolerance. As a rich source of adult MSCs, adipose tissue provides additional advantages of easy accessibility and good safety profile. In recent years, the stromal vascular fraction (SVF) isolated from adipose tissues following enzymatic or mechanical processing without in vitro culture and expansion has demonstrated immunomodulatory and proangiogenic properties. Furthermore, the secretome of AD-MSCs has been utilized in transplantation field as a potential "cell-free" therapeutics. This article reviews recent studies that employ these adipose-derived therapeutics, including AD-MSCs, SVF, and secretome, in various aspects of organ and tissue allotransplantation. Most reports validate their efficacies in prolonging allograft survival. Specifically, the SVF and secretome have performed well for graft preservation and pretreatment, potentially through their proangiogenic and antioxidative capacities. In contrast, AD-MSCs were suitable for peri-transplantation immunosuppression. The proper combination of AD-MSCs, lymphodepletion and conventional immunosuppressants could consistently induce donor-specific tolerance to vascularized composite allotransplants (VCA). For each type of transplantation, optimizing the choice of therapeutics, timing, dose, and frequency of administration may be required. Future progress in the application of adipose-derived therapeutics to induce transplantation tolerance will be further benefited by continued research into their mechanisms of action and the development of standardized protocols for isolation methodologies, cell culture, and efficacy evaluation.
Collapse
Affiliation(s)
- Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- *Correspondence: Hui-Yun Cheng,
| | - Madonna Rica Anggelia
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chan Wei
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
15
|
Cotransplantation With Adipose Tissue-derived Stem Cells Improves Engraftment of Transplanted Hepatocytes. Transplantation 2022; 106:1963-1973. [PMID: 35404871 PMCID: PMC9521584 DOI: 10.1097/tp.0000000000004130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Hepatocyte transplantation is expected to be an alternative therapy to liver transplantation; however, poor engraftment is a severe obstacle to be overcome. The adipose tissue-derived stem cells (ADSCs) are known to improve engraftment of transplanted pancreatic islets, which have many similarities to the hepatocytes. Therefore, we examined the effects and underlying mechanisms of ADSC cotransplantation on hepatocyte engraftment. METHODS Hepatocytes and ADSCs were cotransplanted into the renal subcapsular space and livers of syngeneic analbuminemic rats, and the serum albumin level was quantified to evaluate engraftment. Immunohistochemical staining and fluorescent staining to trace transplanted cells in the liver were also performed. To investigate the mechanisms, cocultured supernatants were analyzed by a multiplex assay and inhibition test using neutralizing antibodies for target factors. RESULTS Hepatocyte engraftment at both transplant sites was significantly improved by ADSC cotransplantation ( P < 0.001, P < 0.001). In the renal subcapsular model, close proximity between hepatocytes and ADSCs was necessary to exert this effect. Unexpectedly, ≈50% of transplanted hepatocytes were attached by ADSCs in the liver. In an in vitro study, the hepatocyte function was significantly improved by ADSC coculture supernatant ( P < 0.001). The multiplex assay and inhibition test demonstrated that hepatocyte growth factor, vascular endothelial growth factor, and interleukin-6 may be key factors for the abovementioned effects of ADSCs. CONCLUSIONS The present study revealed that ADSC cotransplantation can improve the engraftment of transplanted hepatocytes. This effect may be based on crucial factors, such as hepatocyte growth factor, vascular endothelial growth factor, and interleukin-6, which are secreted by ADSCs.
Collapse
|
16
|
Nakafusa Y, Nitta N, Ishii K, Shirasu N, Iwamoto T, Nemoto T, Nakamura M, Goto M, Iwata H, Taniguchi M, Yasunami Y. Acceptance of Murine Islet Allografts Without Immunosuppression in Inguinal Subcutaneous White Adipose Tissue Pretreated With bFGF. Diabetes 2022; 71:1721-1734. [PMID: 35604856 DOI: 10.2337/db21-0684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 04/24/2022] [Indexed: 11/13/2022]
Abstract
Prevention of immune rejection without immunosuppression is the ultimate goal of transplant immunobiology. One way to achieve this in cellular transplantation, such as with islet transplantation, is to create a favorable local environment at the transplant site. In the current study, we found that C57BL/6 mice with streptozotocin-induced diabetes remained normoglycemic for >1 year after transplantation of BALB/c islets without immunosuppression when the inguinal subcutaneous white adipose tissue (ISWAT) was the site of transplantation and when the site was pretreated with basic fibroblast growth factor. Mechanistically, mesenchymal stem cells (MSCs) expanded in the ISWAT after the treatment was found to produce transforming growth factor-β (TGF-β), and prevention of islet allograft rejection could be achieved by cotransplantation with syngeneic MSCs isolated from the ISWAT after the treatment, which was abolished by anti-TGF-β antibody treatment. Importantly, TGF-β-producing cells remained present at the site of cotransplantation up to the end of observation period at 240 days after transplantation. These findings indicate that prevention of islet allograft rejection without immunosuppression is feasible with the use of syngeneic TGF-β-producing MSCs expanded in the ISWAT after the treatment with bFGF, providing a novel strategy for prevention of islet allograft rejection without immunosuppression.
Collapse
Affiliation(s)
- Yuki Nakafusa
- Research Institute for Islet Biology, Fukuoka University Central Research Organization, Fukuoka, Japan
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoyoshi Nitta
- Research Institute for Islet Biology, Fukuoka University Central Research Organization, Fukuoka, Japan
| | - Kazunari Ishii
- Department of Microbiology and Immunology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Naoto Shirasu
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Takahiro Iwamoto
- Research Institute for Islet Biology, Fukuoka University Central Research Organization, Fukuoka, Japan
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Takayuki Nemoto
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Hiroo Iwata
- Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Kobe, Japan
| | - Masaru Taniguchi
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Yohichi Yasunami
- Research Institute for Islet Biology, Fukuoka University Central Research Organization, Fukuoka, Japan
| |
Collapse
|
17
|
Torres-Castro K, Azimi MS, Varhue WB, Honrado C, Peirce SM, Swami NS. Biophysical quantification of reorganization dynamics of human pancreatic islets during co-culture with adipose-derived stem cells. Analyst 2022; 147:2731-2738. [DOI: 10.1039/d2an00222a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Reorganization dynamics of human islets during co-culture with adipose stem cells depends on islet size and the heterogeneity can be assessed based on biomechanical opacity of individual islets.
Collapse
Affiliation(s)
- Karina Torres-Castro
- Department of Electrical & Computer Engineering, University of Virginia, Charlottesville, Virginia-22904, USA
| | - Mohammad S. Azimi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia-22904, USA
| | - Walter B. Varhue
- Department of Electrical & Computer Engineering, University of Virginia, Charlottesville, Virginia-22904, USA
| | - Carlos Honrado
- Department of Electrical & Computer Engineering, University of Virginia, Charlottesville, Virginia-22904, USA
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia-22904, USA
| | - Nathan S. Swami
- Department of Electrical & Computer Engineering, University of Virginia, Charlottesville, Virginia-22904, USA
| |
Collapse
|
18
|
Three-dimensional Vascularized β-cell Spheroid Tissue Derived From Human Induced Pluripotent Stem Cells for Subcutaneous Islet Transplantation in a Mouse Model of Type 1 Diabetes. Transplantation 2022; 106:48-59. [PMID: 34905762 DOI: 10.1097/tp.0000000000003745] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Islet transplantation is an effective replacement therapy for type 1 diabetes (T1D) patients. However, shortage of donor organ for allograft is obstacle for further development of the treatment. Subcutaneous transplantation with stem cell-derived β-cells might overcome this, but poor vascularity in the site is burden for success in the transplantation. We investigated the effect of subcutaneous transplantation of vascularized β-cell spheroid tissue constructed 3-dimensionally using a layer-by-layer (LbL) cell-coating technique in a T1D model mouse. METHODS We used MIN6 cells to determine optimal conditions for the coculture of β-cell spheroids, normal human dermal fibroblasts, and human umbilical vein endothelial cells, and then, under those conditions, we constructed vascularized spheroid tissue using human induced pluripotent stem cell-derived β-cells (hiPS β cells). The function of insulin secretion of the vascularized hiPS β-cell spheroid tissue was evaluated in vitro. Furthermore, the function was investigated in T1D model NOD/SCID mice subcutaneously transplanted with the tissue. RESULTS In vitro, the vascularized hiPS β-cell spheroid tissue exhibited enhanced insulin secretion. The vascularized hiPS β-cell spheroid tissue also significantly decreased blood glucose levels in diabetic immunodeficient mice when transplanted subcutaneously. Furthermore, host mouse vessels were observed in the explanted vascularized hiPS β-cell spheroid tissue. CONCLUSIONS Vascularized hiPS β-cell spheroid tissue decreased blood glucose levels in the diabetic mice. This therapeutic effect was suggested due to host angiogenesis in the graft. This method could lead to a promising regenerative treatment for T1D patients.
Collapse
|
19
|
Fathi I, Imura T, Inagaki A, Nakamura Y, Nabawi A, Goto M. Decellularized Whole-Organ Pre-vascularization: A Novel Approach for Organogenesis. Front Bioeng Biotechnol 2021; 9:756755. [PMID: 34746108 PMCID: PMC8567193 DOI: 10.3389/fbioe.2021.756755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/04/2021] [Indexed: 01/15/2023] Open
Abstract
Introduction: Whole-organ decellularization is an attractive approach for three-dimensional (3D) organ engineering. However, progress with this approach is hindered by intra-vascular blood coagulation that occurs after in vivo implantation of the re-cellularized scaffold, resulting in a short-term graft survival. In this study, we explored an alternative approach for 3D organ engineering through an axial pre-vascularization approach and examined its suitability for pancreatic islet transplantation. Methods: Whole livers from male Lewis rats were decellularized through sequential arterial perfusion of detergents. The decellularized liver scaffold was implanted into Lewis rats, and an arteriovenous bundle was passed through the scaffold. At the time of implantation, fresh bone marrow preparation (BM; n = 3), adipose-derived stem cells (ADSCs; n = 4), or HBSS (n = 4) was injected into the scaffold through the portal vein. After 5 weeks, around 2,600 islet equivalents (IEQs) were injected through the portal vein of the scaffold. The recipient rats were rendered diabetic by the injection of 65 mg/kg STZ intravenously 1 week before islet transplantation and were followed up after transplantation by measuring the blood glucose and body weight for 30 days. Intravenous glucose tolerance test was performed in the cured animals, and samples were collected for immunohistochemical (IHC) analyses. Micro-computed tomography (CT) images were obtained from one rat in each group for representation. Results: Two rats in the BM group and one in the ADSC group showed normalization of blood glucose levels, while one rat from each group showed partial correction of blood glucose levels. In contrast, no rats were cured in the HBSS group. Micro-CT showed evidence of sprouting from the arteriovenous bundle inside the scaffold. IHC analyses showed insulin-positive cells in all three groups. The number of von-Willebrand factor-positive cells in the islet region was higher in the BM and ADSC groups than in the HBSS group. The number of 5-bromo-2'-deoxyuridine-positive cells was significantly lower in the BM group than in the other two groups. Conclusions: Despite the limited numbers, the study showed the promising potential of the pre-vascularized whole-organ scaffold as a novel approach for islet transplantation. Both BM- and ADSCs-seeded scaffolds were superior to the acellular scaffold.
Collapse
Affiliation(s)
- Ibrahim Fathi
- Division of Transplantation and Regenerative Medicine, Tohoku University, Sendai, Japan
- Department of Surgery, University of Alexandria, Alexandria, Egypt
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University, Sendai, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University, Sendai, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ayman Nabawi
- Department of Surgery, University of Alexandria, Alexandria, Egypt
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Tohoku University, Sendai, Japan
- Department of Surgery, Tohoku University, Sendai, Japan
| |
Collapse
|
20
|
Tissue Engineering Strategies for Improving Beta Cell Transplantation Outcome. CURRENT TRANSPLANTATION REPORTS 2021. [DOI: 10.1007/s40472-021-00333-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Abstract
Purpose of Review
Beta cell replacement therapy as a form of islet transplantation is a promising alternative therapy with the possibility to make selected patients with type 1 diabetes (T1D) insulin independent. However, this technique faces challenges such as extensive activation of the host immune system post-transplantation, lifelong need for immunosuppression, and the scarcity of islet donor pancreas. Advancement in tissue engineering strategies can improve these challenges and allow for a more widespread application of this therapy. This review will discuss the recent development and clinical translation of tissue engineering strategies in beta cell replacement therapy.
Recent Findings
Tissue engineering offers innovative solutions for producing unlimited glucose responsive cells and fabrication of appropriate devices/scaffolds for transplantation applications. Generation of pancreatic organoids with supporting cells in biocompatible biomaterials is a powerful technique to improve the function of insulin-producing cell clusters. Fabrication of physical barriers such as encapsulation strategies can protect the cells from the host immune system and allow for graft retrieval, although this strategy still faces major challenges to fully restore physiological glucose regulation.
Summary
The three main components of tissue engineering strategies including the generation of stem cell-derived insulin-producing cells and organoids and the possibilities for therapeutic delivery of cell-seeded devices to extra-hepatic sites need to come together in order to provide safe and functional insulin-producing devices for clinical beta cell replacement therapy.
Collapse
|
21
|
The Transplantation Resistance of Type II Diabetes Mellitus Adipose-Derived Stem Cells Is Due to G6PC and IGF1 Genes Related to the FoxO Signaling Pathway. Int J Mol Sci 2021; 22:ijms22126595. [PMID: 34205470 PMCID: PMC8235161 DOI: 10.3390/ijms22126595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022] Open
Abstract
In cases of patients with rapidly progressive diabetes mellitus (DM), autologous stem cell transplantation is considered as one of the regenerative treatments. However, whether the effects of autonomous stem cell transplantation on DM patients are equivalent to transplantation of stem cells derived from healthy persons is unclear. This study revealed that adipose-derived mesenchymal stem cells (ADSC) derived from type II DM patients had lower transplantation efficiency, proliferation potency, and stemness than those derived from healthy persons, leading to a tendency to induce apoptotic cell death. To address this issue, we conducted a cyclopedic mRNA analysis using a next-generation sequencer and identified G6PC3 and IGF1, genes related to the FoxO signaling pathway, as the genes responsible for lower performance. Moreover, it was demonstrated that the lower transplantation efficiency of ADSCs derived from type II DM patients might be improved by knocking down both G6PC3 and IGF1 genes. This study clarified the difference in transplantation efficiency between ADSCs derived from type II DM patients and those derived from healthy persons and the genes responsible for the lower performance of the former. These results can provide a new strategy for stabilizing the quality of stem cells and improving the therapeutic effects of regenerative treatments on autonomous stem cell transplantation in patients with DM.
Collapse
|
22
|
Pan T, Shen H, Yuan S, Lu G, Zhang Y, Wang H, Zhao Y, Sun X, Liu Q. Combined Transplantation With Human Mesenchymal Stem Cells Improves Retinal Rescue Effect of Human Fetal RPE Cells in Retinal Degeneration Mouse Model. Invest Ophthalmol Vis Sci 2021; 61:9. [PMID: 32639552 PMCID: PMC7425709 DOI: 10.1167/iovs.61.8.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Purpose We verified whether fetal RPE (fRPE) cells and mesenchymal stem cells (MSCs) cotransplantation can combine the features of these two cell types and alleviate retinal degeneration in a retinal degenerative disease mouse model. Methods Tail vein injection of sodium iodate (NaIO3) was conducted to establish the retinal degenerative disease mouse model. MSCs and fRPE cells were transplanted either separately or combined in the subretinal space of retinal degenerative disease animals. ERG, optical coherence tomography, histologic, and immunofluorescence analyses were performed. Furthermore, the expression level of Crx, rhodopsin, Iba1, F4/80, Caspase 3, nerve growth factor, and brain-derived neurotrophic factor were assessed to investigate the mechanisms involved in cell transplantation effects. Results Cotransplantation of fRPE and MSC cells promoted significant improvements in ERG results and in the survival rate of transplanted cells. In addition, MSC and fRPE cell cotransplantation resulted in an increase in the thickness of the total retina, as well as in the outer and inner nuclear layers. Combined transplantation also upregulated the expression level of Crx and rhodopsin and downregulated caspase 3 expression, highlighting its better photoreceptor rescue effect in relation to the single cell type transplantation. Finally, combined transplantation suppressed the expression of Iba1 and F4/80 factors while increasing the endogenous expression of nerve growth factor and brain-derived nerve growth factor neurotrophic factors. These data suggest that MSC and fRPE cell cotransplantation is able to suppress immunoreactions and promote neurotrophic factor excretion. Conclusions Combined transplantation of MSCs and fRPE cells results in a better retinal rescue effect than single cell type transplantation in NaIO3-induced retinopathy.
Collapse
|
23
|
Zhang CJ, Ma Y, Jin ZB. The road to restore vision with photoreceptor regeneration. Exp Eye Res 2020; 202:108283. [PMID: 33010290 DOI: 10.1016/j.exer.2020.108283] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 09/13/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
Neuroretinal diseases are the predominant cause of irreversible blindness worldwide, mainly due to photoreceptor loss. Currently, there are no radical treatments to fully reverse the degeneration or even stop the disease progression. Thus, it is urgent to develop new biological therapeutics for these diseases on the clinical side. Stem cell-based treatments have become a promising therapeutic for neuroretinal diseases through the replacement of damaged cells with photoreceptors and some allied cells. To date, considerable efforts have been made to regenerate the diseased retina based on stem cell technology. In this review, we overview the current status of stem cell-based treatments for photoreceptor regeneration, including the major cell sources derived from different stem cells in pre-clinical or clinical trial stages. Additionally, we discuss herein the major challenges ahead for and potential new strategy toward photoreceptor regeneration.
Collapse
Affiliation(s)
- Chang-Jun Zhang
- Laboratory for Stem Cell & Retinal Regeneration, The Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ya Ma
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China.
| |
Collapse
|
24
|
Razavi M, Ren T, Zheng F, Telichko A, Wang J, Dahl JJ, Demirci U, Thakor AS. Facilitating islet transplantation using a three-step approach with mesenchymal stem cells, encapsulation, and pulsed focused ultrasound. Stem Cell Res Ther 2020; 11:405. [PMID: 32948247 PMCID: PMC7501701 DOI: 10.1186/s13287-020-01897-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/06/2020] [Accepted: 08/24/2020] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The aim of this study was to examine the effect of a three-step approach that utilizes the application of adipose tissue-derived mesenchymal stem cells (AD-MSCs), encapsulation, and pulsed focused ultrasound (pFUS) to help the engraftment and function of transplanted islets. METHODS In step 1, islets were co-cultured with AD-MSCs to form a coating of AD-MSCs on islets: here, AD-MSCs had a cytoprotective effect on islets; in step 2, islets coated with AD-MSCs were conformally encapsulated in a thin layer of alginate using a co-axial air-flow method: here, the capsule enabled AD-MSCs to be in close proximity to islets; in step 3, encapsulated islets coated with AD-MSCs were treated with pFUS: here, pFUS enhanced the secretion of insulin from islets as well as stimulated the cytoprotective effect of AD-MSCs. RESULTS Our approach was shown to prevent islet death and preserve islet functionality in vitro. When 175 syngeneic encapsulated islets coated with AD-MSCs were transplanted beneath the kidney capsule of diabetic mice, and then followed every 3 days with pFUS treatment until day 12 post-transplantation, we saw a significant improvement in islet function with diabetic animals re-establishing glycemic control over the course of our study (i.e., 30 days). In addition, our approach was able to enhance islet engraftment by facilitating their revascularization and reducing inflammation. CONCLUSIONS This study demonstrates that our clinically translatable three-step approach is able to improve the function and viability of transplanted islets.
Collapse
Affiliation(s)
- Mehdi Razavi
- Department of Radiology, Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA, 94304, USA
- Biionix™ (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, 32816, USA
| | - Tanchen Ren
- Department of Radiology, Bio-Acoustic MEMS in Medicine Laboratory (BAMM), Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Fengyang Zheng
- Department of Radiology, Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA, 94304, USA
| | - Arsenii Telichko
- Department of Radiology, Dahl Ultrasound Laboratory, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Jing Wang
- Department of Radiology, Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA, 94304, USA
| | - Jeremy J Dahl
- Department of Radiology, Dahl Ultrasound Laboratory, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Utkan Demirci
- Department of Radiology, Bio-Acoustic MEMS in Medicine Laboratory (BAMM), Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Avnesh S Thakor
- Department of Radiology, Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA, 94304, USA.
| |
Collapse
|
25
|
White Adipose Tissue as a Site for Islet Transplantation. TRANSPLANTOLOGY 2020. [DOI: 10.3390/transplantology1010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Although islet transplantation is recognized as a useful cellular replacement therapy for severe diabetes, surgeons face difficulties in islet engraftment. The transplant site is a pivotal factor that influences the engraftment. Although the liver is the current representative site for clinical islet transplantation, it is not the best site because of limitations in immunity, inflammation, and hypoxia. White adipose tissue, including omentum, is recognized as a useful candidate site for islet transplantation. Its effectiveness has been evaluated in not only various basic and translational studies using small and large animals but also in some recent clinical trials. In this review, we attempt to shed light on the characteristics and usefulness of white adipose tissue as a transplant site for islets.
Collapse
|
26
|
White Adipose Tissue as a Site for Islet Transplantation. TRANSPLANTOLOGY 2020. [DOI: 10.3390/transplantology1020006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although islet transplantation is recognized as a useful cellular replacement therapy for severe diabetes, surgeons face difficulties in islet engraftment. The transplant site is a pivotal factor that influences the engraftment. Although the liver is the current representative site for clinical islet transplantation, it is not the best site because of limitations in immunity, inflammation, and hypoxia. White adipose tissue, including omentum, is recognized as a useful candidate site for islet transplantation. Its effectiveness has been evaluated in not only various basic and translational studies using small and large animals but also in some recent clinical trials. In this review, we attempt to shed light on the characteristics and usefulness of white adipose tissue as a transplant site for islets.
Collapse
|
27
|
Lee YN, Yi HJ, Kim YH, Lee S, Oh J, Okano T, Shim IK, Kim SC. Evaluation of Multi-Layered Pancreatic Islets and Adipose-Derived Stem Cell Sheets Transplanted on Various Sites for Diabetes Treatment. Cells 2020; 9:1999. [PMID: 32878048 PMCID: PMC7563383 DOI: 10.3390/cells9091999] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 12/31/2022] Open
Abstract
Islet cell transplantation is considered an ideal treatment for insulin-deficient diabetes, but implantation sites are limited and show low graft survival. Cell sheet technology and adipose-derived stem cells (ADSCs) can be useful tools for improving islet cell transplantation outcomes since both can increase implantation efficacy and graft survival. Herein, the optimal transplantation site in diabetic mice was investigated using islets and stem cell sheets. We constructed multi-layered cell sheets using rat/human islets and human ADSCs. Cell sheets were fabricated using temperature-responsive culture dishes. Islet/ADSC sheet (AI sheet) group showed higher viability and glucose-stimulated insulin secretion than islet-only group. Compared to islet transplantation alone, subcutaneous AI sheet transplantation showed better blood glucose control and CD31+ vascular traits. Because of the adhesive properties of cell sheets, AI sheets were easily applied on liver and peritoneal surfaces. Liver or peritoneal surface grafts showed better glucose control, weight gain, and intraperitoneal glucose tolerance test (IPGTT) profiles than subcutaneous site grafts using both rat and human islets. Stem cell sheets increased the therapeutic efficacy of islets in vivo because mesenchymal stem cells enhance islet function and induce neovascularization around transplanted islets. The liver and peritoneal surface can be used more effectively than the subcutaneous site in future clinical applications.
Collapse
Affiliation(s)
- Yu Na Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (Y.N.L.); (H.-J.Y.); (Y.H.K.); (S.L.); (J.O.)
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hye-Jin Yi
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (Y.N.L.); (H.-J.Y.); (Y.H.K.); (S.L.); (J.O.)
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Yang Hee Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (Y.N.L.); (H.-J.Y.); (Y.H.K.); (S.L.); (J.O.)
- Regenerative Medicine Research Center, Dalim Tissen Co., Ltd., 31, Yeonhui-ro, Mapo-gu, Seoul 03982, Korea
| | - Song Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (Y.N.L.); (H.-J.Y.); (Y.H.K.); (S.L.); (J.O.)
| | - Jooyun Oh
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (Y.N.L.); (H.-J.Y.); (Y.H.K.); (S.L.); (J.O.)
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA;
| | - In Kyong Shim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (Y.N.L.); (H.-J.Y.); (Y.H.K.); (S.L.); (J.O.)
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Song Cheol Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Department of Surgery & Department of Biomedical Engineering, AMIST, University of Ulsan College of Medicine &Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
28
|
Yamane K, Anazawa T, Tada S, Fujimoto N, Inoguchi K, Emoto N, Nagai K, Masui T, Okajima H, Takaori K, Sumi S, Uemoto S. Mitomycin C treatment improves pancreatic islet graft longevity in intraportal islet transplantation by suppressing proinflammatory response. Sci Rep 2020; 10:12086. [PMID: 32694579 PMCID: PMC7374693 DOI: 10.1038/s41598-020-69009-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
The in vitro culture period prior to cell transplantation (i.e. pancreatic islet transplantation) enables cell modification and is thus advantageous. However, the islet preconditioning method has not been fully explored. Here we present a simple approach for islet preconditioning that uses the antibiotic mitomycin C (MMC), which has antitumor activity, to reduce islet immunogenicity and prevent proinflammatory events in an intraportal islet transplantation model. Freshly isolated mice islets were treated for 30 min with 10 μg/mL MMC or not, cultured for 20 h and transplanted into the livers of syngeneic or allogeneic diabetic mouse recipients. In the allogeneic model, MMC preconditioning significantly prolonged graft survival without requiring immunosuppressants. In vitro, MMC treatment suppressed the expression of proinflammatory cytokines in islet allografts, while immunohistochemical studies revealed the suppression of inflammatory cell infiltration into MMC-treated allografts relative to untreated allografts. Furthermore, MMC preconditioning significantly suppressed the mRNA expression of proinflammatory cytokines into the transplant site and induced the differentiation of regulatory T cells with the ability to suppress CD4+ T cell-mediated immune responses. In conclusion, islet preconditioning with MMC prolonged graft survival in an intraportal islet transplantation model by suppressing proinflammatory events and inducing potentially regulatory lymphocytes.
Collapse
Affiliation(s)
- Kei Yamane
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Takayuki Anazawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan.
| | - Seiichiro Tada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Nanae Fujimoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Kenta Inoguchi
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Norio Emoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Kazuyuki Nagai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Toshihiko Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Hideaki Okajima
- Department of Paediatric Surgery, Kanazawa Medical University, Kanazawa, 9200293, Japan
| | - Kyoichi Takaori
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| | - Shoichiro Sumi
- Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 6068507, Japan
| | - Shinji Uemoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 6068507, Japan
| |
Collapse
|
29
|
Loretelli C, Assi E, Seelam AJ, Ben Nasr M, Fiorina P. Cell therapy for type 1 diabetes. Expert Opin Biol Ther 2020; 20:887-897. [PMID: 32299257 DOI: 10.1080/14712598.2020.1748596] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Type 1 diabetes (T1D) is a lifelong condition resulting from autoimmune destruction of insulin-producing β-cells. Islet or whole-pancreas transplantation is limited by the shortage of donors and need for chronic immune suppression. Novel strategies are needed to prevent β-cell loss and to rescue production of endogenous insulin. AREAS COVERED This review covers the latest advances in cell-based therapies for the treatment and prevention of T1D. Topics include adoptive transfer of cells with increased immunoregulatory potential for β-cell protection, and β-cell replacement strategies such as generation of insulin-producing β-like cells from unlimited sources. EXPERT OPINION Cell therapy provides an opportunity to prevent or reverse T1D. Adoptive transfer of autologous cells having enhanced immunomodulatory properties can suppress autoimmunity and preserve β-cells. Such therapies have been made possible by a combination of genome-editing techniques and transplantation of tolerogenic cells. In-vitro modified autologous hematopoietic stem cells and tolerogenic dendritic cells may protect endogenous and newly generated β-cells from a patient's autoimmune response without hampering immune surveillance for infectious agents and malignant cellular transformations. However, methods to generate cells that meet quality and safety standards for clinical applications require further refinement.
Collapse
Affiliation(s)
- Cristian Loretelli
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy
| | - Emma Assi
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy
| | - Andy Joe Seelam
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy
| | - Moufida Ben Nasr
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy.,Nephrology Division, Boston Children's Hospital, Harvard Medical School , Boston, MA, USA
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center "Romeo Ed Enrica Invernizzi", Department of Biomedical and Clinical Science L. Sacco, Università Degli Studi Di Milano , Milan, Italy.,Nephrology Division, Boston Children's Hospital, Harvard Medical School , Boston, MA, USA.,Division of Endocrinology, ASST Fatebenefratelli-Sacco , Milan, Italy
| |
Collapse
|
30
|
Kuppan P, Seeberger K, Kelly S, Rosko M, Adesida A, Pepper AR, Korbutt GS. Co‐transplantation of human adipose‐derived mesenchymal stem cells with neonatal porcine islets within a prevascularized subcutaneous space augments the xenograft function. Xenotransplantation 2020; 27:e12581. [DOI: 10.1111/xen.12581] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/19/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Purushothaman Kuppan
- Alberta Diabetes Institute University of Alberta Edmonton AB Canada
- Department of Surgery University of Alberta Edmonton AB Canada
| | - Karen Seeberger
- Alberta Diabetes Institute University of Alberta Edmonton AB Canada
- Department of Surgery University of Alberta Edmonton AB Canada
| | - Sandra Kelly
- Alberta Diabetes Institute University of Alberta Edmonton AB Canada
- Department of Surgery University of Alberta Edmonton AB Canada
| | - Mandy Rosko
- Alberta Diabetes Institute University of Alberta Edmonton AB Canada
- Department of Surgery University of Alberta Edmonton AB Canada
| | - Adetola Adesida
- Department of Surgery University of Alberta Edmonton AB Canada
| | - Andrew R. Pepper
- Alberta Diabetes Institute University of Alberta Edmonton AB Canada
- Department of Surgery University of Alberta Edmonton AB Canada
| | - Gregory S. Korbutt
- Alberta Diabetes Institute University of Alberta Edmonton AB Canada
- Department of Surgery University of Alberta Edmonton AB Canada
| |
Collapse
|
31
|
Domingues CC, Kundu N, Kropotova Y, Ahmadi N, Sen S. Antioxidant-upregulated mesenchymal stem cells reduce inflammation and improve fatty liver disease in diet-induced obesity. Stem Cell Res Ther 2019; 10:280. [PMID: 31477174 PMCID: PMC6720095 DOI: 10.1186/s13287-019-1393-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/09/2019] [Accepted: 08/22/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The incidence of obesity and diabetes is increasing rapidly. Optimal management is still elusive. Obesity associated with type 2 diabetes is known to cause adipose tissue inflammation, increase oxidative stress, and cause white fat hyperplasia and mitochondrial dysfunction. In this study, we investigated whether mitochondrial and cytosolic antioxidant-upregulated mesenchymal stem cell (MSC) delivery reduces oxidative stress and subsequently improves glucose tolerance, reduce systemic inflammation, and improves fatty liver disease in diet-induced obese (DIO) mouse models. METHODS Antioxidant genes Sod2 (mitochondrial) and catalase (cytosolic) or null (control) were upregulated in human adipose tissue-derived MSCs using adenoviral constructs. Modified MSCs were then delivered intraperitoneally into mice that were fed a 45% or 60% high-fat diet (HFD), and animals were followed for 4 weeks. RESULTS Over 4 weeks, body weight remained stable; however, we noted a significant reduction in liver fat content by histological analysis and liver triglyceride assay. Triglyceride assay (p < 0.01) confirmed reduced liver fat accumulation in animals that received either Sod2- or Cat-MSCs. There was a lower plasma level of inflammatory marker TNFα, measured in mice that were fed either 45% or 60% HFD and received Sod2- or Cat-MSCs, indicating reduced systemic inflammation. Ucp1 mRNA was upregulated approximately 100-1000-fold for omental fat and 10-100-fold for pericardial fat compared to the Null-MSC-receiving group. Pcgc1a and Prdm16 mRNA upregulation was also noted particularly for pericardial fat. Glucose tolerance showed a positive improvement trend with a lower area under the curve (AUC) values for both Sod2- and Cat-MSCs groups in comparison to control. For mice fed with 60% HFD and that received Sod2-MSCs, glucose levels were significantly lower than control (*p < 0.05) at a time point of 60 min in the glycemic curve during glucose tolerance test. CONCLUSION Reduction of oxidative stress post-antioxidant-upregulated MSC delivery, intraperitoneally, reduces systemic inflammation and fat accumulation in the liver. There is evidence of an increase in browning of white adipose tissue depots with concomitant improvement of glucose tolerance in a weight-independent fashion. Antioxidant-upregulated MSC delivery may be a safe yet effective therapy for obesity and prediabetes and improves related complication such as non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Cleyton C Domingues
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Nabanita Kundu
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Yana Kropotova
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Neeki Ahmadi
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Sabyasachi Sen
- Department of Medicine, The George Washington University, Washington, DC, USA. .,School of Medicine and Health Science, 2300 I Street NW, Washington, DC, 20037, USA.
| |
Collapse
|
32
|
Takahashi H, Sakata N, Yoshimatsu G, Hasegawa S, Kodama S. Regenerative and Transplantation Medicine: Cellular Therapy Using Adipose Tissue-Derived Mesenchymal Stromal Cells for Type 1 Diabetes Mellitus. J Clin Med 2019; 8:249. [PMID: 30781427 PMCID: PMC6406504 DOI: 10.3390/jcm8020249] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the autoimmune targeting of pancreatic β-cells, and, in the advanced stage, severe hypoinsulinemia due to islet destruction. In patients with T1DM, continuous exogenous insulin therapy cannot be avoided. However, an insufficient dose of insulin easily induces extreme hyperglycemia or diabetic ketoacidosis, and intensive insulin therapy may cause hypoglycemic symptoms including hypoglycemic shock. While these insulin therapies are efficacious in most patients, some additional therapies are warranted to support the control of blood glucose levels and reduce the risk of hypoglycemia in patients who respond poorly despite receiving appropriate treatment. There has been a recent gain in the popularity of cellular therapies using mesenchymal stromal cells (MSCs) in various clinical fields, owing to their multipotentiality, capacity for self-renewal, and regenerative and immunomodulatory potential. In particular, adipose tissue-derived MSCs (ADMSCs) have become a focus in the clinical setting due to the abundance and easy isolation of these cells. In this review, we outline the possible therapeutic benefits of ADMSC for the treatment of T1DM.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Naoaki Sakata
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Suguru Hasegawa
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|
33
|
Ren G, Rezaee M, Razavi M, Taysir A, Wang J, Thakor AS. Adipose tissue-derived mesenchymal stem cells rescue the function of islets transplanted in sub-therapeutic numbers via their angiogenic properties. Cell Tissue Res 2019; 376:353-364. [PMID: 30707291 DOI: 10.1007/s00441-019-02997-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/17/2019] [Indexed: 02/07/2023]
Abstract
A significant proportion of islets are lost following transplantation due to hypoxia and inflammation. We hypothesize that adipose tissue-derived mesenchymal stem cells (AD-MSCs) can rescue a sub-therapeutic number of transplanted islets by helping them establish a new blood supply and reducing inflammation. Diabetic mice received syngeneic transplantation with 75 (minimal), 150 (sub-therapeutic), or 225 (therapeutic) islets, with or without 1 × 106 mouse AD-MSCs. Fasting blood glucose (FBG) values were measured over 6 weeks with tissue samples collected for islet structure and morphology (H&E, insulin/glucagon staining). Histological and immunohistochemical analyses of islets were also performed at 2 weeks in animals transplanted with a sub-therapeutic number of islets, with and without AD-MSCs, to determine new blood vessel formation, the presence of pro-angiogenic factors facilitating revascularization, and the degree of inflammation. AD-MSCs had no beneficial effect on FBG values when co-transplanted with a minimal or therapeutic number of islets. However, AD-MSCs significantly reduced FBG values and restored glycemic control in diabetic animals transplanted with a sub-therapeutic number of islets. Islets co-transplanted with AD-MSCs preserved their native morphology and organization and exhibited less aggregation when compared to islets transplanted alone. In the sub-therapeutic group, AD-MSCs significantly increased islet revascularization and the expression of angiogenic factors including hepatocyte growth factor (HGF) and angiopoietin-1 (Ang-1) while also reducing inflammation. AD-MSCs can rescue the function of islets when transplanted in a sub-therapeutic number, for at least 6 weeks, via their ability to maintain islet architecture while concurrently facilitating islet revascularization and reducing inflammation.
Collapse
Affiliation(s)
- Gang Ren
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA
| | - Melika Rezaee
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA.,Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA
| | - Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA
| | - Ahmed Taysir
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA.
| |
Collapse
|
34
|
Gamble A, Pawlick R, Pepper AR, Bruni A, Adesida A, Senior PA, Korbutt GS, Shapiro AMJ. Improved islet recovery and efficacy through co-culture and co-transplantation of islets with human adipose-derived mesenchymal stem cells. PLoS One 2018; 13:e0206449. [PMID: 30419033 PMCID: PMC6231609 DOI: 10.1371/journal.pone.0206449] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/13/2018] [Indexed: 02/07/2023] Open
Abstract
Islet transplantation is an established clinical procedure for select patients with type 1 diabetes and severe hypoglycemia to stabilize glycemic control. Post-transplant, substantial beta cell mass is lost, necessitating multiple donors to maintain euglycemia. A potential strategy to augment islet engraftment is the co-transplantation of islets with multipotent mesenchymal stem cells to capitalize upon their pro-angiogenic and anti-inflammatory properties. Herein, we examine the in vitro and in vivo effect of co-culturing murine islets with human adipose-derived mesenchymal stem cells (Ad-MSCs). Islets co-cultured with Ad-MSCs for 48 hours had decreased cell death, superior viability as measured by membrane integrity, improved glucose stimulated insulin secretion and reduced apoptosis compared to control islets. These observations were recapitulated with human islets, albeit tested in a limited capacity. Recipients of marginal mouse islet mass grafts, co-transplanted with Ad-MSCs without a co-culture period, did not reverse to normoglycemia as efficiently as islets alone. However, utilizing a 48-hour co-culture period, marginal mouse islets grafts with Ad-MSCs achieved a superior percent euglycemia rate when compared to islets cultured and transplanted alone. A co-culture period of human islets with human Ad-MSCs may have a clinical benefit improving engraftment outcomes.
Collapse
Affiliation(s)
- Anissa Gamble
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Edmonton, AB, Canada
| | - Rena Pawlick
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Andrew R. Pepper
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Antonio Bruni
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Adetola Adesida
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Peter A. Senior
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Gregory S. Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - A. M. James Shapiro
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
35
|
Perez-Basterrechea M, Esteban MM, Vega JA, Obaya AJ. Tissue-engineering approaches in pancreatic islet transplantation. Biotechnol Bioeng 2018; 115:3009-3029. [PMID: 30144310 DOI: 10.1002/bit.26821] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
Abstract
Pancreatic islet transplantation is a promising alternative to whole-pancreas transplantation as a treatment of type 1 diabetes mellitus. This technique has been extensively developed during the past few years, with the main purpose of minimizing the complications arising from the standard protocols used in organ transplantation. By using a variety of strategies used in tissue engineering and regenerative medicine, pancreatic islets have been successfully introduced in host patients with different outcomes in terms of islet survival and functionality, as well as the desired normoglycemic control. Here, we describe and discuss those strategies to transplant islets together with different scaffolds, in combination with various cell types and diffusible factors, and always with the aim of reducing host immune response and achieving islet survival, regardless of the site of transplantation.
Collapse
Affiliation(s)
- Marcos Perez-Basterrechea
- Unidad de Terapia Celular y Medicina Regenerativa, Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain.,Plataforma de Terapias Avanzadas, Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Manuel M Esteban
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | - Jose A Vega
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Alvaro J Obaya
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
36
|
Navaei-Nigjeh M, Moloudizargari M, Baeeri M, Gholami M, Lotfibakhshaiesh N, Soleimani M, Vasheghani-Farahani E, Ai J, Abdollahi M. Reduction of marginal mass required for successful islet transplantation in a diabetic rat model using adipose tissue-derived mesenchymal stromal cells. Cytotherapy 2018; 20:1124-1142. [PMID: 30068495 DOI: 10.1016/j.jcyt.2018.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/10/2018] [Accepted: 06/06/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Adipose tissue-derived mesenchymal stromal cells (AT-MSCs), widely known as multipotent progenitors, release several cytokines that support cell survival and repair. There are in vitro and in vivo studies reporting the regenerative role of AT-MSCs possibly mediated by their protective effects on functional islet cells as well as their capacity to differentiate into insulin-producing cells (IPCs). METHODS On such a basis, our goal in the present study was to use three different models including direct and indirect co-cultures and islet-derived conditioned medium (CM) to differentiate AT-MSCs into IPCs and to illuminate the molecular mechanisms of the beneficial impact of AT-MSCs on pancreatic islet functionality. Furthermore, we combined in vitro co-culture of islets and AT-MSCs with in vivo assessment of islet graft function to assess whether co-transplantation of islets with AT-MSCs can reduce marginal mass required for successful islet transplantation and prolong graft function in a diabetic rat model. RESULTS Our findings demonstrated that AT-MSCs are suitable for creating a microenvironment favorable for the repair and longevity of the pancreas β cells through the improvement of islet survival and maintenance of cell morphology and insulin secretion due to their potent properties in differentiation. Most importantly, hybrid transplantation of islets with AT-MSCs significantly promoted survival, engraftment and insulin-producing function of the graft and reduced the islet mass required for reversal of diabetes. CONCLUSIONS This strategy might be of therapeutic potential solving the problem of donor islet material loss that currently limits the application of allogeneic islet transplantation as a more widespread therapy for type 1 diabetes.
Collapse
Affiliation(s)
- Mona Navaei-Nigjeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Science, Tehran, Iran
| | - Milad Moloudizargari
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Science, Tehran, Iran
| | - Mahdi Gholami
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Science, Tehran, Iran
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Science, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Kawamoto K, Ohashi T, Konno M, Nishida N, Koseki J, Matsui H, Sakai D, Kudo T, Eguchi H, Satoh T, Doki Y, Mori M, Ishii H. Cell-free culture conditioned medium elicits pancreatic β cell lineage-specific epigenetic reprogramming in mice. Oncol Lett 2018; 16:3255-3259. [PMID: 30127922 DOI: 10.3892/ol.2018.9008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 03/09/2017] [Indexed: 12/19/2022] Open
Abstract
There are several obstacles to overcome prior to achieving cellular reprogramming of pancreatic β cells in vitro and in vivo. The present study demonstrated that the transfer of epigenetic phenotypes was achieved in the cell-free conditioned medium (CM) of pancreatic insulinoma MIN6 cell cultures. The comparison of a subpopulation of MIN6, m14 and m9 cells indicated that MIN6-m14 cells were more prone to cellular reprogramming. Epigenetic profiling revealed that the transcription factor pancreas/duodenum homeobox protein 1 (Pdx1) was differentially associated among the clones. The culture of differentiated adipocytes in the CM of MIN6-m14 cells resulted in the induction of insulin mRNA expression, and was accompanied by epigenetic events of Pdx1 binding. The epigenetic profiling indicated that Pdx1 is preferentially associated with a previously uncharacterized region of the endoplasmic reticulum (ER) disulfide oxidase, ER oxidoreductin 1 gene. Therefore, the results of the present study indicated that the CM of MIN6 cells was able to induce a pancreatic β cell-like phenotype in differentiated adipocytes. These data provide additional support for the utility of cell-free CM for cellular reprogramming.
Collapse
Affiliation(s)
- Koichi Kawamoto
- Department of Gastroenterological Surgery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan.,Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tomofumi Ohashi
- Department of Gastroenterological Surgery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masamitsu Konno
- Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Naohiro Nishida
- Department of Gastroenterological Surgery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan.,Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Jun Koseki
- Department of Cancer Profiling Discovery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hidetoshi Matsui
- Department of Mathematical Sciences, Faculty of Mathematics, Kyushu University, Fukuoka 819-0395, Japan
| | - Daisuke Sakai
- Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Toshihiro Kudo
- Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Taroh Satoh
- Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan.,Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan.,Department of Cancer Profiling Discovery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan.,Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan.,Department of Cancer Profiling Discovery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hideshi Ishii
- Department of Cancer Frontier Science, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan.,Department of Cancer Profiling Discovery, Osaka University School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
38
|
Li Q, Xia S, Yin Y, Guo Y, Chen F, Jin P. miR-5591-5p regulates the effect of ADSCs in repairing diabetic wound via targeting AGEs/AGER/JNK signaling axis. Cell Death Dis 2018; 9:566. [PMID: 29752466 PMCID: PMC5948214 DOI: 10.1038/s41419-018-0615-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 04/03/2018] [Accepted: 04/18/2018] [Indexed: 12/24/2022]
Abstract
Advanced glycation end products/advanced glycation end products receptor (AGEs/AGER) interaction triggers reactive oxygen species (ROS) generation and activates downstream signal pathways and induces apoptosis in endothelial progenitor cells. A number of studies have revealed the involvement of microRNAs (miRNAs) in regulating intracellular ROS production and apoptosis. However, few studies explore the role of miRNAs in regulating the effect of adipose tissue-derived stem cells (ADSCs) in repairing diabetic wound and the associated cellular mechanisms remain unclear. In this study, ADSCs were exposed to AGEs, then siRNA for AGER was transfected into ADSCs. We found that AGEs/AGER axis induced ROS generation and apoptosis in ADSCs. AGEs treatment downregulated miR-5591-5p in ADSCs, which directly targeted AGER. miR-5591-5p suppressed AGEs/AGER axis-mediated ROS generation and apoptosis in ADSCs in vitro. In addition, miR-5591-5p promoted cell survival and enhanced the ability of ADSCs for repairing cutaneous wound in vivo. Furthermore, we confirmed that c-jun kinase (JNK) signal was involved in the inhibitory effect of miR-5591-5p on AGEs/AGER axis-induced ROS generation and apoptosis in ADSCs. Thus, these results indicated that miR-5591-5p targeting AGEs/AGER/JNK signaling axis possibly regulates the effect of ADSCs in repairing diabetic wound.
Collapse
Affiliation(s)
- Qiang Li
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Jiangsu, Xuzhou, China
| | - Sizhan Xia
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Jiangsu, Xuzhou, China
| | - Yating Yin
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Jiangsu, Xuzhou, China
| | - Yanping Guo
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Jiangsu, Xuzhou, China
| | - Feifei Chen
- Jiangsu Center for the Collaboration and Innovation of Cancer, Xuzhou Medical University, Jiangsu, Xuzhou, China.
| | - Peisheng Jin
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Jiangsu, Xuzhou, China. .,Jiangsu Center for the Collaboration and Innovation of Cancer, Xuzhou Medical University, Jiangsu, Xuzhou, China.
| |
Collapse
|
39
|
Mizushima T, Fukata T, Takeyama H, Takahashi H, Haraguchi N, Nishimura J, Hata T, Matsuda C, Yamamoto H, Doki Y, Mori M. The features of adipose-derived stem cells in patients with inflammatory bowel diseases. Surg Today 2017; 48:352-358. [PMID: 29027585 DOI: 10.1007/s00595-017-1591-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE The treatment outcomes for inflammatory bowel disease (IBD) have been improving, but the development of better therapies is needed. Stem cell therapy is promising, but little is known about the differences in adipose-derived stem cells (ADSCs) between IBD patients and healthy individuals. METHODS ADSCs were isolated from subcutaneous adipose tissue (SAT) in IBD (Crohn's disease, 3; ulcerative colitis, 2) and non-IBD (colorectal cancer, 5; breast cancer, 1) patients. We also analyzed the effects of tumor necrosis factor (TNF)-α on murine ADSCs. RESULTS The numbers of stromal vascular fraction (SVF) cells per gram of SAT were 7.72 ± 3.03 × 105 in IBD and 8.51 ± 8.80 × 105 in non-IBD patients (p > 0.05). The proportions of ADSCs in SVF cells were 4.98 ± 2.61% in IBD and 1.02 ± 0.67% in non-IBD patients. The numbers of ADSCs per gram of SAT were 4.16 ± 2.96 × 104 in IBD and 0.88 ± 1.04 × 104 in non-IBD patients. The proportions and numbers of ADSCs were significantly higher in IBD patients than in non-IBD patients. TNF-α significantly facilitated the proliferation and motility of murine ADSCs. CONCLUSION These results showed the potential advantage of freshly isolated autologous ADSCs in IBD patients.
Collapse
Affiliation(s)
- Tsunekazu Mizushima
- Department of Therapeutics for Inflammatory Bowel Diseases, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Tadafumi Fukata
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Takeyama
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hidekazu Takahashi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naotsugu Haraguchi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Junichi Nishimura
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Taishi Hata
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Chu Matsuda
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masaki Mori
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
40
|
Matsushita K, Dzau VJ. Mesenchymal stem cells in obesity: insights for translational applications. J Transl Med 2017; 97:1158-1166. [PMID: 28414326 DOI: 10.1038/labinvest.2017.42] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 02/24/2017] [Indexed: 12/11/2022] Open
Abstract
Obesity is now a major public health problem worldwide. Lifestyle modification to reduce the characteristic excess body adiposity is important in the treatment of obesity, but effective therapeutic intervention is still needed to control what has become an obesity epidemic. Unfortunately, many anti-obesity drugs have been withdrawn from market due to adverse side effects. Bariatric surgery therefore remains the most effective therapy for severe cases, although such surgery is invasive and researchers continue to seek new control strategies for obesity. Mesenchymal stem cells (MSCs) are a major source of adipocyte generation, and studies have been conducted into the potential roles of MSCs in treating obesity. However, despite significant progress in stem cell research and its potential applications for obesity, adipogenesis is a highly complex process and the molecular mechanisms governing MSC adipogenesis remain ill defined. In particular, successful clinical application of MSCs will require extensive identification and characterization of the transcriptional regulators controlling MSC adipogenesis. Since obesity is associated with the incidence of multiple important comorbidities, an in-depth understanding of the relationship between MSC adipogenesis and the comorbidities of obesity is also necessary to evaluate the potential of effective and safe MSC-based therapies for obesity. In addition, brown adipogenesis is an attractive topic from the viewpoint of therapeutic innovation and future research into MSC-based brown adipogenesis could lead to a novel breakthrough. Ongoing stem cell studies and emerging research fields such as epigenetics are expected to elucidate the complicated mechanisms at play in MSC adipogenesis and develop novel MSC-based therapeutic options for obesity. This review discusses the current understanding of MSCs in adipogenesis and their potential clinical applications for obesity.
Collapse
Affiliation(s)
- Kenichi Matsushita
- Division of Cardiology, Second Department of Internal Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
41
|
Takeyama H, Mizushima T, Uemura M, Haraguchi N, Nishimura J, Hata T, Matsuda C, Takemasa I, Ikenaga M, Murata K, Yamamoto H, Doki Y, Mori M. Adipose-Derived Stem Cells Ameliorate Experimental Murine Colitis via TSP-1-Dependent Activation of Latent TGF-β. Dig Dis Sci 2017; 62:1963-1974. [PMID: 28434074 DOI: 10.1007/s10620-017-4578-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 04/09/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Adipose tissue-derived stem cells (ASCs) have been investigated as therapeutic tools for a variety of autoimmune diseases, including inflammatory diseases. However, the mechanisms underlying the immunomodulatory properties of ASCs are not well understood. Here, we investigated the mechanism of regulatory T cell (Treg) induction in ASC therapy in a murine model of inflammatory bowel disease. METHODS Acute colitis was induced in mice using dextran sulfate sodium and ASCs administered intraperitoneally. Tregs and CD103+ dendritic cells were analyzed in the mesenteric lymph nodes (MLNs), spleen, and colonic lamina propria (CLP). Activation of latent TGF-β by ASCs was analyzed in vitro using ELISA. siRNA technology was used to create ASCs in which TSP-1 or integrinαv was knocked down in order to investigate the involvement of these proteins in the activation of latent TGF-β. In addition, TSP-1-knockdown ASCs were administered to mice with colitis to assess their clinical efficacy in vivo. RESULTS Systemic administration of ASCs significantly lessened the clinical and histopathological severity of colitis. ASCs were distributed throughout the lymphatic system in the MLNs and spleen. Tregs were increased in the MLNs and CLP, but CD103+ dendritic cells were not significantly altered. The ASCs activated latent TGF-β. TSP-1 knockdown impaired TGF-β activation in vitro and abrogated the therapeutic effects of the ASCs in vivo. Furthermore, Tregs were not increased in the MLNs and CLP from mice treated with TSP-1-knockdown ASCs. CONCLUSIONS These results demonstrate that ASCs induce Tregs by activating latent TGF-β via TSP-1, independent of CD103+ dendritic cell induction.
Collapse
Affiliation(s)
- Hiroshi Takeyama
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Tsunekazu Mizushima
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan.
| | - Mamoru Uemura
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Naotsugu Haraguchi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Junichi Nishimura
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Taishi Hata
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Chu Matsuda
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Ichiro Takemasa
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Masakazu Ikenaga
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Kohei Murata
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Hirofumi Yamamoto
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Doki
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Masaki Mori
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
42
|
Schive SW, Mirlashari MR, Hasvold G, Wang M, Josefsen D, Gullestad HP, Korsgren O, Foss A, Kvalheim G, Scholz H. Human Adipose-Derived Mesenchymal Stem Cells Respond to Short-Term Hypoxia by Secreting Factors Beneficial for Human Islets In Vitro and Potentiate Antidiabetic Effect In Vivo. CELL MEDICINE 2017; 9:103-116. [PMID: 28713640 DOI: 10.3727/215517917x693401] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adipose-derived mesenchymal stem cells (ASCs) release factors beneficial for islets in vitro and protect against hyperglycemia in rodent models of diabetes. Oxygen tension has been shown to induce metabolic changes and alter ASCs' release of soluble factors. The effects of hypoxia on the antidiabetic properties of ASCs have not been explored. To investigate this, we incubated human ASCs for 48 h in 21% (normoxia) or 1% O2 (hypoxia) and compared viability, cell growth, surface markers, differentiation capability, and soluble factors in the conditioned media (CM). Human islets were exposed to CM from ASCs incubated in either normoxia or hypoxia, and islet function and apoptosis after culture with or without proinflammatory cytokines were measured. To test hypoxic preconditioned ASCs' islet protective effects in vivo, ASCs were incubated for 48 h in normoxia or hypoxia before being injected into Balb/c Rag 1-/- immunodeficient mice with streptozotocin-induced insulitis. Progression of diabetes and insulin content of pancreas were measured. We found that incubation in hypoxia was well tolerated by ASCs and that levels of VEGF-A, FGF-2, and bNGF were elevated in CM from ASCs incubated in hypoxia compared to normoxia, while levels of HGF, IL-8, and CXCL1 were reduced. CM from ASCs incubated in hypoxia significantly improved human islet function and reduced apoptosis after culture, and reduced cytokine-induced apoptosis. In our mouse model, pancreas insulin content was higher in both groups receiving ASCs compared to control, but the mice receiving preconditioned ASCs had lower random and fasting blood glucose, as well as improved oral glucose tolerance compared to untreated mice. In conclusion, our in vitro results indicate that the islet protective potential of ASCs improves in hypoxia, and we give insight into factors involved in this. Finally we show that hypoxic preconditioning potentiates ASCs' antidiabetic effect in vivo.
Collapse
Affiliation(s)
- Simen W Schive
- Department of Transplant Medicine, Oslo University Hospital, Oslo, Norway.,†Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.,‡Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Mohammad Reza Mirlashari
- §Section for Cell Therapy, Oslo University Hospital, Oslo, Norway.,¶Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Grete Hasvold
- #Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Mengyu Wang
- §Section for Cell Therapy, Oslo University Hospital, Oslo, Norway
| | - Dag Josefsen
- §Section for Cell Therapy, Oslo University Hospital, Oslo, Norway
| | | | - Olle Korsgren
- ††Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Aksel Foss
- Department of Transplant Medicine, Oslo University Hospital, Oslo, Norway.,†Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.,‡Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gunnar Kvalheim
- §Section for Cell Therapy, Oslo University Hospital, Oslo, Norway
| | - Hanne Scholz
- Department of Transplant Medicine, Oslo University Hospital, Oslo, Norway.,†Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.,‡Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
43
|
Combined transplantation of human mesenchymal stem cells and human retinal progenitor cells into the subretinal space of RCS rats. Sci Rep 2017; 7:199. [PMID: 28298640 PMCID: PMC5428026 DOI: 10.1038/s41598-017-00241-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/15/2017] [Indexed: 11/08/2022] Open
Abstract
Retinitis pigmentosa (RP) is one of hereditary retinal diseases characterized by the loss of photoreceptors. Cell transplantation has been clinically applied to treat RP patients. Human retinal progenitor cells (HRPCs) and human bone marrow-derived mesenchymal stem cells (HBMSCs) are the two commonly and practically used stem cells for transplantation. Since combined transplantation could be a promising way to integrate the advantages of both stem cell types, we transplanted HRPCs and HBMSCs into the subretinal space (SRS) of Royal College of Surgeons (RCS) rats. We report that HRPCs/HBMSCs combined transplantation maintains the electroretinogram results much better than HRPCs or HBMSCs single transplantations. The thickness of outer nuclear layer also presented a better outcome in the combined transplantation. Importantly, grafted cells in the combination migrated better, both longitudinally and latitudinally, than single transplantation. The photoreceptor differentiation of grafted cells in the retina of RCS rats receiving combined transplantation also showed a higher ratio than single transplantation. Finally, activation of microglia and the gliosis of Müller cells were more effectively suppressed in combined transplantation, indicating better immunomodulatory and anti-gliosis effects. Taken together, combining the transplantation of HRPCs and HBMSCs is a more effective strategy in stem cell-based therapy for retinal degenerative diseases.
Collapse
|
44
|
Publisher's note. Regen Ther 2016. [DOI: 10.1016/j.reth.2016.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
45
|
Mesenchymal Stem Cells and Metabolic Syndrome: Current Understanding and Potential Clinical Implications. Stem Cells Int 2016; 2016:2892840. [PMID: 27313625 PMCID: PMC4903149 DOI: 10.1155/2016/2892840] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 04/06/2016] [Accepted: 05/04/2016] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is an obesity-based, complicated clinical condition that has become a global epidemic problem with a high associated risk for cardiovascular disease and mortality. Dyslipidemia, hypertension, and diabetes or glucose dysmetabolism are the major factors constituting metabolic syndrome, and these factors are interrelated and share underlying pathophysiological mechanisms. Severe obesity predisposes individuals to metabolic syndrome, and recent data suggest that mesenchymal stem cells (MSCs) contribute significantly to adipocyte generation by increasing the number of adipocytes. Accordingly, an increasing number of studies have examined the potential roles of MSCs in managing obesity and metabolic syndrome. However, despite the growing bank of experimental and clinical data, the efficacy and the safety of MSCs in the clinical setting are still to be optimized. It is thus hoped that ongoing and future studies can elucidate the roles of MSCs in metabolic syndrome and lead to MSC-based therapeutic options for affected patients. This review discusses current understanding of the relationship between MSCs and metabolic syndrome and its potential implications for patient management.
Collapse
|
46
|
Li Q, Guo Y, Chen F, Liu J, Jin P. Stromal cell-derived factor-1 promotes human adipose tissue-derived stem cell survival and chronic wound healing. Exp Ther Med 2016; 12:45-50. [PMID: 27347016 PMCID: PMC4906949 DOI: 10.3892/etm.2016.3309] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 02/04/2015] [Indexed: 12/31/2022] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) hold great potential for the stem cell-based therapy of cutaneous wound healing. Stromal cell-derived factor-1 (SDF-1) activates CXC chemokine receptor (CXCR)4+ and CXCR7+ cells and plays an important role in wound healing. Increasing evidence suggests a critical role for SDF-1 in cell apoptosis and the survival of mesenchymal stem cells. However, the function of SDF-1 in the apoptosis and wound healing ability of ADSCs is not well understood. The aim of this study was to analyze the effect of SDF-1 on the apoptosis and therapeutic effect of ADSCs in cutaneous chronic wounds in vitro and in vivos. By flow cytometric analysis, it was found that hypoxia and serum free promoted the apoptosis of ADSCs. When pretreated with SDF-1, the apoptosis of ADSCs induced by hypoxia and serum depletion was partly recovered. Furthermore, in vivo experiments established that the post-implantation cell survival and chronic wound healing ability of ADSCs were increased following pretreatment with SDF-1 in a diabetic mouse model of chronic wound healing. To explore the potential mechanism underlying the effect of SDF-1 on ADSC apoptosis, western blot analysis was employed and the results indicate that SDF-1 may protect against cell apoptosis in hypoxic and serum-free conditions through activation of the caspase signaling pathway in ADSCs. This study provides evidence that SDF-1 pretreatment can increase the therapeutic effect of ADSCs in cutaneous chronic wounds in vitro and in vivo.
Collapse
Affiliation(s)
- Qiang Li
- Plastic Surgery Department, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China; Department of Ear Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Beijing 100144, P.R. China
| | - Yanping Guo
- Plastic Surgery Department, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Feifei Chen
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jing Liu
- Plastic Surgery Department, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Peisheng Jin
- Plastic Surgery Department, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
47
|
A hybrid of cells and pancreatic islets toward a new bioartificial pancreas. Regen Ther 2016; 3:68-74. [PMID: 31245475 PMCID: PMC6581840 DOI: 10.1016/j.reth.2016.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/31/2016] [Accepted: 02/12/2016] [Indexed: 01/30/2023] Open
Abstract
Cell surface engineering using single-stranded DNA-poly(ethylene glycol)-conjugated phospholipid (ssDNA-PEG-lipid) is useful for inducing cell-cell attachment two and three dimensionally. In this review, we summarize our recent techniques for cell surface engineering and their applications to islet transplantation. Because any DNA sequence can be immobilized onto the cell surface by hydrophobic interactions between ssDNA-PEG-lipid and the cellular membrane without impairing cell function, a cell-cell hybrid can be formed through the DNA hybridization. With this technique, it would be possible to create three-dimensional hybrid structures of pancreatic islets coated with various accessory cells, such as patients' own cells, mesenchymal and adipose-derived stem cells, endothelial progenitor cells, neural crest stem cells or regulatory T cells, which might significantly improve the outcome of islet transplantation in diabetic patients.
Collapse
|
48
|
Staels W, De Groef S, Heremans Y, Coppens V, Van Gassen N, Leuckx G, Van de Casteele M, Van Riet I, Luttun A, Heimberg H, De Leu N. Accessory cells for β-cell transplantation. Diabetes Obes Metab 2016; 18:115-24. [PMID: 26289770 DOI: 10.1111/dom.12556] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/22/2015] [Accepted: 08/13/2015] [Indexed: 12/16/2022]
Abstract
Despite recent advances, insulin therapy remains a treatment, not a cure, for diabetes mellitus with persistent risk of glycaemic alterations and life-threatening complications. Restoration of the endogenous β-cell mass through regeneration or transplantation offers an attractive alternative. Unfortunately, signals that drive β-cell regeneration remain enigmatic and β-cell replacement therapy still faces major hurdles that prevent its widespread application. Co-transplantation of accessory non-islet cells with islet cells has been shown to improve the outcome of experimental islet transplantation. This review will highlight current travails in β-cell therapy and focuses on the potential benefits of accessory cells for islet transplantation in diabetes.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Cell Separation/trends
- Cells, Cultured
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/surgery
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/surgery
- Endothelial Progenitor Cells/cytology
- Endothelial Progenitor Cells/immunology
- Endothelial Progenitor Cells/pathology
- Endothelial Progenitor Cells/transplantation
- Graft Rejection/immunology
- Graft Rejection/metabolism
- Graft Rejection/prevention & control
- Graft Survival
- Humans
- Immune Tolerance
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/transplantation
- Islets of Langerhans Transplantation/adverse effects
- Islets of Langerhans Transplantation/immunology
- Mesenchymal Stem Cell Transplantation/adverse effects
- Mesenchymal Stem Cell Transplantation/trends
- Neural Crest/cytology
- Neural Crest/immunology
- Neural Crest/pathology
- Neural Crest/transplantation
- Stem Cell Transplantation/adverse effects
- Stem Cell Transplantation/trends
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- T-Lymphocytes, Regulatory/transplantation
- Transplantation, Autologous/adverse effects
- Transplantation, Autologous/trends
- Transplantation, Heterotopic/adverse effects
- Transplantation, Heterotopic/trends
- Transplantation, Homologous/adverse effects
- Transplantation, Homologous/trends
Collapse
Affiliation(s)
- W Staels
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
- Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium
| | - S De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Y Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - V Coppens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - N Van Gassen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - G Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Van de Casteele
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - I Van Riet
- Department Hematology Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - A Luttun
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - H Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - N De Leu
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Endocrinology, UZ Brussel, Brussels, Belgium
- Department of Endocrinology, ASZ Aalst, Aalst, Belgium
| |
Collapse
|
49
|
Cao M, Pan Q, Dong H, Yuan X, Li Y, Sun Z, Dong X, Wang H. Adipose-derived mesenchymal stem cells improve glucose homeostasis in high-fat diet-induced obese mice. Stem Cell Res Ther 2015; 6:208. [PMID: 26519255 PMCID: PMC4628312 DOI: 10.1186/s13287-015-0201-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/03/2015] [Accepted: 10/08/2015] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Effective therapies for obesity and diabetes are still lacking. The aim of this study was to evaluate whether a single intravenous infusion of syngeneic adipose-derived mesenchymal stem cells (ASCs) can reduce obesity, lower insulin resistance, and improve glucose homeostasis in a high-fat diet-induced obese (DIO) mouse model. METHODS Seven-week-old C57BL/6 mice were fed a high-fat diet for 20 weeks to generate the DIO mouse model. Mice were given a single intravenous infusion of ex vivo expanded syngeneic ASCs at 2 × 10(6) cells per mouse. DIO or CHOW mice injected with saline were used as controls. Body weights, blood glucose levels, glucose, and insulin tolerance test results were obtained before and 2 and 6 weeks after cell infusion. Triglyceride (TG), high-density lipoprotein (HDL), and insulin levels in serum were measured. Expressions of genes related to insulin resistance, including peroxisome proliferator-activated receptor γ (PPARγ) and insulin receptor (InsR), and inflammation (IL-6, F4/80, and nucleotide-binding oligomerization domain containing 2, or NOD2), were measured in livers at mRNA level by real-time-polymerase chain reaction analysis. Beta-cell mass in pancrheases from CHOW, DIO, and DIO + ASC mice was quantified. GFP(+) ASCs were injected, and the presence of GFP(+) cells in livers and pancreases was determined. RESULTS DIO mice that had received ASCs showed reduced body weights, reduced blood glucose levels, and increased glucose tolerance. ASC treatment was found to reduce TG levels and increase serum HDL levels. In livers, less fat cell deposition was observed, as were increased expression of InsR and PPARγ and reduction in expressions of IL-6 and F4/80. Treated mice showed well-preserved pancreatic β-cell mass with reduced expression of F4/80 and TNF-α compared with DIO controls. GFP(+) cells were found in liver and pancreas tissues at 1 and 2 weeks after cell injection. CONCLUSIONS ASC therapy is effective in lowering blood glucose levels and increasing glucose tolerance in DIO mice. The protective effects of ASCs arise at least in part from suppression of inflammation in the liver. In addition, ASCs are associated with better-preserved pancreatic β-cell mass.
Collapse
Affiliation(s)
- Mingjun Cao
- Colleges of Life Sciences, Qingdao Agricultural University, 700 Chenyang Road, Chenyang, Shandong, 266109, P.R. China.
| | - Qingjie Pan
- College of Animal Science and Veterinary Medicine, 700 Chenyang Road, Chenyang, Shandong, 266109, P.R. China.
| | - Huansheng Dong
- College of Animal Science and Veterinary Medicine, 700 Chenyang Road, Chenyang, Shandong, 266109, P.R. China.
| | - Xinxu Yuan
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 1 Hayden Dr., Petersburg, VA, 23806, USA.
| | - Yang Li
- Colleges of Life Sciences, Qingdao Agricultural University, 700 Chenyang Road, Chenyang, Shandong, 266109, P.R. China.
| | - Zhen Sun
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Ave, Charleston, SC, 29425, USA.
| | - Xiao Dong
- Colleges of Life Sciences, Qingdao Agricultural University, 700 Chenyang Road, Chenyang, Shandong, 266109, P.R. China.
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, BSB 641, 173 Ashley Ave, Charleston, SC, 29425, USA.
| |
Collapse
|
50
|
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
|