Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Bioconjug Chem ; 29(9): 2945-2953, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29985588

ABSTRACT

The microenvironment of pancreatic islets gets disrupted during enzyme digestion and causes islets to remain in a vulnerable state, leading to poor outcome in the initial days of transplantation. To avoid immune invasion while allowing the reconstruction of the microenvironment of the transplanted site, we propose immunoisolation polymers, which can nanoencapsulate islets quickly without cytotoxicity. Here, nonhuman primate (NHP) islets were nanoencapsulated with hyperbranched polyethylene glycol (hb-PEG) and heparin by layer-by-layer technology and transplanted into the kidney subcapsular space of diabetic C57BL/6 mice. An immunosuppressive drug protocol was applied to increase the survival time until the animals were sacrificed. The recipients of NHP islets exhibited high nonfasting blood glucose level (BGL) for 2-3 weeks, which was normalized afterward. Immunohistochemical (IHC) analysis revealed an immature vascular basement membrane and cell surface integrins directly associated with poor initial insulin production. The transplanted grafts regained their own microenvironment within a month without any outside stimuli. No lymphocyte infiltration was observed in the grafts at any time. Humoral and cell-mediated immune responses were prominently diminished by the hb-PEG/Heparin nanoencapsulated islets. Immunoisolation accompanied by an immunosuppressive drug protocol protects islets by helping them avoid immunogenesis while at the same time allowing them to reconstruct their microenvironment.


Subject(s)
Blood Glucose/metabolism , Cellular Microenvironment , Heparin/chemistry , Islets of Langerhans Transplantation/methods , Nanotechnology , Polyethylene Glycols/chemistry , Animals , Antibody Formation , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/therapy , Immunity, Cellular , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL
2.
Biomaterials ; 171: 164-177, 2018 07.
Article in English | MEDLINE | ID: mdl-29698867

ABSTRACT

Intraportal pancreatic islet transplantation incurs huge cell losses during its early stages due to instant blood-mediated inflammatory reactions (IBMIRs), which may also drive regulation of the adaptive immune system. Therefore, a method that evades IBMIR will improve clinical islet transplantation. We used a layer-by-layer approach to shield non-human primate (NHP) islets with polyethylene glycol (nano-shielded islets, NSIs) and polyethylene glycol plus heparin (heparin nano-shielded islets; HNSIs). Islets ranging from 10,000 to 20,000 IEQ/kg body weight were transplanted into 19 cynomolgus monkeys (n = 4, control; n = 5, NSI; and n = 10, HNSI). The mean C-peptide positive graft survival times were 68.5, 64 and 108 days for the control, NSI and HNSI groups, respectively (P = 0.012). HNSI also reduced the factors responsible for IBMIR in vitro. Based on these data, HNSIs in conjunction with clinically established immunosuppressive drug regimens will result in superior outcomes compared to those achieved with the current protocol for clinical islet transplantation.


Subject(s)
Heparin/chemistry , Islets of Langerhans/physiology , Nanoparticles/chemistry , Polymers/chemistry , Allografts/physiology , Animals , Graft Survival , Humans , Liver/pathology , Lymphocyte Subsets/metabolism , Macaca fascicularis , Polyethylene Glycols/chemistry
3.
Diabetes ; 67(3): 473-485, 2018 03.
Article in English | MEDLINE | ID: mdl-29298810

ABSTRACT

Highly angiogenic bone marrow mononuclear cell-derived spheroids (BM-spheroids), formed by selective proliferation of the CD31+CD14+CD34+ monocyte subset via three-dimensional (3D) culture, have had robust angiogenetic capacity in rodent syngeneic renal subcapsular islet transplantation. We wondered whether the efficacy of BM-spheroids could be demonstrated in clinically relevant intraportal islet transplantation models without increasing the risk of portal thrombosis. The thrombogenic potential of intraportally infused BM-spheroids was compared with that of mesenchymal stem cells (MSCs) and MSC-derived spheroids (MSC-spheroids). The angiogenic efficacy and persistence in portal sinusoids of BM-spheroids were examined in rodent syngeneic and primate allogeneic intraportal islet transplantation models. In contrast to MSCs and MSC-spheroids, intraportal infusion of BM-spheroids did not evoke portal thrombosis. BM-spheroids had robust angiogenetic capacity in both the rodent and primate intraportal islet transplantation models and improved posttransplant glycemic outcomes. MRI and intravital microscopy findings revealed the persistence of intraportally infused BM-spheroids in portal sinusoids. Intraportal cotransplantation of allogeneic islets with autologous BM-spheroids in nonhuman primates further confirmed the clinical feasibility of this approach. In conclusion, cotransplantation of BM-spheroids enhances intraportal islet transplantation outcome without portal thrombosis in mice and nonhuman primates. Generating BM-spheroids by 3D culture prevented the rapid migration and disappearance of intraportally infused therapeutic cells.


Subject(s)
Bone Marrow Transplantation/adverse effects , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation/adverse effects , Leukocytes, Mononuclear/transplantation , Liver/immunology , Spheroids, Cellular/transplantation , Transplantation, Heterotopic/adverse effects , Animals , Biomarkers/blood , Biomarkers/metabolism , Cell Tracking , Cells, Cultured , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Islets of Langerhans Transplantation/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Liver/metabolism , Liver/pathology , Macaca fascicularis , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Portal Vein , Spheroids, Cellular/cytology , Spheroids, Cellular/immunology , Streptozocin , Thrombosis/etiology , Thrombosis/immunology , Thrombosis/pathology , Thrombosis/prevention & control , Transplantation, Isogeneic/adverse effects
4.
Xenotransplantation ; 25(1)2018 01.
Article in English | MEDLINE | ID: mdl-29135052

ABSTRACT

BACKGROUND: Porcine islet xenotransplantation is considered an attractive alternative treatment for type 1 diabetes mellitus. However, it is largely limited because of initial rejection due to Instant Blood-Mediated Inflammatory Reaction (IBMIR), oxidative stress, and inflammatory responses. Recently, soluble tumor necrosis factor-ɑ receptor type I (sTNF-αR) and heme oxygenase (HO)-1 genes (HO-1/sTNF-αR) have been shown to improve the viability and functionality of porcine islets after transplantation. METHODS: In this study, genetically modified mesenchymal stem cells (MSCs) expressing the HO-1/sTNF-αR genes (HO-1/sTNF-αR-MSC) were developed using an adenoviral system, and porcine islet viability and function were confirmed by in vitro tests such as GSIS, AO/PI, and the ADP/ATP ratio after coculturing with HO-1/sTNF-αR-MSCs. Subsequently, isolated porcine islets were transplanted underneath the kidney capsule of diabetic humanized mice without MSCs, with MSCs or with HO-1/sTNF-αR-MSCs. RESULTS: According to the results, the HO-1/sTNF-αR-MSC-treated group exhibited improved survival of porcine islets and could reverse hyperglycemia more than porcine islets not treated with MSCs or islets cotransplanted with MSCs. Moreover, the HO-1/sTNF-αR-MSC group maintained its morphological characteristics and the insulin secretion pattern of transplanted porcine islets similar to endogenous islets in immunocompetent humanized mice. CONCLUSIONS: Our results suggest that HO-1/sTNF-αR-MSCs are efficient tools for porcine islet xenotransplantation, and this study may provide basic information for pre-clinical animal models and future clinical trials of porcine islet xenotransplantation.


Subject(s)
Graft Survival , Heme Oxygenase-1/genetics , Heterografts/immunology , Membrane Proteins/genetics , Mesenchymal Stem Cells/cytology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Coculture Techniques , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/therapy , Graft Survival/immunology , Humans , Immunoglobulin Fc Fragments/immunology , Islets of Langerhans/immunology , Islets of Langerhans Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Mice, Transgenic , Transplantation, Heterologous/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...