Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Bioeng Biotechnol ; 10: 886483, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35651551

RESUMO

Pancreatic islet transplantation improves metabolic control and prevents complications in patients with brittle type 1 diabetes (T1D). However, chronic immunosuppression is required to prevent allograft rejection and recurrence of autoimmunity. Islet encapsulation may eliminate the need for immunosuppression. Here, we analyzed in parallel two microencapsulation platforms that provided long-term diabetes reversal in preclinical T1D models, alginate single and double capsules versus polyethylene glycol conformal coating, to identify benefits and weaknesses that could inform the design of future clinical trials with microencapsulated islets. We performed in vitro and in vivo functionality assays with human islets and analyzed the explanted grafts by immunofluorescence. We quantified the size of islets and capsules, measured capsule permeability, and used these data for in silico simulations of islet functionality in COMSOL Multiphysics. We demonstrated that insulin response to glucose stimulation is dependent on capsule size, and the presence of permselective materials augments delays in insulin secretion. Non-coated and conformally coated islets could be transplanted into the fat pad of diabetic mice, resulting in comparable functionality and metabolic control. Mac-2+ cells were found in conformally coated grafts, indicating possible host reactivity. Due to their larger volume, alginate capsules were transplanted in the peritoneal cavity. Despite achieving diabetes reversal, changes in islet composition were found in retrieved capsules, and recipient mice experienced hypoglycemia indicative of hyperinsulinemia induced by glucose retention in large capsules as the in silico model predicted. We concluded that minimal capsule size is critical for physiological insulin secretion, and anti-inflammatory modulation may be beneficial for small conformal capsules.

2.
Xenotransplantation ; 28(3): e12673, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33522023

RESUMO

BACKGROUND: Alginate-encapsulated islet xenografts have restored normoglycemia in diabetic animals for various periods of time. Plausible mechanisms of graft failure in vivo include immune rejection and hypoxia. We sought to understand the effects of encapsulated adult porcine islet (API) dosage on the peritoneal dissolved oxygen (DO) level in correlation to the achieved glycemic regulation in diabetic mice. METHODS: Adult porcine islets encapsulated in barium alginate were transplanted intraperitoneally in streptozotocin diabetic BALB/c mice at 6000 and 4000 islet equivalents (IEQ) and in normal mice at 500 IEQ; APIs encapsulated in calcium alginate were transplanted at 6000 IEQ in diabetic mice. In all cases, cell-free barium alginate capsules containing a perfluorocarbon emulsion were co-implanted for DO measurements using 19 F NMR spectroscopy. Blood glucose levels and peritoneal DO were measured over 60 days or until graft failure. Explanted capsules were evaluated microscopically and histologically. RESULTS: Both barium and calcium alginate-encapsulated APIs at 6000 IEQ reversed diabetes until day 60; barium alginate-encapsulated APIs at 4000 IEQ also reversed diabetes but with a higher failure rate. Transplanted APIs significantly reduced the peritoneal DO, approximately in a dose-dependent manner. The number of viable islets and the insulin content per capsule decreased over time. Capsules retrieved from normoglycemic mice exhibited minimal host cell adherence. CONCLUSIONS: Transplantation of encapsulated APIs can reduce peritoneal DO to severely hypoxic levels. Although normoglycemia could be maintained within the study period, the DO levels suggest that hypoxia is a factor contributing to loss of islet viability and insulin secretion with time in mice.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Alginatos , Animais , Sobrevivência de Enxerto , Camundongos , Camundongos Endogâmicos BALB C , Oxigênio , Estreptozocina , Suínos , Transplante Heterólogo
3.
Xenotransplantation ; 27(4): e12577, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31903659

RESUMO

BACKGROUND: Our goal was to identify clinically relevant immunotherapies that synergize with microencapsulation to protect adult porcine islet (API) xenografts in diabetic NOD mice. We have shown previously that dual costimulatory blockade (CTLA4-Ig plus anti-CD154 mAb) combined with encapsulation protects APIs long-term in NOD mice. Since no anti-CD154 mAbs currently are approved for use in humans, we tested the efficacy of other targeted immunosuppression regimens that might be used for diabetic patients receiving encapsulated islets. METHODS: Microencapsulated APIs were transplanted i.p. in diabetic NOD mice given either no immunosuppression or combinations immunosuppressive reagents. Graft function was monitored by blood glucose levels, i.p. glucose tolerance tests, and histology. Mechanisms of rejection were investigated by phenotyping host peritoneal cells and measuring graft site cytokine and chemokine levels. RESULTS: New immunosuppressive therapies were compared to CTLA4-Ig plus anti-CD154 mAb, used here as a control. The most effective was triple treatment with CTLA4-Ig, anti-CD154 mAb, and intracapsular CXCL12, and the next most effective was a non-depleting anti-CD4 mAb (YTS177.9) plus intracapsular CXCL12. Three additional regimens (CTLA4-Ig plus YTS177.9, YTS177.9 alone, and anti-OX40-Ligand mAb alone) significantly prolonged encapsulated API function. Dual treatment with CTLA4-Ig plus anti-CD40 mAb was as effective as CTLA4-Ig plus anti-CD154 mAb. Five other monotherapies and three combination therapies did not augment encapsulated API survival. Most peritoneal cytokines and chemokines were either absent or minimal. At necropsy, the capsules were intact, not fibrosed, and clean when function was maintained, but were coated with host cells if rejection had occurred. CONCLUSIONS: Multiple different immunotherapies which specifically inhibit CD4+ T cells, modulate T-cell trafficking, or interfere with antigen presentation can substitute for anti-CD154 mAb to prolong encapsulated islet xenograft function in diabetic NOD mice.


Assuntos
Diabetes Mellitus Experimental , Terapia de Imunossupressão/métodos , Transplante das Ilhotas Pancreáticas , Transplante Heterólogo , Animais , Ligante de CD40 , Diabetes Mellitus Experimental/cirurgia , Rejeição de Enxerto , Sobrevivência de Enxerto , Xenoenxertos , Camundongos , Camundongos Endogâmicos NOD , Suínos
4.
Transplantation ; 104(2): 259-269, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31385927

RESUMO

BACKGROUND: We have utilized a noninvasive technique for measuring the partial pressure of oxygen (pO2) in alginate microcapsules implanted intraperitoneally in healthy nonhuman primates (NHPs). Average pO2 is important for determining if a transplant site and capsules with certain passive diffusion characteristics can support the islet viability, metabolic activity, and dose necessary to reverse diabetes. METHODS: Perfluoro-15-crown-5-ether alginate capsules were infused intraperitoneally into 3 healthy NHPs. Peritoneal pO2 levels were measured on days 0 and 7 using fluorine-19 magnetic resonance relaxometry and a fiber-optic probe. Fluorine-19 MRI was used to determine the locations of capsules within the peritoneal space on days 0 and 7. Gross and histologic evaluations of the capsules were used to assess their biocompatibility postmortem. RESULTS: At day 0 immediately after infusion of capsules equilibrated to room air, capsules were concentrated near the infusion site, and the pO2 measurement using magnetic resonance relaxometry was 147 ± 9 mm Hg. On day 7 after capsules were dispersed throughout the peritoneal cavity, the pO2 level was 61 ± 11 mm Hg. Measurements using the fiber-optic oxygen sensor were 132 ± 7.5 mm Hg (day 0) and 89 ± 6.1 mm Hg (day 7). Perfluoro-15-crown-5-ether capsules retrieved on day 7 were intact and free-floating without host cell attachment, although the numbers of peritoneal CD20 B cells, CD4 and CD8 T cells, and CD14 macrophages increased consistent with a mild foreign body reaction. CONCLUSIONS: The peritoneal pO2 of normal NHPs is relatively low and we predict would decrease further when encapsulated islets are transplanted intraperitoneally.


Assuntos
Alginatos/farmacologia , Diabetes Mellitus Experimental/cirurgia , Imagem por Ressonância Magnética de Flúor-19/métodos , Transplante das Ilhotas Pancreáticas/métodos , Consumo de Oxigênio/fisiologia , Oxigênio/metabolismo , Cavidade Peritoneal/cirurgia , Animais , Cápsulas , Diabetes Mellitus Experimental/metabolismo , Feminino , Sobrevivência de Enxerto , Macaca mulatta , Pressão Parcial
5.
Xenotransplantation ; 25(6): e12450, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30117193

RESUMO

BACKGROUND: Xenogeneic donors would provide an unlimited source of islets for the treatment of type 1 diabetes (T1D). The goal of this study was to assess the function of microencapsulated adult porcine islets (APIs) transplanted ip in streptozotocin (STZ)-diabetic non-human primates (NHPs) given targeted immunosuppression. METHODS: APIs were encapsulated in: (a) single barium-gelled alginate capsules or (b) double alginate capsules with an inner, islet-containing compartment and a durable, biocompatible outer alginate layer. Immunosuppressed, streptozotocin-diabetic NHPs were transplanted ip with encapsulated APIs, and graft function was monitored by measuring blood glucose, %HbA1c, and porcine C-peptide. At graft failure, explanted capsules were assessed for biocompatibility and durability plus islet viability and functionality. Host immune responses were evaluated by phenotyping peritoneal cell populations, quantitation of peritoneal cytokines and chemokines, and measurement of anti-porcine IgG and IgM plus anti-Gal IgG. RESULTS: NHP recipients had reduced hyperglycemia, decreased exogenous insulin requirements, and lower percent hemoglobin A1c (%HbA1c) levels. Porcine C-peptide was detected in plasma of all recipients, but these levels diminished with time. However, relatively high levels of porcine C-peptide were detected locally in the peritoneal graft site of some recipients at sacrifice. IV glucose tolerance tests demonstrated metabolic function, but the grafts eventually failed in all diabetic NHPs regardless of the type of encapsulation or the host immunosuppression regimen. Explanted microcapsules were intact, "clean," and free-floating without evidence of fibrosis at graft failure, and some reversed diabetes when re-implanted ip in diabetic immunoincompetent mice. Histology of explanted capsules showed scant evidence of a host cellular response, and viable islets could be found. Flow cytometric analyses of peritoneal cells and peripheral blood showed similarly minimal evidence of a host immune response. Preformed anti-porcine IgG and IgM antibodies were present in recipient plasma, but these levels did not rise post-transplant. Peritoneal graft site cytokine or chemokine levels were equivalent to normal controls, with the exception of minimal elevation observed for IL-6 or IL-1ß, GRO-α, I-309, IP-10, and MCP-1. However, we found central necrosis in many of the encapsulated islets after graft failure, and explanted islets expressed endogenous markers of hypoxia (HIF-1α, osteopontin, and GLUT-1), suggesting a role for non-immunologic factors, likely hypoxia, in graft failure. CONCLUSIONS: With donor xenoislet microencapsulation and host immunosuppression, APIs corrected hyperglycemia after ip transplantation in STZ-diabetic NHPs in the short term. The islet xenografts lost efficacy gradually, but at graft failure, some viable islets remained, substantial porcine C-peptide was detected in the peritoneal graft site, and there was very little evidence of a host immune response. We postulate that chronic effects of non-immunologic factors, such as in vivo hypoxic and hyperglycemic conditions, damaged the encapsulated islet xenografts. To achieve long-term function, new approaches must be developed to prevent this damage, for example, by increasing the oxygen supply to microencapsulated islets in the ip space.


Assuntos
Diabetes Mellitus Experimental/induzido quimicamente , Composição de Medicamentos , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Transplante Heterólogo , Animais , Composição de Medicamentos/métodos , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Xenoenxertos/imunologia , Terapia de Imunossupressão/métodos , Transplante das Ilhotas Pancreáticas/imunologia , Primatas , Estreptozocina/farmacologia , Suínos
6.
J Tissue Eng Regen Med ; 10(3): 222-32, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23281223

RESUMO

Stem cell-based therapies hold great promise as a clinically viable approach for vascular regeneration. Preclinical studies have been very encouraging and early clinical trials have suggested favourable outcomes. However, significant challenges remain in terms of optimizing cell retention and maintenance of the paracrine effects of implanted cells. To address these issues, we have proposed the use of a cellular encapsulation approach to enhance vascular regeneration. We contained human mesenchymal stem cells (hMSCs) in biocompatible alginate microcapsules for therapeutic treatment in the setting of murine hindlimb ischaemia. This approach supported the paracrine pro-angiogenic activity of hMSCs, prevented incorporation of hMSCs into the host tissue and markedly enhanced their therapeutic effect. While injection of non-encapsulated hMSCs resulted in a 22 ± 10% increase in vascular density and no increase in perfusion, treatment with encapsulated hMSCs resulted in a 70 ± 8% increase in vascular density and 21 ± 7% increase in perfusion. The described cellular encapsulation strategy may help to better define the mechanisms responsible for the beneficial effects of cell-based therapies and provide a therapeutic strategy for inducing vascular growth in the adult. As hMSCs are relatively easy to isolate from patients, and alginate is biocompatible and already used in clinical applications, therapeutic cell encapsulation for vascular repair represents a highly translatable platform for cell-based therapy in humans.


Assuntos
Alginatos/farmacologia , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Animais , Cápsulas , Sobrevivência Celular/efeitos dos fármacos , Células Imobilizadas/citologia , Células Imobilizadas/efeitos dos fármacos , Ácido Glucurônico/farmacologia , Ácidos Hexurônicos/farmacologia , Membro Posterior/efeitos dos fármacos , Membro Posterior/patologia , Humanos , Isquemia/patologia , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Nus , Permeabilidade , Cicatrização/efeitos dos fármacos
7.
Xenotransplantation ; 21(2): 127-39, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24635017

RESUMO

BACKGROUND: Our goal was to improve islet transplantation as a therapy for patients with type I diabetes mellitus. Because human donor islets are scarce, we are studying islet xenografts in the diabetic NOD mouse model. We hypothesize that optimal xenoislet survival will be achieved by the combination of donor islet immunoisolation with recipient immunosuppression. We and others have studied adult and neonatal porcine islets as sources of tissue for microencapsulated islet xenografts, but we believe it is also advantageous to consider using islets from fish, which can be raised in large numbers relatively quickly and economically. Therefore, in this study, we have evaluated the function of microencapsulated xenogeneic piscine (tilapia) islets transplanted intraperitoneally (IP) in NOD mice in the presence of CD4(+) T-cell depletion and/or costimulatory blockade. METHODS: Spontaneously diabetic NOD mice or streptozotocin (STZ)-diabetic NOD-SCID mice were transplanted IP with microencapsulated tilapia islets. Recipient immunosuppression included anti-CD4 mAb, CTLA4-Ig, anti-CD80 mAb, anti-CD86 mAb, or anti-CD154 mAb, alone or in combination. Graft function was evaluated by blood glucose (BG) levels, intravenous (IV) and oral glucose tolerance tests (GTTs), histologic and immunohistochemical analyses of grafts, and flow cytometric analysis of peritoneal cells. RESULTS: Encapsulated tilapia islets normalized random BG levels for up to 210 days in NOD-SCID mice. In diabetic NOD mice, encapsulated tilapia islets were rejected on day 11 ± 4 with a peritoneal infiltrate of macrophages, eosinophils, B cells, occasional neutrophils, but few T cells. Immunohistochemical staining demonstrated the presence of murine IgG on tilapia islets within capsules of rejecting, non-immunosuppressed mice, as well as murine IgG-positive lymphocytes in the layer of host cells surrounding those capsules. These findings suggested that our barium (Ba)-gelled alginate capsules are permeable to IgG and that anti-piscine antibodies may be involved in the rejection of encapsulated tilapia islets in untreated mice. No single immunosuppressive agent prolonged encapsulated tilapia islet survival in NOD mice, but the combination of CTLA4-Ig plus anti-CD154 mAb extended tilapia islet graft survival until rejection at 119 ± 20 days and inhibited host cell recruitment to the peritoneal cavity. Triple treatment with CTLA4-Ig, anti-CD154 mAb, and anti-CD4 mAb allowed graft survival for 157 ± 35 days with little evidence of a host cellular reaction. IV and oral glucose tolerance tests (GTTs) of recipients with functioning xenografts demonstrated remarkably normal metabolic function. CONCLUSIONS: We conclude that microencapsulated tilapia islets can survive long term with excellent metabolic control in diabetic mice given targeted immunosuppression, suggesting that cross-species physiological incompatibility may not compromise the applicability of this novel approach for future clinical applications. We predict that an improved microcapsule that prevents the entrance of IgG will enhance tilapia islet survival in this model, possibly allowing the application of this technique with limited or no immunosuppression.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/cirurgia , Transplante Heterólogo , Animais , Rejeição de Enxerto/prevenção & controle , Imunossupressores/uso terapêutico , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Suínos , Tilápia , Transplante Heterólogo/métodos
8.
Surgery ; 154(6): 1385-93; discussion 1393, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24206618

RESUMO

BACKGROUND: Human pituitary adenomas express folate receptors (FR); therefore, we hypothesized that parathyroid (PT) tumors also might express FR, whereas normal human thyroids might not. The purpose of our study was to characterize the functionality of FRs on human PT tumors, with the goal of developing an imaging tool that would concentrate in PT more than in the thyroid. METHODS: Human PTs and thyroids were evaluated for FR expression by immunohistochemistry. Expression of genes for FRα and FRß was measured with the Illumina Human HT-12 Expression Bead Chips and verified by quantitative reverse-transcription polymerase chain reaction. Folate incorporation by PT cells versus normal thyroid cells was determined by incubation with (99m)Technetium ((99m)Tc)(CO)3-folate and (99m)Tc-Etarfolatide, and uptake was determined by gamma counting. Specific targeting of FRs was demonstrated by blocking with cold folate. A549 cells and Jurkat cells served as FR-negative controls, and KB cells and HeLa cells were FR-positive controls. RESULTS: On immunohistochemistry and Western blotting, human PT cells expressed FRs, whereas human thyroid cells did not. The FRα gene was expressed in all PTs analyzed, and the FRß gene was expressed by most. Uptake of (99m)Tc(CO)3-folate was increased in PT cells versus thyroid cells. There was dose-dependent uptake of (99m)Tc-etarfolatide, and uptake was inhibited by preincubation with cold folate, confirming FR-mediated binding. CONCLUSION: This is the first report of the expression and functionality of FRs on human PT cells. These findings suggest that (99m)Tc-folate holds potential for localization of PT tumors preoperatively and their treatment.


Assuntos
Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo , Receptor 2 de Folato/genética , Receptor 2 de Folato/metabolismo , Glândulas Paratireoides/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetulus , Ácido Fólico/análogos & derivados , Ácido Fólico/metabolismo , Expressão Gênica , Células HeLa , Humanos , Células Jurkat , Células KB , Compostos de Organotecnécio/metabolismo , Glândulas Paratireoides/diagnóstico por imagem , Neoplasias das Paratireoides/diagnóstico por imagem , Neoplasias das Paratireoides/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Cintilografia , Compostos Radiofarmacêuticos/metabolismo , Glândula Tireoide/diagnóstico por imagem , Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Neoplasias da Glândula Tireoide/metabolismo
9.
J Am Heart Assoc ; 2(5): e000367, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-24113327

RESUMO

BACKGROUND: Stem cells for cardiac repair have shown promise in preclinical trials, but lower than expected retention, viability, and efficacy. Encapsulation is one potential strategy to increase viable cell retention while facilitating paracrine effects. METHODS AND RESULTS: Human mesenchymal stem cells (hMSC) were encapsulated in alginate and attached to the heart with a hydrogel patch in a rat myocardial infarction (MI) model. Cells were tracked using bioluminescence (BLI) and cardiac function measured by transthoracic echocardiography (TTE) and cardiac magnetic resonance imaging (CMR). Microvasculature was quantified using von Willebrand factor staining and scar measured by Masson's Trichrome. Post-MI ejection fraction by CMR was greatly improved in encapsulated hMSC-treated animals (MI: 34 ± 3%, MI + Gel: 35 ± 3%, MI + Gel + hMSC: 39 ± 2%, MI + Gel + encapsulated hMSC: 56 ± 1%; n = 4 per group; P < 0.01). Data represent mean ± SEM. By TTE, encapsulated hMSC-treated animals had improved fractional shortening. Longitudinal BLI showed greatest hMSC retention when the cells were encapsulated (P < 0.05). Scar size at 28 days was significantly reduced in encapsulated hMSC-treated animals (MI: 12 ± 1%, n = 8; MI + Gel: 14 ± 2%, n = 7; MI + Gel + hMSC: 14 ± 1%, n = 7; MI+Gel+encapsulated hMSC: 7 ± 1%, n = 6; P < 0.05). There was a large increase in microvascular density in the peri-infarct area (MI: 121 ± 10, n = 7; MI + Gel: 153 ± 26, n = 5; MI + Gel + hMSC: 198 ± 18, n = 7; MI + Gel + encapsulated hMSC: 828 ± 56 vessels/mm2, n = 6; P < 0.01). CONCLUSIONS: Alginate encapsulation improved retention of hMSCs and facilitated paracrine effects such as increased peri-infarct microvasculature and decreased scar. Encapsulation of MSCs improved cardiac function post-MI and represents a new, translatable strategy for optimization of regenerative therapies for cardiovascular diseases.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Infarto do Miocárdio/cirurgia , Animais , Técnicas de Cultura de Células/métodos , Sobrevivência de Enxerto , Humanos , Masculino , Ratos
10.
Transplantation ; 88(2): 160-9, 2009 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-19623010

RESUMO

BACKGROUND: The long-term metabolic function of microencapsulated xenogeneic adult porcine islets (API) was assessed in a murine model of type 1 diabetes mellitus. METHODS: API were encapsulated in barium-gelled alginate and transplanted intraperitoneally in diabetic nonobese diabetic (NOD) mice given no immunosuppression or given costimulatory blockade (CoB; CTLA4-Ig+anti-CD154 mAb). Control mice received nonencapsulated API under the kidney capsule. Graft function was monitored by measurement of random blood glucose levels, serum glycosylated hemoglobin (HbA1c), serum porcine C peptide, in vivo glucose tolerance tests, and histologic analyses of host pancreas and graft biopsies. Host immune responses to the islet xenografts were characterized by phenotyping peritoneal cellular infiltrates and by measuring serum antiporcine antibody levels. RESULTS: Without immunosuppression, nonencapsulated API functioned for less than 1 week, and microencapsulated API functioned for 35+/-14 days before rejection, associated with both a cellular and a humoral immune response. With continuous CoB, nonencapsulated API functioned for 27+/-4 days, whereas microencapsulated API functioned for >450 days with measurable levels of serum porcine C peptide, near normal in vivo glucose tolerance tests and HbA1c levels, and intact microcapsules containing viable, insulin-positive porcine islets. CONCLUSIONS: Microencapsulated API restored normoglycemia for more than 1 year in spontaneously diabetic NODs given dual CoB. To our knowledge, this is the first study to document long-term normalized HbA1c, porcine C peptide, and near normal glucose tolerance in immunosuppressed diabetic NOD mice transplanted intraperitoneally with microencapsulated API. Our study suggests that transplantation of microencapsulated porcine islet xenografts may be a future treatment for patients with type 1 diabetes mellitus.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Tolerância ao Transplante/imunologia , Transplante Heterólogo/imunologia , Animais , Peptídeo C/sangue , Teste de Tolerância a Glucose , Hemoglobinas Glicadas/análise , Rejeição de Enxerto/imunologia , Camundongos , Camundongos Endogâmicos NOD , Suínos
11.
J Diabetes Sci Technol ; 2(5): 760-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19885258

RESUMO

BACKGROUND: If alginate microcapsules are to be used clinically for therapeutic cell transplants, capsule formulations must be designed to enhance optimal biocompatibility and immune acceptance. METHODS: Microcapsules were generated using highly purified, endotoxin-free, ultra-low viscosity, high mannuronic acid alginate. The capsules differed with respect to gelling cation (50 mM barium or 100 mM calcium), alginate concentration (2.0% or 3.3%), alginate density (homogeneous or inhomogeneous), and the presence or absence poly-L-lysine (PLL) coating. Four types of empty capsules were implanted intraperitoneally (i.p.) in normal NOD mice, and their biocompatibility was evaluated after various time periods in vivo. Encapsulated adult porcine islets (APIs) were transplanted i.p. in diabetic NOD mice, and immune acceptance was evaluated by graft survival times, host cell adherence to capsule surfaces, and flow cytometric analysis of peritoneal host cells. RESULTS: All empty alginate capsules were biocompatible in vivo, but barium-gelled alginate capsules without PLL were clearly the most biocompatible, since 99% of these empty capsules had no host cell adherence up to 9 months in vivo. In diabetic NOD mice, APIs functioned significantly longer in barium-alginate capsules without PLL than in calcium-alginate capsules with PLL and had strikingly less host cell adherence, although large numbers of host cells (predominantly macrophages and eosinophils) infiltrated the peritoneal cavities of recipients with APIs in both types of capsules. Addition of PLL coatings to barium-alginate capsules dramatically decreased graft survival. CONCLUSIONS: Inhomogeneous barium-gelled alginate capsules without PLL are the optimal candidates for clinical trials, based on their enhanced biocompatibility and immune acceptance in vivo.

12.
Biochem Biophys Res Commun ; 340(1): 236-43, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16375863

RESUMO

Our goal is to develop effective islet grafts for treating type 1 diabetes. Since human islets are scarce, we evaluated the efficacy of a microencapsulated insulin-secreting conditionally transformed allogeneic beta-cell line (betaTC-tet) in non-obese diabetic mice treated with tetracycline to inhibit cell growth. Relatively low serum levels of tetracycline controlled proliferation of betaTC-tet cells without inhibiting effective control of hyperglycemia in recipients. There was no significant host cellular reaction to the allografts or host cell adherence to microcapsules, and host cytokine levels were similar to those of sham-operated controls. We conclude that encapsulated allogeneic beta-cell lines may be clinically relevant, because they effectively restore euglycemia and do not elicit a strong cellular immune response following transplantation. To our knowledge, this is the first extensive characterization of the kinetics of host cellular and cytokine responses to an encapsulated islet cell line in an animal model of type 1 diabetes.


Assuntos
Técnicas de Cultura de Células/métodos , Citocinas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/cirurgia , Reação Enxerto-Hospedeiro/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Animais , Linhagem Celular , Diabetes Mellitus Tipo 1/patologia , Camundongos
13.
Transplantation ; 79(4): 409-18, 2005 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-15729166

RESUMO

BACKGROUND: Transplantation of human islets has been successful clinically. Since human islets are scarce, we are studying microencapsulated porcine islet xenografts in nonobese diabetic (NOD) mice. We have evaluated the cellular immune response in NOD mice with and without dual costimulatory blockade. METHODS: Alginate-poly-L-lysine-encapsulated adult porcine islets were transplanted i.p. in untreated diabetic NODs and NODs treated with CTLA4-Ig to block CD28/B7 and with anti-CD154 mAb to inhibit CD40/CD40-ligand interactions. Groups of mice were sacrificed on subsequent days; microcapsules were evaluated by histology; peritoneal cells were analyzed by FACS; and peritoneal cytokines were quantified by ELISA. Controls included immunoincompetent NOD-Scids and diabetic NODs given sham surgery or empty microcapsules. RESULTS: Within 20 days, encapsulated porcine islets induced accumulation of large numbers of macrophages, eosinophils, and significant numbers of CD4 and CD8 T cells at the graft site, and all grafts were rejected. During rejection, IFNgamma, IL-12 and IL-5 were significantly elevated over sham-operated controls, whereas IL-2, TNFalpha, IL-4, IL-6, IL-10, IL-1beta and TGFbeta were unchanged. Treatment with CTLA4-Ig and anti-CD154 prevented graft destruction in all animals during the 26 days of the experiment, dramatically inhibited recruitment of host inflammatory cells, and inhibited peritoneal IFNgamma and IL-5 concentrations while delaying IL-12 production. CONCLUSIONS: When two different pathways of T cell costimulation were blocked, T cell-dependent inflammatory responses were inhibited, and survival of encapsulated islet xenografts was significantly prolonged. These findings suggest synergy between encapsulation of donor islets and simultaneous blockade of two host costimulatory pathways in prolonging xenoislet transplant survival.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígeno B7-1/fisiologia , Antígenos CD28/fisiologia , Antígenos CD40/fisiologia , Ligante de CD40/fisiologia , Imunoconjugados/farmacologia , Terapia de Imunossupressão , Transplante das Ilhotas Pancreáticas/imunologia , Transplante Heterólogo/imunologia , Abatacepte , Animais , Citocinas/biossíntese , Feminino , Sobrevivência de Enxerto , Masculino , Camundongos , Camundongos Endogâmicos NOD , Suínos
14.
Cell Transplant ; 11(7): 695-705, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12518896

RESUMO

Our goal is to develop effective islet xenografts for treating human diabetes. We have studied microencapsulated neonatal porcine islet cell clusters (ICCs) transplanted intraperitoneally in spontaneously diabetic NOD mice, where they function to maintain normoglycemia in the autoimmune host. Nonencapsulated neonatal porcine ICCs functioned for 4.5 +/- 0.5 days before being rejected; encapsulation prolonged graft function to 17 +/- 2 days. CTLA4-Ig treatment did not enhance the survival of nonencapsulated ICCs. However, CTLA4-Ig treatment significantly extended the function of encapsulated ICCs to 73 +/- 5 days. Histological analyses demonstrated a profuse pericapsular cellular reaction associated with rejection of encapsulated islet xenografts in untreated mice, while this reaction was significantly reduced in CTLA4-Ig-treated mice. To study mechanisms of xenograft rejection in this model, we analyzed proliferative responses to neonatal porcine ICCs and cytokines present in the peritoneal cavities of transplanted mice. Spleen cells from both CTLA4-Ig-treated and untreated rejecting NODs exhibited vigorous proliferation in the absence of antigenic stimulation, suggesting prior activation in vivo, while splenocytes from CTLA4-Ig-treated NODs with functioning grafts had low proliferative levels, equal to controls. Islet-specific proliferation was not detected in islet-rejecting mice, perhaps due to their high background levels. With the exception of elevated IL-6 levels, empty capsules did not provoke a significant peritoneal cytokine response compared with sham surgery or untransplanted control mice. However, IL-5, IL-12, TGF-beta, and IL-1beta were significantly elevated in NODs receiving encapsulated neonatal porcine ICCs compared with untransplanted controls. There were no significant differences between peritoneal cytokine concentrations in CTLA4-Ig-treated mice with long-term functioning grafts compared to mice that rejected grafts at earlier time points. We conclude that the combination of donor islet microencapsulation and brief treatment of the recipient with co-stimulatory blockade delays sensitization of the host, possibly by altering mechanism(s) for recruitment and/or activation of host effector cells.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Rejeição de Enxerto/tratamento farmacológico , Sobrevivência de Enxerto/efeitos dos fármacos , Imunoconjugados/farmacologia , Imunossupressores/farmacologia , Transplante das Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/efeitos dos fármacos , Abatacepte , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Feminino , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/imunologia , Reação Enxerto-Hospedeiro/imunologia , Imunização , Imunoconjugados/imunologia , Imunoconjugados/uso terapêutico , Imunossupressores/imunologia , Imunossupressores/uso terapêutico , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Sus scrofa , Transplante Heterólogo/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...