Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.142
Filtrar
1.
BMJ Open Diabetes Res Care ; 12(2)2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38485229

RESUMO

INTRODUCTION: Static incubation (static glucose-stimulated insulin secretion, sGSIS) is a measure of islet secretory function. The Stimulation Index (SI; insulin produced in high glucose/insulin produced in low glucose) is currently used as a product release criterion of islet transplant potency. RESEARCH DESIGN AND METHODS: Our hypothesis was that the Delta, insulin secreted in high glucose minus insulin secreted in low glucose, would be more predictive. To evaluate this hypothesis, sGSIS was performed on 32 consecutive human islet preparations, immobilizing the islets in a slurry of Sepharose beads to minimize mechanical perturbation. Simultaneous full-mass subrenal capsular transplants were performed in chemically induced diabetic immunodeficient mice. Logistic regression analysis was used to determine optimal cut-points for diabetes reversal time and the Fisher Exact Test was used to assess the ability of the Delta and the SI to accurately classify transplant outcomes. Receiver operating characteristic curve analysis was performed on cut-point grouped data, assessing the predictive power and optimal cut-point for each sGSIS potency metric. Finally, standard Kaplan-Meier-type survival analysis was conducted. RESULTS: In the case of the sGSIS the Delta provided a superior islet potency metric relative to the SI.ConclusionsThe sGSIS Delta value is predicitive of time to diabetes reversal in the full mass human islet transplant bioassay.


Assuntos
Diabetes Mellitus , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Humanos , Camundongos , Animais , Secreção de Insulina , Glucose/farmacologia , Glucose/metabolismo , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/fisiologia , Diabetes Mellitus/metabolismo , Insulina/metabolismo , Bioensaio
2.
Biomater Sci ; 12(4): 821-836, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38168805

RESUMO

Islet transplantation holds significant promise as a curative approach for type 1 diabetes (T1D). However, the transition of islet transplantation from the experimental phase to widespread clinical implementation has not occurred yet. One major hurdle in this field is the challenge of insufficient vascularization and subsequent early loss of transplanted islets, especially in non-intraportal transplantation sites. The establishment of a fully functional vascular system following transplantation is crucial for the survival and secretion function of islet grafts. This vascular network not only ensures the delivery of oxygen and nutrients, but also plays a critical role in insulin release and the timely removal of metabolic waste from the grafts. This review summarizes recent advances in effective strategies to improve graft revascularization and enhance islet survival. These advancements include the local release and regulation of angiogenic factors (e.g., vascular endothelial growth factor, VEGF), co-transplantation of vascular fragments, and pre-vascularization of the graft site. These innovative approaches pave the way for the development of effective islet transplantation therapies for individuals with T1D.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Humanos , Ilhotas Pancreáticas/metabolismo , Diabetes Mellitus Tipo 1/cirurgia , Materiais Biocompatíveis , Fator A de Crescimento do Endotélio Vascular/metabolismo , Transplante das Ilhotas Pancreáticas/fisiologia , Neovascularização Fisiológica
3.
Horm Metab Res ; 56(4): 279-285, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37956864

RESUMO

Ferroptosis was recently identified as a non-apoptotic, iron-dependent cell death mechanism that is involved in various pathologic conditions. There is first evidence for its significance also in the context of islet isolation and transplantation. Transplantation of pancreatic human islets is a viable treatment strategy for patients with complicated diabetes mellitus type 1 (T1D) that suffer from severe hypoglycemia. A major determinant for functional outcome is the initial islet mass transplanted. Efficient islet isolation procedures and measures to minimize islet loss are therefore of high relevance. To this end, better understanding and subsequent targeted inhibition of cell death during islet isolation and transplantation is an effective approach. In this study, we aimed to elucidate the mechanism of ferroptosis in pancreatic islets. Using a rodent model, isolated islets were characterized relating to the effects of experimental induction (RSL3) and inhibition (Fer1) of ferroptotic pathways. Besides viability, survival, and function, the study focused on characteristic ferroptosis-associated intracellular changes such as MDA level, iron concentration and the expression of ACSL4. The study demonstrates that pharmaceutical induction of ferroptosis by RSL3 causes enhancement of oxidative stress and leads to an increase of intracellular iron, zinc and MDA concentration, as well as the expression of ACSL4 protein. Consequently, a massive reduction of islet function, viability, and survival was found. Fer1 has the potential to inhibit and attenuate these cellular changes and thereby protect the islets from cell death.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Humanos , Transplante das Ilhotas Pancreáticas/efeitos adversos , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Diabetes Mellitus Tipo 1/metabolismo , Morte Celular , Ferro
4.
Stem Cell Rev Rep ; 20(3): 839-844, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38153636

RESUMO

BACKGROUND: Insular allograft for unstable type 1 diabetes and autograft in pancreatectomy patients are nowadays considered established procedures with precise indications and predictable outcomes. The clinical outcome of islet transplantation is similar to that of pancreas transplantation, avoiding the complications associated with organ transplantation. OBJECTIVE: We hypothesised that transplantation of islets of Langerhans within an endocrine organ could better promote their engraftment and function. This could help to resolve or ameliorate known pathological conditions such as unstable type 1 diabetes and complicated type 2 diabetes. RATIONALE: Pancreatic islet transplantation is currently performed almost exclusively in the liver. The liver provides a sufficiently favourable environment, although not entirely. The hepatic parenchyma has a lower oxygen tension than the pancreatic parenchyma and the vascular structure of the liver is not typical of an exclusively endocrine organ. Moreover, islet transplantation into the liver is not without complications, including hematoma or portal vein thrombosis. PROPOSED PROJECT: The thyroid gland is the endocrine gland proposed as a 'container'. In fact, it has all the characteristics of 'physio-compatibility' which can address the objectives assumed. It is indeed an ideal site because it is an easily accessible anatomical site that allows islets to be implanted using ultrasound-guided transcutaneous inoculation technique. Moreover, it has physiological and anatomical endocrine affinities with pancreatic islets and, if necessary, it can be removed, using hormone supplementation or replacement therapy. CONCLUSIONS: The thyroid gland may be proposed as an ideal site for islet implantation due to its anatomical and physiocompatibility characteristics.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Transplante de Pâncreas , Humanos , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/cirurgia , Glândula Tireoide , Pancreatectomia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/cirurgia , Ilhotas Pancreáticas/cirurgia , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia
5.
Tissue Cell ; 79: 101919, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36137362

RESUMO

Type 1 diabetes is an autoimmune disease that emerges with the destruction of beta cells of pancreatic Langerhans islets. Three different therapeutical approaches have been developed so far; pancreas transplantation, islet transplantation, and cell-based therapies. Bioengineering cell sheets for tissue generating is one of the latest approaches that have been used to construct cell-sheets instead of single cells so that it mimics the in vivo environments more. In this study, extra-hepatic functional islet tissue was constructed by transferring the 3-D beta cells and GFP labelled MSCs MSC sheets to the subcutaneous site of rats with STZ-induced diabetes. rBM-MSCs and beta cells were cultured on the 6-well PIPAAm culture dishes. Obtained rBM-MSCs as two-cell sheets and beta cells cultured in droplets with matrigel has transplanted into the dorsal subcutaneous area of diabetic rats. Fasting blood glucose levels and body weights were evaluated for 30 days after transplantation. Immunocytochemistry analysis for the anti-apoptotic, anti-inflammatory, and angiogenetic effects of MSCs on the 30th day of subcutaneous cell transplantation. All recipient rats transplanted with beta-cells with MSCs returned toward normoglycemia by day 5 and remained at this level for 30 days. Immunocytochemical analyses supported that the MSCs and beta cells preserved their viability and function. MSCs secrete cytokines and growth factors TGF-ß and IL-6; MSCs of the important features of the anti-apoptotic and anti-inflammatory properties, thanks to apoptosis of beta cells preserve graft explained by inhibition. In transplantation of MSCs induced angiogenesis and neovascularization, PECAM-1 and GFP positive simultaneously determining endothelial cells was observed indicating. Subcutaneous 3D beta-cell transplantation would be possible with the MSC-sheets as a feeder layer of beta cells. The beta-cell line is glucose-sensitive and has a high insulin release potential, and can be used as an alternative to islets in in vivo transplant studies.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ratos , Animais , Células Endoteliais/metabolismo , Células Secretoras de Somatostatina/metabolismo , Transplante das Ilhotas Pancreáticas/fisiologia , Bioengenharia , Insulina/metabolismo
6.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(1): 1-7, 2022 Jan 28.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-35545357

RESUMO

OBJECTIVES: Heparin is mainly used as an anticoagulant in clinic, and it also has a certain anti-inflammatory effect. At present, after portal vein islet transplantation in diabetic patients, heparin is mainly infused through the peripheral veins of the limbs to achieve the purpose of anticoagulation and protection of the graft, rather than through the portal vein. In this study, animal experiments were conducted to investigate the effect of heparin infusion via the portal vein and marginal ear vein on the instant blood-mediated inflammatory reaction (IBMIR) after portal vein islet transplantation, which is the choice of anticoagulation methods for clinical islet transplantation to provide a basis for decision-making. METHODS: A total of 50 neonatal pigs (Xeno-1 type, 3-5 days) were selected. Islets were isolated and purified from the pancreas of neonatal pigs. Ten non-diabetic Landrace pigs (1.5-2.0 months) served as recipients, and 12 000 IEQ/kg neonatal porcine islets were transplanted into the liver through the portal vein. All recipients received bolus injection of 50 U/kg of heparin 10 minutes before transplantation. After the bolus injection of heparin, the experimental group received heparin via the portal vein [10 U/(kg·h), 5 recipients], and the control group received heparin via the marginal ear vein [10 U/(kg·h), 5 recipients]. The superior vena cava blood was collected from the 2 groups pre-operation at 1, 3, 24 h post-operation of the transplantation. The portal vein blood was collected from the experimental group at 1 and 3 h after the transplantation as well. The levels of complement C3a, C5a, thrombin-antithrombin complex (TAT), ß-thromboglobulin (ß-TG), and D-dimer as well as activated partial thromboplastin time (APTT) in superior vena cava blood from 1 and 3 h post-transplantation were detected in the 2 groups, and the levels of anti-Xa and anti-IIa in the portal vein and superior vena cava blood from 1 and 3 h post-transplantation in the experimental group were detected. Twenty four hours after the transplantation, the liver tissues in the 2 groups were collected for pathological examination to observe the inflammatory cell infiltration and peripheral thrombosis around the islets graft in liver. RESULTS: Before transplantation, there was no statistically significant difference in C3a, C5a, TAT, ß-TG, D-dimer levels and APTT between the 2 groups (all P>0.05). At 1 and 3 h after transplantation, the C3a, TAT, and D-dimer levels in the experimental group were significant decreased than those in the control groups (all P<0.05), and at 3 h after transplantation the C5a was significant decreased than that in the control group (P<0.05). At 1 and 3 h after transplantation, the anti-Xa and anti-IIa levels in the portal vein blood were significantly increased than those in the superior vena cava blood in the experimental group (all P<0.05). Pathological results showed the presence of islet cell clusters in the liver blood vessels. The thrombus formation and neutrophil infiltration around islet graft was not obvious in the experimental group, while massive thrombus formation and neutrophil infiltration in the control group. CONCLUSIONS: Compared with marginal ear vein infusion of heparin, the direct infusion of heparin in the portal vein has a certain inhibitory effect on complement system, coagulation system activation and inflammatory cell infiltration in portal vein islet transplantation, which may attenuate the occurrence of IBMIR.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico , Heparina/uso terapêutico , Humanos , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Veia Porta , Suínos , Veia Cava Superior
7.
Nat Med ; 28(2): 272-282, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35115708

RESUMO

Human pluripotent stem-cell-derived islets (hPSC-islets) are a promising cell resource for diabetes treatment1,2. However, this therapeutic strategy has not been systematically assessed in large animal models physiologically similar to humans, such as non-human primates3. In this study, we generated islets from human chemically induced pluripotent stem cells (hCiPSC-islets) and show that a one-dose intraportal infusion of hCiPSC-islets into diabetic non-human primates effectively restored endogenous insulin secretion and improved glycemic control. Fasting and average pre-prandial blood glucose levels significantly decreased in all recipients, accompanied by meal or glucose-responsive C-peptide release and overall increase in body weight. Notably, in the four long-term follow-up macaques, average hemoglobin A1c dropped by over 2% compared with peak values, whereas the average exogenous insulin requirement reduced by 49% 15 weeks after transplantation. Collectively, our findings show the feasibility of hPSC-islets for diabetic treatment in a preclinical context, marking a substantial step forward in clinical translation of hPSC-islets.


Assuntos
Diabetes Mellitus Experimental , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Glicemia , Diabetes Mellitus Experimental/terapia , Humanos , Insulina , Transplante das Ilhotas Pancreáticas/fisiologia , Primatas
8.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-928999

RESUMO

OBJECTIVES@#Heparin is mainly used as an anticoagulant in clinic, and it also has a certain anti-inflammatory effect. At present, after portal vein islet transplantation in diabetic patients, heparin is mainly infused through the peripheral veins of the limbs to achieve the purpose of anticoagulation and protection of the graft, rather than through the portal vein. In this study, animal experiments were conducted to investigate the effect of heparin infusion via the portal vein and marginal ear vein on the instant blood-mediated inflammatory reaction (IBMIR) after portal vein islet transplantation, which is the choice of anticoagulation methods for clinical islet transplantation to provide a basis for decision-making.@*METHODS@#A total of 50 neonatal pigs (Xeno-1 type, 3-5 days) were selected. Islets were isolated and purified from the pancreas of neonatal pigs. Ten non-diabetic Landrace pigs (1.5-2.0 months) served as recipients, and 12 000 IEQ/kg neonatal porcine islets were transplanted into the liver through the portal vein. All recipients received bolus injection of 50 U/kg of heparin 10 minutes before transplantation. After the bolus injection of heparin, the experimental group received heparin via the portal vein [10 U/(kg·h), 5 recipients], and the control group received heparin via the marginal ear vein [10 U/(kg·h), 5 recipients]. The superior vena cava blood was collected from the 2 groups pre-operation at 1, 3, 24 h post-operation of the transplantation. The portal vein blood was collected from the experimental group at 1 and 3 h after the transplantation as well. The levels of complement C3a, C5a, thrombin-antithrombin complex (TAT), β-thromboglobulin (β-TG), and D-dimer as well as activated partial thromboplastin time (APTT) in superior vena cava blood from 1 and 3 h post-transplantation were detected in the 2 groups, and the levels of anti-Xa and anti-IIa in the portal vein and superior vena cava blood from 1 and 3 h post-transplantation in the experimental group were detected. Twenty four hours after the transplantation, the liver tissues in the 2 groups were collected for pathological examination to observe the inflammatory cell infiltration and peripheral thrombosis around the islets graft in liver.@*RESULTS@#Before transplantation, there was no statistically significant difference in C3a, C5a, TAT, β-TG, D-dimer levels and APTT between the 2 groups (all P>0.05). At 1 and 3 h after transplantation, the C3a, TAT, and D-dimer levels in the experimental group were significant decreased than those in the control groups (all P<0.05), and at 3 h after transplantation the C5a was significant decreased than that in the control group (P<0.05). At 1 and 3 h after transplantation, the anti-Xa and anti-IIa levels in the portal vein blood were significantly increased than those in the superior vena cava blood in the experimental group (all P<0.05). Pathological results showed the presence of islet cell clusters in the liver blood vessels. The thrombus formation and neutrophil infiltration around islet graft was not obvious in the experimental group, while massive thrombus formation and neutrophil infiltration in the control group.@*CONCLUSIONS@#Compared with marginal ear vein infusion of heparin, the direct infusion of heparin in the portal vein has a certain inhibitory effect on complement system, coagulation system activation and inflammatory cell infiltration in portal vein islet transplantation, which may attenuate the occurrence of IBMIR.


Assuntos
Animais , Humanos , Anticoagulantes/uso terapêutico , Heparina/uso terapêutico , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/fisiologia , Veia Porta , Suínos , Veia Cava Superior
9.
Methods Mol Biol ; 2224: 87-98, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33606208

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease, where insulin-producing ß-cells in the pancreas are inappropriately recognized and destroyed by immune cells. Islet transplantation is the most successful cell-based therapy for T1D individuals who experience frequent and severe life-threatening hypoglycemia. However, this therapy is extremely restricted owing to the limited availability of donor pancreas. In recent years, significant progress has been made in generating ß-cells from stem/progenitor cells using different approaches of in vitro differentiation. The insulin production from such in vitro generated ß-cells is still far less than that observed in islet ß-cells. We employed a novel strategy to improve the efficiency of progenitor cell differentiation by performing partial mouse pancreas resection after transplanting in vitro generated insulin-producing cells under the kidney capsule of these mice. Pancreas resection (pancreatectomy) has been shown to induce regenerative pathways, leading to regeneration of almost the entire resected pancreas over 3-5 weeks in mice. We found that in our method, regenerating mouse pancreas promotes better graft differentiation/maturation and insulin production from transplanted cells. In this chapter, we detail the protocols used for transplantation of in vitro differentiated cells in immunocompromised mice, partial pancreatectomy in host (NOD scid) mice, and assessment of graft function. We believe that our protocols provide a solid platform for further studies aimed at understanding growth/differentiation molecules secreted from regenerating pancreas that promote graft maturation.


Assuntos
Diferenciação Celular/fisiologia , Pâncreas/fisiologia , Animais , Diabetes Mellitus Tipo 1/fisiopatologia , Células Secretoras de Insulina/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pancreatectomia/métodos , Células-Tronco/fisiologia
10.
J Biomed Mater Res B Appl Biomater ; 108(2): 343-352, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31013399

RESUMO

Optimal function of immunoisolated islets requires adequate supply of oxygen to metabolically active insulin producing beta-cells. Using mathematical modeling, we investigated the influence of the pO2 on islet insulin secretory capacity and evaluated conditions that could lead to the development of tissue anoxia, modeled for a 300 µm islet in a 500 µm microcapsule or a 500 µm planar, slab-shaped macrocapsule. The pO2 was used to assess the part of islets that contributed to insulin secretion. Assuming a 500 µm macrocapsule with a 300 µm islet, with oxygen consumption rate (OCR) of 100-300 nmol min-1 mg-1 DNA, islets did not develop any necrotic core. The nonfunctional zone (with no insulin secretion if pO2 < 0.1 mmHg) was 0.3% for human islets (OCR ~100 nmol/min/mg DNA) and 35% for porcine islets (OCR ~300 nmol/min/mg DNA). The OCR of the islet preparation is profoundly affected by islet size, with optimal size of <250 µm in diameter (human) or <150 µm (porcine). Our data suggest that microcapsules afford superior oxygen delivery to encapsulated islets than macrocapsules, and optimal islet function can be achieved by encapsulating multiple, small (<150 µm) islets with OCR of ~100 nmol min-1 mg-1 DNA (human islets) or ~200 nmol min-1 mg-1 DNA (porcine islets).


Assuntos
Cápsulas/metabolismo , Transplante das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/metabolismo , Oxigênio/metabolismo , Animais , DNA/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina/fisiologia , Modelos Biológicos , Modelos Teóricos , Consumo de Oxigênio/fisiologia , Suínos
11.
Sci Rep ; 9(1): 3918, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30850640

RESUMO

The anterior chamber of the eye (ACE) has emerged as a promising clinical islet transplantation site because of its multiple advantages over the conventional intra-hepatic portal site. This includes reduced surgical invasiveness and increased islet graft survival rate. It also allows for enhanced accessibility and monitoring of the islets. Although the ACE is initially an immuno-privileged site, this privilege is disrupted once the islet grafts are re-vascularized. Given that the ACE is a confined space, achieving graft immune tolerance through local immunosuppressive drug delivery is therefore feasible. Here, we show that islet rejection in the ACE of mice can be significantly suppressed through local delivery of rapamycin by carefully designed sustained-release microparticles. In this 30-day study, allogeneic islet grafts with blank microparticles were completely rejected 18 days post-transplantation into mice. Importantly, allogeneic islet grafts co-injected with rapamycin releasing microparticles into a different eye of the same recipient were preserved much longer, with some grafts surviving for more than 30 days. Hence, islet allograft survival was enhanced by a localized and prolonged delivery of an immunosuppressive drug. We envisage that this procedure will relieve diabetic transplant recipients from harsh systemic immune suppression, while achieving improved glycemic control and reduced insulin dependence.


Assuntos
Câmara Anterior , Rejeição de Enxerto/prevenção & controle , Imunossupressores/administração & dosagem , Transplante das Ilhotas Pancreáticas/métodos , Sirolimo/administração & dosagem , Aloenxertos , Animais , Câmara Anterior/efeitos dos fármacos , Câmara Anterior/imunologia , Câmara Anterior/cirurgia , Preparações de Ação Retardada , Diabetes Mellitus/imunologia , Diabetes Mellitus/cirurgia , Sistemas de Liberação de Medicamentos , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Transplante das Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Modelos Animais , Tolerância ao Transplante/efeitos dos fármacos
12.
Stem Cells Transl Med ; 8(3): 209-214, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30417988

RESUMO

We examined data and patterns in clinical islet transplant studies registered on ClinicalTrials.gov (CTgov) for treatment of type 1 diabetes (T1D), with a goal of extracting insights to apply in the design of a pluripotent stem cell-derived islet therapy. Clinical islet transplantation, as a cell therapy (rather than solid organ transplant) is a unique precedent for stem cell-based islet therapies. Registration activity shows that the field is not growing significantly, and newer registrations suggest that the reasons for stagnation include need for a more optimal site of infusion/transplantation, and especially a need for better immune protective strategies to advance a more effective and durable therapy for T1D. Stem Cells Translational Medicine 2019;8:209&214.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Transplante das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/citologia , Células-Tronco/citologia , Transplantes/citologia , Diferenciação Celular/fisiologia , Ensaios Clínicos como Assunto , Humanos
13.
Curr Stem Cell Res Ther ; 14(1): 52-56, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30207243

RESUMO

In the field of stem cell therapy, research on the application of mesenchymal stem cells (MSCs) has flourished because of the various functions. On the other hand, research on the method of cell transplantation has developed from the administration of cell suspensions to cell-sheet engineering and 3D technology. In the trend, a cell transplantation platform named CellSaic, which is a combination of xeno-free recombinant scaffolds in a cell aggregate-like shape, was developed. CellSaic is the cell transplantation platform that can prevent the central necrosis within cell aggregates by arranging the cells and petaloid pieces of recombinant peptide (RCP) in a mosaic. The prevention of central necrosis is the most significant advantage over other 3D culture systems. This review details the unique characteristics of CellSaic including safety examination results and describes its future application for MSC transplantation. Particularly, in the application of MSCs, it has been reported that the MSC CellSaics increased the effect on improving various symptoms compared with MSCs only in the application of the therapy to inflammatory bowel disease (IBD), cerebral infarction, bone cartilage regeneration in joints, and islet transplantation. In accordance with the "One Health" concept, it is anticipated that this technology is expected to contribute to companion animal therapy and human therapy in the future.


Assuntos
Agregação Celular/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Fragmentos de Peptídeos/fisiologia , Engenharia Tecidual , Animais , Cartilagem/crescimento & desenvolvimento , Adesão Celular/fisiologia , Infarto Cerebral/terapia , Colite/terapia , Citocinas/metabolismo , Humanos , Doenças Inflamatórias Intestinais/terapia , Transplante das Ilhotas Pancreáticas/fisiologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Camundongos , Necrose/fisiopatologia , Neoplasias Pancreáticas/terapia , Transplantes/fisiologia
14.
Cell Immunol ; 329: 10-16, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29661473

RESUMO

Silk fibroin is a novel biomaterial for enhancing transplanted islet cell function and survival. This study investigated whether silk fibroin may have unique properties that improve islet function in the face of inflammatory-mediated stress during transplantation. Murine islet function was tested in vitro with either silk fibroin or alginate and challenged with inflammatory cytokines. The glucose-stimulated insulin secretion index for all conditions decreased with inflammatory cytokines, but was better preserved for islets exposed to silk compared to those exposed to alginate or medium. GLUT2 transporter expression on the cell surface of islets exposed to silk was increased compared to alginate or medium alone. Upon cytokine stress, a greater percentage of islet cells exposed to silk expressed GLUT2 on their surface. We conclude that preconditioning islets with silk fibroin stimulates islet cell surface GLUT2 expression, an increase, which persists under inflammatory stress, and may improve islet engraftment and function after transplantation.


Assuntos
Fibroínas/metabolismo , Fibroínas/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Alginatos/farmacologia , Animais , Fibroínas/fisiologia , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Inflamação , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Seda/fisiologia , Estresse Fisiológico/efeitos dos fármacos
15.
Sci Rep ; 8(1): 6508, 2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29695723

RESUMO

Transplantation of encapsulated islets can cure diabetes without immunosuppression, but oxygen supply limitations can cause failure. We investigated a retrievable macroencapsulation device wherein islets are encapsulated in a planar alginate slab and supplied with exogenous oxygen from a replenishable gas chamber. Translation to clinically-useful devices entails reduction of device size by increasing islet surface density, which requires increased gas chamber pO2. Here we show that islet surface density can be substantially increased safely by increasing gas chamber pO2 to a supraphysiological level that maintains all islets viable and functional. These levels were determined from measurements of pO2 profiles in islet-alginate slabs. Encapsulated islets implanted with surface density as high as 4,800 islet equivalents/cm3 in diabetic rats maintained normoglycemia for more than 7 months and provided near-normal intravenous glucose tolerance tests. Nearly 90% of the original viable tissue was recovered after device explantation. Damaged islets failed after progressively shorter times. The required values of gas chamber pO2 were predictable from a mathematical model of oxygen consumption and diffusion in the device. These results demonstrate feasibility of developing retrievable macroencapsulated devices small enough for clinical use and provide a firm basis for design of devices for testing in large animals and humans.


Assuntos
Sobrevivência Celular/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiologia , Oxigênio/metabolismo , Alginatos/metabolismo , Animais , Glicemia/metabolismo , Glicemia/fisiologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Teste de Tolerância a Glucose/métodos , Sobrevivência de Enxerto/fisiologia , Terapia de Imunossupressão/métodos , Masculino , Consumo de Oxigênio/fisiologia , Ratos , Ratos Endogâmicos Lew
16.
Islets ; 10(1): 40-49, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29315020

RESUMO

Pancreatic islet transplantation is being extensively researched as an alternative treatment for type 1 diabetic patients. This treatment is currently limited by temporal mismatch, between the availability of pancreas and isolated islets from deceased organ donor, and the recipient's need for freshly isolated islets. To solve this issue, cryopreservation of islets may offer the potential to bank islets for transplant on demand. Cryopreservation, however, introduces an overwhelmingly harsh environment to the ever-so-fragile islets. After exposure to the freezing and thawing, islets are usually either apoptotic, non-functional, or non-viable. Several studies have proposed various techniques that could lead to increased cell survival and function following a deep freeze. The purpose of this article is to critically review the techniques of islet cryopreservation, with the goal of highlighting optimization parameters that can lead to the most viable and functional islet upon recovery and/or transplant.


Assuntos
Criopreservação/métodos , Ilhotas Pancreáticas , Animais , História do Século XIX , História do Século XX , História do Século XXI , Humanos , Transplante das Ilhotas Pancreáticas/história , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia
17.
Med Sci (Paris) ; 32(4): 401-7, 2016 Apr.
Artigo em Francês | MEDLINE | ID: mdl-27137698

RESUMO

Islet transplantation has set the ground for diabetes cell therapy and is still undergoing various developments that might improve clinical outcomes. Alternative sources for ß-cell replacement strategies are now led by human pluripotent stem cells that demonstrate near-normal ß-cell features after in vitro differentiation and which can reverse diabetes in mice. Yet, their propensity for tumor formation is still unresolved. The adult pancreas is suggested as a reservoir of facultative progenitors that could represent adequate candidates for ß-cell engineering, either in vivo through pharmacological treatment or after expansion in culture. This review focuses on the latest developments in protocols aiming at de novo production of functional ß cells.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/tendências , Diabetes Mellitus/terapia , Adulto , Animais , Diferenciação Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Humanos , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas/métodos , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Pâncreas/citologia , Pâncreas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Células-Tronco Pluripotentes/transplante , Regeneração
18.
Diabetes ; 65(5): 1208-18, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26936960

RESUMO

Diabetes is associated with loss of functional pancreatic ß-cells, and restoration of ß-cells is a major goal for regenerative therapies. Endogenous regeneration of ß-cells via ß-cell replication has the potential to restore cellular mass; however, pharmacological agents that promote regeneration or expansion of endogenous ß-cells have been elusive. The regenerative capacity of ß-cells declines rapidly with age, due to accumulation of p16(INK4a), resulting in limited capacity for adult endocrine pancreas regeneration. Here, we show that transforming growth factor-ß (TGF-ß) signaling via Smad3 integrates with the trithorax complex to activate and maintain Ink4a expression to prevent ß-cell replication. Importantly, inhibition of TGF-ß signaling can result in repression of the Ink4a/Arf locus, resulting in increased ß-cell replication in adult mice. Furthermore, small molecule inhibitors of the TGF-ß pathway promote ß-cell replication in human islets transplanted into NOD-scid IL-2Rg(null) mice. These data reveal a novel role for TGF-ß signaling in the regulation of the Ink4a/Arf locus and highlight the potential of using small molecule inhibitors of TGF-ß signaling to promote human ß-cell replication.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/antagonistas & inibidores , Células Secretoras de Insulina/efeitos dos fármacos , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Benzamidas/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/agonistas , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Dioxóis/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Receptores de Fatores de Crescimento Transformadores beta/agonistas , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Regeneração/efeitos dos fármacos , Bancos de Tecidos , Fator de Crescimento Transformador beta1/metabolismo , Transplante Heterólogo , Transplante Heterotópico
19.
Diabetes ; 65(2): 451-62, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26581595

RESUMO

Clinical islet transplantation has become an established treatment modality for selected patients with type 1 diabetes. However, a large proportion of transplanted islets is lost through multiple factors, including immunosuppressant-related toxicity, often requiring more than one donor to achieve insulin independence. On the basis of the cytoprotective capabilities of antifreeze proteins (AFPs), we hypothesized that supplementation of islets with synthetic AFP analog antiaging glycopeptide (AAGP) would enhance posttransplant engraftment and function and protect against tacrolimus (Tac) toxicity. In vitro and in vivo islet Tac exposure elicited significant but reversible reduction in insulin secretion in both mouse and human islets. Supplementation with AAGP resulted in improvement of islet survival (Tac(+) vs. Tac+AAGP, 31.5% vs. 67.6%, P < 0.01) coupled with better insulin secretion (area under the curve: Tac(+) vs. Tac+AAGP, 7.3 vs. 129.2 mmol/L/60 min, P < 0.001). The addition of AAGP reduced oxidative stress, enhanced insulin exocytosis, improved apoptosis, and improved engraftment in mice by decreasing expression of interleukin (IL)-1ß, IL-6, keratinocyte chemokine, and tumor necrosis factor-α. Finally, transplant efficacy was superior in the Tac+AAGP group and was similar to islets not exposed to Tac, despite receiving continuous treatment for a limited time. Thus, supplementation with AAGP during culture improves islet potency and attenuates long-term Tac-induced graft dysfunction.


Assuntos
Proteínas Anticongelantes/farmacologia , Imunossupressores/toxicidade , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/efeitos dos fármacos , Tacrolimo/toxicidade , Animais , Apoptose/efeitos dos fármacos , Exocitose , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Insulina/metabolismo , Secreção de Insulina , Interleucinas/metabolismo , Ilhotas Pancreáticas/lesões , Ilhotas Pancreáticas/fisiologia , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia
20.
Cell Transplant ; 25(1): 83-95, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25853639

RESUMO

A mathematical nonlinear regression model of several parameters (baseline insulin intake, posttransplant 2-h postprandial blood glucose, and stimulated C-peptide) from type 1 diabetics with HbA1c <6.5% who do not require insulin therapy and have no hypoglycemic instances was developed for accurately predicting supplemental insulin requirements in the posttransplant period. An insulin deficit threshold of 0.018 U/kg/day was defined as the average first-year calculated insulin deficit (CID), above which HbA1c rose to >6.5% during year 2 of the posttransplant period. When insulin-untreated subjects were divided into two groups based on whether the average CID was smaller (group I) or greater (group II) than the insulin deficit threshold, HbA1c was found to be similar in the two groups in year 1, but increased significantly in group II to above 6.5% (with mean glucose of 121.9 mg/dl) but remained below 6.5% in group I subjects (with mean glucose of 108.7 mg/dl) in year 2 of the follow-up period. The greater insulin deficit in group II was also associated with a higher susceptibility to hyperglycemia during periods of low serum Rapamune and Prograf levels (combined levels below 11.2 and 4.7 ng/ml, respectively). Although the differences between predicted insulin requirement (PIR) and actual empirical insulin intake in the insulin-treated subjects were generally small, they were nonetheless sufficient to identify over- and underinsulinization at each follow-up visit for all subjects (n = 14 subjects, 135 observations). The newly developed model can effectively identify underinsulinized islet transplant recipients at risk for graft dysfunction due to inadequate supplemental insulin intake or those potentially susceptible to graft function loss due to inadequate immunosuppression. While less common following islet cell therapy, the model can also identify overinsulinized subjects who may be at risk for hypoglycemia.


Assuntos
Insulina/uso terapêutico , Transplante das Ilhotas Pancreáticas/fisiologia , Feminino , Hemoglobinas Glicadas/metabolismo , Humanos , Terapia de Imunossupressão , Masculino , Pessoa de Meia-Idade , Dinâmica não Linear , Reprodutibilidade dos Testes , Sirolimo/sangue , Tacrolimo/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...