Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 12(3): 611-623, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30773486

RESUMO

The transplantation of human embryonic stem cell (hESC)-derived insulin-producing ß cells for the treatment of diabetes is finally approaching the clinical stage. However, even with state-of-the-art differentiation protocols, a significant percentage of undefined non-endocrine cell types are still generated. Most importantly, there is the potential for carry-over of non-differentiated cell types that may produce teratomas. We sought to modify hESCs so that their differentiated progeny could be selectively devoid of tumorigenic cells and enriched for cells of the desired phenotype (in this case, ß cells). Here we report the generation of a modified hESC line harboring two suicide gene cassettes, whose expression results in cell death in the presence of specific pro-drugs. We show the efficacy of this system at enriching for ß cells and eliminating tumorigenic ones both in vitro and in vivo. Our approach is innovative inasmuch as it allows for the preservation of the desired cells while eliminating those with the potential to develop teratomas.


Assuntos
Carcinogênese/patologia , Células-Tronco Embrionárias Humanas/patologia , Células Secretoras de Insulina/patologia , Animais , Carcinogênese/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Teratoma/genética , Teratoma/patologia
2.
PLoS One ; 11(6): e0157245, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27285580

RESUMO

BACKGROUND: Allogeneic human islet transplantation is an effective therapy for the treatment of patients with Type 1 Diabetes (T1D). The low number of islet transplants performed worldwide and the different transplantation protocols used limit the identification of the most effective therapeutic options to improve the efficacy of this approach. METHODS: We present a retrospective analysis on the data collected from 44 patients with T1D who underwent islet transplantation at our institute between 2000 and 2007. Several variables were included: recipient demographics and immunological characteristics, donor and transplant characteristics, induction protocols, and additional medical treatment received. Immunosuppression was induced with anti-CD25 (Daclizumab), alone or in association with anti-tumor necrosis factor alpha (TNF-α) treatments (Etanercept or Infliximab), or with anti-CD52 (Alemtuzumab) in association with anti-TNF-α treatments (Etanercept or Infliximab). Subsets of patients were treated with Filgrastim for moderate/severe neutropenia and/or Exenatide for post prandial hyperglycemia. RESULTS: The analysis performed indicates a negative association between graft survival (c-peptide level ≥ 0.3 ng/ml) and islet infusion volume, with the caveat that, the progressive reduction of infusion volumes over the years has been paralleled by improved immunosuppressive protocols. A positive association is instead suggested between graft survival and administration of Exenatide and Filgrastim, alone or in combination. CONCLUSION: This retrospective analysis may be of assistance to further improve long-term outcomes of protocols for transplant of islets and other organs.


Assuntos
Filgrastim/uso terapêutico , Sobrevivência de Enxerto/efeitos dos fármacos , Fármacos Hematológicos/uso terapêutico , Hipoglicemiantes/uso terapêutico , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/efeitos dos fármacos , Peptídeos/uso terapêutico , Peçonhas/uso terapêutico , Adulto , Idoso , Anticorpos Monoclonais Humanizados/uso terapêutico , Daclizumabe , Exenatida , Humanos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/etiologia , Imunoglobulina G/uso terapêutico , Imunossupressores/uso terapêutico , Ilhotas Pancreáticas/fisiologia , Transplante das Ilhotas Pancreáticas/efeitos adversos , Pessoa de Meia-Idade , Neutropenia/tratamento farmacológico , Neutropenia/etiologia , Estudos Retrospectivos , Transplante Homólogo
3.
J Diabetes Res ; 2015: 737586, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26347378

RESUMO

The nonobese diabetic (NOD) mouse represents a well-established experimental model analogous to human type 1 diabetes mellitus (T1D) as it is characterized by progressive autoimmune destruction of pancreatic ß-cells. Experiments were designed to investigate the impact of moderate-intensity training on T1D immunomodulation and inflammation. Under a chronic exercise regime, NOD mice were trained on a treadmill for 12 weeks (12 m/min for 30 min, 5 d/wk) while age-matched, control animals were left untrained. Prior to and upon completion of the training period, fed plasma glucose and immunological soluble factors were monitored. Both groups showed deteriorated glycemic profiles throughout the study although trained mice tended to be more compensated than controls after 10 weeks of training. An exercise-induced weight loss was detected in the trained mice with respect to the controls from week 6. After 12 weeks, IL-6 and MIP-1ß were decreased in the trained animals compared to their baseline values and versus controls, although not significantly. Morphometric analysis of pancreata revealed the presence of larger infiltrates along with decreased α-cells areas in the control mice compared to trained mice. Exercise may exert positive immunomodulation of systemic functions with respect to both T1D and inflammation, but only in a stringent therapeutic window.


Assuntos
Diabetes Mellitus Tipo 1/sangue , Hiperglicemia/patologia , Inflamação/patologia , Condicionamento Físico Animal , Animais , Glicemia/análise , Peso Corporal , Quimiocina CCL4/sangue , Diabetes Mellitus Tipo 1/genética , Feminino , Hiperglicemia/terapia , Imuno-Histoquímica , Inflamação/terapia , Células Secretoras de Insulina/citologia , Interleucina-6/sangue , Camundongos , Camundongos Endogâmicos NOD , Pâncreas/metabolismo
4.
Biotechnol Bioeng ; 112(9): 1916-26, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25786390

RESUMO

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.


Assuntos
Fibrina/química , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Becaplermina , Proliferação de Células/efeitos dos fármacos , Humanos , Hidrogéis/química , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-sis/química , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/farmacologia , Fator A de Crescimento do Endotélio Vascular/química
5.
Eur J Immunol ; 44(11): 3307-19, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25113564

RESUMO

By restraining T-cell activation and promoting Treg-cell expansion, myeloid-derived suppressor cells (MDSCs) and tolerogenic DCs can control self-reactive and antigraft effector T cells in autoimmunity and transplantation. Their therapeutic use and characterization, however, is limited by their scarce availability in the peripheral blood of tumor-free donors. In the present study, we describe and characterize a novel population of human myeloid suppressor cells, named fibrocytic MDSC, which are differentiated from umbilical cord blood precursors by 4-day culture with FDA-approved cytokines (recombinant human-GM-CSF and recombinant human-G-CSF). This MDSC subset, characterized by the expression of MDSC-, DC-, and fibrocyte-associated markers, promotes Treg-cell expansion and induces normoglycemia in a xenogeneic mouse model of Type 1 diabetes. In order to exert their protolerogenic function, fibrocytic MDSCs require direct contact with activated T cells, which leads to the production and secretion of IDO. This new myeloid subset may have an important role in the in vitro and in vivo production of Treg cells for the treatment of autoimmune diseases, and in either the prevention or control of allograft rejection.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Ativação Linfocitária/imunologia , Células Mieloides/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular , Proliferação de Células , Diabetes Mellitus Tipo 1/imunologia , Feminino , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Rejeição de Enxerto/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células HEK293 , Humanos , Tolerância Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Mieloides/citologia , Proteínas Recombinantes/farmacologia , Linfócitos T Reguladores/transplante
6.
Pancreas ; 43(8): 1317-22, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25036907

RESUMO

OBJECTIVES: The effects of glucocorticoid during culture on human islet cells have been controversial. Exendin-4 (EX) enhances the insulin secretion and significantly improves clinical outcomes in islet cell transplantation. In this study, we examined the effects of glucocorticoids and EX on human islet cells during pretransplant culture. METHODS: Methylprednisolone (MP) and/or EX were added to the standard culture medium for clinical islet cell transplantation. Islets were cultured for 24 hours with 3 different conditions (control, no additives; MP alone; and MP + EX). ß-Cell fractional viability, cellular composition, multiple cytokine/chemokine production, multiple phosphorylation proteins, and glucose-induced insulin secretion were evaluated. RESULTS: Viable ß-cell survival in MP and MP + EX group was significantly higher than in the control group. Exendin-4 prevented MP-induced reduction of insulin secretion. Methylprednisolone supplementation to the culture medium decreased cytokine and chemokine production. Moreover, extracellular signal-regulated kinase 1/2 phosphorylation was significantly increased by MP and MP + EX. CONCLUSIONS: Glucocorticoid supplementation into culture media significantly decreased the cytokine/chemokine production and increased the extracellular signal-regulated kinase 1/2 phosphorylation, resulting in the improvement of human ß-cell survival. In addition, EX maintained the insulin secretion suppressed by MP. The supplementation of MP and EX together could be a useful strategy to create suitable human islets for transplantation.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Sobrevivência Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Exenatida , Glucose/farmacologia , Humanos , Mediadores da Inflamação , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metilprednisolona/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Peptídeos/farmacologia , Fosfoproteínas/análise , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Peçonhas/farmacologia
7.
Genom Data ; 2: 389-92, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26484135

RESUMO

Myeloid-derived suppressor cells (MDSCs) have been shown to control self-reactive and anti-graft effector T-cells in autoimmunity and transplantation, but their therapeutic use is limited by their scarce availability in the peripheral blood of tumor-free donors. We isolated and characterized a novel population of myeloid suppressor cells, named fibrocytic MDSC (f-MDSC), which are differentiated from umbilical cord blood (UCB) precursors (Zoso et al., 2014). This MDSC subset promotes regulatory T-cell expansion and induces normoglycemia in a xenogeneic model of type 1 diabetes. Here we describe in details the experimental design and the bioinformatics analyses of the gene expression dataset used to investigate the molecular mechanisms at the base of MDSC tolerogenic and suppressive properties. We also provide an R code to easily access the data and perform the quality controls and basic analyses relevant to this dataset. Raw and pre-processed data are available at Gene Expression Omnibus under accession GSE52376.

8.
Stem Cells Transl Med ; 3(3): 277-89, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24375542

RESUMO

The possibility of using human embryonic stem (hES) cell-derived ß cells as an alternative to cadaveric islets for the treatment of type 1 diabetes is now widely acknowledged. However, current differentiation methods consistently fail to generate meaningful numbers of mature, functional ß cells. In order to address this issue, we set out to explore the role of oxygen modulation in the maturation of pancreatic progenitor (PP) cells differentiated from hES cells. We have previously determined that oxygenation is a powerful driver of murine PP differentiation along the endocrine lineage of the pancreas. We hypothesized that targeting physiological oxygen partial pressure (pO2) levels seen in mature islets would help the differentiation of PP cells along the ß-cell lineage. This hypothesis was tested both in vivo (by exposing PP-transplanted immunodeficient mice to a daily hyperbaric oxygen regimen) and in vitro (by allowing PP cells to mature in a perfluorocarbon-based culture device designed to carefully adjust pO2 to a desired range). Our results show that oxygen modulation does indeed contribute to enhanced maturation of PP cells, as evidenced by improved engraftment, segregation of α and ß cells, body weight maintenance, and rate of diabetes reversal in vivo, and by elevated expression of pancreatic endocrine makers, ß-cell differentiation yield, and insulin production in vitro. Our studies confirm the importance of oxygen modulation as a key variable to consider in the design of ß-cell differentiation protocols and open the door to future strategies for the transplantation of fully mature ß cells.


Assuntos
Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/terapia , Células-Tronco Embrionárias/efeitos dos fármacos , Hospedeiro Imunocomprometido , Células Secretoras de Insulina/efeitos dos fármacos , Oxigênio/farmacologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Diabetes Mellitus Experimental/patologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Fluorocarbonos/farmacologia , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Glucagon/imunologia , Glucose/metabolismo , Glucose/farmacologia , Sobrevivência de Enxerto , Humanos , Insulina/biossíntese , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/transplante , Camundongos , Camundongos Nus , Consumo de Oxigênio/fisiologia
9.
Stem Cells ; 31(10): 2047-60, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23873634

RESUMO

Regenerative medicine is transitioning into clinical programs using stem/progenitor cell therapies for repair of damaged organs. We summarize those for liver and pancreas, organs that share endodermal stem cell populations, biliary tree stem cells (hBTSCs), located in peribiliary glands. They are precursors to hepatic stem/progenitors in canals of Hering and to committed progenitors in pancreatic duct glands. They give rise to maturational lineages along a radial axis within bile duct walls and a proximal-to-distal axis starting at the duodenum and ending with mature cells in the liver or pancreas. Clinical trials have been ongoing for years assessing effects of determined stem cells (fetal-liver-derived hepatic stem/progenitors) transplanted into the hepatic artery of patients with various liver diseases. Immunosuppression was not required. Control subjects, those given standard of care for a given condition, all died within a year or deteriorated in their liver functions. Subjects transplanted with 100-150 million hepatic stem/progenitor cells had improved liver functions and survival extending for several years. Full evaluations of safety and efficacy of transplants are still in progress. Determined stem cell therapies for diabetes using hBTSCs remain to be explored but are likely to occur following ongoing preclinical studies. In addition, mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) are being used for patients with chronic liver conditions or with diabetes. MSCs have demonstrated significant effects through paracrine signaling of trophic and immunomodulatory factors, and there is limited evidence for inefficient lineage restriction into mature parenchymal or islet cells. HSCs' effects are primarily via modulation of immune mechanisms.


Assuntos
Hepatite/terapia , Transplante de Células-Tronco Mesenquimais , Pancreatite/terapia , Diferenciação Celular , Linhagem da Célula , Hepatite/imunologia , Humanos , Fígado/embriologia , Fígado/imunologia , Fígado/patologia , Células-Tronco Mesenquimais/fisiologia , Pâncreas/embriologia , Pâncreas/imunologia , Pâncreas/patologia , Pancreatite/imunologia , Nicho de Células-Tronco
10.
Stem Cells ; 31(9): 1966-79, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23847135

RESUMO

Peribiliary glands (PBGs) in bile duct walls, and pancreatic duct glands (PDGs) associated with pancreatic ducts, in humans of all ages, contain a continuous, ramifying network of cells in overlapping maturational lineages. We show that proximal (PBGs)-to-distal (PDGs) maturational lineages start near the duodenum with cells expressing markers of pluripotency (NANOG, OCT4, and SOX2), proliferation (Ki67), self-replication (SALL4), and early hepato-pancreatic commitment (SOX9, SOX17, PDX1, and LGR5), transitioning to PDG cells with no expression of pluripotency or self-replication markers, maintenance of pancreatic genes (PDX1), and expression of markers of pancreatic endocrine maturation (NGN3, MUC6, and insulin). Radial-axis lineages start in PBGs near the ducts' fibromuscular layers with stem cells and end at the ducts' lumens with cells devoid of stem cell traits and positive for pancreatic endocrine genes. Biliary tree-derived cells behaved as stem cells in culture under expansion conditions, culture plastic and serum-free Kubota's Medium, proliferating for months as undifferentiated cells, whereas pancreas-derived cells underwent only approximately 8-10 divisions, then partially differentiated towards an islet fate. Biliary tree-derived cells proved precursors of pancreas' committed progenitors. Both could be driven by three-dimensional conditions, islet-derived matrix components and a serum-free, hormonally defined medium for an islet fate (HDM-P), to form spheroids with ultrastructural, electrophysiological and functional characteristics of neoislets, including glucose regulatability. Implantation of these neoislets into epididymal fat pads of immunocompromised mice, chemically rendered diabetic, resulted in secretion of human C-peptide, regulatable by glucose, and able to alleviate hyperglycemia in hosts. The biliary tree-derived stem cells and their connections to pancreatic committed progenitors constitute a biological framework for life-long pancreatic organogenesis.


Assuntos
Sistema Biliar/citologia , Linhagem da Célula , Organogênese , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Células-Tronco/citologia , Adulto , Animais , Antígenos de Neoplasias/metabolismo , Biomarcadores/metabolismo , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Experimental/terapia , Fenômenos Eletrofisiológicos , Molécula de Adesão da Célula Epitelial , Regulação da Expressão Gênica , Humanos , Hiperglicemia/terapia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/ultraestrutura , Transplante das Ilhotas Pancreáticas , Camundongos , Organogênese/genética , Ductos Pancreáticos/citologia , Fenótipo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Esferoides Celulares/ultraestrutura , Nicho de Células-Tronco/genética , Células-Tronco/metabolismo
11.
Cell Transplant ; 22(9): 1723-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23068091

RESUMO

Conventional culture vessels are not designed for physiological oxygen (O2) delivery. Both hyperoxia and hypoxia-commonly observed when culturing cells in regular plasticware-have been linked to reduced cellular function and death. Pancreatic islets, used for the clinical treatment of diabetes, are especially sensitive to sub- and supraphysiological O2 concentrations. A result of current culture standards is that a high percentage of islet preparations are never transplanted because of cell death and loss of function in the 24-48 h postisolation. Here, we describe a new culture system designed to provide quasiphysiological oxygenation to islets in culture. The use of dishes where islets rest atop a perfluorocarbon (PFC)-based membrane, coupled with a careful adjustment of environmental O2 concentration to target the islet physiological pO2 range, resulted in dramatic gains in viability and function. These observations underline the importance of approximating culture conditions as closely as possible to those of the native microenvironment, and fill a widely acknowledged gap in our ability to preserve islet functionality in vitro. As stem cell-derived insulin-producing cells are likely to suffer from the same limitations as those observed in real islets, our findings are especially timely in the context of current efforts to define renewable sources for transplantation.


Assuntos
Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Fluorocarbonos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Oxigênio/administração & dosagem , Oxigênio/metabolismo , Animais , Morte Celular/fisiologia , Sobrevivência Celular/fisiologia , Feminino , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Nus
12.
PLoS One ; 7(12): e50265, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23227162

RESUMO

Two major hurdles need to be surmounted for cell therapy for diabetes: (i) allo-immune rejection of grafted pancreatic islets, or stem/precursor cell-derived insulin-secreting cells; and (ii) continuing auto-immunity against the diabetogenic endogenous target antigen. Nanotherapeutics offer a novel approach to overcome these problems and here we ask if creation of "stealth" islets encapsulated within a thin cage of pegylated material of 100-200 nanometers thick provides a viable option for islet transplantation. The aims of this study were to test islet viability and functionality following encapsulation within the pegylated cage, and functional efficacy in vivo in terms of graft-derived control of normoglycemia in diabetic mice. We first demonstrated that pegylation of the islet surface, plus or minus nanoparticles, improved long-term islet viability in vitro compared to non-pegylated (naked) control islets. Moreover, pegylation of the islets with nanoparticles was compatible with glucose-stimulated insulin secretion and insulin biogenesis. We next looked for functionality of the created "stealth" DBA/2 (H-2(d)) islets in vivo by comparing glycemic profiles across 4 groups of streptozotozin-induced diabetic C57BL/6 (H-2(b)) recipients of (i) naked islets; (ii) pegylated islets; (iii) pegylated islets with nanoparticles (empty); and (iv) pegylated islets with nanoparticles loaded with a cargo of leukemia inhibitory factor (LIF), a factor both promotes adaptive immune tolerance and regulates pancreatic ß cell mass. Without any other treatment, normoglycemia was lost after 17 d (+/-7.5 d) in control group. In striking contrast, recipients in groups (ii), (iii), and (iv) showed long-term (>100 d) normoglycemia involving 30%; 43%, and 57% of the recipients in each respective group. In conclusion, construction of "stealth" islets by pegylation-based nanotherapeutics not only supports islet structure and function, but also effectively isolates the islets from immune-mediated destruction. The added value of nanoparticles to deliver immune modulators plus growth factors such as LIF expands the potential of this novel therapeutic approach to cell therapy for diabetes.


Assuntos
Glicemia/metabolismo , Complexo Principal de Histocompatibilidade/imunologia , Nanomedicina , Transplante de Pâncreas , Pâncreas/imunologia , Polietilenoglicóis/administração & dosagem , Animais , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Microscopia Eletrônica de Varredura
13.
Regen Med ; 7(6 Suppl): 41-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23210810

RESUMO

Diabetes mellitus represents a global epidemic affecting over 350 million patients worldwide and projected by the WHO to surpass the 500 million patient mark within the next two decades. Besides Type 1 and Type 2 diabetes mellitus, the study of the endocrine compartment of the pancreas is of great translational interest, as strategies aimed at restoring its mass could become therapies for glycemic dysregulation, drug-related diabetes following diabetogenic therapies, or hyperglycemic disturbances following the treatment of cancer and nesidioblastosis. Such strategies generally fall under one of the 'three Rs': replacement (islet transplantation and stem cell differentiation); reprogramming (e.g., from the exocrine compartment of the pancreas); and regeneration (replication and induction of endogenous stem cells). As the latter has been extensively reviewed in recent months by us and others, this article focuses on emerging reprogramming and replacement approaches.


Assuntos
Reprogramação Celular/genética , Diabetes Mellitus/terapia , Medicina Regenerativa/métodos , Transplante de Células-Tronco , Animais , Humanos
14.
Stem Cells Transl Med ; 1(1): 59-63, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23197641

RESUMO

Mesenchymal stem cells (MSCs) have already made their mark in the young field of regenerative medicine. Easily derived from many adult tissues, their therapeutic worth has already been validated for a number of conditions. Unlike embryonic stem cells, neither their procurement nor their use is deemed controversial. Here we review the potential use of MSCs for the treatment of type 1 diabetes mellitus, a devastating chronic disease in which the insulin-producing cells of the pancreas (the ß-cells) are the target of an autoimmune process. It has been hypothesized that stem cell-derived ß-cells may be used to replenish the islet mass in diabetic patients, making islet transplantation (a form of cell therapy that has already proven effective at clinically restoring normoglycemia) available to millions of prospective patients. Here we review the most current advances in the design and application of protocols for the differentiation of transplantable ß-cells, with a special emphasis in analyzing MSC potency according to their tissue of origin. Although no single method appears to be ripe enough for clinical trials yet, recent progress in reprogramming (a biotechnological breakthrough that relativizes the thus far insurmountable barriers between embryonal germ layers) bodes well for the rise of MSCs as a potential weapon of choice to develop personalized therapies for type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas , Transplante de Células-Tronco Mesenquimais , Medicina Regenerativa , Animais , Diferenciação Celular , Linhagem da Célula , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Regeneração
15.
Colloids Surf B Biointerfaces ; 98: 26-35, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22652356

RESUMO

Nano-scale emulsification has long been utilized by the food and cosmetics industry to maximize material delivery through increased surface area to volume ratios. More recently, these methods have been employed in the area of biomedical research to enhance and control the delivery of desired agents, as in perfluorocarbon emulsions for oxygen delivery. In this work, we evaluate critical factors for the optimization of PFC emulsions for use in cell-based applications. Cytotoxicity screening revealed minimal cytotoxicity of components, with the exception of one perfluorocarbon utilized for emulsion manufacture, perfluorooctylbromide (PFOB), and specific w% limitations of PEG-based surfactants utilized. We optimized the manufacture of stable nano-scale emulsions via evaluation of: component materials, emulsification time and pressure, and resulting particle size and temporal stability. The initial emulsion size was greatly dependent upon the emulsion surfactant tested, with pluronics providing the smallest size. Temporal stability of the nano-scale emulsions was directly related to the perfluorocarbon utilized, with perfluorotributylamine, FC-43, providing a highly stable emulsion, while perfluorodecalin, PFD, coalesced over time. The oxygen mass transfer, or diffusive permeability, of the resulting emulsions was also characterized. Our studies found particle size to be the critical factor affecting oxygen mass transfer, as increased micelle size resulted in reduced oxygen diffusion. Overall, this work demonstrates the importance of accurate characterization of emulsification parameters in order to generate stable, reproducible emulsions with the desired bio-delivery properties.


Assuntos
Emulsões/efeitos adversos , Emulsões/química , Oxigênio/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fluorocarbonos/química , Hidrocarbonetos Bromados , Camundongos , Tamanho da Partícula
16.
Expert Opin Biol Ther ; 12(6): 731-41, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22506840

RESUMO

INTRODUCTION: The study of the endocrine compartment of the pancreas (the islets of Langerhans) is of great translational interest, as strategies aimed at restoring its mass could become therapies for glycemic dysregulation in type 1 and 2 diabetes mellitus, drug-related diabetes following diabetogenic therapies or hyperglycemic disturbances following the treatment of cancer and nesidioblastosis. Such strategies generally fall under one of the 'three Rs,' namely, replacement (islet transplantation and stem cell differentiation), reprogramming (chiefly from the exocrine compartment of the pancreas) and regeneration (replication and induction of endogenous stem cells). AREAS COVERED: This expert opinion focuses on the latter, as islets are known to regenerate under specific circumstances of physiological (e.g., pregnancy), pathological (e.g., obesity, hyperglycemia, mutations in the glucose-sensing pathway) or experimental (e.g., partial pancreatectomy, cellophane wrapping, partial duct ligation) nature. This review presents the different models of pancreatic regeneration, which encompass the replication of existing beta-cells, reversible epithelial-to-mesenchymal transition and the reactivation of resident stem cells. EXPERT OPINION: Rather than a set mechanism, the pancreas appears to possess a wide range of facultative regeneration pathways. These are discussed in the context of the development of potential strategies aimed at restoring beta-cell function in insulin-dependent diabetes.


Assuntos
Diabetes Mellitus/terapia , Células Secretoras de Insulina/patologia , Regeneração , Medicina Regenerativa/métodos , Células-Tronco/patologia , Engenharia Tecidual , Animais , Diferenciação Celular , Proliferação de Células , Diabetes Mellitus/sangue , Diabetes Mellitus/patologia , Transição Epitelial-Mesenquimal , Humanos , Células Secretoras de Insulina/metabolismo , Células-Tronco/metabolismo
17.
Cancer Res ; 72(6): 1373-83, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22282665

RESUMO

In addition to promoting tumor progression and metastasis by enhancing angiogenesis and invasion, myeloid-derived suppressor cells (MDSC) and tumor-associated macrophage (TAM) also inhibit antitumor T-cell functions and limit the efficacy of immunotherapeutic interventions. Despite the importance of these leukocyte populations, a simple method for their specific depletion has not been developed. In this study, we generated an RNA aptamer that blocks the murine or human IL-4 receptor-α (IL4Rα or CD124) that is critical for MDSC suppression function. In tumor-bearing mice, this anti-IL4Rα aptamer preferentially targeted MDSCs and TAM and unexpectedly promoted their elimination, an effect that was associated with an increased number of tumor-infiltrating T cells and a reduction in tumor growth. Mechanistic investigations of aptamer-triggered apoptosis in MDSCs confirmed the importance of IL4Ra-STAT6 pathway activation in MDSC survival. Our findings define a straightforward strategy to deplete MDSCs and TAMs in vivo, and they strengthen the concept that IL4Rα signaling is pivotal for MDSC survival. More broadly, these findings suggest therapeutic strategies based on IL4Rα signaling blockades to arrest an important cellular mechanism of tumoral immune escape mediated by MDSCs and TAM in cancer.


Assuntos
Apoptose/efeitos dos fármacos , Aptâmeros de Nucleotídeos/farmacologia , Carcinoma/tratamento farmacológico , Progressão da Doença , Neoplasias Mamárias Experimentais/tratamento farmacológico , Receptores de Superfície Celular/antagonistas & inibidores , Animais , Apoptose/imunologia , Carcinoma/imunologia , Linhagem Celular Tumoral , Humanos , Subunidade alfa de Receptor de Interleucina-4/antagonistas & inibidores , Subunidade alfa de Receptor de Interleucina-4/imunologia , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Receptores de Superfície Celular/imunologia , Fator de Transcrição STAT6/metabolismo , Evasão Tumoral/efeitos dos fármacos , Evasão Tumoral/imunologia
18.
Cell Transplant ; 21(6): 1321-39, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22195604

RESUMO

We sought to assess the potential of human cord blood-derived mesenchymal stem cells (CB-MSCs) to derive insulin-producing, glucose-responsive cells. We show here that differentiation protocols based on stepwise culture conditions initially described for human embryonic stem cells (hESCs) lead to differentiation of cord blood-derived precursors towards a pancreatic endocrine phenotype, as assessed by marker expression and in vitro glucose-regulated insulin secretion. Transplantation of these cells in immune-deficient animals shows human C-peptide production in response to a glucose challenge. These data suggest that human cord blood may be a promising source for regenerative medicine approaches for the treatment of diabetes mellitus.


Assuntos
Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Células-Tronco Mesenquimais/citologia , Cordão Umbilical/citologia , Animais , Biomarcadores/metabolismo , Peptídeo C/metabolismo , Diferenciação Celular , Células Cultivadas , Células Endócrinas/citologia , Células Endócrinas/metabolismo , Humanos , Imunofenotipagem , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/transplante , Fatores de Transcrição Maf/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Fatores de Transcrição/metabolismo , Transplante Heterólogo
19.
Cancer Res ; 71(24): 7452-62, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21987727

RESUMO

DNA-based vaccines hold promise to outperform conventional antigen-based vaccines by virtue of many unique features. However, DNA vaccines have thus far fallen short of expectations, due in part to poor targeting of professional antigen-presenting cells (APC) and low immunogenicity. In this study, we describe a new platform for effective and selective delivery of DNA to APCs in vivo that offers intrinsic immune-enhancing characteristics. This platform is based on conjugation of fifth generation polyamidoamine (G5-PAMAM) dendrimers, a DNA-loading surface, with MHC class II-targeting peptides that can selectively deliver these dendrimers to APCs under conditions that enhance their immune stimulatory potency. DNA conjugated with this platform efficiently transfected murine and human APCs in vitro. Subcutaneous administration of DNA-peptide-dendrimer complexes in vivo preferentially transfected dendritic cells (DC) in the draining lymph nodes, promoted generation of high affinity T cells, and elicited rejection of established tumors. Taken together, our findings show how PAMAM dendrimer complexes can be used for high transfection efficiency and effective targeting of APCs in vivo, conferring properties essential to generate effective DNA vaccines.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Dendrímeros/química , Peptídeos/imunologia , Vacinas de DNA/imunologia , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , DNA/genética , DNA/imunologia , DNA/metabolismo , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica/imunologia , Eletricidade Estática , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética
20.
PLoS One ; 6(8): e22364, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21857924

RESUMO

Alongside Pdx1 and Beta2/NeuroD, the transcription factor MafA has been shown to be instrumental in the maintenance of the beta cell phenotype. Indeed, a combination of MafA, Pdx1 and Ngn3 (an upstream regulator of Beta2/NeuroD) was recently reported to lead to the effective reprogramming of acinar cells into insulin-producing beta cells. These experiments set the stage for the development of new strategies to address the impairment of glycemic control in diabetic patients. However, the clinical applicability of reprogramming in this context is deemed to be poor due to the need to use viral vehicles for the delivery of the above factors. Here we describe a recombinant transducible version of the MafA protein (TAT-MafA) that penetrates across cell membranes with an efficiency of 100% and binds to the insulin promoter in vitro. When injected in utero into living mouse embryos, TAT-MafA significantly up-regulates target genes and induces enhanced insulin production as well as cytoarchitectural changes consistent with faster islet maturation. As the latest addition to our armamentarium of transducible proteins (which already includes Pdx1 and Ngn3), the purification and characterization of a functional TAT-MafA protein opens the door to prospective therapeutic uses that circumvent the use of viral delivery. To our knowledge, this is also the first report on the use of protein transduction in utero.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Fatores de Transcrição Maf Maior/metabolismo , Pâncreas/metabolismo , Útero/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Expressão Gênica , Produtos do Gene tat/genética , Produtos do Gene tat/metabolismo , Insulina/genética , Ilhotas Pancreáticas/citologia , Fatores de Transcrição Maf Maior/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/embriologia , Gravidez , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA