Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cell Rev Rep ; 19(3): 601-624, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36434300

RESUMO

Type 1 diabetes (T1D) is a chronic, lifelong metabolic disease. It is characterised by the autoimmune-mediated loss of insulin-producing pancreatic ß cells in the islets of Langerhans (ß-islets), resulting in disrupted glucose homeostasis. Administration of exogenous insulin is the most common management method for T1D, but this requires lifelong reliance on insulin injections and invasive blood glucose monitoring. Replacement therapies with beta cells are being developed as an advanced curative treatment for T1D. Unfortunately, this approach is limited by the lack of donated pancreatic tissue, the difficulties in beta cell isolation and viability maintenance, the longevity of the transplanted cells in vivo, and consequently high costs. Emerging approaches to address these limitations are under intensive investigations, including the production of insulin-producing beta cells from various stem cells, and the development of bioengineered devices including nanotechnologies for improving islet transplantation efficacy without the need for recipients taking toxic anti-rejection drugs. These emerging approaches present promising prospects, while the challenges with the new techniques need to be tackled for ultimately clinical treatment of T1D. This review discussed the benefits and limitations of the cell-based therapies for beta cell replacement as potential curative treatment for T1D, and the applications of bioengineered devices including nanotechnology to overcome the challenges associated with beta cell transplantation.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Humanos , Diabetes Mellitus Tipo 1/terapia , Automonitorização da Glicemia , Glicemia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo
2.
Pharmaceutics ; 15(1)2022 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-36678718

RESUMO

Drug-delivery vehicles have garnered immense interest in recent years due to unparalleled progress made in material science and nanomedicine. However, the development of stimuli-responsive devices with controllable drug-release systems (DRSs) is still in its nascent stage. In this paper, we designed a two-way controlled drug-release system that can be promoted and prolonged, using the external stimulation of near-infrared light (NIR) and protein coating. A hierarchical nanostructure was fabricated using upconversion nanoparticles (UCNPs)-mesoporous silica as the core-shell structure with protein lysozyme coating. The mesoporous silica shell provides abundant pores for the loading of drug molecules and a specific type of photosensitive molecules. The morphology and the physical properties of the nanostructures were thoroughly characterized. The results exhibited the uniform core-shell nanostructures of ~four UCNPs encapsulated in one mesoporous silica nanoparticle. The core-shell nanoparticles were in the spherical shape with an average size of 200 nm, average surface area of 446.54 m2/g, and pore size of 4.6 nm. Using doxorubicin (DOX), a chemotherapy agent as the drug model, we demonstrated that a novel DRS with capacity of smart modulation to promote or inhibit the drug release under NIR light and protein coating, respectively. Further, we demonstrated the therapeutic effect of the designed DRSs using breast cancer cells. The reported novel controlled DRS with dual functionality could have a promising potential for chemotherapy treatment of solid cancers.

3.
Methods Mol Biol ; 2029: 93-102, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31273736

RESUMO

Type 1 diabetes, characterized by autoimmune destruction of pancreatic beta cells, affects 41 million people worldwide. Beta cell replacement therapies have immense potential as a treatment option because pancreatic progenitors derived from human pluripotent stem cells can provide a near limitless supply of transplantable tissue. The key limitation of this approach is the need for lifelong use of immunosuppressive drugs that have undesirable side effects. Microencapsulation is an option for providing protection for transplanted cells from mechanical stress and immune attack. Traditionally, pluripotent cells are differentiated on a 2D matrix before being transferred into an immunoisolation device. Here, we describe a method of differentiating pluripotent stem cells into pancreatic progenitors while the cells are encapsulated in alginate microspheres. This method provides several advantages including the need for fewer steps compared to the traditional approach, protection against mechanical/physical damage during differentiation in bioreactors, and immune-protection of cells once transplanted into the host.


Assuntos
Pâncreas/citologia , Células-Tronco/citologia , Alginatos/química , Diferenciação Celular/fisiologia , Diabetes Mellitus Tipo 1/terapia , Células-Tronco Embrionárias/citologia , Humanos , Células Secretoras de Insulina/citologia , Microesferas , Células-Tronco Pluripotentes/fisiologia
4.
Sci Rep ; 7(1): 10059, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855611

RESUMO

Pericapsular fibrotic overgrowth (PFO) is associated with poor survival of encapsulated islets. A strategy to combat PFO is the use of mesenchymal stem cells (MSC). MSC have anti-inflammatory properties and their potential can be enhanced by stimulation with proinflammatory cytokines. This study investigated whether co-encapsulation or co-transplantation of MSC with encapsulated islets would reduce PFO and improve graft survival. Stimulating MSC with a cytokine cocktail of IFN-γ and TNF-α enhanced their immunosuppressive potential by increasing nitric oxide production and secreting higher levels of immunomodulatory cytokines. In vitro, co-encapsulation with MSC did not affect islet viability but significantly enhanced glucose-induced insulin secretion. In vivo, normoglycemia was achieved in 100% mice receiving islets co-encapsulated with stimulated MSC as opposed to 71.4% receiving unstimulated MSC and only 9.1% receiving encapsulated islets alone. Microcapsules retrieved from both unstimulated and stimulated MSC groups had significantly less PFO with improved islet viability and function compared to encapsulated islets alone. Levels of peritoneal immunomodulatory cytokines IL-4, IL-6, IL-10 and G-CSF were significantly higher in MSC co-encapsulated groups. Similar results were obtained when encapsulated islets and MSC were co-transplanted. In summary, co-encapsulation or co-transplantation of MSC with encapsulated islets reduced PFO and improved the functional outcome of allotransplants.


Assuntos
Composição de Medicamentos/métodos , Sobrevivência de Enxerto/fisiologia , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Alginatos/química , Animais , Células Imobilizadas/citologia , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/imunologia , Citocinas/genética , Citocinas/imunologia , Feminino , Fibrose/prevenção & controle , Expressão Gênica , Insulina/biossíntese , Interferon gama/farmacologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/imunologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Transplante Homólogo , Fator de Necrose Tumoral alfa/farmacologia
5.
Stem Cells Dev ; 26(20): 1505-1519, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28756741

RESUMO

Mitochondrial deoxyribonucleic acid (mtDNA) copy number is tightly regulated during pluripotency and differentiation. There is increased demand of cellular adenosine triphosphate (ATP) during differentiation for energy-intensive cell types such as hepatocytes and neurons to meet the cell's functional requirements. During hepatocyte differentiation, mtDNA copy number should be synchronously increased to generate sufficient ATP through oxidative phosphorylation. Unlike bone marrow mesenchymal cells, mtDNA copy number failed to increase by 28 days of differentiation of human amniotic epithelial cells (hAEC) into hepatocyte-like cells (HLC) despite their expression of some end-stage hepatic markers. This was due to higher levels of DNA methylation at exon 2 of POLGA, the mtDNA-specific replication factor. Treatment with a DNA demethylation agent, 5-azacytidine, resulted in increased mtDNA copy number, reduced DNA methylation at exon 2 of POLGA, and reduced hepatic gene expression. Depletion of mtDNA followed by subsequent differentiation did not increase mtDNA copy number, but reduced DNA methylation at exon 2 of POLGA and increased expression of hepatic and pluripotency genes. We encapsulated hAEC in barium alginate microcapsules and subsequently differentiated them into HLC. Encapsulation resulted in no net increase of mtDNA copy number but a significant reduction in DNA methylation of POLGA. RNAseq analysis showed that differentiated HLC express hepatocyte-specific genes but also increased expression of inflammatory interferon genes. Differentiation in encapsulated cells showed suppression of inflammatory genes as well as increased expression of genes associated with hepatocyte function pathways and networks. This study demonstrates that an increase in classical hepatic gene expression can be achieved in HLC through encapsulation, although they fail to effectively regulate mtDNA copy number.


Assuntos
Âmnio/citologia , Diferenciação Celular/genética , Variações do Número de Cópias de DNA/genética , DNA Mitocondrial/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Hepatócitos/citologia , Carcinogênese/genética , Sobrevivência Celular , Dosagem de Genes , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Humanos , Mitocôndrias/metabolismo
6.
Rev Diabet Stud ; 14(1): 51-78, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28632821

RESUMO

Transplantation of pancreatic islets encapsulated within immuno-protective microcapsules is a strategy that has the potential to overcome graft rejection without the need for toxic immunosuppressive medication. However, despite promising preclinical studies, clinical trials using encapsulated islets have lacked long-term efficacy, and although generally considered clinically safe, have not been encouraging overall. One of the major factors limiting the long-term function of encapsulated islets is the host's immunological reaction to the transplanted graft which is often manifested as pericapsular fibrotic overgrowth (PFO). PFO forms a barrier on the capsule surface that prevents the ingress of oxygen and nutrients leading to islet cell starvation, hypoxia and death. The mechanism of PFO formation is still not elucidated fully and studies using a pig model have tried to understand the host immune response to empty alginate microcapsules. In this review, the varied strategies to overcome or reduce PFO are discussed, including alginate purification, altering microcapsule geometry, modifying alginate chemical composition, co-encapsulation with immunomodulatory cells, administration of pharmacological agents, and alternative transplantation sites. Nanoencapsulation technologies, such as conformal and layer-by-layer coating technologies, as well as nanofiber, thin-film nanoporous devices, and silicone based NanoGland devices are also addressed. Finally, this review outlines recent progress in imaging technologies to track encapsulated cells, as well as promising perspectives concerning the production of insulin-producing cells from stem cells for encapsulation.


Assuntos
Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/citologia , Técnicas de Cultura de Tecidos/métodos , Animais , Cápsulas , Separação Celular/métodos , Separação Celular/tendências , Diabetes Mellitus Tipo 1/terapia , Composição de Medicamentos/métodos , Sobrevivência de Enxerto , Humanos , Ilhotas Pancreáticas/fisiologia , Transplante das Ilhotas Pancreáticas/tendências
7.
J Diabetes Res ; 2016: 6165893, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27631014

RESUMO

Microencapsulated islets are usually injected free-floating into the peritoneal cavity, so the position of the grafts remains elusive after transplantation. This study aims to assess magnetic resonance imaging (MRI) as a noninvasive means to track microencapsulated insulin producing cells following transplantation. Encapsulated insulin producing cells (MIN6 and human islets) were labelled with magnetic microspheres (MM), assessed for viability and insulin secretion, and imaged in vitro using a clinical grade 3 T MRI and in vivo using both clinical grade 3 T and research grade 11.7 T MRI. Fluorescent imaging demonstrated the uptake of MM by both MIN6 and human islets with no changes in cell morphology and viability. MM labelling did not affect the glucose responsiveness of encapsulated MIN6 and islets in vitro. In vivo encapsulated MM-labelled MIN6 normalized sugar levels when transplanted into diabetic mice. In vitro MRI demonstrated that single microcapsules as well as clusters of encapsulated MM-labelled cells could be visualised clearly in agarose gel phantoms. In vivo encapsulated MM-labelled MIN6 could be visualised more clearly within the peritoneal cavity as discrete hypointensities using the high power 11.7 T but not the clinical grade 3 T MRI. This study demonstrates a method to noninvasively track encapsulated insulin producing cells by MM labelling and MRI.


Assuntos
Células Secretoras de Insulina/transplante , Transplante das Ilhotas Pancreáticas , Imageamento por Ressonância Magnética/métodos , Imãs , Microesferas , Transplantes/diagnóstico por imagem , Animais , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Camundongos
8.
Cell Transplant ; 25(7): 1299-317, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26351069

RESUMO

Pericapsular fibrotic overgrowth (PFO) is a problem that thwarts full implementation of cellular replacement therapies involving encapsulation in an immunoprotective material, such as for the treatment of diabetes. Mesenchymal stem cells (MSCs) have inherent anti-inflammatory properties. We postulated that coencapsulation of MSCs with the target cells would reduce PFO. A hepatoinsulinoma cell line (HUH7) was used to model human target cells and was coencapsulated with either human or mouse MSCs at different ratios in alginate microcapsules. Viability of encapsulated cells was assessed in vitro and xenografted either intraperitoneally or subcutaneously into C57BL/6 mice. Graft retrieval was performed at 3 weeks posttransplantation and assessed for PFO. Coencapsulation of human MSCs (hMSCs) or mouse MSCs (mMSCs) with HUH7 at different ratios did not alter cell viability in vitro. In vivo data from intraperitoneal infusions showed that PFO for HUH7 cells coencapsulated with hMSCs and mMSCs in a ratio of 1:1 was significantly reduced by ∼30% and ∼35%, respectively, compared to HUH7 encapsulated alone. PFO for HUH7 cells was reduced by ∼51% when the ratio of mMSC/HUH7 was increased to 2:1. Implanting the microcapsules subcutaneously rather than intraperitoneally substantially reduced PFO in all treatment groups, which was most significant in the mMSC/HUH7 2:1 group with a ∼53% reduction in PFO compared with HUH7 alone. Despite the reduced PFO reaction to the individual microcapsules implanted subcutaneously, all microcapsule treatment groups were contained in a vascularized fibrotic pouch at 3 weeks. The presence of MSCs in microcapsules retrieved from these fibrotic pouches improved graft survival with significantly higher cell viabilities of 83.1 ± 0.6% and 79.1 ± 0.8% seen with microcapsules containing mMSC/HUH7 at 2:1 and 1:1 ratios, respectively, compared to HUH7 alone (51.5 ± 0.7%) transplanted subcutaneously. This study showed that coencapsulation of MSCs with target cells has a dose-dependent effect on reducing PFO and improving graft survival when implanted either intraperitoneally or subcutaneously in a stringent xenotransplantation setting.


Assuntos
Sobrevivência de Enxerto , Células-Tronco Mesenquimais/citologia , Transplante Heterólogo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Células Imobilizadas/citologia , Modelos Animais de Doenças , Feminino , Fibrose , Humanos , Implantes Experimentais , Camundongos , Células-Tronco Multipotentes/citologia , Cavidade Peritoneal/citologia , Tela Subcutânea/patologia
11.
Stem Cells Dev ; 23(8): 866-76, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24295364

RESUMO

Placenta derived human amniotic epithelial cells (hAEC) are an attractive source of stem cells for the generation of hepatocyte-like cells (HLC) for therapeutic applications to treat liver diseases. During hAEC differentiation into HLC, they become increasingly immunogenic, which may result in immune cell-mediated rejection upon transplantation into allogeneic recipients. Placing cells within devices such as alginate microcapsules can prevent immune cell-mediated rejection. The aim of this study was to investigate the characteristics of HLC generated from hAEC and to examine the effects of encapsulation on HLC viability, gene expression, and function. hAEC were differentiated for 4 weeks and evaluated for hepatocyte-specific gene expression and function. Differentiated cells were encapsulated in barium alginate microcapsules and cultured for 7 days and the effect of encapsulation on cell viability, function, and hepatocyte related gene expression was determined. Differentiated cells performed key functions of hepatocytes including urea synthesis, drug-metabolizing cytochrome P450 (CYP)3A4 activity, indocyanine green (ICG) uptake, low-density lipoprotein (LDL) uptake, and exhibited glutathione antioxidant capacity. A number of hepatocyte-related genes involved in fat, cholesterol, bile acid synthesis, and xenobiotic metabolism were also expressed showing that the hAEC had differentiated into HLC. Upon encapsulation, the HLC remained viable for at least 7 days in culture, continued to express genes involved in fat, cholesterol, bile acid, and xenobiotic metabolism and had glutathione antioxidant capacity. CYP3A4 activity and urea synthesis by the encapsulated HLC were higher than that of monolayer HLC cultures. Functional HLC can be derived from hAEC, and HLC can be encapsulated within alginate microcapsules without losing viability or function in vitro.


Assuntos
Âmnio/citologia , Células Epiteliais/fisiologia , Hepatócitos/metabolismo , Alginatos/química , Biomarcadores/metabolismo , Cápsulas , Diferenciação Celular , Sobrevivência Celular , Meios de Cultivo Condicionados , Feminino , Expressão Gênica , Ácido Glucurônico/química , Células Hep G2 , Ácidos Hexurônicos/química , Humanos , Placenta/citologia , Gravidez
12.
PLoS One ; 8(3): e59120, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23554983

RESUMO

Xenotransplantation of microencapsulated fetal pig islet-like cell clusters (FP ICCs) offers a potential cellular therapy for type 1 diabetes. Although microcapsules prevent direct contact of the host immune system with the xenografted tissue, poor graft survival is still an issue. This study aimed to characterise the nature of the host immune cells present on the engrafted microcapsules and effects on encapsulated FP ICCs that were transplanted into immunocompetent mice. Encapsulated FP ICCs were transplanted into the peritoneal cavity of C57BL/6 mice. Grafts retrieved at days 1, 3, 7, 14 and 21 post-transplantation were analysed for pericapsular fibrotic overgrowth (PFO), cell viability, intragraft porcine gene expression, macrophages, myofibroblasts and intraperitoneal murine cytokines. Graft function was assessed ex vivo by insulin secretion studies. Xenogeneic immune response to encapsulated FP ICCs was associated with enhanced intragraft mRNA expression of porcine antigens MIP-1α, IL-8, HMGB1 and HSP90 seen within the first two weeks post-transplantation. This was associated with the recruitment of host macrophages, infiltration of myofibroblasts and collagen deposition leading to PFO which was evident from day 7 post-transplantation. This was accompanied by a decrease in cell viability and loss of FP ICC architecture. The only pro-inflammatory cytokine detected in the murine peritoneal flushing was TNF-α with levels peaking at day 7 post transplantation. This correlated with the onset of PFO at day 7 implying activated macrophages as its source. The anti-inflammatory cytokines detected were IL-5 and IL-4 with levels peaking at days 1 and 7, respectively. Porcine C-peptide was undetectable at all time points post-transplantation. PFO was absent and murine intraperitoneal cytokines were undetectable when empty microcapsules were transplanted. In conclusion, this study demonstrated that the macrophages are direct effectors of the xenogeneic immune response to encapsulated FP ICCs leading to PFO mediated by a combination of both pro- and anti-inflammatory cytokines.


Assuntos
Antígenos/imunologia , Rejeição de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/imunologia , Macrófagos/imunologia , Transplante Heterotópico , Alginatos/química , Animais , Biomarcadores/metabolismo , Movimento Celular , Citocinas/biossíntese , Citocinas/imunologia , Feto , Ácido Glucurônico/química , Rejeição de Enxerto/patologia , Ácidos Hexurônicos/química , Imunocompetência , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Macrófagos/citologia , Camundongos , Miofibroblastos/citologia , Miofibroblastos/imunologia , Cavidade Peritoneal , Suínos , Transplante Heterólogo
13.
Pancreas ; 41(1): 54-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22143343

RESUMO

OBJECTIVE: A challenge in using human embryonic stem cells (hESCs) as the source of surrogate ß cells is the establishment of methods that could effectively direct their differentiation into functional ß cells. The aim of this study was to assess the effect of NANOG gene suppression in differentiating hESCs as a mean of increasing the efficiency with which endoderm-derived pancreatic cells could be generated. METHODS: A homogenous cell population with stable suppression of NANOG was generated in hESC ENVY line using plasmid-based siRNA approach. Pancreatic differentiation was undertaken according to the ontology-based in vitro selection protocol and followed by transplantation into immunodeficiency mice to mature in vivo. RESULTS: We observed up-regulation of definitive endoderm genes, which expand the role of NANOG in blocking definitive endoderm differentiation. The ontology-based differentiation protocol resulted in increased expression of markers essential for pancreatic epithelium development. Transplantation of these cells further revealed a homogenous pancreatic exocrine-like morphology that stained positively for amylase. CONCLUSIONS: The suppression of NANOG displayed an effective differentiation toward endoderm and pancreatic progenitors. Investigation of the factors required for endocrine formation combined with a prolonged in vivo culturing could be further used to increase the ratio of endocrine-exocrine cells fate.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Proteínas de Homeodomínio/metabolismo , Pâncreas/citologia , Amilases/metabolismo , Animais , Western Blotting , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Endoderma/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia de Contraste de Fase , Proteína Homeobox Nanog , Pâncreas/metabolismo , Pâncreas Exócrino/citologia , Pâncreas Exócrino/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Células-Tronco/metabolismo
14.
Diabetes Metab Res Rev ; 27(8): 928-32, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22069287

RESUMO

BACKGROUND: Cellular-based therapies for insulin-dependent diabetes are potential means of achieving and maintaining normal blood glucose levels (BGL) without the need for insulin administration. Islets isolated from donor pancreases have been the most common tissue used to date, but supply is a limiting factor. The use of human embryonic stem cells (hESC) as a therapy became a possibility with the report that these cells could be differentiated to pancreatic progenitors (PP) over 12 days in vitro. Conversion of PP to glucose-responsive insulin-secreting cells can be achieved by transplanting the progenitors in vivo where cell maturation occurs. To date this step has not been shown under in vitro conditions. METHODS: Prior to transplanting, cells are encapsulated in alginate to prevent the immune cells of recipient attacking the graft. The alginate capsules have pores with a molecular weight cut-off of 250 kDa. These are too small to allow entry of immune cells, but large enough for passage of nutrients and insulin. RESULTS: Encapsulated insulin-producing cells survive and function when transplanted, and have been shown to normalize BGL when allografted into diabetic mice. As few as 750 encapsulated human islets are sufficient to normalize BGL of diabetic non-obese diabetic severe combined immunodeficient (NOD/SCID) recipient mice for at least 2 months. The safety of transplanting encapsulated human islets as demonstrated by the lack of major adverse events and infection was recently shown in a first-in-human clinical trial. Finally, fetal porcine islet-like cell clusters, which are akin to PP derived from ESC, mature and normalize BGL of diabetic recipient mice with the same efficiency as non-encapsulated clusters placed under the kidney capsule. CONCLUSION: Transplanting encapsulated PP, derived from hESCs, into diabetic recipients is the strategy that is now being explored in the Australia Diabetes Therapy Project.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Células-Tronco Embrionárias/transplante , Células Secretoras de Insulina/transplante , Alginatos , Animais , Glicemia/metabolismo , Cápsulas , Diferenciação Celular , Ácido Glucurônico , Ácidos Hexurônicos , Humanos , Insulina/uso terapêutico , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Suínos , Transplante Heterólogo
15.
Islets ; 1(1): 62-74, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-21084851

RESUMO

Previously, the insulin producing liver cell line HUH7-ins has been shown to synthesize, store and secrete insulin in response to glucose via secretory granules. The current study characterized the gene expression profile of HUH7-ins with the aim to identify changes possibly involved in the formation of granules. Additionally, experiments were conducted to determine the influence of chromogranin A (CgA) on secretory granule biogenesis (SGB) in HUH7-ins. Expression of 165 genes were significantly changed in HUH7-ins,though interestingly the majority of secretory granule associated genes, such as the chromogranins were unchanged. CgA was over-expressed in glucose unresponsive HUH7-ins cells to test whether CgA played a role in SGB and would restore the regulated secretory phenotype. Over-expression affected neither the storage nor regulated secretion of insulin. These data suggest that SGB may by regulated at a post-transcriptional level with no evidence to indicate that CgA regulates SGB in the cell line HUH7-ins.


Assuntos
Cromogranina A/fisiologia , Perfilação da Expressão Gênica , Células Secretoras de Insulina/metabolismo , Vesículas Secretórias/genética , Linhagem Celular Tumoral , Cromogranina A/genética , Cromogranina A/metabolismo , Regulação da Expressão Gênica/fisiologia , Glucose/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/química , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Vesículas Secretórias/metabolismo , Estudos de Validação como Assunto
16.
Curr Opin Organ Transplant ; 13(6): 633-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19060555

RESUMO

PURPOSE OF REVIEW: The transplantation of human islets has come a long way since the first diabetic person became insulin independent in 1989. The advent of a steroid-free immunosuppressive protocol in 2000 resulted in most recipients becoming insulin independent and remaining so for a year. However, beta-cell function declines thereafter. Strategies to enhance the islet mass transplanted and preserve beta-cell function are necessary. RECENT FINDINGS: This review covers recent advances in determining the selection of appropriate enzymes for islet isolation, use of pancreases from heart-dead donors and techniques for predicting the functional capacity of isolated islets prior to transplantation. Changing the transplantation site away from the liver, where many islets are destroyed by an inflammatory process, is reviewed, and the possibility of seeding islets onto three-dimensional biodegradable scaffolds discussed. A method of preventing apoptosis of the beta cells prior to transplantation is detailed, as is the beneficial effect of using exenatide, after transplantation. Novel techniques to image islets are discussed, and this requires the labelling of the islets prior to implantation. Enhancing the vascularization of islets is shown to enhance functional outcomes. Encapsulation of the islets should obviate the need for using antirejection drugs, and it may be possible to expand beta cells in vitro. SUMMARY: The above strategies are likely to enhance the outcomes of clinical islet transplants.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/efeitos dos fármacos , Imunossupressores/uso terapêutico , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Colagenases/química , Diabetes Mellitus Tipo 1/patologia , Exenatida , Rejeição de Enxerto/patologia , Rejeição de Enxerto/fisiopatologia , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/transplante , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/métodos , Neovascularização Fisiológica , Peptídeos/uso terapêutico , Coloração e Rotulagem/métodos , Termolisina/química , Doadores de Tecidos , Alicerces Teciduais , Peçonhas/uso terapêutico
17.
Stem Cells Dev ; 17(6): 1087-94, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18513165

RESUMO

Recent work indicates that neural progenitors can be isolated from the skin of rodents and humans. The persistence of these cells in accessible adult tissue raises the possibility of their exploitation for research and therapeutic purposes. This study reports on the derivation, culture, and characterization of homogenous canine skin-derived neuroprecursor cells (SKiNPs) from mature animals. Canine tissue was used because naturalistic brain diseases in community-dwelling dogs are emerging as ecologically sound models for a range of neurological conditions. Adult SKiNPs were initially isolated as neurospheres and then cultured for 10-15 passages in an adherent monolayer assay. Serumfree expansion conditions contained B-27, 20 ng/mL EGF, and 40 ng/mL bFGF. Gene expressions by PCR indicated expression of nestin, CD133, NCAM, and FGF2R, but not GFAP. Highly uniform expression of nestin (76 +/- 8.3%), NCAM (84 +/- 3.3%), betaIII-tubulin (96 +/- 4.3%), and CD133 (68 +/- 13.5%) was also observed. Directed differentiation of SKiNPs in the presence of serum induced betaIIItubulin, NSE, NCAM, and MAP2 in >90% of differentiated cells by immunophenotype analysis. Our culture system rapidly induces canine skin cells into neural precursors, maintains nestin expression in more than 75% of proliferating cells, and generates an almost universal neuronal-like phenotype after 7 days of in vitro differentiation. Their biological characteristics are suggestive of transiently amplifying fate-restricted neuroprecursors rather than true neural stem cells. This system may be an effective alternative for autologous neurorestorative cell replacement in canine models for further translational research.


Assuntos
Células-Tronco Adultas/citologia , Diferenciação Celular , Neurônios/citologia , Transplante de Células-Tronco , Células-Tronco Adultas/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Encefalopatias/terapia , Células Cultivadas , Cães , Humanos , Neurônios/metabolismo , Pele , Transplante Autólogo
18.
Regen Med ; 3(1): 33-47, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18154461

RESUMO

In the past 10 years there have been substantial developments in adult stem cell research, and the transplantation of these cells now holds great promise for regenerative medicine, such as in the treatment of Type 1 diabetes. A large proportion of studies have focused on stem cells sourced from hematopoietic tissues: bone marrow, umbilical cord blood and peripheral blood. Attempts to transdifferentiate these cells into insulin-producing cells, both in vivo and in vitro, have produced conflicting results. Although insulin production and normalization of blood glucose levels have been described in some studies, the true mechanism of stem cell plasticity remains in question - are the functional changes seen due to true transdifferentiation or do they result from cell fusion or other factors? There is evidence that stem cell plasticity is a true phenomenon, but whether it will ever be of therapeutic benefit for Type 1 diabetes remains uncertain.


Assuntos
Células-Tronco Adultas , Diabetes Mellitus Tipo 1/terapia , Ilhotas Pancreáticas/patologia , Adulto , Células-Tronco Adultas/citologia , Animais , Diferenciação Celular , Feminino , Humanos , Transplante das Ilhotas Pancreáticas , Masculino , Medicina Regenerativa
19.
Transplantation ; 83(11): 1440-8, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17565317

RESUMO

BACKGROUND: Fetal beta cells are a potential form of cell therapy for type 1 diabetes. To protect transplanted cells from cellular immune attack, microencapsulation using barium alginate can be employed. Whether microencapsulated fetal pancreatic cells will differentiate as occurs with nonencapsulated fetal pancreatic cells is presently unknown. It is suggested that such differentiation would occur in encapsulated cells, similar to previous experiments conducted using encapsulated embryonic stem cells. METHODS: Streptozotocin-induced diabetic severe combined immunodeficient mice were transplanted with 5,000 to 38,000 fetal pig islet-like cell clusters (ICCs) within barium alginate microcapsules of diameter 300, 600, or 1000 microm. Viability, insulin secretion, and content of encapsulated cells were measured prior to transplantation. Blood glucose levels (BGL) were measured twice weekly and porcine C-peptide monthly. Encapsulated cells were recovered from mice at 6 months posttransplantation for analysis. RESULTS: Encapsulated cells became glucose responsive and normalized BGL within 13 to 68 days posttransplantation, with 5,000 to 10,000 ICCs required. Microcapsule diameter did not affect the time required to achieve normoglycemia. BGL remained normal for the 6-month duration of the experiments. After removal of grafts at 25 weeks posttransplantation, glucose stimulated insulin secretion of the explants was enhanced 96-fold, insulin content was enhanced 34-fold, and the percentage of insulin and glucagon positive cells increased 10-fold and threefold, respectively, from the time of transplantation. CONCLUSIONS: This study demonstrates that fetal pancreatic cells differentiate and function normally when placed within barium alginate microcapsules and transplanted.


Assuntos
Diferenciação Celular , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/cirurgia , Transplante de Tecido Fetal/métodos , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/transplante , Alginatos , Animais , Glicemia/metabolismo , Cápsulas , Agregação Celular , Sobrevivência Celular , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/metabolismo , Imunofluorescência , Glucagon/metabolismo , Glucose/farmacologia , Ácido Glucurônico , Ácidos Hexurônicos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/embriologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos SCID , Pâncreas/metabolismo , Coloração e Rotulagem , Fatores de Tempo
20.
Regen Med ; 2(3): 289-300, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17511565

RESUMO

AIM: To generate complex surrogate tissue by transplanting 3D scaffolds seeded with human embryonic stem cells (hESCs) between the liver lobules of severe combined immunodeficient (SCID) mice and to assess the teratoma-forming potential. MATERIALS & METHODS: 3D poly-(lactic-co-glycolic acid) (PLGA) scaffolds coated with laminin were seeded with hESCs and then transplanted between the liver lobules of SCID mice. After a period of in vivo differentiation, the scaffolds were retrieved and analyzed using reverse transcription polymerase chain reaction, immunofluorescent staining and scanning electron microscopy. RESULTS: A proportion of the hESCs within the scaffolds differentiated into cells that produced proteins characteristic of specific tissues, including endoderm and pancreatic markers glucogon-like peptide-1 receptor, islet amyloid polypeptide and Insulin. Markers of hepatic and neuronal lineages were also investigated. Major matrix proteins abundant in multiple tissue types, including collagen I, laminin and collagen IV, were found to be profuse within the scaffold pores. Transplantation of the seeded scaffolds between liver lobules also resulted in extensive vascularization both from host blood vessel incursion and the differentiation of hESCs into endothelial progenitor cells. An investigation of teratoma-forming potential demonstrated that transplantation of 3D scaffolds seeded with hESCs will, under certain conditions, lead to the growth of teratomas. DISCUSSION: Transplantation of 3D scaffolds seeded with hESCs between liver lobules resulted in the development of surrogate tissue containing cells that produced proteins representing the pancreatic, hepatic and neuronal lineages, the assembly of an extracellular matrix structure and the formation of a vasculature. hESCs seeded within 3D scaffolds and transplanted into SCID mice were capable of forming teratomas. However, the formation and progression of teratoma growth is shown to be dependant on both the site of transplantation and the treatment of cells prior to transplantation.


Assuntos
Materiais Biocompatíveis , Células-Tronco Embrionárias/transplante , Próteses e Implantes , Teratoma/etiologia , Engenharia Tecidual , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Células-Tronco Embrionárias/patologia , Humanos , Fígado/citologia , Fígado/patologia , Camundongos , Camundongos SCID , Neurônios/citologia , Neurônios/patologia , Pâncreas/citologia , Pâncreas/patologia , Próteses e Implantes/efeitos adversos , Teratoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA