Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-34884797

RESUMO

The anti-inflammatory role of regulatory B cells (Breg cells) has been associated with IL-35 based on studies of experimental autoimmune uveitis and encephalitis. The role of Breg cells and IL-35+ Breg cells for type 1 diabetes (T1D) remains to be investigated. We studied PBMCs from T1D subjects and healthy controls (HC) and found lowered proportions of Breg cells and IL-35+ Breg cells in T1D. To elucidate the role of Breg cells, the lymphoid organs of two mouse models of T1D were examined. Lower proportions of Breg cells and IL-35+ Breg cells were found in the animal models of T1D compared with control mice. In addition, the systemic administration of recombinant mouse IL-35 prevented hyperglycemia after multiple low dose streptozotocin (MLDSTZ) injections and increased the proportions of Breg cells and IL-35+ Breg cells. A higher proportion of IFN-γ+ cells among Breg cells were found in the PBMCs of the T1D subjects. In the MLDSTZ mice, IL-35 administration decreased the proportions of IFN-γ+ cells among the Breg cells. Our data illustrate that Breg cells may play an important role in the development of T1D and that IL-35 treatment prevents the development of hyperglycemia by maintaining the phenotype of the Breg cells under an experimental T1D condition.


Assuntos
Anti-Inflamatórios/farmacologia , Linfócitos B Reguladores/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Hiperglicemia/prevenção & controle , Interleucinas/farmacologia , Adulto , Animais , Anti-Inflamatórios/sangue , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Hiperglicemia/induzido quimicamente , Interferon gama/sangue , Interleucinas/sangue , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Estreptozocina/toxicidade
2.
Exp Cell Res ; 384(1): 111613, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31494095

RESUMO

The lipotoxicity is considered as one of the risk for diabetes. Here we report C-type lectin domain family 11, member A (Clec11a) as a new regulator in islet playing a protective role in lipotoxicity induced dysfunction. Islet transcriptome sequencing was performed using the high-fat diet induced obesity (DIO) mice model. We found a significant decrease of Clec11a expression in islets of DIO mice compared to normal control mice, which was further confirmed by real-time PCR. Immunostaining demonstrated the localization of the Clec11a protein in mouse islets. Administration of recombinant human Clec11a (rClec11a) protein promoted the proliferation of islet cells and rescued the inhibition of fatty acid on cell proliferation, which involved the activation of Erk signaling pathway. We also found that the rClec11a altered the expression of genes involved in lipid metabolism.


Assuntos
Proliferação de Células/fisiologia , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Ilhotas Pancreáticas/metabolismo , Lectinas Tipo C/metabolismo , Metabolismo dos Lipídeos/fisiologia , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Transdução de Sinais/fisiologia , Transcriptoma/fisiologia
3.
Genome Res ; 25(10): 1521-35, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26430163

RESUMO

Sporadic breast cancer (SBC) is a common disease without robust means of early risk prediction in the population. We studied 282 females with SBC, focusing on copy number aberrations in cancer-free breast tissue (uninvolved margin, UM) outside the primary tumor (PT). In total, 1162 UMs (1-14 per breast) were studied. Comparative analysis between UM(s), PT(s), and blood/skin from the same patient as a control is the core of the study design. We identified 108 patients with at least one aberrant UM, representing 38.3% of cases. Gains in gene copy number were the principal type of mutations in microscopically normal breast cells, suggesting that oncogenic activation of genes via increased gene copy number is a predominant mechanism for initiation of SBC pathogenesis. The gain of ERBB2, with overexpression of HER2 protein, was the most common aberration in normal cells. Five additional growth factor receptor genes (EGFR, FGFR1, IGF1R, LIFR, and NGFR) also showed recurrent gains, and these were occasionally present in combination with the gain of ERBB2. All the aberrations found in the normal breast cells were previously described in cancer literature, suggesting their causative, driving role in pathogenesis of SBC. We demonstrate that analysis of normal cells from cancer patients leads to identification of signatures that may increase risk of SBC and our results could influence the choice of surgical intervention to remove all predisposing cells. Early detection of copy number gains suggesting a predisposition toward cancer development, long before detectable tumors are formed, is a key to the anticipated shift into a preventive paradigm of personalized medicine for breast cancer.


Assuntos
Neoplasias da Mama/genética , Mama/anatomia & histologia , Mutação , Adulto , Idoso , Idoso de 80 Anos ou mais , Mama/patologia , Neoplasias da Mama/patologia , Estudos de Coortes , Análise Mutacional de DNA , Feminino , Dosagem de Genes , Genes erbB-2 , Predisposição Genética para Doença , Genótipo , Humanos , Pessoa de Meia-Idade , Receptor ErbB-2/genética , Receptores de Fatores de Crescimento/genética , Fatores de Risco
4.
Horm Metab Res ; 50(8): 627-639, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30001566

RESUMO

We assessed glucose uptake in different tissues in type 2 diabetes (T2D), prediabetes, and control subjects to elucidate its impact in the development of whole-body insulin resistance and T2D. Thirteen T2D, 12 prediabetes, and 10 control subjects, matched for age and BMI, underwent OGTT and abdominal subcutaneous adipose tissue (SAT) biopsies. Integrated whole-body 18F-FDG PET and MRI were performed during a hyperinsulinemic euglycemic clamp to asses glucose uptake rate (MRglu) in several tissues. MRglu in skeletal muscle, SAT, visceral adipose tissue (VAT), and liver was significantly reduced in T2D subjects and correlated positively with M-values (r=0.884, r=0.574, r=0.707 and r=0.403, respectively). Brain MRglu was significantly higher in T2D and prediabetes subjects and had a significant inverse correlation with M-values (r=-0.616). Myocardial MRglu did not differ between groups and did not correlate with the M-values. A multivariate model including skeletal muscle, brain and VAT MRglu best predicted the M-values (adjusted r2=0.85). In addition, SAT MRglu correlated with SAT glucose uptake ex vivo (r=0.491). In different stages of the development of T2D, glucose uptake during hyperinsulinemia is elevated in the brain in parallel with an impairment in peripheral organs. Impaired glucose uptake in skeletal muscle and VAT together with elevated glucose uptake in brain were independently associated with whole-body insulin resistance, and these tissue-specific alterations may contribute to T2D development.


Assuntos
Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Resistência à Insulina , Gordura Intra-Abdominal/metabolismo , Músculo Esquelético/metabolismo , Adulto , Idoso , Transporte Biológico , Encéfalo/diagnóstico por imagem , Diabetes Mellitus Tipo 2/diagnóstico por imagem , Feminino , Técnica Clamp de Glucose , Humanos , Gordura Intra-Abdominal/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Imagem Corporal Total
5.
Diabetologia ; 58(1): 132-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25257098

RESUMO

AIMS/HYPOTHESIS: Highly blood-perfused islets have been observed to be the most functional islets in the native pancreas. We hypothesised that differences in vascular support of islets in donor pancreases influence their susceptibility to cellular stress and capacity for vascular engraftment after transplantation. METHODS: Highly blood-perfused islets in rats were identified by injection of microspheres into the ascending aorta before islet isolation. Cell death was evaluated after in vitro cytokine or hypoxia exposure, and 2 days post transplantation. One month post transplantation, islet engraftment, including vascular density, blood perfusion and oxygen tension (pO2) in the tissue, was evaluated. RESULTS: Microsphere-containing islets had a similar frequency of cell death during standard culture conditions but increased cell death after exposure to cytokines and hypoxia in comparison with other islets. Two days after transplantation the percentage of apoptotic or necrotic cells was also higher in grafts of such islets and 1 month post transplantation these grafts were composed of substantially more connective tissue. Grafts of highly blood-perfused islets in the native pancreas regained a higher vascular density, blood perfusion and pO2 in comparison with grafts of other islets. CONCLUSIONS/INTERPRETATION: Native islets that are highly blood-perfused regained this feature after transplantation, indicating a superior capacity for revascularisation and post-transplant function. However, the same group of islets was more vulnerable to different kinds of cellular stress, which limited their early survival post transplantation. Preferential death of these most active islets may contribute to the high number of islets needed to provide cure with islet transplantation.


Assuntos
Vasos Sanguíneos/patologia , Sobrevivência de Enxerto , Transplante das Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/irrigação sanguínea , Animais , Apoptose/fisiologia , Células Cultivadas , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Oxigênio/metabolismo , Ratos , Ratos Endogâmicos Lew , Enxerto Vascular/normas
6.
Int J Neurosci ; 125(7): 547-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25077520

RESUMO

PURPOSE: Neural crest stem cells derived from the boundary cap (bNCSCs), markedly promote survival, proliferation and function of insulin producing ß-cells in vitro and in vivo after coculture/transplantation with pancreatic islets [ 1, 2 ]. Recently, we have shown that beneficial effects on ß-cells require cadherin contacts between bNCSCs and ß-cells [ 3, 4 ]. Here we investigated whether hair follicle (HF) NCSCs, a potential source for human allogeneic transplantation, exert similar positive effects on ß-cells. MATERIALS AND METHODS: We established cocultures of HF-NCSCs or bNCSCs from mice expressing enhanced green fluorescent protein together with pancreatic islets from DxRed expressing mice or NMRI mice and compared their migration towards islet cells and effect on proliferation of ß-cells as well as intracellular relations between NCSCs and islets using qRT-PCR analysis and immunohistochemistry. RESULTS: Whereas both types of NCSCs migrated extensively in the presence of islets, only bNCSCs demonstrated directed migration toward islets, induced ß-cell proliferation and increased the presence of cadherin at the junctions between bNCSCs and ß-cells. Even in direct contact between ß-cells and HF-NCSCs, no cadherin expression was detected. CONCLUSIONS: These observations indicate that HF-NCSCs do not confer the same positive effect on ß-cells as demonstrated for bNCSCs. Furthermore, these data suggest that induction of cadherin expression by HF-NCSCs may be useful for their ability to support ß-cells in coculture and after transplantation.


Assuntos
Folículo Piloso/citologia , Ilhotas Pancreáticas/fisiologia , Crista Neural/citologia , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Actinas/genética , Actinas/metabolismo , Animais , Caderinas/metabolismo , Diferenciação Celular , Movimento Celular , Proliferação de Células/fisiologia , Células Cultivadas , Técnicas de Cocultura , Embrião de Mamíferos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Tempo
7.
Diabetologia ; 57(1): 50-3, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24078058

RESUMO

AIMS/HYPOTHESIS: The hormone betatrophin was recently described as a potent stimulator of beta cell proliferation in mice. Insulin resistance, but not insulin deficiency, caused upregulation of betatrophin expression. If these findings were found to be fully applicable in humans, this would open up the possibility of future betatrophin treatment in type 1 diabetes. The present study measured for the first time betatrophin concentrations in humans and tested the hypothesis that there would be no difference in circulating betatrophin concentrations between patients with type 1 diabetes and healthy individuals. METHODS: Betatrophin concentrations in plasma of 33 patients with type 1 diabetes and 24 age-matched healthy controls were measured by ELISA. The study participants were characterised for blood lipids, BMI, plasma glucose and HbA1c, and, for the diabetic patients, their insulin requirements and any residual C-peptide concentrations. RESULTS: Plasma betatrophin concentrations were normally ~300 pg/ml, but were approximately doubled in patients with type 1 diabetes. In the patients, there were no correlations between betatrophin and age, blood lipids, BMI, glucose control or insulin requirement, whereas in controls betatrophin levels increased with age. BMI, blood pressure and triacylglycerol, LDL-cholesterol and HDL-cholesterol levels were similar in patients and healthy controls. CONCLUSIONS/INTERPRETATION: Circulating concentrations of betatrophin are increased in type 1 diabetes in contrast with what was recently described in an insulin-deficient mouse model. However, increased betatrophin concentrations do not protect against loss of C-peptide. Betatrophin treatment in type 1 diabetes would therefore probably not be successful without the use of supraphysiological doses or a combination with immune regulatory treatment.


Assuntos
Diabetes Mellitus Tipo 1/sangue , Hormônios Peptídicos/sangue , Adulto , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Peptídeo C/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Adulto Jovem
8.
Diabetes ; 73(7): 1127-1139, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38603470

RESUMO

Pluripotent stem cell-derived islets (SC-islets) have emerged as a new source for ß-cell replacement therapy. The function of human islet transplants is hampered by excessive cell death posttransplantation; contributing factors include inflammatory reactions, insufficient revascularization, and islet amyloid formation. However, there is a gap in knowledge of the engraftment process of SC-islets. In this experimental study, we investigated the engraftment capability of SC-islets at 3 months posttransplantation and observed that cell apoptosis rates were lower but vascular density was similar in SC-islets compared with human islets. Whereas the human islet transplant vascular structures were a mixture of remnant donor endothelium and ingrowing blood vessels, the SC-islets contained ingrowing blood vessels only. Oxygenation in the SC-islet grafts was twice as high as that in the corresponding grafts of human islets, suggesting better vascular functionality. Similar to the blood vessel ingrowth, reinnervation of the SC-islets was four- to fivefold higher than that of the human islets. Both SC-islets and human islets contained amyloid at 1 and 3 months posttransplantation. We conclude that the vascular and neural engraftment of SC-islets are superior to those of human islets, but grafts of both origins develop amyloid, with potential long-term consequences.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Humanos , Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Animais , Camundongos , Apoptose/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Sobrevivência de Enxerto/fisiologia , Masculino
10.
Free Radic Res ; 57(6-12): 460-469, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37972305

RESUMO

NADPH oxidase 4 (NOX4) inhibition has been reported to mitigate diabetes-induced beta-cell dysfunction and improve survival in vitro, as well as counteract high-fat diet-induced glucose intolerance in mice. We investigated the antidiabetic effects of the selective NOX4 inhibitor GLX7013159 in vivo in athymic diabetic mice transplanted with human islets over a period of 4 weeks. The GLX7013159-treated mice achieved lower blood glucose and water consumption throughout the treatment period. Furthermore, GLX7013159 treatment resulted in improved insulin and c-peptide levels, better insulin secretion capacity, as well as in greatly reduced apoptotic rates of the insulin-positive human cells, measured as colocalization of insulin and cleaved caspase-3. We conclude that the antidiabetic effects of NOX4 inhibition by GLX7013159 are observed also during a prolonged study period in vivo and are likely to be due to an improved survival and function of the human beta-cells.


Assuntos
Diabetes Mellitus Experimental , Insulinas , Humanos , Camundongos , Animais , NADPH Oxidase 4 , Glicemia , Hipoglicemiantes , Insulina , Glucose/farmacologia
11.
J Mol Endocrinol ; 71(1)2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37078556

RESUMO

Beta-cell dysfunction is a hallmark of disease progression in patients with diabetes. Research has been focused on maintaining and restoring beta-cell function during diabetes development. The aims of this study were to explore the expression of C-type lectin domain containing 11A (CLEC11A), a secreted sulphated glycoprotein, in human islets and to evaluate the effects of CLEC11A on beta-cell function and proliferation in vitro. To test these hypotheses, human islets and human EndoC-ßH1 cell line were used in this study. We identified that CLEC11A was expressed in beta-cells and alpha-cells in human islets but not in EndoC-ßH1 cells, whereas the receptor of CLEC11A called integrin subunit alpha 11 was found in both human islets and EndoC-ßH1 cells. Long-term treatment with exogenous recombinant human CLEC11A (rhCLEC11A) accentuated glucose-stimulated insulin secretion, insulin content, and proliferation from human islets and EndoC-ßH1 cells, which was partially due to the accentuated expression levels of transcription factors MAFA and PDX1. However, the impaired beta-cell function and reduced mRNA expression of INS and MAFA in EndoC-ßH1 cells that were caused by chronic palmitate exposure could only be partially improved by the introduction of rhCLEC11A. Based on these results, we conclude that rhCLEC11A promotes insulin secretion, insulin content, and proliferation in human beta-cells, which are associated with the accentuated expression levels of transcription factors MAFA and PDX1. CLEC11A, therefore, may provide a novel therapeutic target for maintaining beta-cell function in patients with diabetes.


Assuntos
Células Secretoras de Insulina , Insulina , Humanos , Secreção de Insulina , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/metabolismo , Proliferação de Células
12.
EJNMMI Res ; 13(1): 107, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38100042

RESUMO

BACKGROUND: Beta-cell replacement methods such as transplantation of isolated donor islets have been proposed as a curative treatment of type 1 diabetes, but widespread application is challenging due to shortages of donor tissue and the need for continuous immunosuppressive treatments. Stem-cell-derived islets have been suggested as an alternative source of beta cells, but face transplantation protocols optimization difficulties, mainly due to a lack of available methods and markers to directly monitor grafts survival, as well as their localization and function. Molecular imaging techniques and particularly positron emission tomography has been suggested as a tool for monitoring the fate of islets after clinical transplantation. The integral membrane protein DGCR2 has been demonstrated to be a potential pancreatic islet biomarker, with specific expression on insulin-positive human embryonic stem-cell-derived pancreatic progenitor cells. The candidate Affibody molecule ZDGCR2:AM106 was radiolabeled with fluorine-18 using a novel click chemistry-based approach. The resulting positron emission tomography tracer [18F]ZDGCR2:AM106 was evaluated for binding to recombinant human DGCR2 and cryosections of stem-cell-derived islets, as well as in vivo using an immune-deficient mouse model transplanted with stem-cell-derived islets. Biodistribution of the [18F]ZDGCR2:AM106 was also assessed in healthy rats and pigs. RESULTS: [18F]ZDGCR2:AM106 was successfully synthesized with high radiochemical purity and yield via a pretargeting approach. [18F]ZDGCR2:AM106 retained binding to recombinant human DCGR2 as well as to cryosectioned stem-cell-derived islets, but in vivo binding to native pancreatic tissue in both rat and pig was low. However, in vivo uptake of [18F]ZDGCR2:AM106 in stem-cell-derived islets transplanted in the immunodeficient mice was observed, albeit only within the early imaging frames after injection of the radiotracer. CONCLUSION: Targeting of DGCR2 is a promising approach for in vivo detection of stem-cell-derived islets grafts by molecular imaging. The synthesis of [18F]ZDGCR2:AM106 was successfully performed via a pretargeting method to label a site-specific covalently bonded fluorine-18 to the Affibody molecule. However, the rapid washout of [18F]ZDGCR2:AM106 from the stem-cell-derived islets graft indicates that dissociation kinetics can be improved. Further studies using alternative binders of similar classes with improved binding potential are warranted.

13.
Nat Biotechnol ; 40(7): 1042-1055, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35241836

RESUMO

Transplantation of pancreatic islet cells derived from human pluripotent stem cells is a promising treatment for diabetes. Despite progress in the generation of stem-cell-derived islets (SC-islets), no detailed characterization of their functional properties has been conducted. Here, we generated functionally mature SC-islets using an optimized protocol and benchmarked them comprehensively against primary adult islets. Biphasic glucose-stimulated insulin secretion developed during in vitro maturation, associated with cytoarchitectural reorganization and the increasing presence of alpha cells. Electrophysiology, signaling and exocytosis of SC-islets were similar to those of adult islets. Glucose-responsive insulin secretion was achieved despite differences in glycolytic and mitochondrial glucose metabolism. Single-cell transcriptomics of SC-islets in vitro and throughout 6 months of engraftment in mice revealed a continuous maturation trajectory culminating in a transcriptional landscape closely resembling that of primary islets. Our thorough evaluation of SC-islet maturation highlights their advanced degree of functionality and supports their use in further efforts to understand and combat diabetes.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Células-Tronco Pluripotentes , Animais , Glucose/metabolismo , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Camundongos , Células-Tronco Pluripotentes/metabolismo
14.
Nat Commun ; 12(1): 4127, 2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-34226552

RESUMO

Gliomas are brain tumors characterized by an immunosuppressive microenvironment. Immunostimulatory agonistic CD40 antibodies (αCD40) are in clinical development for solid tumors, but are yet to be evaluated for glioma. Here, we demonstrate that systemic delivery of αCD40 in preclinical glioma models induces the formation of tertiary lymphoid structures (TLS) in proximity of meningeal tissue. In treatment-naïve glioma patients, the presence of TLS correlates with increased T cell infiltration. However, systemic delivery of αCD40 induces hypofunctional T cells and impairs the response to immune checkpoint inhibitors in pre-clinical glioma models. This is associated with a systemic induction of suppressive CD11b+ B cells post-αCD40 treatment, which accumulate in the tumor microenvironment. Our work unveils the pleiotropic effects of αCD40 therapy in glioma and reveals that immunotherapies can modulate TLS formation in the brain, opening up for future opportunities to regulate the immune response.


Assuntos
Antígenos CD40/imunologia , Glioma/tratamento farmacológico , Estruturas Linfoides Terciárias/imunologia , Animais , Antineoplásicos/farmacologia , Linfócitos B/imunologia , Neoplasias Encefálicas/tratamento farmacológico , Antígeno CD11b , Linhagem Celular Tumoral , Citocinas , Feminino , Expressão Gênica , Glioma/patologia , Humanos , Imunoglobulina G/genética , Imunoterapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides , Fenótipo , Linfócitos T , Microambiente Tumoral/imunologia
15.
Stem Cell Res ; 50: 102114, 2020 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-33340796

RESUMO

Availability of numerous high-quality iPSC lines is needed to overcome donor-associated variability caused by genetic background effects. We generated two human iPSC lines from dermal fibroblasts of two healthy females using Sendai virus reprogramming. Quality assessment of the iPSC lines confirmed the expression of pluripotency markers, trilineage differentiation capacity and absence of exogenous expression of reprogramming factors. Both iPSC lines were genetically stable with a genotype that matched the fibroblast lines of donors. These iPSC lines add to available reference lines as a resource for disease modeling of polygenic and multifactorial diseases, for evaluation of differentiation protocols and toxicology screening.

16.
Cell Transplant ; 18(1): 23-30, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19476206

RESUMO

Pancreatic islets implanted heterotopically (i.e., into the kidney, spleen, or liver) become poorly revascularized following transplantation. We hypothesized that islets implanted into the pancreas would become better revascularized. Islets isolated from transgenic mice expressing enhanced yellow fluorescent protein (EYFP) in all somatic cells were cultured before they were implanted into the pancreas or beneath the renal capsule of athymic mice. Vascular density was evaluated in histological sections 1 month posttransplantation. EYFP was used as reporter for the transgene to identify the transplanted islets. Islet endothelial cells were visualized by staining with the lectin Bandeiraea simplicifolia (BS-1). Capillary numbers in intrapancreatically implanted islets were only slightly lower than those counted in endogenous islets, whereas islets implanted beneath the renal capsule had a markedly lower vascular density. In order to determine if this high graft vascular density at the intrapancreatic site reflected expansion of remnant donor endothelial cells or increased ingrowth of blood vessels from the host, also islets from Tie2-green fluorescent protein (GFP) mice (i.e., islets with fluorescent endothelial cells) were transplanted into the pancreas or beneath the renal capsule of athymic mice. These islet grafts revealed that the new vascular structures formed in the islet grafts contained very few GFP-positive cells, and thus mainly were of recipient origin. The reason(s) for the much better ingrowth of blood vessels at the intrapancreatic site merits further studies, because this may help us form strategies to overcome the barrier for ingrowth of host vessels also into islets in heterotopic implantation sites.


Assuntos
Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/citologia , Animais , Células Endoteliais , Sobrevivência de Enxerto , Humanos , Imuno-Histoquímica , Proteínas Luminescentes/análise , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica/fisiologia
17.
Curr Opin Organ Transplant ; 14(6): 688-93, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19745734

RESUMO

PURPOSE OF REVIEW: To summarize recent studies on the oxygenation of pancreatic islets and its role in islet transplantation. RECENT FINDINGS: Pancreatic islet cells are highly sensitive to hypoxic conditions. Hypoxia contributes to poor islet yield at isolation, as well as inflammatory events and cellular death during culture and early posttransplantation. Use of oxygen carriers, such as semifluorinated alkanes, during pancreas preservation and gas-permeable devices for islet culture and transport has in recent studies proven beneficial. Beta-cell death can be limited posttransplantation by targeting hypoxia-induced cellular pathways that cause apoptotic death. Owing to low revascularization, impaired oxygenation seems to prevail in intraportally transplanted islets. Means to improve revascularization, oxygenation and function of transplanted islets can be achieved not only by stimulating angiogenic factors, but also by decrease of angiostatic factors such as thrombospondin-1 in islets for transplantation. Moreover, bone-marrow-derived cells, such as mesenchymal stem cells and hematopoietic stem cells, can induce or contribute to increased revascularization. SUMMARY: Low oxygenation of islets contributes to cellular death and dysfunction during preparation of islets for transplantation, as well as posttransplantation. Interventions at these different steps to ensure adequate oxygenation have the potential to improve the results of clinical islet transplantation.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Consumo de Oxigênio , Oxigênio/metabolismo , Animais , Morte Celular , Hipóxia Celular , Humanos , Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/métodos , Técnicas de Cultura de Tecidos , Coleta de Tecidos e Órgãos
18.
J Endocr Soc ; 3(8): 1608-1616, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31404404

RESUMO

Low-oxygenated and dormant islets with a capacity to become activated when needed may play a crucial role in the complex machinery behind glucose homeostasis. We hypothesized that low-oxygenated islets, when not functionally challenged, do not rapidly cycle between activation and inactivation but are a stable population that remain low-oxygenated. As this was confirmed, we aimed to characterize these islets with regard to cell composition, vascular density, and endocrine cell proliferation. The 2-nitroimidazole low-oxygenation marker pimonidazole was administered as a single or repeated dose to Wistar Furth rats. The stability of oxygen status of islets was evaluated by immunohistochemistry as the number of islets with incorporated pimonidazole adducts after one or repeated pimonidazole injections. Adjacent sections were evaluated for islet cell composition, vascular density, and endocrine cell proliferation. Single and repeated pimonidazole injections over an 8-hour period yielded accumulation of pimonidazole adducts in the same islets. An average of 30% of all islets was in all cases positively stained for pimonidazole adducts. These islets showed a similar endocrine cell composition as other islets but had lower vascular density and ß-cell proliferation. In conclusion, low-oxygenated islets were found to be a stable subpopulation of islets for at least 8 hours. Although they have previously been observed to be less functionally active, their islet cell composition was similar to that of other islets. Consistent with their lower oxygenation, they had fewer blood vessels than other islets. Notably, ß-cell regeneration preferentially occurred in better-oxygenated islets.

19.
Cell Transplant ; 28(11): 1455-1460, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31359771

RESUMO

Beta cell replacement is an exciting field where new beta cell sources and alternative sites are widely explored. The liver has been the implantation site of choice in the clinic since the advent of islet transplantation. However, in most cases, repeated islet transplantation is needed to achieve normoglycemia in diabetic recipients. This study aimed to investigate whether there are differences in islet survival and engraftment between a first and a second transplantation, performed 1 week apart, to the liver. C57BL/6 mice were accordingly transplanted twice with an initial infusion of syngeneic islets expressing green fluorescent protein (GFP). The second islet transplant was performed 1 week later and consisted of islets isolated from non-GFP C57BL/6-mice. Animals were sacrificed either 1 day or 1 month after the second transplantation. A control group received a saline infusion instead of GFP-expressing islets, 1 week later obtained a standard non-GFP islet transplant, and was subsequently sacrificed 1 month later. Islet engraftment in the liver was assessed by immunohistochemistry and serum was analyzed for angiogenic factors induced by the first islet transplantation. Almost 70% of islets found in the liver following repeated islet transplantation originated from the second transplantation. The vascular density in the transplanted non-GFP-expressing islets did not differ depending on whether their transplantation was preceded by a primary islet transplantation or saline administration only nor did angiogenic factors in serum prior to the transplantation of non-GFP islets differ between animals that had received a previous islet transplantation or a saline infusion. We conclude that first islet transplantation creates, by unknown mechanisms, favorable conditions for the survival of a second transplant to the liver.


Assuntos
Sobrevivência de Enxerto , Transplante das Ilhotas Pancreáticas/métodos , Animais , Células Cultivadas , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Transplante Homólogo/métodos
20.
Ups J Med Sci ; 124(4): 228-237, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31623497

RESUMO

Background: Murine boundary cap-derived neural crest stem cells (NCSCs) are capable of enhancing islet function by stimulating beta cell proliferation as well as increasing the neural and vascular density in the islets both in vitro and in vivo. This study aimed to isolate NCSC-like cells from human bone marrow.Methods: CD271 magnetic cell separation and culture techniques were used to purify a NCSC-enriched population of human bone marrow. Analyses of the CD271+ and CD271- fractions in terms of protein expression were performed, and the capacity of the CD271+ bone marrow cells to form 3-dimensional spheres when grown under non-adherent conditions was also investigated. Moreover, the NCSC characteristics of the CD271+ cells were evaluated by their ability to migrate toward human islets as well as human islet-like cell clusters (ICC) derived from pluripotent stem cells.Results: The CD271+ bone marrow population fulfilled the criterion of being multipotent stem cells, having the potential to differentiate into glial cells, neurons as well as myofibroblasts in vitro. They had the capacity to form 3-dimensional spheres as well as an ability to migrate toward human islets, further supporting their NCSC identity. Additionally, we demonstrated similar migration features toward stem cell-derived ICC.Conclusion: The results support the NCSC identity of the CD271-enriched human bone marrow population. It remains to investigate whether the human bone marrow-derived NCSCs have the ability to improve transplantation efficacy of not only human islets but stem cell-derived ICC as well.


Assuntos
Técnicas de Cultura de Células , Separação Celular/métodos , Ilhotas Pancreáticas/citologia , Crista Neural/citologia , Células-Tronco Pluripotentes/citologia , Adapaleno/metabolismo , Adulto , Idoso , Células da Medula Óssea/citologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Humanos , Transplante das Ilhotas Pancreáticas , Masculino , Pessoa de Meia-Idade , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa