Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabet Med ; 41(6): e15279, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38185936

RESUMO

AIMS: Evidence is accumulating of the therapeutic benefits of mesenchymal stromal cells (MSCs) in diabetes-related conditions. We have identified a novel population of stromal cells within islets of Langerhans - islet stellate cells (ISCs) - which have a similar morphology to MSCs. In this study we characterize mouse ISCs and compare their morphology and function to MSCs to determine whether ISCs may also have therapeutic potential in diabetes. METHODS: ISCs isolated from mouse islets were compared to mouse bone marrow MSCs by analysis of cell morphology; expression of cell-surface markers and extracellular matrix (ECM) components; proliferation; apoptosis; paracrine activity; and differentiation into adipocytes, chondrocytes and osteocytes. We also assessed the effects of co-culture with ISCs or MSCs on the insulin secretory capacity of islet beta cells. RESULTS: Although morphological similar, ISCs were functionally distinct from MSCs. Thus, ISCs were less proliferative and more apoptotic; they had different expression levels of important paracrine factors; and they were less efficient at differentiation down multiple lineages. Co-culture of mouse islets with ISCs enhanced glucose induced insulin secretion more effectively than co-culture with MSCs. CONCLUSIONS: ISCs are a specific sub-type of islet-derived stromal cells that possess biological behaviors distinct from MSCs. The enhanced beneficial effects of ISCs on islet beta cell function suggests that they may offer a therapeutic target for enhancing beta cell functional survival in diabetes.


Assuntos
Diferenciação Celular , Técnicas de Cocultura , Células Secretoras de Insulina , Ilhotas Pancreáticas , Células-Tronco Mesenquimais , Animais , Camundongos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/citologia , Diferenciação Celular/fisiologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Células Estreladas do Pâncreas/metabolismo , Células Estreladas do Pâncreas/fisiologia , Proliferação de Células/fisiologia , Insulina/metabolismo , Células Cultivadas , Secreção de Insulina/fisiologia , Camundongos Endogâmicos C57BL , Masculino , Apoptose/fisiologia
2.
Diabet Med ; 40(12): e15227, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37728506

RESUMO

AIMS: Human islet transplantation as a therapy for type 1 diabetes is compromised by the loss of functional beta cells in the immediate post-transplantation period. Mesenchymal stromal cells (MSCs) and MSC-derived secretory peptides improve the outcomes of islet transplantation in rodent models of diabetes. Here, we utilized a mouse model for human islet transplantation and assessed the effects of a cocktail of MSC-secreted peptides (screened by MSC-secretome for human islet GPCRs) on the functional survival of human islets. METHODS: Human islets from nine donors (Age: 36-57; BMI: 20-35) were treated with a cocktail of human recombinant annexin A1 (ANXA1), stromal cell-derived factor-1 (SDF-1/CXCL12) and complement component C3 (C3a). Glucose-stimulated insulin secretion (GSIS) was assessed in static incubation, and cytokine-induced apoptosis was assessed by measuring caspase 3/7 activity. mRNA expression levels were determined by qPCR. Human islet function in vivo was assessed using a novel model for human islet transplantation into a T1D mouse model. Human islet function in vivo was assessed using islet transplantation under the kidney capsule of immunodeficient mice prior to STZ destruction of endogenous mouse beta cells to model T1DM. RESULTS: Pretreatment with a cocktail of MSC-secreted peptides increased GSIS in vitro and protected against cytokine-induced apoptosis in human islets isolated from nine donors. Animals transplanted with either treated or untreated human islets remained normoglycaemic for up to 28 days after STZ-administration to ablate the endogenous mouse beta cells, whereas non-transplanted animals showed significantly increased blood glucose immediately after STZ administration. Removal of the human islet graft by nephrectomy resulted in rapid increases in blood glucose to similar levels as the non-transplanted controls. Pretreating human islets with the MSC-derived cocktail significantly improved glucose tolerance in graft recipients, consistent with enhanced functional survival of the treated islets in vivo. CONCLUSION: Pretreating human islets before transplantation with a defined cocktail of MSC-derived molecules could be employed to improve the quality of human islets for transplantation therapy for type 1 diabetes.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Adulto , Pessoa de Meia-Idade , Ilhotas Pancreáticas/metabolismo , Insulina/metabolismo , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Experimental/terapia , Transplante das Ilhotas Pancreáticas/métodos , Células-Tronco Mesenquimais/metabolismo , Glucose/farmacologia , Glucose/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças
3.
Front Microbiol ; 13: 1004679, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36386661

RESUMO

The intestinal microbiota plays an important role in host metabolism via production of dietary metabolites. Microbiota imbalances are linked to type 2 diabetes (T2D), but dietary modification of the microbiota may promote glycemic control. Using a rodent model of T2D and an in vitro gut model system, this study investigated whether differences in gut microbiota between control mice and mice fed a high-fat, high-fructose (HFHFr) diet influenced the production of phenolic acid metabolites following fermentation of wholegrain (WW) and control wheat (CW). In addition, the study assessed whether changes in metabolite profiles affected pancreatic beta cell function. Fecal samples from control or HFHFr-fed mice were fermented in vitro with 0.1% (w/v) WW or CW for 0, 6, and 24 h. Microbiota composition was determined by bacterial 16S rRNA sequencing and phenolic acid (PA) profiles by UPLC-MS/MS. Cell viability, apoptosis and insulin release from pancreatic MIN6 beta cells and primary mouse islets were assessed in response to fermentation supernatants and selected PAs. HFHFr mice exhibited an overall dysbiotic microbiota with an increase in abundance of proteobacterial taxa (particularly Oxalobacteraceae) and Lachnospiraceae, and a decrease in Lactobacillus. A trend toward restoration of diversity and compositional reorganization was observed following WW fermentation at 6 h, although after 24 h, the HFHFr microbiota was monodominated by Cupriavidus. In parallel, the PA profile was significantly altered in the HFHFr group compared to controls with decreased levels of 3-OH-benzoic acid, 4-OH-benzoic acid, isoferulic acid and ferulic acid at 6 h of WW fermentation. In pancreatic beta cells, exposure to pre-fermentation supernatants led to inhibition of insulin release, which was reversed over fermentation time. We conclude that HFHFr mice as a model of T2D are characterized by a dysbiotic microbiota, which is modulated by the in vitro fermentation of WW. The differences in microbiota composition have implications for PA profile dynamics and for the secretory capacity of pancreatic beta cells.

4.
Diabet Med ; 39(12): e14962, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36151994

RESUMO

AIMS: Beta cell endoplasmic reticulum (ER) stress can cause cellular death and dysfunction and has been implicated in the pathogenesis of diabetes. Animal models of beta cell ER stress are critical in further understanding this and for testing novel diabetes therapeutics. The KINGS mouse is a model of beta cell ER stress driven by a heterozygous mutation in Ins2. In this study, we investigated how beta cell ER stress in the KINGS mouse drives diabetes. METHODS: We investigated whether the unfolded protein response (UPR) was activated in islets isolated from male and female KINGS mice and whether this impacted beta cell mass and turnover. RESULTS: Whilst the UPR was up-regulated in KINGS islets, with increased protein expression of markers of all three UPR arms, this was not associated with a mass loss of beta cells; beta cell apoptosis rates did not increase until after the development of overt diabetes, and did not lead to substantial changes in beta cell mass. CONCLUSION: We propose that the KINGS mouse represents a model where beta cell maladaptive UPR signalling drives diabetes development without causing mass beta cell loss.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina , Feminino , Masculino , Camundongos , Humanos , Animais , Estresse do Retículo Endoplasmático/fisiologia , Células Secretoras de Insulina/metabolismo , Resposta a Proteínas não Dobradas , Diabetes Mellitus/metabolismo , Apoptose
5.
Diabetes Obes Metab ; 24(11): 2241-2252, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35815375

RESUMO

AIM: To establish the impact of sex, dosing route, fasting duration and acute habituation stress on glucose tolerance test (GTT) measurements used in the preclinical evaluation of potential glucose-modulating therapeutics. METHODS: Adult male and female C57Bl/6J mice, implanted with HD-XG glucose telemetry devices, were fasted for 16 hours or 6 hours following acute habituation stress due to whole cage change, cage change with retention of used bedding or no cage change prior to intraperitoneal (IP) GTTs. To evaluate protocol refinement and sex on the ability of the GTT to detect drug effects, we administered 250 mg/kg oral metformin or 10 nmol/kg IP exendin-4 using optimized protocols. RESULTS: Female mice were less sensitive to human intervention when initiating fasting. Following a 6-hour fast, retention of bedding whilst changing the cage base promotes quicker stabilization of basal blood glucose in both sexes. Prolonged fasting for 16 hours resulted in an exaggerated GTT response but induced pronounced basal hypoglycaemia. Following GTT protocol optimization the effect of exendin-4 and metformin was equivalent in both sexes, with females showing a more modest but more reproducible GTT response. CONCLUSIONS: Variations in GTT protocol have profound effects on glucose homeostasis. Protocol refinement and/or the use of females still allows for detection of drug effects, providing evidence that more severe phenotypes are not an essential prerequisite when characterizing/validating new drugs.


Assuntos
Glicemia , Metformina , Adulto , Animais , Exenatida , Feminino , Glucose , Teste de Tolerância a Glucose , Humanos , Masculino , Metformina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL
6.
Diabet Med ; 38(12): e14705, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34596274

RESUMO

Mice are used extensively in preclinical diabetes research to model various aspects of blood glucose homeostasis. Careful experimental design is vital for maximising welfare and improving reproducibility of data. Alongside decisions regarding physiological characteristics of the animal cohort (e.g., sex, strain and age), experimental protocols must also be carefully considered. This includes choosing relevant end points of interest and understanding what information they can provide and what their limitations are. Details of experimental protocols must, therefore, be carefully planned during the experimental design stage, especially considering the impact of researcher interventions on preclinical end points. Indeed, in line with the 3Rs of animal research, experiments should be refined where possible to maximise welfare. The role of welfare may be particularly pertinent in preclinical diabetes research as blood glucose concentrations are directly altered by physiological stress responses. Despite the potential impact of variations in experimental protocols, there is distinct lack of standardisation and consistency throughout the literature with regards to several experimental procedures including fasting, cage changing and glucose tolerance test protocol. This review firstly highlights practical considerations with regard to the choice of end points in preclinical diabetes research and the potential for novel technologies such as continuous glucose monitoring and glucose clamping techniques to improve data resolution. The potential influence of differing experimental protocols and in vivo procedures on both welfare and experimental outcomes is then discussed with focus on standardisation, consistency and full disclosure of methods.


Assuntos
Pesquisa Biomédica/métodos , Automonitorização da Glicemia/métodos , Glicemia/metabolismo , Diabetes Mellitus Experimental/diagnóstico , Diabetes Mellitus/diagnóstico , Animais , Diabetes Mellitus/sangue , Diabetes Mellitus Experimental/sangue , Teste de Tolerância a Glucose , Camundongos
7.
Diabet Med ; 38(12): e14711, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34614258

RESUMO

Diabetes mellitus is characterised by hyperglycaemia, which results from an absolute or relative lack of insulin. Chronic and acute hyperglycaemia are associated with a range of health complications and an overall increased risk of mortality. Mouse models are vital in understanding the pathogenesis of this disease and its complications, as well as for developing new diabetes therapeutics. However, for experimental questions to be suitably tested, it is critical that factors inherent to the animal model are considered, as these can have profound impacts on experimental outcome, data reproducibility and robustness. In this review, we discuss key considerations relating to model choice, physiological characteristics (such as age, sex and genetic background) and husbandry practices and explore the impact of these on common experimental readouts used in preclinical diabetes research.


Assuntos
Pesquisa Biomédica/métodos , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 2/terapia , Gerenciamento Clínico , Resistência à Insulina/fisiologia , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Camundongos
8.
Mol Metab ; 53: 101285, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34224919

RESUMO

OBJECTIVE: Members of the adhesion G protein-coupled receptor (aGPCR) subfamily are important actors in metabolic processes, with GPR56 (ADGRG1) emerging as a possible target for type 2 diabetes therapy. GPR56 can be activated by collagen III, its endogenous ligand, and by a synthetic seven amino-acid peptide (TYFAVLM; P7) contained within the GPR56 Stachel sequence. However, the mechanisms regulating GPR56 trafficking dynamics and agonist activities are not yet clear. METHODS: Here, we introduced SNAPf-tag into the N-terminal segment of GPR56 to monitor GPR56 cellular activity in situ. Confocal and super-resolution microscopy were used to investigate the trafficking pattern of GPR56 in native MIN6 ß-cells and in MIN6 ß-cells where GPR56 had been deleted by CRISPR-Cas9 gene editing. Insulin secretion, changes in intracellular calcium, and ß-cell apoptosis were determined by radioimmunoassay, single-cell calcium microfluorimetry, and measuring caspase 3/7 activities, respectively, in MIN6 ß-cells and human islets. RESULTS: SNAP-tag labelling indicated that GPR56 predominantly underwent constitutive internalisation in the absence of an exogenous agonist, unlike GLP-1R. Collagen III further stimulated GPR56 internalisation, whereas P7 was without significant effect. The overexpression of GPR56 in MIN6 ß-cells did not affect insulin secretion. However, it was associated with reduced ß-cell apoptosis, while the deletion of GPR56 made MIN6 ß-cells more susceptible to cytokine-induced apoptosis. P7 induced a rapid increase in the intracellular calcium in MIN6 ß-cells (in a GPR56-dependent manner) and human islets, and it also caused a sustained and reversible increase in insulin secretion from human islets. Collagen III protected human islets from cytokine-induced apoptosis, while P7 was without significant effect. CONCLUSIONS: These data indicate that GPR56 exhibits both agonist-dependent and -independent trafficking in ß-cells and suggest that while GPR56 undergoes constitutive signalling, it can also respond to its ligands when required. We have also identified that constitutive and agonist-dependent GPR56 activation is coupled to protect ß-cells against apoptosis, offering a potential therapeutic target to maintain ß-cell mass in type 2 diabetes.


Assuntos
Células Secretoras de Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células HEK293 , Humanos , Microscopia Confocal , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética
9.
Mol Metab ; 43: 101112, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33157254

RESUMO

OBJECTIVE: Metabolic diseases are an increasing problem in society with the brain-metabolic axis as a master regulator of the human body for sustaining homeostasis under metabolic stress. However, metabolic inflammation and disease will trigger sustained activation of the hypothalamic-pituitary-adrenal axis. In this study, we investigated the role of metabolic stress on progenitor cells in the hypothalamic-pituitary-adrenal axis. METHODS: In vitro, we applied insulin and leptin to murine progenitor cells isolated from the pituitary and adrenal cortex and examined the role of these hormones on proliferation and differentiation. In vivo, we investigated two different mouse models of metabolic disease, obesity in leptin-deficient ob/ob mice and obesity achieved via feeding with a high-fat diet. RESULTS: Insulin was shown to lead to enhanced proliferation and differentiation of both pituitary and adrenocortical progenitors. No alterations in the progenitors were noted in our chronic metabolic stress models. However, hyperactivation of the hypothalamic-pituitary-adrenal axis was observed and the expression of the appetite-regulating genes Npy and Agrp changed in both the hypothalamus and adrenal. CONCLUSIONS: It is well-known that chronic stress and stress hormones such as glucocorticoids can induce metabolic changes including obesity and diabetes. In this article, we show for the first time that this might be based on an early sensitization of stem cells of the hypothalamic-pituitary-adrenal axis. Thus, pituitary and adrenal progenitor cells exposed to high levels of insulin are metabolically primed to a hyper-functional state leading to enhanced hormone production. Likewise, obese animals exhibit a hyperactive hypothalamic-pituitary-adrenal axis leading to adrenal hyperplasia. This might explain how stress in early life can increase the risk for developing metabolic syndrome in adulthood.


Assuntos
Insulina/metabolismo , Obesidade/metabolismo , Células-Tronco/fisiologia , Estresse Fisiológico/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Insulina/farmacologia , Leptina/metabolismo , Leptina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Camundongos Transgênicos , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/fisiologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
10.
Diabetes ; 69(12): 2667-2677, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32994272

RESUMO

Animal models are important tools in diabetes research because ethical and logistical constraints limit access to human tissue. ß-Cell dysfunction is a common contributor to the pathogenesis of most types of diabetes. Spontaneous hyperglycemia was developed in a colony of C57BL/6J mice at King's College London (KCL). Sequencing identified a mutation in the Ins2 gene, causing a glycine-to-serine substitution at position 32 on the B chain of the preproinsulin 2 molecule. Mice with the Ins2 +/G32S mutation were named KCL Ins2 G32S (KINGS) mice. The same mutation in humans (rs80356664) causes dominantly inherited neonatal diabetes. Mice were characterized, and ß-cell function was investigated. Male mice became overtly diabetic at ∼5 weeks of age, whereas female mice had only slightly elevated nonfasting glycemia. Islets showed decreased insulin content and impaired glucose-induced insulin secretion, which was more severe in males. Transmission electron microscopy and studies of gene and protein expression showed ß-cell endoplasmic reticulum (ER) stress in both sexes. Despite this, ß-cell numbers were only slightly reduced in older animals. In conclusion, the KINGS mouse is a novel model of a human form of diabetes that may be useful to study ß-cell responses to ER stress.


Assuntos
Diabetes Mellitus/genética , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/fisiologia , Células Secretoras de Insulina/metabolismo , Animais , Ecossistema , Feminino , Teste de Tolerância a Glucose , Humanos , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos , Mutação , Polimorfismo de Nucleotídeo Único
11.
Sci Rep ; 10(1): 15741, 2020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-32978479

RESUMO

Regenerative medicine approaches to enhancing beta cell growth and survival represent potential treatments for diabetes. It is known that growth factors such as insulin, IGF-1 and HGF support beta cell growth and survival, but in people with type 2 diabetes the destructive effects of metabolic stress predominate and beta cell death or dysfunction occurs. In this study we explore the novel hypothesis that regulation of growth factor receptor trafficking can be used to promote islet beta cell survival. Growth factor signalling is dependent on the presence of cell surface receptors. Endosomal trafficking and subsequent recycling or degradation of these receptors is controlled by the Rab GTPase family of proteins. We show that Rab7a siRNA inhibition enhances IGF-1 and HGF signalling in beta cells and increases expression of the growth factor receptors IGF-1R and c-Met. Furthermore, Rab7a inhibition promotes beta cell growth and islet survival, and protects against activation of apoptosis and autophagy pathways under conditions of metabolic stress. This study therefore demonstrates that Rab7a-mediated trafficking of growth factor receptors controls beta cell survival. Pharmaceutical Rab7a inhibition may provide a means to promote beta cell survival in the context of metabolic stress and prevent the onset of type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Ilhotas Pancreáticas/citologia , Proteínas Proto-Oncogênicas c-met/metabolismo , RNA Interferente Pequeno/farmacologia , Receptor IGF Tipo 1/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Apoptose , Autofagia , Proliferação de Células , Células Cultivadas , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Transporte Proteico/efeitos dos fármacos , Estresse Fisiológico , Regulação para Cima , Proteínas rab de Ligação ao GTP/antagonistas & inibidores , proteínas de unión al GTP Rab7
12.
Methods Mol Biol ; 2128: 1-10, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32180182

RESUMO

Mouse models of diabetes are important tools used in preclinical diabetes research. However, when working with these models, it is important to consider factors that could influence experimental outcome. This is particularly important given the wide variety of models available, each with specific characteristics that could be influenced by extrinsic or intrinsic factors. Blood glucose concentrations, a commonly used and valid endpoint in these models, are particularly susceptible to manipulation by these factors. These include potential effects of intrinsic factors such as strain, sex, and age and extrinsic factors such as husbandry practices and experimental protocols. These variables should therefore be taken into consideration when the model is chosen and the experiments are designed. This chapter outlines common variables that can impact the phenotype of a model, as well as describes the methods used for assessing onset of diabetes and monitoring diabetic mice.


Assuntos
Criação de Animais Domésticos/métodos , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/urina , Fatores Etários , Idade de Início , Animais , Glicemia/análise , Diabetes Mellitus Experimental/diagnóstico , Diabetes Mellitus Experimental/tratamento farmacológico , Técnicas e Procedimentos Diagnósticos , Feminino , Glicosúria , Insulina/administração & dosagem , Masculino , Camundongos , Camundongos Mutantes , Fenótipo , Fatores Sexuais , Fatores de Tempo , Urinálise/métodos
13.
Methods Mol Biol ; 2128: 135-147, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32180191

RESUMO

Streptozotocin (STZ) selectively destroys beta cells and is widely used to induce experimental diabetes in rodents. Rodent beta cells are very sensitive to the toxic effects of STZ, while human beta cells are highly resistant to STZ. Taking advantage of this characteristic, here, we describe two protocols for the induction of STZ-diabetes. In the first model, hyperglycemia is induced prior to islet transplantation, whereas in the second model, STZ is injected after islet transplantation. The former model has many applications and thus is the most commonly used method. However, when implanting human islets into mice, there are clear benefits to administering STZ after the transplantation. It reduces the cost and burden of experiments and the number of human islets needed for transplantation and improves the welfare and survival of animals used in the experiments. In both methods, a key step in the experimental protocol is to remove the graft-bearing kidney at the end of the experiment and monitor onset of hyperglycemia. This can be used to demonstrate that the glycemic control of the animal is due to the engrafted islets and not regeneration of endogenous beta cells. This chapter outlines protocols of administering streptozotocin pre- and post-islet transplantation in mice as well as nephrectomy to remove the graft-bearing kidney.


Assuntos
Diabetes Mellitus Experimental/induzido quimicamente , Transplante das Ilhotas Pancreáticas/métodos , Nefrectomia/métodos , Animais , Glicemia/análise , Feminino , Humanos , Hiperglicemia/sangue , Células Secretoras de Insulina/efeitos dos fármacos , Rim/cirurgia , Masculino , Camundongos , Estreptozocina/administração & dosagem
14.
Methods Mol Biol ; 2128: 225-239, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32180197

RESUMO

Measurement of blood glucose concentration is a common end point in studies using animal models of diabetes. Usually a blood glucose meter is used to measure non-fasted blood glucose concentrations, typically at frequencies of between 1 and 7 times per week. This process involves pricking the tip of the tail to collect a small blood sample (0.5-5 µL), which could potentially cause a stress response and affect blood glucose concentrations. Moreover, with blood glucose concentrations constantly fluctuating in response to feeding and activity, a single-point measurement can easily misrepresent the actual glycemic control of the animal. In this chapter, we discuss the use of continuous glucose monitoring in mice by radio-telemetry which allows second-by-second changes in blood glucose to be captured without restraining the mouse. Glucose excursions rather than single-point measurements may prove more useful in detecting effects of treatments, and lack of handling may avoid stress responses causing artefacts. We outline what is involved in implanting such devices into mice including some practical tips to maximize success.


Assuntos
Automonitorização da Glicemia/instrumentação , Glicemia/análise , Monitorização Fisiológica/métodos , Implantação de Prótese/métodos , Telemetria/instrumentação , Telemetria/métodos , Animais , Estado de Consciência/fisiologia , Diabetes Mellitus Experimental , Locomoção/fisiologia , Próteses e Implantes
15.
Stem Cells ; 38(4): 574-584, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31912945

RESUMO

Pretransplant islet culture is associated with the loss of islet cell mass and insulin secretory function. Insulin secretion from islet ß-cells is primarily controlled by mitochondrial ATP generation in response to elevations in extracellular glucose. Coculture of islets with mesenchymal stromal cells (MSCs) improves islet insulin secretory function in vitro, which correlates with superior islet graft function in vivo. This study aimed to determine whether the improved islet function is associated with mitochondrial transfer from MSCs to cocultured islets. We have demonstrated mitochondrial transfer from human adipose MSCs to human islet ß-cells in coculture. Fluorescence imaging showed that mitochondrial transfer occurs, at least partially, through tunneling nanotube (TNT)-like structures. The extent of mitochondrial transfer to clinically relevant human islets was greater than that to experimental mouse islets. Human islets are subjected to more extreme cellular stressors than mouse islets, which may induce "danger signals" for MSCs, initiating the donation of MSC-derived mitochondria to human islet ß-cells. Our observations of increased MSC-mediated mitochondria transfer to hypoxia-exposed mouse islets are consistent with this and suggest that MSCs are most effective in supporting the secretory function of compromised ß-cells. Ensuring optimal MSC-derived mitochondria transfer in preculture and/or cotransplantation strategies could be used to maximize the therapeutic efficacy of MSCs, thus enabling the more widespread application of clinical islet transplantation.


Assuntos
Diabetes Mellitus Experimental/terapia , Células Secretoras de Insulina/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Células-Tronco Mesenquimais/metabolismo , Mitocôndrias/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos
16.
Methods Mol Biol ; 2076: 265-280, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31586334

RESUMO

Islet transplantation is a potential treatment for Type 1 diabetes; however, improvements need to be made before it could become clinically widely available. In preclinical studies, the mouse is often used to model islet transplantation, with most studies aiming to improve transplantation outcome by manipulating the islets prior to transplantation or by treating the recipient mouse. Here, we describe the process of islet transplantation in the mouse, including how one can make the mouse diabetic, isolate donor islets, and transplant the islets into two different sites. Finally, we discuss how to assess the outcome of the transplantation in order to determine whether the experimental intervention has been beneficial.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Animais , Glicemia , Separação Celular/métodos , Diabetes Mellitus Experimental , Rejeição de Enxerto , Sobrevivência de Enxerto , Xenoenxertos , Insulina/sangue , Ilhotas Pancreáticas/citologia , Transplante das Ilhotas Pancreáticas/efeitos adversos , Transplante das Ilhotas Pancreáticas/métodos , Camundongos , Transplante Homólogo , Resultado do Tratamento
17.
Nat Commun ; 10(1): 3350, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31350390

RESUMO

The liver parenchyma is composed of hepatocytes and bile duct epithelial cells (BECs). Controversy exists regarding the cellular origin of human liver parenchymal tissue generation during embryonic development, homeostasis or repair. Here we report the existence of a hepatobiliary hybrid progenitor (HHyP) population in human foetal liver using single-cell RNA sequencing. HHyPs are anatomically restricted to the ductal plate of foetal liver and maintain a transcriptional profile distinct from foetal hepatocytes, mature hepatocytes and mature BECs. In addition, molecular heterogeneity within the EpCAM+ population of freshly isolated foetal and adult human liver identifies diverse gene expression signatures of hepatic and biliary lineage potential. Finally, we FACS isolate foetal HHyPs and confirm their hybrid progenitor phenotype in vivo. Our study suggests that hepatobiliary progenitor cells previously identified in mice also exist in humans, and can be distinguished from other parenchymal populations, including mature BECs, by distinct gene expression profiles.


Assuntos
Fígado/citologia , Transcrição Gênica , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Molécula de Adesão da Célula Epitelial/genética , Molécula de Adesão da Célula Epitelial/metabolismo , Feto/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Análise de Célula Única , Células-Tronco/citologia , Células-Tronco/metabolismo
18.
Stem Cells Transl Med ; 8(9): 935-944, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31066521

RESUMO

Islet transplantation has the potential to cure type 1 diabetes, but current transplantation protocols are not optimal and there is extensive loss of islet ß-cell insulin secretory function during the immediate post-transplantation period. Studies using experimental models of diabetes have shown that the coculture of islets with mesenchymal stromal cells (MSCs) prior to transplantation improves graft function, but several variables differed among research groups (e.g., type of MSCs used and the treatment conditions). We have therefore assessed the effects of MSCs on mouse and human islets by investigating the importance of tissue source for MSCs, the coculture protocol configuration and length, the effect of activated MSCs, and different ß-cell secretory stimuli. MSCs derived from adipose tissue (aMSCs) were the most effective at supporting ß-cell insulin secretion in both mouse and human islets, in a direct contact coculture configuration. Preculture with aMSCs enhanced both phases of glucose-induced insulin secretion and further enhanced secretory responses to the non-nutrients carbachol and arginine. These effects required a coculture period of 48-72 hours and were not dependent on activation of the MSCs. Thus, direct contact coculture with autologous, adipose-derived MSCs for a minimum of 48 hours before implantation is likely to be an effective addition to human islet transplantation protocols. Stem Cells Translational Medicine 2019;8:935&944.


Assuntos
Ilhotas Pancreáticas/citologia , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/citologia , Animais , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Técnicas de Cocultura , Glucose/farmacologia , Humanos , Secreção de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Pâncreas/citologia , Fator de Necrose Tumoral alfa/farmacologia
19.
Stem Cells Transl Med ; 8(2): 124-137, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30456803

RESUMO

Recent advancements in the production of hepatocytes from human pluripotent stem cells (hPSC-Heps) afford tremendous possibilities for treatment of patients with liver disease. Validated current good manufacturing practice (cGMP) lines are an essential prerequisite for such applications but have only recently been established. Whether such cGMP lines are capable of hepatic differentiation is not known. To address this knowledge gap, we examined the proficiency of three recently derived cGMP lines (two hiPSC and one hESC) to differentiate into hepatocytes and their suitability for therapy. hPSC-Heps generated using a chemically defined four-step hepatic differentiation protocol uniformly demonstrated highly reproducible phenotypes and functionality. Seeding into a 3D poly(ethylene glycol)-diacrylate fabricated inverted colloid crystal scaffold converted these immature progenitors into more advanced hepatic tissue structures. Hepatic constructs could also be successfully encapsulated into the immune-privileged material alginate and remained viable as well as functional upon transplantation into immune competent mice. This is the first report we are aware of demonstrating cGMP-compliant hPSCs can generate cells with advanced hepatic function potentially suitable for future therapeutic applications. Stem Cells Translational Medicine 2019;8:124&14.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/normas , Hepatócitos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Técnicas de Cultura de Células/normas , Diferenciação Celular/fisiologia , Linhagem Celular , Humanos , Fígado/citologia , Camundongos
20.
Cytotherapy ; 20(12): 1427-1436, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30377040

RESUMO

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) enhance islet function both in vitro and in vivo, at least in part by secreting ligands that activate islet G-protein coupled receptors (GPCRs). We assessed whether pre-treatment with a defined "cocktail" of MSC-secreted GPCR ligands enhances islet functional survival in vitro and improves the outcomes of islet transplantation in an experimental model of diabetes. METHODS: Isolated islets were cultured for 48 h with ANXA1, SDF-1 or C3a, alone or in combination. Glucose-stimulated insulin secretion (GSIS) and cytokine-induced apoptosis were measured immediately after the 48 h culture period and at 24 h or 72 h following removal of the ligands from the culture media. Islets were syngeneically transplanted underneath the kidney capsule of streptozotocin-induced diabetic C57BL/6 mice and blood glucose levels monitored for 28 days. RESULTS: Pre-culturing islets with a cocktail of ANXA1/SDF-1/C3a potentiated GSIS and protected islet cells from cytokine-induced apoptosis in vitro. These effects were maintained for up to 72 h after the removal of the factors from the culture medium, suggesting a sustained protection of islet graft functional survival during the immediate post-transplantation period. Islets pre-treated with the cocktail of MSC secretory factors were more effective in reducing blood glucose in diabetic mice, consistent with their improved functional survival in vivo. DISCUSSION: Pre-culturing islets with a cocktail of MSC secretory products offers a well-defined, cell-free approach to improve clinical islet transplantation outcomes while avoiding many of the safety, regulatory and logistical hurdles of incorporating MSCs into transplantation protocols.


Assuntos
Quimiocina CXCL12/farmacologia , Complemento C3a/farmacologia , Transplante das Ilhotas Pancreáticas/métodos , Células-Tronco Mesenquimais/metabolismo , Animais , Anexina A1/genética , Anexina A1/metabolismo , Anexina A1/farmacologia , Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Complemento C3a/genética , Complemento C3a/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Glucose/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Receptores Acoplados a Proteínas G/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...