Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Metabolism ; 151: 155762, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38122893

RESUMO

BACKGROUND: Obesity and type 2 diabetes frequently have metabolic dysfunction-associated steatotic liver disease (MASLD) including steatohepatitis (MASH). In obesity, the liver may adapt its oxidative capacity, but the role of mitochondrial turnover in MASLD remains uncertain. METHODS: This cross-sectional study compared individuals with class III obesity (n = 8/group) without (control, OBE CON; NAFLD activity score: 0.4 ± 0.1) or with steatosis (OBE MASL, 2.3 ± 0.4), or MASH (OBE MASH, 5.3 ± 0.3, p < 0.05 vs. other groups). Hepatic mitochondrial ultrastructure was assessed by transmission electron microscopy, mitochondrial respiration by high-resolution respirometry, biomarkers of mitochondrial quality control and endoplasmic reticulum (ER) stress by Western Blot. RESULTS: Mitochondrial oxidative capacity was 31 % higher in OBE MASL, but 25 % lower in OBE MASH (p < 0.05 vs. OBE CON). OBE MASH showed ~1.5fold lower mitochondrial number, but ~1.2-1.5fold higher diameter and area (p < 0.001 vs. other groups). Biomarkers of autophagy (p62), mitophagy (PINK1, PARKIN), fission (DRP-1, FIS1) and fusion (MFN1/2, OPA1) were reduced in OBE MASH (p < 0.05 vs. OBE CON). OBE MASL showed lower p62, p-PARKIN/PARKIN, and p-DRP-1 (p < 0.05 vs. OBE CON). OBE MASL and MASH showed higher ER stress markers (PERK, ATF4, p-eIF2α-S51/eIF2α; p < 0.05 vs. OBE CON). Mitochondrial diameter associated inversely with fusion/fission biomarkers and with oxidative capacity, but positively with H2O2. CONCLUSION: Humans with hepatic steatosis already exhibit impaired mitochondrial turnover, despite upregulated oxidative capacity, and evidence for ER stress. In MASH, oxidative stress likely mediates progressive decline of mitochondrial turnover, ultrastructure and respiration indicating that mitochondrial quality control is key for energy metabolism and may have potential for targeting MASH. ClinGovTrial:NCT01477957.


Assuntos
Diabetes Mellitus Tipo 2 , Fígado Gorduroso , Hepatopatia Gordurosa não Alcoólica , Humanos , Estudos Transversais , Peróxido de Hidrogênio , Mitofagia , Obesidade/complicações , Obesidade/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Biomarcadores
2.
Nat Cell Biol ; 25(1): 20-29, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36543979

RESUMO

Impaired proinsulin-to-insulin processing in pancreatic ß-cells is a key defective step in both type 1 diabetes and type 2 diabetes (T2D) (refs. 1,2), but the mechanisms involved remain to be defined. Altered metabolism of sphingolipids (SLs) has been linked to development of obesity, type 1 diabetes and T2D (refs. 3-8); nonetheless, the role of specific SL species in ß-cell function and demise is unclear. Here we define the lipid signature of T2D-associated ß-cell failure, including an imbalance of specific very-long-chain SLs and long-chain SLs. ß-cell-specific ablation of CerS2, the enzyme necessary for generation of very-long-chain SLs, selectively reduces insulin content, impairs insulin secretion and disturbs systemic glucose tolerance in multiple complementary models. In contrast, ablation of long-chain-SL-synthesizing enzymes has no effect on insulin content. By quantitatively defining the SL-protein interactome, we reveal that CerS2 ablation affects SL binding to several endoplasmic reticulum-Golgi transport proteins, including Tmed2, which we define as an endogenous regulator of the essential proinsulin processing enzyme Pcsk1. Our study uncovers roles for specific SL subtypes and SL-binding proteins in ß-cell function and T2D-associated ß-cell failure.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Proinsulina/genética , Proinsulina/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Esfingolipídeos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Insulina/metabolismo , Homeostase , Proteínas de Transporte/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo
3.
Endocrinology ; 159(4): 1748-1761, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29481597

RESUMO

The Rab guanosine triphosphatase-activating protein (RabGAP) TBC1D1 has been shown to be a key regulator of glucose and lipid metabolism in skeletal muscle. Its function in pancreatic islets, however, is not yet fully understood. Here, we aimed to clarify the specific impact of TBC1D1 on insulin secretion and substrate use in pancreatic islets. We analyzed the dynamics of glucose-stimulated insulin secretion (GSIS) and lipid metabolism in isolated islets from Tbc1d1-deficient (D1KO) mice. To further investigate the underlying cellular mechanisms, we conducted pharmacological studies in these islets. In addition, we determined morphology and number of both pancreatic islets and insulin vesicles in ß-cells using light and transmission electron microscopy. Isolated pancreatic islets from D1KO mice exhibited substantially increased GSIS compared with wild-type (WT) controls. This was attributed to both enhanced first and second phase of insulin secretion, and this enhanced secretion persisted during repetitive glucose stimuli. Studies with sulfonylureas or KCl in isolated islets demonstrated that TBC1D1 exerts its function via a signaling pathway at the level of membrane depolarization. In line, ultrastructural analysis of isolated pancreatic islets revealed both higher insulin-granule density and number of docked granules in ß-cells from D1KO mice compared with WT controls. Like in skeletal muscle, lipid use in isolated islets was enhanced upon D1KO, presumably as a result of a higher mitochondrial fission rate and/or higher mitochondrial activity. Our results clearly demonstrate a dual role of TBC1D1 in controlling substrate metabolism of the pancreatic islet.


Assuntos
Ácidos Graxos/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Metabolismo dos Lipídeos/genética , Animais , Proteínas Ativadoras de GTPase/genética , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Knockout
4.
Nat Commun ; 7: 13560, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27995929

RESUMO

An important question is how growing tissues establish a blood vessel network. Here we study vascular network formation in pancreatic islets, endocrine tissues derived from pancreatic epithelium. We find that depletion of integrin-linked kinase (ILK) in the pancreatic epithelial cells of mice results in glucose intolerance due to a loss of the intra-islet vasculature. In turn, blood vessels accumulate at the islet periphery. Neither alterations in endothelial cell proliferation, apoptosis, morphology, Vegfa expression and VEGF-A secretion nor 'empty sleeves' of vascular basement membrane are found. Instead, biophysical experiments reveal that the biomechanical properties of pancreatic islet cells, such as their actomyosin-mediated cortex tension and adhesive forces to endothelial cells, are significantly changed. These results suggest that a sorting event is driving the segregation of endothelial and epithelial cells and indicate that the epithelial biomechanical properties determine whether the blood vasculature invades or envelops a growing epithelial tissue.


Assuntos
Epitélio/irrigação sanguínea , Epitélio/fisiologia , Ilhotas Pancreáticas/irrigação sanguínea , Proteínas Serina-Treonina Quinases/fisiologia , Actomiosina/fisiologia , Animais , Membrana Basal/fisiologia , Fenômenos Biomecânicos , Adesão Celular/fisiologia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Células Epiteliais/fisiologia , Feminino , Intolerância à Glucose/fisiopatologia , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
PLoS One ; 10(9): e0138535, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26422139

RESUMO

A hallmark feature of type 1 and type 2 diabetes mellitus is the progressive dysfunction and loss of insulin-producing pancreatic beta cells, and inflammatory cytokines are known to trigger beta cell death. Here we asked whether the anti-oxidant protein DJ-1 encoded by the Parkinson's disease gene PARK7 protects islet cells from cytokine- and streptozotocin-mediated cell death. Wild type and DJ-1 knockout mice (KO) were treated with multiple low doses of streptozotocin (MLDS) to induce inflammatory beta cell stress and cell death. Subsequently, glucose tolerance tests were performed, and plasma insulin as well as fasting and random blood glucose concentrations were monitored. Mitochondrial morphology and number of insulin granules were quantified in beta cells. Moreover, islet cell damage was determined in vitro after streptozotocin and cytokine treatment of isolated wild type and DJ-1 KO islets using calcein AM/ethidium homodimer-1 staining and TUNEL staining. Compared to wild type mice, DJ-1 KO mice became diabetic following MLDS treatment. Insulin concentrations were substantially reduced, and fasting blood glucose concentrations were significantly higher in MLDS-treated DJ-1 KO mice compared to equally treated wild type mice. Rates of beta cell apoptosis upon MLDS treatment were twofold higher in DJ-1 KO mice compared to wild type mice, and in vitro inflammatory cytokines led to twice as much beta cell death in pancreatic islets from DJ-1 KO mice versus those of wild type mice. In conclusion, this study identified the anti-oxidant protein DJ-1 as being capable of protecting pancreatic islet cells from cell death induced by an inflammatory and cytotoxic setting.


Assuntos
Citocinas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Proteínas Oncogênicas/metabolismo , Peroxirredoxinas/metabolismo , Animais , Morte Celular , Citocinas/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Proteínas Oncogênicas/genética , Peroxirredoxinas/genética , Proteína Desglicase DJ-1 , Vesículas Secretórias/genética , Vesículas Secretórias/metabolismo
6.
PLoS One ; 10(9): e0137043, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26331477

RESUMO

BACKGROUND: Glomerular podocytes are highly differentiated cells that are key components of the kidney filtration units. The podocyte cytoskeleton builds the basis for the dynamic podocyte cytoarchitecture and plays a central role for proper podocyte function. Recent studies implicate that immunosuppressive agents including the mTOR-inhibitor everolimus have a protective role directly on the stability of the podocyte actin cytoskeleton. In contrast, a potential stabilization of microtubules by everolimus has not been studied so far. METHODS: To elucidate mechanisms underlying mTOR-inhibitor mediated cytoskeletal rearrangements, we carried out microarray gene expression studies to identify target genes and corresponding pathways in response to everolimus. We analyzed the effect of everolimus in a puromycin aminonucleoside experimental in vitro model of podocyte injury. RESULTS: Upon treatment with puromycin aminonucleoside, microarray analysis revealed gene clusters involved in cytoskeletal reorganization, cell adhesion, migration and extracellular matrix composition to be affected. Everolimus was capable of protecting podocytes from injury, both on transcriptional and protein level. Rescued genes included tubulin beta 2B class IIb (TUBB2B) and doublecortin domain containing 2 (DCDC2), both involved in microtubule structure formation in neuronal cells but not identified in podocytes so far. Validating gene expression data, Western-blot analysis in cultured podocytes demonstrated an increase of TUBB2B and DCDC2 protein after everolimus treatment, and immunohistochemistry in healthy control kidneys confirmed a podocyte-specific expression. Interestingly, Tubb2bbrdp/brdp mice revealed a delay in glomerular podocyte development as showed by podocyte-specific markers Wilm's tumour 1, Podocin, Nephrin and Synaptopodin. CONCLUSIONS: Taken together, our study suggests that off-target, non-immune mediated effects of the mTOR-inhibitor everolimus on the podocyte cytoskeleton might involve regulation of microtubules, revealing a potential novel role of TUBB2B and DCDC2 in glomerular podocyte development.


Assuntos
Everolimo/farmacologia , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/efeitos dos fármacos , Podócitos/efeitos dos fármacos , Tubulina (Proteína)/genética , Animais , Adesão Celular , Linhagem Celular Transformada , Humanos , Rim/metabolismo , Camundongos , Camundongos Mutantes , Microtúbulos/metabolismo , Podócitos/metabolismo , Transcriptoma
7.
Diabetes ; 63(3): 1148-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24319115

RESUMO

Immune-mediated processes have been implicated in the pathogenesis of diabetic polyneuropathy. Langerhans cells (LCs) are the sole dendritic cell type located in the healthy epidermis and exert tolerogenic immune functions. We aimed to determine whether alterations in cutaneous LC density and intraepidermal nerve fiber density (IENFD) are present in patients with recently diagnosed type 2 diabetes. Skin biopsy specimens from the distal leg from 96 type 2 diabetic patients and 75 healthy control subjects were used for quantification of LC density and IENFD. LCs and IENFs were labeled using immunohistochemistry. Nerve conduction studies, quantitative sensory testing, and neurological examination were used to assess peripheral nerve function. LC density was markedly reduced in the diabetic group compared with the control group, but did not correlate with reduced IENFD or peripheral nerve function. Multivariate linear regression analysis revealed a strong association between LC density and whole-body insulin sensitivity in women but not in men with diabetes. Prospective studies should establish whether the pronounced reduction of cutaneous LCs detected in recently diagnosed type 2 diabetes could promote a cutaneous immunogenic imbalance toward inflammation predisposing to polyneuropathy and foot ulcers.


Assuntos
Diabetes Mellitus Tipo 2/imunologia , Células de Langerhans/patologia , Pele/imunologia , Adulto , Idoso , Contagem de Células , Diabetes Mellitus Tipo 2/sangue , Feminino , Hemoglobinas Glicadas/análise , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Fibras Nervosas/patologia , Condução Nervosa
8.
Arch Physiol Biochem ; 112(1): 49-58, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16754203

RESUMO

New Zealand Obese (NZO) male mice develop a polygenic juvenile-onset obesity and maturity onset hyperinsulinemia. Approximately 50% transit to chronic hyperglycemia. Here we report on the proliferation of beta cells in relation to both the individual's metabolic status and structural parameters of the endocrine pancreas. Proliferating beta cells were quantified in pancreas sections by immunoenzymatic double staining of Ki-67 protein, as a marker for proliferating cells, and endocrine non-beta cells in order to distinguish them from beta cells. In normoglycemic NZO/Hl males Ki-67 labelling indices (IKi-67) of beta cells varied between 0.14 and 1.5%, and correlated significantly with both serum insulin levels and beta cell size. There was no correlation with the glycemic status. In diabetic males, beta cell size was increased. IKi-67 varied between 1 and 3%. The data suggest that the secretory activity of beta cells triggered by glucose, entailed changes in both beta cell hypertrophy and proliferation. As shown by morphometric measurements, beta cell expansion in diabetic mice was limited, in spite of high IKi-67 values. This suggested increased death rates of beta cells.


Assuntos
Hiperglicemia/patologia , Hiperinsulinismo/patologia , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/patologia , Obesidade/patologia , Animais , Apoptose , Proliferação de Células , Diabetes Mellitus Tipo 1/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Insulina/análise , Células Secretoras de Insulina/química , Antígeno Ki-67/análise , Masculino , Camundongos , Camundongos Mutantes , Camundongos Obesos
9.
Lab Invest ; 82(7): 843-53, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12118086

RESUMO

We report the first combined light and electron microscopic analysis of the pancreas during the development of type 2 diabetes in the New Zealand Obese (NZO) mouse. As in most other polygenic rodent models of type 2 diabetes, hyperglycemia associated with beta cell destruction is male sex-limited. Increasing degrees of hyperinsulinemia and transition to diabetes were clearly reflected by the islet volume fraction, by the beta cell granulation state, and by ultrastructural changes, primarily of the endoplasmic reticulum. One of the unusual histopathologic features of NZO mice of both sexes was the presence of B-lymphocyte enriched leukocytic aggregates in the pancreas. Immunocytochemical analysis of the pancreas of 52-week-old diabetic males indicated enrichment for CD19(+) B lymphocytes. Staining of adjacent sections for CD3 and CD5 indicated CD5 coexpression on some of the CD19(+) cells, suggesting the presence of the B1-B subset associated with generation of natural autoantibodies in other autoimmune-prone New Zealand mouse strains. In addition, plasma cells in peri-insular leukocytic infiltrates were identified by electron microscopy. Hence, although autoimmunity has previously proven to be a secondary manifestation of beta cell destruction in most rodent models of type 2 diabetes, the present observations suggest that B lymphocyte function, in association with male gender, may contribute to the development of insulin resistance and chronic hyperglycemia in the NZO model.


Assuntos
Diabetes Mellitus Tipo 1/patologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Subpopulações de Linfócitos/imunologia , Envelhecimento/imunologia , Animais , Diabetes Mellitus Tipo 1/imunologia , Hiperglicemia/imunologia , Hiperglicemia/patologia , Masculino , Camundongos , Camundongos Mutantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA