Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Neurochem ; 157(6): 1930-1945, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33539571

RESUMO

Ketogenic diets (KDs) alter brain metabolism. Multiple mechanisms may account for their effects, and different brain regions may variably respond. Here, we considered how a KD affects brain neuron and astrocyte transcription. We placed male C57Bl6/N mice on either a 3-month KD or chow diet, generated enriched neuron and astrocyte fractions, and used RNA-Seq to assess transcription. Neurons from KD-treated mice generally showed transcriptional pathway activation while their astrocytes showed a mix of transcriptional pathway suppression and activation. The KD especially affected pathways implicated in mitochondrial and endoplasmic reticulum function, insulin signaling, and inflammation. An unbiased analysis of KD-associated expression changes strongly implicated transcriptional pathways altered in AD, which prompted us to explore in more detail the potential molecular relevance of a KD to AD. Our results indicate a KD differently affects neurons and astrocytes, and provide unbiased evidence that KD-induced brain effects are potentially relevant to neurodegenerative diseases such as AD.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Dieta Cetogênica/métodos , Corpos Cetônicos/metabolismo , Neurônios/metabolismo , Transcrição Gênica/fisiologia , Animais , Dieta Cetogênica/tendências , Corpos Cetônicos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
J Biol Chem ; 294(4): 1363-1379, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30523150

RESUMO

The addition of a single ß-d-GlcNAc sugar (O-GlcNAc) by O-GlcNAc-transferase (OGT) and O-GlcNAc removal by O-GlcNAcase (OGA) maintain homeostatic O-GlcNAc levels on cellular proteins. Changes in protein O-GlcNAcylation regulate cellular differentiation and cell fate decisions, but how these changes affect erythropoiesis, an essential process in blood cell formation, remains unclear. Here, we investigated the role of O-GlcNAcylation in erythropoiesis by using G1E-ER4 cells, which carry the erythroid-specific transcription factor GATA-binding protein 1 (GATA-1) fused to the estrogen receptor (GATA-1-ER) and therefore undergo erythropoiesis after ß-estradiol (E2) addition. We observed that during G1E-ER4 differentiation, overall O-GlcNAc levels decrease, and physical interactions of GATA-1 with both OGT and OGA increase. RNA-Seq-based transcriptome analysis of G1E-ER4 cells differentiated in the presence of the OGA inhibitor Thiamet-G (TMG) revealed changes in expression of 433 GATA-1 target genes. ChIP results indicated that the TMG treatment decreases the occupancy of GATA-1, OGT, and OGA at the GATA-binding site of the lysosomal protein transmembrane 5 (Laptm5) gene promoter. TMG also reduced the expression of genes involved in differentiation of NB4 and HL60 human myeloid leukemia cells, suggesting that O-GlcNAcylation is involved in the regulation of hematopoietic differentiation. Sustained treatment of G1E-ER4 cells with TMG before differentiation reduced hemoglobin-positive cells and increased stem/progenitor cell surface markers. Our results show that alterations in O-GlcNAcylation disrupt transcriptional programs controlling erythropoietic lineage commitment, suggesting a role for O-GlcNAcylation in regulating hematopoietic cell fate.


Assuntos
Acetilglucosamina/metabolismo , Diferenciação Celular , Células Eritroides/citologia , Hematopoese , Homeostase , Células Mieloides/citologia , N-Acetilglucosaminiltransferases/metabolismo , Células Cultivadas , Células Eritroides/metabolismo , Fator de Transcrição GATA1/metabolismo , Humanos , Células Mieloides/fisiologia
3.
J Alzheimers Dis ; 97(4): 1793-1806, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38306050

RESUMO

Background: Some epidemiologic studies associate traumatic brain injury (TBI) with Alzheimer's disease (AD). Objective: To test whether a TBI-induced acceleration of age-related mitochondrial change could potentially mediate the reported TBI-AD association. Methods: We administered unilateral controlled cortical impact (CCI) or sham injuries to 5-month-old C57BL/6J and tau transgenic rTg4510 mice. In the non-transgenics, we assessed behavior (1-5 days, 1 month, and 15 months), lesion size (1 and 15 months), respiratory chain enzymes (1 and 15 months), and mitochondrial DNA copy number (mtDNAcn) (1 and 15 months) after CCI/sham. In the transgenics we quantified post-injury mtDNAcn and tangle burden. Results: In the non-transgenics CCI caused acute behavioral deficits that improved or resolved by 1-month post-injury. Protein-normalized complex I and cytochrome oxidase activities were not significantly altered at 1 or 15 months, although complex I activity in the CCI ipsilesional cortex declined during that period. Hippocampal mtDNAcn was not altered by injury at 1 month, increased with age, and rose to the greatest extent in the CCI contralesional hippocampus. In the injured then aged transgenics, the ipsilesional hippocampus contained less mtDNA and fewer tangles than the contralesional hippocampus; mtDNAcn and tangle counts did not correlate. Conclusions: As mice age their brains increase mtDNAcn as part of a compensatory response that preserves mitochondrial function, and TBI enhances this response. TBI may, therefore, increase the amount of compensation required to preserve late-life mitochondrial function. If TBI does modify AD risk, altering the trajectory or biology of aging-related mitochondrial changes could mediate the effect.


Assuntos
Doença de Alzheimer , Lesões Encefálicas Traumáticas , Camundongos , Animais , Camundongos Endogâmicos C57BL , Lesões Encefálicas Traumáticas/patologia , Encéfalo/patologia , Mitocôndrias/patologia , DNA Mitocondrial/genética , Camundongos Transgênicos , Modelos Animais de Doenças
4.
Clin Immunol ; 148(2): 149-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23770626

RESUMO

In insulin dependent diabetes mellitus (T1D), self-reactive T cells infiltrate pancreatic islets and induce beta cell destruction and dysregulation of blood glucose. A goal is to control only the self-reactive T cells, leaving the remainder of the T cell population free to protect the host. One approach is blockade of the second signal for T cell activation while allowing the first (antigen-specific) signal to occur. This work proposes that small peptides that block interaction of second signals delivered through the counter receptors LFA-1:ICAM-1 will induce attacking T cells (receiving the antigen signal) to become anergic or undergo apoptosis. In NOD mice, the peptides eliminated T cell reactivity against pancreatic antigens and reduced cellular infiltration into islets, which retained stronger density of insulin staining at five weeks after cessation of therapy. In in vitro studies the peptides induced nonresponsiveness during activation of T cells from mice and from human peripheral blood.


Assuntos
Células Secretoras de Insulina/fisiologia , Molécula 1 de Adesão Intercelular/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Peptídeos/farmacologia , Linfócitos T/fisiologia , Envelhecimento , Animais , Glicemia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Células Secretoras de Insulina/imunologia , Molécula 1 de Adesão Intercelular/genética , Antígeno-1 Associado à Função Linfocitária/genética , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T/imunologia
5.
Pharmacol Res ; 76: 58-66, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23891763

RESUMO

The ultimate treatment goal of diabetes is to preserve and restore islet cell function. Treatment of certain diabetic animal models with incretins has been reported to preserve and possibly enhance islet function and promote islet cell growth. The studies reported here detail islet cell anatomy in animals chronically treated with the incretin analog, liraglutide. Our aim was to quantitatively and qualitatively analyze islet cells from diabetic animals treated with vehicle (control) or liraglutide to determine whether normal islet cell anatomy is maintained or enhanced with pharmaceutical treatment. We harvested pancreata from liraglutide and vehicle-treated Zucker Diabetic Fatty (ZDF) rats to examine islet structure and function and obtain isolated islets. Twelve-week-old male rats were assigned to 3 groups: (1) liraglutide-treated diabetic, (2) vehicle-treated diabetic, and (3) lean non-diabetic. Liraglutide was given SC twice daily for 9 weeks. As expected, liraglutide treatment reduced body weight by 15% compared to the vehicle-treated animals, eventually to levels that were not different from lean controls. At the termination of the study, blood glucose was significantly less in the liraglutide-treated rats compared to vehicle treated controls (485.8±22.5 and 547.2±33.1mg/dl, respectively). Insulin content/islet (measured by immunohistochemistry) was 34.2±0.7 pixel units in vehicle-treated rats, and 54.9±0.6 in the liraglutide-treated animals. Glucose-stimulated insulin secretion from isolated islets (measured as the stimulation index) was maintained in the liraglutide-treated rats, but not in the vehicle-treated. However, liraglutide did not preserve normal islet architecture. There was a decrease in the glucagon-positive area/islet and in the α-cell numbers/area with liraglutide treatment (6.5 cells/field), compared to vehicle (17.9 cells/field). There was an increase in ß-cell numbers, the ß- to α-cell ratio that was statistically higher in the liraglutide-treated rats (24.3±4.4) compared to vehicle (9.1±2.8). Disrupted mitochondria were more commonly observed in the α-cells (51.9±10.3% of cells) than in the ß-cells (27.2±4.4%) in the liraglutide-treated group. While liraglutide enhanced or maintained growth and function of certain islet cells, the overall ratio of α- to ß-cells was decreased and there was an absolute reduction in islet α-cell content. There was selective disruption of intracellular α-cell organelles, representing an uncoupling of the bihormonal islet signaling that is required for normal metabolic regulation. The relevance of the findings to long-term liraglutide treatment in people with diabetes is unknown and should be investigated in appropriately designed clinical studies.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Células Secretoras de Glucagon/efeitos dos fármacos , Hipoglicemiantes/uso terapêutico , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Animais , Glicemia/análise , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Glucagon/análise , Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/administração & dosagem , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Hipoglicemiantes/administração & dosagem , Insulina/análise , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Liraglutida , Masculino , Ratos , Ratos Zucker
6.
J Alzheimers Dis ; 92(2): 591-604, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36776072

RESUMO

BACKGROUND: Mitochondria can trigger Alzheimer's disease (AD)-associated molecular phenomena, but how mitochondria impact apolipoprotein E (APOE; apoE) is not well known. OBJECTIVE: Consider whether and how mitochondrial biology influences APOE and apoE biology. METHODS: We measured APOE expression in human SH-SY5Y neuronal cells with different forms of mitochondrial dysfunction including total, chronic mitochondrial DNA (mtDNA) depletion (ρ0 cells); acute, partial mtDNA depletion; and toxin-induced mitochondrial dysfunction. We further assessed intracellular and secreted apoE protein levels in the ρ0 cells and interrogated the impact of transcription factors and stress signaling pathways known to influence APOE expression. RESULTS: SH-SY5Y ρ0 cells exhibited a 65-fold increase in APOE mRNA, an 8-fold increase in secreted apoE protein, and increased intracellular apoE protein. Other models of primary mitochondrial dysfunction including partial mtDNA-depletion, toxin-induced respiratory chain inhibition, and chemical-induced manipulations of the mitochondrial membrane potential similarly increased SH-SY5Y cell APOE mRNA. We explored potential mediators and found in the ρ0 cells knock-down of the C/EBPα and NFE2L2 (Nrf2) transcription factors reduced APOE mRNA. The activity of two mitogen-activated protein kinases, JNK and ERK, also strongly influenced ρ0 cell APOE mRNA levels. CONCLUSION: Primary mitochondrial dysfunction either directly or indirectly activates APOE expression in a neuronal cell model by altering transcription factors and stress signaling pathways. These studies demonstrate mitochondrial biology can influence the biology of the APOE gene and apoE protein, which are implicated in AD.


Assuntos
Doença de Alzheimer , Neuroblastoma , Humanos , Neuroblastoma/metabolismo , Mitocôndrias/metabolismo , DNA Mitocondrial/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Fatores de Transcrição/metabolismo , Doença de Alzheimer/metabolismo , RNA Mensageiro/metabolismo , Biologia , Linhagem Celular Tumoral
7.
Biochim Biophys Acta ; 1812(11): 1532-41, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21839170

RESUMO

NADH-cytochrome b5 oxidoreductase (Ncb5or) is an endoplasmic reticulum (ER)-associated redox enzyme involved in fatty acid metabolism, and phenotypic abnormalities of Ncb5or(-/-) mice include diabetes and lipoatrophy. These mice are lean and insulin-sensitive but become hyperglycemic at age 7 weeks as a result of ß-cell dysfunction and loss. Here we examine early cellular and molecular events associated with manifestations of ß-cell defects in Ncb5or(-/-) mice. We observe lower islet ß-cell content in pancreata at age 4 weeks and prominent ER distention in ß-cells by age 5 weeks. Ultrastructural changes progress rapidly in severity from age 5 to 6 weeks, and their frequency rises from 10% of ß-cells at 5 weeks to 33% at 6 weeks. These changes correlate temporally with the onset of diabetes. ER stress responses and lipid load in Ncb5or(-/-) ß-cells were assessed with isolated islets from mice at age 5 weeks. Expression levels of the stress marker protein Grp78/BiP and of phosphorylated eIF2α protein were found to be reduced, although their transcript levels did not decline. This pattern stands in contrast to the canonical unfolded protein response. Ncb5or(-/-) ß-cells also accumulated higher intracellular levels of palmitate and other free fatty acids and exhibited greater reactive oxygen species production than wild-type cells. An alloxan-susceptible genetic background was found to confer accelerated onset of diabetes in Ncb5or(-/-) mice. These findings provide the first direct evidence that manifestations of diabetes in lean Ncb5or(-/-) mice involve saturated free fatty acid overload of ß-cells and ER and oxidative stress responses.


Assuntos
Citocromo-B(5) Redutase/fisiologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Retículo Endoplasmático/patologia , Células Secretoras de Insulina/patologia , Estresse Oxidativo , Aloxano , Animais , Western Blotting , Diabetes Mellitus Experimental/metabolismo , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Ácidos Graxos Insaturados/metabolismo , Feminino , Imunofluorescência , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Oxirredução , Palmitatos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Resposta a Proteínas não Dobradas
8.
Mitochondrion ; 64: 136-144, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35398304

RESUMO

In studies with human participants, exosome-based biospecimens can facilitate unique biomarker assessments. As exosome cargos can include mitochondrial components, there is interest in using exosomes to inform the status of an individual's mitochondria. Here, we evaluated whether targeted pharmacologic manipulations could influence the quantity of exosomes shed by cells, and whether these manipulations could impact their mitochondrial cargos. We treated human SH-SY5Y cells with bafilomycin A1, which interferes with general autophagy and mitophagy by inhibiting lysosome acidification and lysosome-autophagosome fusion; deferiprone (DFP), which enhances receptor-mediated mitophagy; or both. Exosome fractions from treated cells were harvested from the cell medium and analyzed for content including mitochondria-derived components. We found bafilomycin increased particle yields, and a combination of bafilomycin plus DFP consistently increased particle yields and mitochondria-associated content. Specifically, the exosome fractions from the bafilomycin plus DFP-treated cells contained more mitochondrial DNA (mtDNA), mtDNA-derived mRNA transcripts, and citrate synthase protein. Our data suggest pharmacologic manipulations that enhance mitophagy initiation, while inhibiting the lysosomal digestion of autophagosomes and multivesicular bodies, could potentially enhance the sensitivity of exosome-based biomarker assays intended to inform the status of an individual's mitochondria.


Assuntos
Exossomos , Humanos , Autofagia , Biomarcadores/metabolismo , DNA Mitocondrial/metabolismo , Lisossomos/genética , Lisossomos/metabolismo , Mitocôndrias/metabolismo
9.
Cardiovasc Diabetol ; 9: 56, 2010 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-20860788

RESUMO

BACKGROUND: Early markers of diabetic autonomic neuropathy (DAN) in an electrocardiogram (ECG) include elevated R wave amplitudes, widening of QTc intervals and decreased heart rate variability (HRV). The severity of DAN has a direct relationship with mortality risk. Aerobic exercise training is a common recommendation for the delay and possible reversal of cardiac dysfunction. Limited research exists on ECG measures for the evaluation of aerobic exercise training in Zucker Diabetic Fatty (ZDF) rat, a model of type 2 diabetes. The objective of this study was to assess whether aerobic exercise training may attenuate diabetes induced ECG changes. METHODS: Male ZDF (obese fa/fa) and control Zucker (lean fa/+) rats were assigned to 4 groups: sedentary control (SC), sedentary diabetic (SD), exercised control (EC) and exercised diabetic (ED). The exercised groups began 7 weeks of treadmill training after the development of diabetes in the ED group. Baseline (prior to the training) and termination measurements included body weight, heart weight, blood glucose and glycated hemoglobin levels and ECG parameters. One way repeated measures ANOVA (group) analyzed within and between subject differences and interactions. Pearson coefficients and descriptive statistics described variable relationships and animal characteristics. RESULTS: Diabetes caused crucial changes in R wave amplitudes (p < 0.001), heart rate variability (p < 0.01), QT intervals (p < 0.001) and QTc intervals (p < 0.001). R wave amplitude augmentation in SD rats from baseline to termination was ameliorated by exercise, resulting in R wave amplitude changes in ED animals similar to control rats. Aerobic exercise training neither attenuated QT or QTc interval prolongation nor restored decreases in HRV in diabetic rats. CONCLUSION: This study revealed alterations in R wave amplitudes, HRV, QT and QTc intervals in ZDF rats. Of these changes, aerobic exercise training was able to correct R wave amplitude changes. In addition, exercise has beneficial effect in this diabetic rat model in regards to ECG correlates of left ventricular mass.


Assuntos
Neuropatias Diabéticas/fisiopatologia , Eletrocardiografia , Cardiopatias/diagnóstico , Cardiopatias/fisiopatologia , Obesidade/fisiopatologia , Condicionamento Físico Animal/fisiologia , Animais , Peso Corporal/fisiologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/fisiopatologia , Diabetes Mellitus Tipo 2/terapia , Neuropatias Diabéticas/complicações , Neuropatias Diabéticas/terapia , Modelos Animais de Doenças , Cardiopatias/etiologia , Frequência Cardíaca/fisiologia , Masculino , Obesidade/complicações , Ratos , Ratos Zucker
10.
Exp Neurol ; 305: 97-107, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29649429

RESUMO

Insulin is known to have neurotrophic properties and loss of insulin support to sensory neurons may contribute to peripheral diabetic neuropathy (PDN). Here, genetically-modified mice were generated in which peripheral sensory neurons lacked the insulin receptor (SNIRKO mice) to determine whether disrupted sensory neuron insulin signaling plays a crucial role in the development of PDN and whether SNIRKO mice develop symptoms of PDN due to reduced insulin neurotrophic support. Our results revealed that SNIRKO mice were euglycemic and never displayed significant changes in a wide range of sensorimotor behaviors, nerve conduction velocity or intraepidermal nerve fiber density. However, SNIRKO mice displayed elevated serum insulin levels, glucose intolerance, and increased insulin content in the islets of Langerhans of the pancreas. These results contribute to the growing idea that sensory innervation of pancreatic islets is key to regulating islet function and that a negative feedback loop of sensory neuron insulin signaling keeps this regulation in balance. Our results suggest that a loss of insulin receptors in sensory neurons does not lead to peripheral nerve dysfunction. The SNIRKO mice will be a powerful tool to investigate sensory neuron insulin signaling and may give a unique insight into the role that sensory neurons play in modifying islet physiology.


Assuntos
Deleção de Genes , Insulina/metabolismo , Pâncreas/metabolismo , Receptor de Insulina/deficiência , Células Receptoras Sensoriais/metabolismo , Animais , Glicemia/metabolismo , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Pâncreas/citologia , Receptor de Insulina/genética
11.
Artigo em Inglês | MEDLINE | ID: mdl-27554916

RESUMO

Early screens for new diabetes drugs rely on monolayers of ß-cells, which are known to be poor predictors of the in vivo response. Previously, we developed a method to create uniform islet spheroids from freshly-dispersed human donor tissue for drug screening. While the human engineered islets worked well to reduce donor-to-donor variability, it is difficult and expensive to obtain sufficient high-quality human islets for drug testing. Thus, this study utilized a genetically-modified ß-cell culture line (INS-1832/13) in 2D and as 3D spheroids and compared the results to human islet tissue formed into spheroids using a high-throughput 384-well format. In response to increasing concentrations of glucose, all 3 groups increased insulin release, but the cultured ß-cells (2D and 3D) were more sensitive to glucose (EC50 5.85mM for 2D ß-cells, 16.24mM for 3D ß-cell spheroids) than the human islet spheroids (EC50 53.69mM). The order of responses to glybenclamide was human spheroids >3D ß-cell culture >2D ß-cell culture. In response to caffeine, the ß-cells in 2D or 3D were more responsive compared to the human islet spheroids (EC50 0.39 and 0.31mM for 2D and 3D ß-cells respectively). When exposed to inhibitors of insulin secretion (nifedipine and diazoxide), the responses were more similar between groups. Z' calculations, indicative of assay quality, determined that the 3D ß-cell spheroids reached the criteria of an excellent to ideal drug screen assay more consistently than the other test models. In conclusion, 3D ß-cell spheroids from a cultured cell line can be used in HTS assays that, according to reference drugs tested here, are sensitive and predictive of the in vivo response.


Assuntos
Descoberta de Drogas/métodos , Ensaios de Triagem em Larga Escala/métodos , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Esferoides Celulares/efeitos dos fármacos , Adulto , Técnicas de Cultura de Células , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Glucose/farmacologia , Humanos , Hipoglicemiantes/química , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Esferoides Celulares/metabolismo
12.
Exp Biol Med (Maywood) ; 241(7): 697-705, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26946532

RESUMO

Fetal hemoglobin is a major genetic modifier of the phenotypic heterogeneity in patients with sickle cell disease and certain ß-thalassemias. Normal levels of fetal hemoglobin postnatally are approximately 1% of total hemoglobin. Patients who have hereditary persistence of fetal hemoglobin, characterized by elevated synthesis of γ-globin in adulthood, show reduced disease pathophysiology. Hereditary persistence of fetal hemoglobin is caused by ß-globin locus deletions (deletional hereditary persistence of fetal hemoglobin) or γ-globin gene promoter point mutations (non-deletional hereditary persistence of fetal hemoglobin). Current research has focused on elucidating the pathways involved in the maintenance/reactivation of γ-globin in adult life. To better understand these pathways, we generated new ß-globin locus yeast artificial chromosome transgenic mice bearing the (A)γ-globin -175 T > C or -195 C > G hereditary persistence of fetal hemoglobin mutations to model naturally occurring hereditary persistence of fetal hemoglobin. Adult -175 and -195 mutant ß-YAC mice displayed a hereditary persistence of fetal hemoglobin phenotype, as measured at the mRNA and protein levels. The molecular basis for these phenotypes was examined by chromatin immunoprecipitation of transcription factor/co-factor binding, including YY1, PAX1, TAL1, LMO2, and LDB1. In -175 HPFH versus wild-type samples, the occupancy of LMO2, TAL1 and LDB1 proteins was enriched in HPFH mice (5.8-fold, 5.2-fold and 2.7-fold, respectively), a result that concurs with a recent study in cell lines showing that these proteins form a complex with GATA-1 to mediate long-range interactions between the locus control region and the (A)γ-globin gene. Both hereditary persistence of fetal hemoglobin mutations result in a gain of (A)γ-globin activation, in contrast to other hereditary persistence of fetal hemoglobin mutations that result in a loss of repression. The mice provide additional tools to study γ-globin gene expression and may reveal new targets for selectively activating fetal hemoglobin.


Assuntos
Hemoglobina Fetal/genética , Globinas beta/genética , Anemia Falciforme/sangue , Anemia Falciforme/genética , Animais , Cromossomos Artificiais de Levedura/genética , Modelos Animais de Doenças , Hemoglobina Fetal/análise , Citometria de Fluxo , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Transgênicos/genética , Reação em Cadeia da Polimerase em Tempo Real
13.
Neurosci Res ; 52(4): 371-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15936837

RESUMO

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is currently a leading neurotoxic agent used for producing Parkinsonism in laboratory animals. The MPTP neurotoxicity in humans is irreversible and the consequential clinical and neurochemical features closely resemble those of the idiopathic Parkinson's disease. Therefore, handling of MPTP in laboratory may pose neurotoxic risk among researchers and animal caretakers. While it is well recognized that systemic administration of MPTP will cause Parkinsonian-like symptoms in humans and animals, it is not known whether similar neurological toxicity is transmittable and would develop in normal subjects housed closely with the MPTP-treated animals. In the present study, we treated mice daily with MPTP hydrochloride (30mg/kg, s.c.) for 5 consecutive days. In the same cage, a non-treated mouse (cagemate) was kept allowing for close physical interaction, free contact with the excreta, and sharing of food and water. Seventy-two hours after the treatment, the MPTP-treated mice and MPTP-exposed cagemates were analyzed for dopaminergic neurotoxicity comparing with the MPTP non-exposed control animals. We detected a significant number of TUNEL-positive cells, loss of tyrosine hydroxylase immunoreactivity in the substantia nigra, and depletion of dopamine in the striatum of MPTP-treated mice. However, these neurotoxic indices were not detected in the MPTP-exposed cagemates or MPTP non-exposed controls. Following each MPTP injection, approximately 42% of the chemical was excreted within 3h through the urine largely in the form of MPTP N-oxide, which is not expected to cross the blood-brain barrier and to cause dopaminergic toxicity in the brain when administered peripherally. These observations suggest that MPTP injections in mice do not transmit and cause Parkinsonian-like dopaminergic neurotoxicity in the neighboring normal cagemates through direct physical contact and exposure from the contaminated cage, food, water, and excreta.


Assuntos
Transtornos Parkinsonianos/patologia , Substância Negra/efeitos dos fármacos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/urina , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Western Blotting/métodos , Cromatografia Líquida de Alta Pressão/métodos , Modelos Animais de Doenças , Dopamina/metabolismo , Imuno-Histoquímica/métodos , Marcação In Situ das Extremidades Cortadas/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Parkinsonianos/metabolismo , Estatísticas não Paramétricas , Substância Negra/metabolismo , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/metabolismo
14.
Tissue Eng Part A ; 19(5-6): 604-12, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23004004

RESUMO

Isolated islets can provide a source of tissue for research, transplantation, and drug discovery to develop therapies for diabetes. Empirical modeling of islet diffusion barriers demonstrated that only the outermost layers of cells were exposed to glucose and sufficient oxygen levels, resulting in core cell death. Islets under a diameter of 100 µm exhibited a lower diffusion barrier, superior survival rates, and improved functional properties. Utilizing these observations, we engineered optimal islets by dispersing them into single cells and reaggregating them over several days in a micromold. These custom-designed micromolds contained conical-shaped recesses that enhanced reaggregation of cells into a defined geometry. The engineered islets, or Kanslets, were all under 100 µm in diameter, and had the same general cellular composition as native islets. Kanslets continued to produce new insulin molecules and had microvilli on the islet surface, much like native islets. The engineered islets had a statistically higher viability (percent of live cells), and increased glucose diffusion compared to native islets. In addition, they remained responsive to varying glucose levels by secreting insulin. When transplanted into diabetic rats, engineered islets performed reduced random blood glucose to normal levels within 48 h. Optimally, engineering islets may be a suitable alternative to utilizing native, isolated islet tissue for a variety of applications. Reaggregating tissue in an optimized manner using our engineered micromold approach has immense impact for three-dimensional tissue production and its subsequent use in research, drug discovery, and the clinic.


Assuntos
Ilhotas Pancreáticas/citologia , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Glicemia/metabolismo , Agregação Celular , Difusão , Ilhotas Pancreáticas/ultraestrutura , Transplante das Ilhotas Pancreáticas , Tamanho do Órgão , Proinsulina/metabolismo , Ratos , Sobrevivência de Tecidos
15.
Islets ; 5(2): 87-94, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23648896

RESUMO

For the past 30 years, data have suggested that unique islet populations exist, based on morphology and glucose sensitivity. Yet little has been done to determine the mechanism of these functional differences. The purpose of this study was to determine whether human islets were comprised functionally unique populations, and to elucidate a possible mechanism. Islets or pancreatic sections from 29 human donors were analyzed. Islets were isolated and measured for insulin secretion, cell composition and organization, insulin and glucagon granule density and insulin content. Insulin secretion was significantly greater in small compared with large islets. In sectioned human pancreata, ß-cells comprised a higher proportion of the total endocrine cells in small islets (63%) than large islets (39%). A higher percentage of ß-cells in small islets contacted blood vessels (44%) compared with large islets (31%). Total insulin content of isolated human islets was significantly greater in the small (1323 ± 512 µIU/IE) compared with large islets (126 ± 48 µIU/IE). There was less immunostaining for insulin in the large islets from human pancreatic sections, especially in the core of the islet, compared with small islets. The results suggest that differences in insulin secretion between large and small islets may be due to a higher percentage of ß-cells in small islets with more ß-cells in contact with blood vessels and a higher concentration of insulin/ß-cell in small islets.


Assuntos
Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestrutura , Insulina/metabolismo , Ilhotas Pancreáticas/ultraestrutura , Regulação para Cima , Adulto , Contagem de Células , Feminino , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/ultraestrutura , Humanos , Hiperglicemia/metabolismo , Imuno-Histoquímica , Secreção de Insulina , Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/crescimento & desenvolvimento , Ilhotas Pancreáticas/metabolismo , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Proinsulina/metabolismo , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura , Somatostatina , Células Secretoras de Somatostatina/metabolismo , Células Secretoras de Somatostatina/ultraestrutura , Bancos de Tecidos
16.
J Diabetes Res ; 2013: 965832, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762878

RESUMO

Type 1 diabetes (T1D) is characterized by hyperglycemia due to lost or damaged islet insulin-producing ß -cells. Rodent models of T1D result in hyperglycemia, but with different forms of islet deterioration. This study focused on 1 toxin-induced and 2 autoimmune rodent models of T1D: BioBreeding Diabetes Resistant rats, nonobese diabetic mice, and Dark Agouti rats treated with streptozotocin. Immunochemistry was used to evaluate the insulin levels in the ß -cells, cell composition, and insulitis. T1D caused complete or significant loss of ß -cells in all animal models, while increasing numbers of α -cells. Lymphocytic infiltration was noted in and around islets early in the progression of autoimmune diabetes. The loss of lymphocytic infiltration coincided with the absence of ß -cells. In all models, the remaining α - and δ -cells regrouped by relocating to the islet center. The resulting islets were smaller in size and irregularly shaped. Insulin injections subsequent to induction of toxin-induced diabetes significantly preserved ß -cells and islet morphology. Diabetes in animal models is anatomically heterogeneous and involves important changes in numbers and location of the remaining α - and δ -cells. Comparisons with human pancreatic sections from healthy and diabetic donors showed similar morphological changes to the diabetic BBDR rat model.


Assuntos
Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Hiperglicemia/patologia , Ilhotas Pancreáticas/patologia , Animais , Glicemia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Feminino , Humanos , Hiperglicemia/imunologia , Hiperglicemia/fisiopatologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/fisiopatologia , Masculino , Camundongos , Ratos
17.
PLoS One ; 8(3): e58767, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23536823

RESUMO

Nicotinamide phosphoribosyltransferase (Nampt) is a rate-limiting enzyme in the mammalian NAD+ biosynthesis of a salvage pathway and exists in 2 known forms, intracellular Nampt (iNampt) and a secreted form, extracellular Nampt (eNampt). eNampt can generate an intermediate product, nicotinamide mononucleotide (NMN), which has been reported to support insulin secretion in pancreatic islets. Nampt has been reported to be expressed in the pancreas but islet specific expression has not been adequately defined. The aim of this study was to characterize Nampt expression, secretion and regulation by glucose in human islets. Gene and protein expression of Nampt was assessed in human pancreatic tissue and isolated islets by qRT-PCR and immunofluorescence/confocal imaging respectively. Variable amounts of Nampt mRNA were detected in pancreatic tissue and isolated islets. Immunofluorescence staining for Nampt was found in the exocrine and endocrine tissue of fetal pancreas. However, in adulthood, Nampt expression was localized predominantly in beta cells. Isolated human islets secreted increasing amounts of eNampt in response to high glucose (20 mM) in a static glucose-stimulated insulin secretion assay (GSIS). In addition to an increase in eNampt secretion, exposure to 20 mM glucose also increased Nampt mRNA levels but not protein content. The secretion of eNampt was attenuated by the addition of membrane depolarization inhibitors, diazoxide and nifedipine. Islet-secreted eNampt showed enzymatic activity in a reaction with increasing production of NAD+/NADH over time. In summary, we show that Nampt is expressed in both exocrine and endocrine tissue early in life but in adulthood expression is localized to endocrine tissue. Enzymatically active eNampt is secreted by human islets, is regulated by glucose and requires membrane depolarization.


Assuntos
Regulação da Expressão Gênica , Ilhotas Pancreáticas/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Ativação Enzimática , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glucagon/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Humanos , Lactente , Recém-Nascido , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Nicotinamida Fosforribosiltransferase/metabolismo , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Ligação Proteica , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adulto Jovem
18.
J Appl Physiol (1985) ; 113(5): 817-26, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22797313

RESUMO

One of the fundamental biochemical defects underlying the complications of diabetic cardiovascular system is elevation of diacylglycerol (DAG) and its effects on protein kinase C (PKC) signaling. It has been noted that exercise training attenuates poor cardiac performance in Type 1 diabetes. However, the role of PKC signaling in exercise-induced alleviation of cardiac abnormalities in diabetes is not clear. We investigated the possibility that exercise training modulates PKC-ßII signaling to elicit its beneficial effects on the diabetic heart. bio-breeding diabetic resistant rats, a model reminiscent of Type 1 diabetes in humans, were randomly assigned to four groups: 1) nonexercised nondiabetic (NN); 2) nonexercised diabetic (ND); 3) exercised nondiabetic; and 4) exercised diabetic. Treadmill training was initiated upon the onset of diabetes. At the end of 8 wk, left ventricular (LV) hemodynamic assessment revealed compromised function in ND compared with the NN group. LV myocardial histology revealed increased collagen deposition in ND compared with the NN group, while electron microscopy showed a reduction in the viable mitochondrial fraction. Although the PKC-ßII levels and activity were unchanged in the diabetic heart, the DAG levels were increased. With exercise training, the deterioration of LV structure and function in diabetes was attenuated. Notably, improved cardiac performance in training was associated with a decrease in myocardial DAG levels in diabetes. Exercise-induced benefits on cardiac performance in diabetes may be mediated by prevention of an increase in myocardial DAG levels.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Diglicerídeos/antagonistas & inibidores , Diglicerídeos/metabolismo , Miocárdio/metabolismo , Condicionamento Físico Animal/fisiologia , Função Ventricular Esquerda/fisiologia , Sequência de Aminoácidos , Animais , Diglicerídeos/biossíntese , Masculino , Dados de Sequência Molecular , Condicionamento Físico Animal/métodos , Proteína Quinase C/fisiologia , Proteína Quinase C beta , Distribuição Aleatória , Ratos
19.
Exp Diabetes Res ; 2012: 671673, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23197975

RESUMO

KU-32 is a novel, novobiocin-based Hsp90 inhibitor that protects against neuronal glucotoxicity and reverses multiple clinical indices of diabetic peripheral neuropathy in a rodent model. However, any drug with potential for treating diabetic complications must also have no adverse effects on the function of pancreatic islets. Thus, the goal of the current study was to assess the effect of KU-32 on the in vitro viability and function of human islets. Treating human islets with KU-32 for 24 hours showed no toxicity as assessed using the alamarBlue assay. Confocal microscopy confirmed that with a minimum of 2-day exposure, KU-32 improved cellular viability by blocking apoptosis. Functionally, isolated human islets released more glucose-stimulated insulin when preincubated in KU-32. However, diabetic BKS-db/db mice, a model for type 2 diabetes, administered KU-32 for 10 weeks did not show any significant changes in blood glucose and insulin levels, despite having greater insulin staining/beta cell in the pancreas compared to untreated BKS db/db mice. In summary, KU-32 did not harm isolated human islets and may even be protective. However, the effect does not appear significant enough to alter the in vivo metabolic parameters of diabetic mice.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Neuropatias Diabéticas/tratamento farmacológico , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Novobiocina/análogos & derivados , Adulto , Animais , Apoptose/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/patologia , Neuropatias Diabéticas/sangue , Neuropatias Diabéticas/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Insulina/sangue , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Microscopia Confocal , Pessoa de Meia-Idade , Novobiocina/farmacologia , Novobiocina/toxicidade , Fatores de Tempo , Técnicas de Cultura de Tecidos
20.
Islets ; 3(1): 6-13, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21325888

RESUMO

The existence of morphologically distinct populations of islets in the pancreas was described over 60 years ago. Unfortunately, little attention has been paid to possible functional differences between islet subpopulations until recently. We demonstrated that one population, the small islets, were superior to large islets in a number of functional aspects. However, that work did not determine whether these differences were inherent, or whether they arose because of the challenge of isolation procedures. Nor, were there data to explain the differences in insulin secretion. We utilized immunohistochemistry, immunofluorescence, ELISA, and transmission electron microscopy to compare the unique characteristics found in isolated rat islet populations in situ and after isolation. Insulin secretion of small isolated islets was significantly higher compared to large islets, which correlated with higher insulin content/area in small islets (in situ), a higher density of insulin secretory granules, and greater insulin content/volume in isolated islets. Specifically, the core b-cells of the large islets contained less insulin/cell with a lower insulin granule density than peripheral b-cells. When insulin secretion was normalized for total insulin content, large and small islets released the same percentage of total insulin. Small islets had a higher density of cells/area than large islets in vitro and in situ. The data provide a possible explanation for the inferior insulin secretion from large islets, as they have a lower total cell density and the b-cells of the core contain less insulin/cell.


Assuntos
Insulina/análise , Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/citologia , Animais , Contagem de Células , Separação Celular , Tamanho Celular , Células Cultivadas , Imuno-Histoquímica , Hibridização In Situ , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Concentração Osmolar , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA