Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Hum Mol Genet ; 23(24): 6528-41, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25027327

RESUMO

Brain bioenergetic function declines in some neurodegenerative diseases, this may influence other pathologies and administering bioenergetic intermediates could have therapeutic value. To test how one intermediate, oxaloacetate (OAA) affects brain bioenergetics, insulin signaling, inflammation and neurogenesis, we administered intraperitoneal OAA, 1-2 g/kg once per day for 1-2 weeks, to C57Bl/6 mice. OAA altered levels, distributions or post-translational modifications of mRNA and proteins (proliferator-activated receptor-gamma coactivator 1α, PGC1 related co-activator, nuclear respiratory factor 1, transcription factor A of the mitochondria, cytochrome oxidase subunit 4 isoform 1, cAMP-response element binding, p38 MAPK and adenosine monophosphate-activated protein kinase) in ways that should promote mitochondrial biogenesis. OAA increased Akt, mammalian target of rapamycin and P70S6K phosphorylation. OAA lowered nuclear factor κB nucleus-to-cytoplasm ratios and CCL11 mRNA. Hippocampal vascular endothelial growth factor mRNA, doublecortin mRNA, doublecortin protein, doublecortin-positive neuron counts and neurite length increased in OAA-treated mice. (1)H-MRS showed OAA increased brain lactate, GABA and glutathione thereby demonstrating metabolic changes are detectable in vivo. In mice, OAA promotes brain mitochondrial biogenesis, activates the insulin signaling pathway, reduces neuroinflammation and activates hippocampal neurogenesis.


Assuntos
Hipocampo/efeitos dos fármacos , Insulina/metabolismo , Renovação Mitocondrial/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Ácido Oxaloacético/administração & dosagem , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas do Domínio Duplacortina , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica , Glutationa/metabolismo , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Inflamação/prevenção & controle , Injeções Intraperitoneais , Insulina/genética , Ácido Láctico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Renovação Mitocondrial/genética , Neurogênese/genética , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fator 1 Nuclear Respiratório/genética , Fator 1 Nuclear Respiratório/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ácido gama-Aminobutírico/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
J Biol Chem ; 289(21): 14719-30, 2014 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-24713701

RESUMO

Mitochondrial impairment is commonly found in many diseases such as diabetes, cancer, and Alzheimer disease. We demonstrate that the enzymes responsible for the addition or removal of the O-GlcNAc modification, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively, are critical regulators of mitochondrial function. Using a SILAC (stable isotope labeling of amino acids in cell culture)-based proteomics screen, we quantified the changes in mitochondrial protein expression in OGT- and OGA-overexpressing cells. Strikingly, overexpression of OGT or OGA showed significant decreases in mitochondria-localized proteins involved in the respiratory chain and the tricarboxylic acid cycle. Furthermore, mitochondrial morphology was altered in these cells. Both cellular respiration and glycolysis were reduced in OGT/OGA-overexpressing cells. These data demonstrate that alterations in O-GlcNAc cycling profoundly affect energy and metabolite production.


Assuntos
Acetilglucosamina/metabolismo , Ciclo do Ácido Cítrico , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Linhagem Celular Tumoral , Transporte de Elétrons , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glicólise , Humanos , Immunoblotting , Espectrometria de Massas , Microscopia Eletrônica , Mitocôndrias/genética , Mitocôndrias/ultraestrutura , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Proteômica/métodos , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo
3.
Hum Mol Genet ; 22(19): 3931-46, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23740939

RESUMO

Bioenergetic dysfunction occurs in Alzheimer's disease (AD) and mild cognitive impairment (MCI), a clinical syndrome that frequently precedes symptomatic AD. In this study, we modeled AD and MCI bioenergetic dysfunction by transferring mitochondria from MCI, AD and control subject platelets to mtDNA-depleted SH-SY5Y cells. Bioenergetic fluxes and bioenergetics-related infrastructures were characterized in the resulting cytoplasmic hybrid (cybrid) cell lines. Relative to control cybrids, AD and MCI cybrids showed changes in oxygen consumption, respiratory coupling and glucose utilization. AD and MCI cybrids had higher ADP/ATP and lower NAD+/NADH ratios. AD and MCI cybrids exhibited differences in proteins that monitor, respond to or regulate cell bioenergetic fluxes including HIF1α, PGC1α, SIRT1, AMPK, p38 MAPK and mTOR. Several endpoints suggested mitochondrial mass increased in the AD cybrid group and probably to a lesser extent in the MCI cybrid group, and that the mitochondrial fission-fusion balance shifted towards increased fission in the AD and MCI cybrids. As many of the changes we observed in AD and MCI cybrid models are also seen in AD subject brains, we conclude reduced bioenergetic function is present during very early AD, is not brain-limited and induces protean retrograde responses that likely have both adaptive and mal-adaptive consequences.


Assuntos
Doença de Alzheimer/metabolismo , Disfunção Cognitiva/metabolismo , Mitocôndrias/fisiologia , Mitocôndrias/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Idoso , Idoso de 80 Anos ou mais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Estudos de Casos e Controles , Linhagem Celular , DNA Mitocondrial/metabolismo , Metabolismo Energético , Humanos , Células Híbridas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pessoa de Meia-Idade , Mitocôndrias/enzimologia , Mitocôndrias/genética , Dinâmica Mitocondrial , Consumo de Oxigênio , Proteínas de Ligação a RNA , Sirtuína 1/genética , Sirtuína 1/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
4.
J Bioenerg Biomembr ; 47(1-2): 1-11, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25104046

RESUMO

Diet composition may affect energy metabolism in a tissue-specific manner. Using C57Bl/6J mice, we tested the effect of ketosis-inducing and non-inducing high fat diets on genes relevant to brain bioenergetic infrastructures, and on proteins that constitute and regulate that infrastructure. At the end of a one-month study period the two high fat diets appeared to differentially affect peripheral insulin signaling, but brain insulin signaling was not obviously altered. Some bioenergetic infrastructure parameters were similarly impacted by both high fat diets, while other parameters were only impacted by the ketogenic diet. For both diets, mRNA levels for CREB, PGC1α, and NRF2 increased while NRF1, TFAM, and COX4I1 mRNA levels decreased. PGC1ß mRNA increased and TNFα mRNA decreased only with the ketogenic diet. Brain mtDNA levels fell in both the ketogenic and non-ketogenic high fat diet groups, although TOMM20 and COX4I1 protein levels were maintained, and mRNA and protein levels of the mtDNA-encoded COX2 subunit were also preserved. Overall, the pattern of changes observed in mice fed ketogenic and non-ketogenic high fat diets over a one month time period suggests these interventions enhance some aspects of the brain's aerobic infrastructure, and may enhance mtDNA transcription efficiency. Further studies to determine which diet effects are due to changes in brain ketone body levels, fatty acid levels, glucose levels, altered brain insulin signaling, or other factors such as adipose tissue-associated hormones are indicated.


Assuntos
Encéfalo/metabolismo , Dieta Cetogênica/efeitos adversos , Gorduras na Dieta/efeitos adversos , Metabolismo Energético/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Animais , Encéfalo/patologia , DNA Mitocondrial/metabolismo , Camundongos , Fatores de Tempo
5.
Biochim Biophys Acta ; 1830(4): 2891-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23313167

RESUMO

BACKGROUND: Although some reciprocal glycolysis-respiration relationships are well recognized, the relationship between reduced glycolysis flux and mitochondrial respiration has not been critically characterized. METHODS: We concomitantly measured the extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) of SH-SY5Y neuroblastoma cells under free and restricted glycolysis flux conditions. RESULTS: Under conditions of fixed energy demand ECAR and OCR values showed a reciprocal relationship. In addition to observing an expected Crabtree effect in which increasing glucose availability raised the ECAR and reduced the OCR, a novel reciprocal relationship was documented in which reducing the ECAR via glucose deprivation or glycolysis inhibition increased the OCR. Substituting galactose for glucose, which reduces net glycolysis ATP yield without blocking glycolysis flux, similarly reduced the ECAR and increased the OCR. We further determined how reduced ECAR conditions affect proteins that associate with energy sensing and energy response pathways. ERK phosphorylation, SIRT1, and HIF1a decreased while AKT, p38, and AMPK phosphorylation increased. CONCLUSIONS: These data document a novel intracellular glycolysis-respiration effect in which restricting glycolysis flux increases mitochondrial respiration. GENERAL SIGNIFICANCE: Since this effect can be used to manipulate cell bioenergetic infrastructures, this particular glycolysis-respiration effect can practically inform the development of new mitochondrial medicine approaches.


Assuntos
Respiração Celular , Glicólise , Neuroblastoma/metabolismo , Linhagem Celular Tumoral , Humanos , Neuroblastoma/patologia , Consumo de Oxigênio , Proteínas Proto-Oncogênicas c-akt/fisiologia
6.
Biochim Biophys Acta ; 1822(4): 522-6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22265987

RESUMO

Dietary restriction (DR) extends lifespan across multiple species including mouse. Antioxidant plant extracts rich in polyphenols have also been shown to increase lifespan. We hypothesized that polyphenols might potentiate DR-induced lifespan extension. Twenty week old C57BL/6 mice were placed on one of three diets: continuous feeding (control), alternate day chow (Intermittent fed, IF), or IF supplemented with polyphenol antioxidants (PAO) from blueberry, pomegranate, and green tea extracts (IF+PAO). Both IF and IF+PAO groups outlived the control group and the IF+PAO group outlived the IF group (all p<0.001). In the brain, IF induced the expression of inflammatory genes and p38 MAPK phosphorylation, while the addition of PAO reduced brain inflammatory gene expression and p38 MAPK phosphorylation. Our data indicate that while IF overall promotes longevity, some aspects of IF-induced stress may paradoxically lessen this effect. Polyphenol compounds, in turn, may potentiate IF-induced longevity by minimizing specific components of IF-induced cell stress.


Assuntos
Restrição Calórica , Expectativa de Vida , Polifenóis/farmacologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
J Neurochem ; 127(1): 91-100, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23927032

RESUMO

The effects of exercise are not limited to muscle, and its ability to mitigate some chronic diseases is under study. A more complete understanding of how exercise impacts non-muscle tissues might facilitate design of clinical trials and exercise mimetics. Here, we focused on lactate's ability to mediate changes in liver and brain bioenergetic-associated parameters. In one group of experiments, C57BL/6 mice underwent 7 weeks of treadmill exercise sessions at intensities intended to exceed the lactate threshold. Over time, the mice dramatically increased their lactate threshold. To ensure that plasma lactate accumulated during the final week, the mice were run to exhaustion. In the liver, mRNA levels of gluconeogenesis-promoting genes increased. While peroxisome proliferator-activated receptor-gamma co-activator 1 alpha (PGC-1α) expression increased, there was a decrease in PGC-1ß expression, and overall gene expression changes favored respiratory chain down-regulation. In the brain, PGC-1α and PGC-1ß were unchanged, but PGC-1-related co-activator expression and mitochondrial DNA copy number increased. Brain tumor necrosis factor alpha expression fell, whereas vascular endothelial growth factor A expression rose. In another group of experiments, exogenously administered lactate was found to reproduce some but not all of these observed liver and brain changes. Our data suggest that lactate, an exercise byproduct, could mediate some of the effects exercise has on the liver and the brain, and that lactate itself can act as a partial exercise mimetic.


Assuntos
Encéfalo/metabolismo , Ácido Láctico/farmacologia , Fígado/metabolismo , Condicionamento Físico Animal/fisiologia , Animais , Glicemia/metabolismo , Peso Corporal/fisiologia , Encéfalo/efeitos dos fármacos , Química Encefálica/fisiologia , DNA Mitocondrial/biossíntese , DNA Mitocondrial/genética , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Dosagem de Genes , Imuno-Histoquímica , Insulina/sangue , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 1 Nuclear Respiratório/metabolismo , Reação em Cadeia da Polimerase , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Neurobiol Dis ; 51: 3-12, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22266017

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects a staggering percentage of the aging population and causes memory loss and cognitive decline. Mitochondrial abnormalities can be observed systemically and in brains of patients suffering from AD, and may account for part of the disease phenotype. In this review, we summarize some of the key findings that indicate mitochondrial dysfunction is present in AD-affected subjects, including cytochrome oxidase deficiency, endophenotype data, and altered mitochondrial morphology. Special attention is given to recently described perturbations in mitochondrial autophagy, fission-fusion dynamics, and biogenesis. We also briefly discuss how mitochondrial dysfunction may influence amyloidosis in Alzheimer's disease, why mitochondria are a valid therapeutic target, and strategies for addressing AD-specific mitochondrial dysfunction.


Assuntos
Doença de Alzheimer/patologia , Homeostase/fisiologia , Mitocôndrias/patologia , Dinâmica Mitocondrial/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Humanos , Mitocôndrias/metabolismo
9.
Exp Physiol ; 98(1): 207-19, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22613742

RESUMO

To assess the effects of exercise on liver and brain bioenergetic infrastructures, we exposed C57BL/6 mice to 6 weeks of moderate-intensity treadmill exercise. During the training period, fasting blood glucose was lower in exercised mice than in sedentary mice, but serum insulin levels were not reduced. At week 6, trained mice showed a paradoxical decrease in plasma lactate during exercise, which was accompanied by an increase in the liver monocarboxylate transporter 2 protein level (∼30%, P < 0.05). Exercise increased liver peroxisomal proliferator-activated receptor-γ coactivator 1α expression (approximately twofold, P < 0.001), NAD-dependent deacetylase sirtuin-1 protein (∼30%, P < 0.05), p38 protein (∼15%, P < 0.05), cytochrome c oxidase subunit 4 isoform 1 protein (∼50%, P < 0.05) and AMP-activated protein kinase phosphorylation (∼40%, P < 0.05). Despite this, liver mitochondrial DNA copy number (∼30%, P = 0.05), mitochondrial transcription factor A expression (∼15%, P < 0.05), cytochrome c oxidase subunit 2 expression (∼10%, P < 0.05), cAMP-response element binding protein phosphorylation (∼60%, P < 0.05) and brain-derived neurotrophic factor expression (∼40%, P < 0.05) were all reduced, while cytochrome oxidase and citrate synthase activities were unchanged. The only altered brain parameter observed was a reduction in tumour necrosis factor α expression (∼35%, P < 0.05); tumour necrosis factor α expression was unchanged in liver. Our data suggest that lactate produced by exercising muscle modifies the liver bioenergetic infrastructure, and enhanced liver uptake may in turn limit the ability of exercise-generated lactate to modify brain bioenergetics. Also, it appears that, at least in the liver, a dissociated mitochondrial biogenesis, in which some components are strategically enhanced while others are minimized, can occur.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Fígado/metabolismo , Mitocôndrias/fisiologia , Condicionamento Físico Animal/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Glicemia/metabolismo , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Proteína de Ligação a CREB/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Ácido Láctico/sangue , Ácido Láctico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transportadores de Ácidos Monocarboxílicos/sangue , Fator 1 Nuclear Respiratório/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosforilação , Sirtuína 1/metabolismo , Transativadores/biossíntese , Fatores de Transcrição , Fator de Necrose Tumoral alfa , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Cartilage ; : 19476035231168387, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37086007

RESUMO

OBJECTIVE: Functional polymorphisms of interleukin 16 (IL16) have been reported to be closely related to the risk of osteoarthritis (OA). However, how IL16 affects OA remains unclear. In this study, the role of IL16 in OA and the possible mechanisms were examined. METHODS: We established a meniscal/ligament injury (MLI) post-traumatic OA model in Sprague Dawley rats and an IL1ß-induced ADTC5 cells OA model. We detected the expression of IL16, novel-miR-81, MMP3, and MMP13 by quantitative real-time polymerase chain reaction. Western blot was performed to detect the expression of IL16, MMP3, and MMP13. The association between IL16 and novel-miR-81 was confirmed by luciferase reporter assay. Hematoxylin and eosin staining, Safranin O and Fast Green staining, and immunohistochemical staining were performed to clarify the effect of intra-articular injection of novel-miR-81 agomir in rats OA model. RESULTS: IL16 was upregulated in OA model. Knockdown of IL16 and overexpression of novel-miR-81 downregulated the expression of MMP3 and MMP13. Importantly, IL16 was a key target of novel-miR-81. Intra-articular injection of novel-miR-81 agomir could attenuate OA progression in rats OA model. CONCLUSION: Novel-miR-81 targeted IL16 to relieve OA, suggesting that novel-miR-81and IL16 may be new therapeutic targets for OA.

11.
J Biol Chem ; 286(13): 11141-54, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21300801

RESUMO

The endoplasmic reticulum-associated NADH cytochrome b(5) oxidoreductase (Ncb5or) is widely distributed in animal tissues. Ncb5or(-/-) mice develop diabetes at age 7 weeks and have increased susceptibility to the diabetogenic oxidant streptozotocin. Ncb5or deficiency also results in lipoatrophy and increased hepatocyte sensitivity to cytotoxic effects of saturated fatty acids. Here we investigate the mechanisms of these phenomena in prediabetic Ncb5or(-/-) mice and find that, despite increased rates of fatty acid uptake and synthesis and higher stearoyl-CoA desaturase (SCD) expression, Ncb5or(-/-) liver accumulates less triacylglycerol (TAG) than wild type (WT). Increased fatty acid catabolism and oxidative stress are evident in Ncb5or(-/-) hepatocytes and reflect increased mitochondrial content, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) expression, fatty acid oxidation rates, oxidative stress response gene expression, and oxidized glutathione content. Ncb5or(-/-) hepatocytes readily incorporate exogenous fatty acids into TAG but accumulate more free fatty acids (FFA) and have greater palmitate-induced oxidative stress responses and cell death than WT, all of which are alleviated by co-incubation with oleate via TAG channeling. A high fat diet rich in palmitate and oleate stimulates both lipogenesis and fatty acid catabolism in Ncb5or(-/-) liver, resulting in TAG levels similar to WT but increased intracellular FFA accumulation. Hepatic SCD-specific activity is lower in Ncb5or(-/-) than in WT mice, although Ncb5or(-/-) liver has a greater increase in Scd1 mRNA and protein levels. Together, these findings suggest that increased FFA accumulation and catabolism and oxidative stress are major consequences of Ncb5or deficiency in liver.


Assuntos
Citocromo-B(5) Redutase , Hepatócitos/enzimologia , Ácido Oleico/metabolismo , Estresse Oxidativo , Ácido Palmítico/metabolismo , Animais , Diabetes Mellitus/enzimologia , Diabetes Mellitus/genética , Diabetes Mellitus Lipoatrófica , Regulação Enzimológica da Expressão Gênica/genética , Lipodistrofia Generalizada Congênita/enzimologia , Lipodistrofia Generalizada Congênita/genética , Camundongos , Camundongos Knockout , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição , Triglicerídeos/metabolismo
12.
Cell Reprogram ; 24(1): 9-20, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35180001

RESUMO

Bone marrow mesenchymal stem cells (BMSCs) differentiate into chondrocytes under appropriate conditions, providing a method for the treatment of bone- and joint-related diseases. Previously, we found that mulberry (Morus nigra) promoted the chondrogenic differentiation of BMSCs. Although the mechanism of action and active ingredients remain unknown, several studies describe the involvement of micro-RNAs. We obtained BMSCs from the bone marrow of Sprague Dawley rats. Cell Counting Kit-8 assays showed that maclurin (25 µg/mL) treatment was not toxic to BMSCs, and compared with untreated controls, maclurin upregulated Sox9 and Col2a expression. Quantitative-PCR revealed that miR-203a-3p levels decreased significantly during chondrogenic differentiation of BMSCs promoted by maclurin. Compared with treatment with an miR-203a-3p inhibitor, miR-203a-3p mimic inhibited expression of Sox9 and Col2a as evidenced by immunofluorescence staining and Western blotting. Smad1 was identified as a key target gene of miR-203a-3p according to biological-prediction software, and miR-203a-3p negatively regulated its transcription and translation in the dual-luciferase reporter gene assay and Western blotting. Sox9 and Col2a expression was downregulated following transfection of short interfering Smad1 (siSmad1) plasmids into BMSCs. We elucidated how maclurin promotes the chondrogenic differentiation of BMSCs by regulating miR-203a-3p/Smad1, which provides a strategy for future exploration of osteoarthritis therapy through cell transplantation.


Assuntos
Células-Tronco Mesenquimais , MicroRNAs , Animais , Células da Medula Óssea , Diferenciação Celular/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Lectinas de Plantas , Ratos , Ratos Sprague-Dawley , Proteína Smad1/metabolismo
13.
Biochim Biophys Acta ; 1802(1): 228-34, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19682571

RESUMO

Mitochondrial dysfunction has severe cellular consequences, and is linked to aging and neurological disorders in humans. Impaired energy supply or Ca(2+) buffering, increased ROS production, or control of apoptosis by mitochondria may contribute to the progressive decline of long-lived postmitotic cells. Mitochondrial biogenesis refers to the process via which cells increase their individual mitochondrial mass. Mitochondrial biogenesis may represent an attempt by cells to increase their aerobic set point, or an attempt to maintain a pre-existing aerobic set point in the face of declining mitochondrial function. Neuronal mitochondrial biogenesis itself has been poorly studied, but investigations from other tissues and model systems suggest a series of transcription factors, transcription co-activators, and signal transduction proteins should function to regulate mitochondrial number and mass within neurons. We review data pertinent to the mitochondrial biogenesis field, and discuss implications for brain aging and neurodegenerative disease research efforts.


Assuntos
Encéfalo/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Envelhecimento , Animais , Encéfalo/patologia , Humanos , Proteínas Mitocondriais/metabolismo , Doenças Neurodegenerativas/metabolismo , Espécies Reativas de Oxigênio/metabolismo
14.
J Neurochem ; 117(1): 154-63, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21244426

RESUMO

Dietary restriction (DR) has recognized health benefits that may extend to brain. We examined how DR affects bioenergetics-relevant enzymes and signaling pathways in the brains of C57BL/6 mice. Five-month-old male mice were placed in ad libitum or one of two repeated fasting and refeeding (RFR) groups, an alternate day (intermittent fed; IF) or alternate day plus antioxidants (blueberry, pomegranate, and green tea extracts) (IF + AO) fed group. During the 24-h fast blood glucose levels initially fell but stabilized within 6 h of starting the fast, thus avoiding frank hypoglycemia. DR in general appeared to enhance insulin sensitivity. After six weeks brain AKT and glycogen synthase kinase 3 beta phosphorylation were lower in the RFR mice, suggesting RFR reduced brain insulin-signaling pathway activity. Pathways that mediate mitochondrial biogenesis were not activated; AMP kinase phosphorylation, silent information regulator 2 phosphorylation, peroxisomal proliferator-activated receptor-gamma coactivator 1 alpha levels, and cytochrome oxidase subunit 4 levels did not change. ATP levels also did not decline, which suggests the RFR protocols did not directly impact brain bioenergetics. Antioxidant supplementation did not affect the brain parameters we evaluated. Our data indicate in young adult male C57BL/6 mice, RFR primarily affects brain energy metabolism by reducing brain insulin signaling, which potentially results indirectly as a consequence of reduced peripheral insulin production.


Assuntos
Encéfalo/metabolismo , Jejum/metabolismo , Insulina/fisiologia , Transdução de Sinais/fisiologia , Fatores Etários , Animais , Glicemia/metabolismo , Metabolismo Energético/fisiologia , Insulina/biossíntese , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo
15.
Anat Rec (Hoboken) ; 304(3): 531-540, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32589363

RESUMO

MicroRNAs (miRNAs) participate in the repair of skin trauma. Our previous study indicated that loureirin A promoted hair follicle stem cells (HFSCs) to repair skin epidermis. However, the mechanism of miRNA-mediated regulation of loureirin A-induced HFSC differentiation remained to be explored. In the present study, HFSCs from rat vibrissa were identified by immunofluorescence in vitro. Microarray and quantitative real time polymerase chain reaction analyses demonstrated that miR-203a-3p was upregulated in differentiated HFSCs induced by loureirin A. The expression of cytoskeletal keratin (CK) 5 and involucrin was promoted by miR-203a-3p mimics while repressed by a miR-203a-3p inhibitor. Smad1 was identified as a key target of miR-203a-3p using target prediction tools. Luciferase reporter gene test confirmed a special target association between miR-203a-3p and Smad1. Short interfering Smad1 was transfected into HFSCs, and the expression levels of CK5 and involucrin were upregulated. Thus, it can be inferred that miR-203a-3p negatively regulated the expression of Smad1 and promoted the differentiation of loureirin A-induced HFSCs. Bone morphogenetic protein (BMP) signal inhibition and Wnt activation coregulate skin injury repair. BMP/Smad1 signaling is involved in maintaining the characteristics of HFSCs and inhibiting their differentiation. Our results showed that miR-203a-3p reduces Smad1 to release BMP inhibition. Taken together, miR-203a-3p/Smad1 is a potential therapeutic molecular target in skin wound healing, and may play an active role in wound repair and regenerative medicine.


Assuntos
Diferenciação Celular/fisiologia , Chalconas/farmacologia , Folículo Piloso/metabolismo , MicroRNAs/metabolismo , Proteína Smad1/metabolismo , Células-Tronco/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Folículo Piloso/efeitos dos fármacos , Masculino , MicroRNAs/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos , Proteínas Wnt/metabolismo
16.
FEBS Lett ; 580(14): 3539-44, 2006 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-16730715

RESUMO

Glucocorticoids (GC) are common components in chemotherapeutic protocols for lymphoid malignancies. GC-induced apoptosis requires the intrinsic, BCL-2 family-regulated pathway. Treatment of CCRF-CEM (T cell acute lymphoblastic leukemia) cells with the GC, dexamethasone (Dex), activates p38-mitogen activated protein kinase (p38-MAPK) and then induces mRNA transcription and synthesis levels of BIM, a BH3-only pro-apoptotic BCL-2 family member. Dex-induced apoptosis is dramatically inhibited by downregulation of BIM by shRNA or by pretreatment with a p38-MAPK inhibitor, SB203580, which also reduces BIM induction. These findings indicate that BIM induction through p38-MAPK activation is a critical pathway in GC-induced cell death.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , Leucemia-Linfoma de Células T do Adulto/patologia , Proteínas de Membrana/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Sequência de Bases , Proteína 11 Semelhante a Bcl-2 , Western Blotting , Linhagem Celular Tumoral , Primers do DNA , Ativação Enzimática , Humanos , Leucemia-Linfoma de Células T do Adulto/enzimologia
17.
Artigo em Inglês | MEDLINE | ID: mdl-25832828

RESUMO

Rasagiline, a monoamine oxidase B inhibitor, slowed disease progression in the SOD1 mouse, and in a case series of patients with amyotrophic lateral sclerosis (ALS). Here we determine whether rasagiline is safe and effective in ALS compared to historical placebo controls, and whether it alters mitochondrial biomarkers. We performed a prospective open-label, multicenter screening trial of 36 ALS patients treated with 2 mg oral rasagiline daily for 12 months. Outcomes included the slope of deterioration of the revised ALS Functional Rating Scale (ALSFRS-R), adverse event monitoring, time to treatment failure, and exploratory biomarkers. Participants experienced no serious drug-related adverse events, and the most common adverse event was nausea (11.1%). Rasagiline did not improve the rate of decline in the ALSFRS-R; however, differences in symptom duration compared to historical placebo controls differentially affected ALSFRS-R slope estimates. Rasagiline changed biomarkers over 12 months, such that the mitochondrial membrane potential increased (JC-1 red/green fluorescent ratio 1.92, p = 0.0001) and apoptosis markers decreased (Bcl-2/Bax ratio 0.24, p < 0.0001). In conclusion, engagement of exploratory biomarkers and questions about comparability of baseline characteristics lead us to recommend a further placebo-controlled trial.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Indanos/uso terapêutico , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Idoso , Apoptose/efeitos dos fármacos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Estudos Retrospectivos , Estatísticas não Paramétricas , Fatores de Tempo , Resultado do Tratamento , Proteína X Associada a bcl-2/metabolismo
18.
J Alzheimers Dis ; 33(4): 1135-46, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23099815

RESUMO

Alzheimer's disease (AD) patients have reduced brain acetylcholine and reversing this deficit yields clinical benefits. In this study we explored how increased cholinergic tone impacts cell bioenergetics, which are also perturbed in AD. We treated SH-SY5Y neuroblastoma cells with carbachol, a cholinergic agonist, and tested for bioenergetic flux and bioenergetic infrastructure changes. Carbachol rapidly increased both oxidative phosphorylation and glycolysis fluxes. ATP levels rose slightly, as did cell energy demand, and AMPK phosphorylation occurred. At least some of these effects depended on muscarinic receptor activation, ER calcium release, and ER calcium re-uptake. Our data show that increasing cholinergic signaling enhances cell bioenergetics, and reveal mechanisms that mediate this effect. Phenomena we observed could potentially explain why cholinesterase inhibitor therapy increases AD brain glucose utilization and N-acetyl aspartate levels. The question of whether cholinesterase inhibitors have a disease modifying effect in AD has long been debated; our data suggest a theoretical mechanism through which such an effect could potentially arise.


Assuntos
Agonistas Colinérgicos/farmacologia , Metabolismo Energético/fisiologia , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Acetilcolina/metabolismo , Carbacol/farmacologia , Linhagem Celular Tumoral , Metabolismo Energético/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Glicólise/fisiologia , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Transdução de Sinais/efeitos dos fármacos
19.
Adv Pharmacol ; 64: 83-126, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22840745

RESUMO

Mitochondria from persons with Alzheimer's disease (AD) differ from those of age-matched control subjects. Differences in mitochondrial morphology and function are well documented, and are not brain-limited. Some of these differences are present during all stages of AD, and are even seen in individuals who are without AD symptoms and signs but who have an increased risk of developing AD. This chapter considers the status of mitochondria in AD subjects, the potential basis for AD subject mitochondrial perturbations, and the implications of these perturbations. Data from multiple lines of investigation, including epidemiologic, biochemical, molecular, and cytoplasmic hybrid studies, are reviewed. The possibility that mitochondria could potentially constitute a reasonable AD therapeutic target is discussed, as are several potential mitochondrial medicine treatment strategies.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Mitocôndrias/patologia , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/patologia , Doença de Alzheimer/metabolismo , Animais , DNA Mitocondrial , Humanos , Doenças Mitocondriais/metabolismo , Oxirredução , Estresse Oxidativo
20.
J Alzheimers Dis ; 27(3): 483-90, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21841246

RESUMO

Biomarker studies demonstrate inheritance of glucose hypometabolism and increased amyloid-ß deposition in adult offspring of mothers, but not fathers, affected by late-onset Alzheimer's disease (LOAD). The underlying genetic mechanisms are unknown. We investigated whether cognitively normal (NL) individuals with a maternal history of LOAD (MH) have reduced platelet mitochondrial cytochrome oxidase activity (COX, electron transport chain complex IV) compared to those with paternal (PH) or negative family history (NH). Thirty-six consecutive NL individuals (age 55 ± 15 y, range 27-71 y, 56% female, CDR = 0, MMSE ≥28, 28% APOE-4 carriers), including 12 NH, 12 PH, and 12 MH, received a blood draw to measure platelet mitochondrial COX activity. Citrate synthase activity (CS) was measured as a reference. Groups were comparable for clinical and neuropsychological measures. We found that after correcting for CS, COX activity was reduced by 29% in MH compared to NH, and by 30% in MH compared to PH (p ≤ 0.006). Results remained significant controlling for age, gender, education, and APOE. No differences were found between PH and NH. COX measures discriminated MH from the other groups with accuracy ≥75%, and relative risk ≥3 (p ≤ 0.005). Among NL with LOAD-parents, only those with MH showed reduced COX activity in platelet mitochondria compared to PH and NH. The association between maternal history of LOAD and systemic COX reductions suggests transmission via mitochondrial DNA, which is exclusively maternally inherited in humans.


Assuntos
Doença de Alzheimer/enzimologia , Regulação para Baixo/genética , Complexo IV da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Mitocôndrias/enzimologia , Mães , Adulto , Idade de Início , Idoso , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Biomarcadores/sangue , Plaquetas/enzimologia , Estudos de Coortes , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Ativação Enzimática/genética , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Mitocôndrias/patologia , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA