Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(16)2022 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-36012191

RESUMO

Diabetes mellitus type 2 (T2DM) has been associated with alterations in the male reproductive tract, especially in the epididymis. Although it is known that T2DM alters epididymal physiology, disturbing mitochondrial function and favoring oxidative stress, the mechanisms remain unknown. Sirtuin 1 (SIRT1), peroxisome proliferators-activated receptor γ coactivator 1α (PGC-1α), and sirtuin 3 (SIRT3) are key regulators of mitochondrial function and inducers of antioxidant defenses. In this study, we hypothesized that the epididymal SIRT1/PGC-1α/SIRT3 axis mediates T2DM-induced epididymis dysfunction by controlling the oxidative profile. Using 7 Goto-Kakizaki (GK) rats (a non-obese model that spontaneously develops T2DM early in life), and 7 age-matched Wistar control rats, we evaluated the protein levels of SIRT1, PGC-1α, and SIRT3, as well as the expression of mitochondrial respiratory complexes. The activities of epididymal glutathione peroxidase (GPx), glutathione reductase (GR), superoxide dismutase (SOD), and catalase (CAT) were determined, as well as the epididymal antioxidant capacity. We also evaluated protein nitration, carbonylation, and lipid peroxidation in the epididymis. The T2DM rats presented with hyperglycemia and glucose intolerance. Epididymal levels of SIRT1, PGC-1α, and SIRT3 were decreased, as well as the expression of the mitochondrial complexes II, III, and V, in the T2DM rats. We found a significant decrease in the activities of SOD, CAT, and GPx, consistent with the lower antioxidant capacity and higher protein nitration and lipid peroxidation detected in the epididymis of the T2DM rats. In sum, T2DM disrupted the epididymal SIRT1/PGC-1α/SIRT3 pathway, which is associated with a compromised mitochondrial function. This resulted in a decline of the antioxidant defenses and an increased oxidative damage in that tissue, which may be responsible for the impaired male reproductive function observed in diabetic men.


Assuntos
Diabetes Mellitus Tipo 2 , Sirtuína 3 , Animais , Antioxidantes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Epididimo/metabolismo , Humanos , Masculino , Estresse Oxidativo/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Ratos , Ratos Wistar , Sirtuína 1/metabolismo , Sirtuína 3/metabolismo , Superóxido Dismutase/metabolismo
2.
Clin Sci (Lond) ; 135(9): 1103-1126, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33899910

RESUMO

Poor maternal nutrition in pregnancy affects fetal development, predisposing offspring to cardiometabolic diseases. The role of mitochondria during fetal development on later-life cardiac dysfunction caused by maternal nutrient reduction (MNR) remains unexplored. We hypothesized that MNR during gestation causes fetal cardiac bioenergetic deficits, compromising cardiac mitochondrial metabolism and reserve capacity. To enable human translation, we developed a primate baboon model (Papio spp.) of moderate MNR in which mothers receive 70% of control nutrition during pregnancy, resulting in intrauterine growth restriction (IUGR) offspring and later exhibiting myocardial remodeling and heart failure at human equivalent ∼25 years. Term control and MNR baboon offspring were necropsied following cesarean-section, and left ventricle (LV) samples were collected. MNR adversely impacted fetal cardiac LV mitochondria in a sex-dependent fashion. Increased maternal plasma aspartate aminotransferase, creatine phosphokinase (CPK), and elevated cortisol levels in MNR concomitant with decreased blood insulin in male fetal MNR were measured. MNR resulted in a two-fold increase in fetal LV mitochondrial DNA (mtDNA). MNR resulted in increased transcripts for several respiratory chain (NDUFB8, UQCRC1, and cytochrome c) and adenosine triphosphate (ATP) synthase proteins. However, MNR fetal LV mitochondrial complex I and complex II/III activities were significantly decreased, possibly contributing to the 73% decreased ATP content and increased lipid peroxidation. MNR fetal LV showed mitochondria with sparse and disarranged cristae dysmorphology. Conclusion: MNR disruption of fetal cardiac mitochondrial fitness likely contributes to the documented developmental programming of adult cardiac dysfunction, indicating a programmed mitochondrial inability to deliver sufficient energy to cardiac tissues as a chronic mechanism for later-life heart failure.


Assuntos
Transtornos da Nutrição Fetal/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Mitocôndrias Cardíacas/metabolismo , Nucleotídeos de Adenina/metabolismo , Animais , Feminino , Transtornos da Nutrição Fetal/patologia , Mitocôndrias Cardíacas/ultraestrutura , Estresse Oxidativo , Papio , Gravidez
3.
Int J Mol Sci ; 21(5)2020 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-32143329

RESUMO

Alzheimer's disease (AD) is the most common form of dementia worldwide, being characterized by the deposition of senile plaques, neurofibrillary tangles (enriched in the amyloid beta (Aß) peptide and hyperphosphorylated tau (p-tau), respectively) and memory loss. Aging, type 2 diabetes (T2D) and female sex (especially after menopause) are risk factors for AD, but their crosslinking mechanisms remain unclear. Most clinical trials targeting AD neuropathology failed and it remains incurable. However, evidence suggests that effective anti-T2D drugs, such as the GLP-1 mimetic and neuroprotector liraglutide, can be also efficient against AD. Thus, we aimed to study the benefits of a peripheral liraglutide treatment in AD female mice. We used blood and brain cortical lysates from 10-month-old 3xTg-AD female mice, treated for 28 days with liraglutide (0.2 mg/kg, once/day) to evaluate parameters affected in AD (e.g., Aß and p-tau, motor and cognitive function, glucose metabolism, inflammation and oxidative/nitrosative stress). Despite the limited signs of cognitive changes in mature female mice, liraglutide only reduced their cortical Aß1-42 levels. Liraglutide partially attenuated brain estradiol and GLP-1 and activated PKA levels, oxidative/nitrosative stress and inflammation in these AD female mice. Our results support the earlier use of liraglutide as a potential preventive/therapeutic agent against the accumulation of the first neuropathological features of AD in females.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Hipoglicemiantes/farmacologia , Inflamação/metabolismo , Liraglutida/farmacologia , Fragmentos de Peptídeos/metabolismo , Animais , Comportamento Animal , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Estradiol/metabolismo , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glicólise , Aprendizagem em Labirinto , Transtornos da Memória , Camundongos , Emaranhados Neurofibrilares/metabolismo , Estresse Nitrosativo , Estresse Oxidativo , Fenótipo , Placa Amiloide/metabolismo
4.
J Neurosci ; 37(10): 2776-2794, 2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28123081

RESUMO

Transcriptional deregulation and changes in mitochondrial bioenergetics, including pyruvate dehydrogenase (PDH) dysfunction, have been described in Huntington's disease (HD). We showed previously that the histone deacetylase inhibitors (HDACIs) trichostatin A and sodium butyrate (SB) ameliorate mitochondrial function in cells expressing mutant huntingtin. In this work, we investigated the effect of HDACIs on the regulation of PDH activity in striatal cells derived from HD knock-in mice and YAC128 mice. Mutant cells exhibited decreased PDH activity and increased PDH E1alpha phosphorylation/inactivation, accompanied by enhanced protein levels of PDH kinases 1 and 3 (PDK1 and PDK3). Exposure to dichloroacetate, an inhibitor of PDKs, increased mitochondrial respiration and decreased production of reactive oxygen species in mutant cells, emphasizing PDH as an interesting therapeutic target in HD. Treatment with SB and sodium phenylbutyrate, another HDACI, recovered cell viability and overall mitochondrial metabolism in mutant cells. Exposure to SB also suppressed hypoxia-inducible factor-1 (HIF-1α) stabilization and decreased the transcription of the two most abundant PDK isoforms, PDK2 and PDK3, culminating in increased PDH activation in mutant cells. Concordantly, PDK3 knockdown improved mitochondrial function, emphasizing the role of PDK3 inactivation on the positive effects achieved by SB treatment. YAC128 mouse brain presented higher mRNA levels of PDK1-3 and PDH phosphorylation and decreased energy levels that were significantly ameliorated after SB treatment. Furthermore, enhanced motor learning and coordination were observed in SB-treated YAC128 mice. These results suggest that HDACIs, particularly SB, promote the activity of PDH in the HD brain, helping to counteract HD-related deficits in mitochondrial bioenergetics and motor function.SIGNIFICANCE STATEMENT The present work provides a better understanding of mitochondrial dysfunction in Huntington's disease (HD) by showing that the pyruvate dehydrogenase (PDH) complex is a promising therapeutic target. In particular, the histone deacetylase inhibitor sodium butyrate (SB) may indirectly (through reduced hypoxia-inducible factor 1 alpha stabilization) decrease the expression of the most abundant PDH kinase isoforms (e.g., PDK3), ameliorating PDH activity and mitochondrial metabolism and further affecting motor behavior in HD mice, thus constituting a promising agent for HD neuroprotective treatment.


Assuntos
Inibidores de Histona Desacetilases/administração & dosagem , Doença de Huntington/tratamento farmacológico , Doença de Huntington/metabolismo , Neurônios/enzimologia , Fármacos Neuroprotetores/administração & dosagem , Complexo Piruvato Desidrogenase/metabolismo , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Neurônios/efeitos dos fármacos , Resultado do Tratamento
5.
Biochim Biophys Acta ; 1837(3): 335-44, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24361842

RESUMO

Pre-diabetes, a risk factor for type 2 diabetes development, leads to metabolic changes at testicular level. Peroxisome proliferator-activated receptor γ coactivator 1 α (PGC-1α) and Sirtuin 3 (Sirt3) are pivotal in mitochondrial function. We hypothesized that pre-diabetes disrupts testicular PGC-1α/Sirt3 axis, compromising testicular mitochondrial function. Using a high-energy-diet induced pre-diabetic rat model, we evaluated testicular levels of PGC-1α and its downstream targets, nuclear respiratory factors 1 (NRF-1) and 2 (NRF-2), mitochondrial transcription factor A (TFAM) and Sirt3. We also assessed mitochondrial DNA (mtDNA) content, mitochondrial function, energy levels and oxidative stress parameters. Protein levels were quantified by Western Blot, mtDNA content was determined by qPCR. Mitochondrial complex activity and oxidative stress parameters were spectrophotometrically evaluated. Adenine nucleotide levels, adenosine and its metabolites (inosine and hypoxanthine) were determined by reverse-phase HPLC. Pre-diabetic rats showed increased blood glucose levels and impaired glucose tolerance. Both testicular PGC-1α and Sirt3 levels were decreased. NRF-1, NRF-2 and TFAM were not altered. Testicular mtDNA content was decreased. Mitochondrial complex I activity was increased, whereas mitochondrial complex III activity was decreased. Adenylate energy charge was decreased in pre-diabetic rats, as were ATP and ADP levels. Conversely, AMP levels were increased, evidencing a decreased ATP/AMP ratio. Concerning to oxidative stress pre-diabetes decreased testicular antioxidant capacity and increased lipid and protein oxidation. In sum, pre-diabetes compromises testicular mitochondrial function by repressing PGC-1α/Sirt3 axis and mtDNA copy number, declining respiratory capacity and increasing oxidative stress. This study gives new insights into overall testicular bioenergetics at this prodromal stage of disease.


Assuntos
Metabolismo Energético/fisiologia , Estresse Oxidativo/fisiologia , Estado Pré-Diabético/fisiopatologia , Sirtuína 3/metabolismo , Testículo/metabolismo , Fatores de Transcrição/metabolismo , Difosfato de Adenosina/metabolismo , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Glicemia/metabolismo , Western Blotting , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Insulina/sangue , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Fator 1 Nuclear Respiratório/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Reação em Cadeia da Polimerase , Estado Pré-Diabético/sangue , Ratos , Ratos Wistar
6.
J Bioenerg Biomembr ; 47(1-2): 119-31, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25217852

RESUMO

The integrity of mitochondrial function is essential to cell life. It follows that disturbances of mitochondrial function will lead to disruption of cell function, expressed as disease or even death. Considering that neuronal uncoupling proteins (UCPs) decrease reactive oxygen species (ROS) production at the expense of energy production, it is important to understand the underlying mechanisms by which UCPs control the balance between the production of adenosine triphosphate (ATP) and ROS in the context of normal physiological activity and in pathological conditions. Here we review the current understanding of neuronal UCPs-mediated respiratory uncoupling process by performing a survey in their physiology and regulation. The latest findings regarding neuronal UCPs physiological roles and their involvement and interest as potential targets for therapeutic intervention in brain diseases will also be exploited.


Assuntos
Encefalopatias/metabolismo , Encéfalo/metabolismo , Canais Iônicos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Neurônios/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/patologia , Encefalopatias/tratamento farmacológico , Encefalopatias/patologia , Metabolismo Energético , Humanos , Mitocôndrias/patologia , Neurônios/patologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Desacopladora 1
7.
IUBMB Life ; 64(12): 951-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23129399

RESUMO

Alterations of the insulin signaling cascade underlie cognitive decline and the development of several neurodegenerative diseases. In recent years, a great interest has been put in studying the interaction between diabetes and Alzheimer's disease (AD). In fact, evidence shows that both diseases present several biochemical similarities including defects in the insulin signaling pathway. Here, we give an overview of the main functions of insulin in the central nervous system. The impact of insulin signaling impairment in brain aging and AD is also discussed. Finally, we present evidence supporting the notion that insulin is a link between diabetes and AD.


Assuntos
Envelhecimento , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus/metabolismo , Insulina/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Encéfalo/patologia , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Expressão Gênica , Glucose/metabolismo , Humanos , Insulina/genética , Resistência à Insulina , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neurônios/metabolismo , Neurônios/patologia , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Fatores de Risco , Transdução de Sinais
8.
Rev Port Cardiol ; 41(12): 979-984, 2022 12.
Artigo em Inglês, Português | MEDLINE | ID: mdl-36153294

RESUMO

INTRODUCTION: Heart disease and cancer are the two leading causes of morbidity and mortality worldwide. Advances in cancer screening and management have led to longer survival and better quality of life. Despite this progress, many cancer patients experience cardiovascular complications during and after cancer treatment. This study describes the experience of a cardio-oncology program at tertiary academic hospital. METHODS: In this retrospective observational study, cancer patients referred to the CHULN cardio-oncology consultation (COC) between January 2016 and December of 2019 were included. Data collected included: patient demographics, cancer type, reason for referral, cardiovascular risk factors, cardiac and oncologic treatments and clinical outcomes. RESULTS: A total of 520 patients (mean age: 65 ± 14 years; 65% women) were referred to the COC. The main reasons for referral were suspected heart failure (26%), pre-high risk chemotherapy assessment (20%) and decreased LVEF (15%). Pre-existing cardiovascular risk factors were common (79%) and 309 (59%) were taking cardiac medications. The most common type of malignancy was breast cancer (216, 41%) followed by gastrointestinal (139, 27%). More than half received anthracycline-based regimens (303, 58%). Most patients (401; 77%) successfully completed cancer therapy. At the time of last data collection, the majority of patients were alive (430, 83%). Cardiac-related mortality was observed in 16%. CONCLUSIONS: The close collaboration between cardiology and oncology teams and timely cardiac monitoring was the key to the majority of patients to completing their prescribed cancer therapy.


Assuntos
Neoplasias da Mama , Cardiopatias , Neoplasias , Humanos , Feminino , Pessoa de Meia-Idade , Idoso , Masculino , Qualidade de Vida , Oncologia , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Antraciclinas/efeitos adversos , Cardiopatias/complicações , Neoplasias da Mama/tratamento farmacológico , Centros de Atenção Terciária , Cardiotoxicidade/etiologia
9.
ACS Chem Neurosci ; 13(15): 2252-2260, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35868251

RESUMO

Polypharmacology is a new trend in amyotrophic lateral sclerosis (ALS) therapy and an effective way of addressing a multifactorial etiology involving excitotoxicity, mitochondrial dysfunction, oxidative stress, and microglial activation. Inspired by a reported clinical trial, we converted a riluzole (1)-rasagiline (2) combination into single-molecule multi-target-directed ligands. By a ligand-based approach, the highly structurally integrated hybrids 3-8 were designed and synthesized. Through a target- and phenotypic-based screening pipeline, we identified hit compound 6. It showed monoamine oxidase A (MAO-A) inhibitory activity (IC50 = 6.9 µM) rationalized by in silico studies as well as in vitro brain permeability. By using neuronal and non-neuronal cell models, including ALS-patient-derived cells, we disclosed for 6 a neuroprotective/neuroinflammatory profile similar to that of the parent compounds and their combination. Furthermore, the unexpected MAO inhibitory activity of 1 (IC50 = 8.7 µM) might add a piece to the puzzle of its anti-ALS molecular profile.


Assuntos
Esclerose Lateral Amiotrófica , Fármacos Neuroprotetores , Esclerose Lateral Amiotrófica/tratamento farmacológico , Humanos , Indanos , Ligantes , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Riluzol/farmacologia , Riluzol/uso terapêutico
10.
Free Radic Biol Med ; 146: 372-382, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31751762

RESUMO

Abnormal protein homeostasis (proteostasis), dysfunctional mitochondria, and aberrant redox signalling are often associated in neurodegenerative disorders, such as Huntington's (HD), Alzheimer's and Parkinson's diseases. It remains incompletely understood, however, how changes in redox signalling affect proteostasis mechanisms, including protein degradation pathways and unfolded protein responses (UPR). Here we address this open question by investigating the interplay between redox signalling and proteostasis in a mouse model of HD, and by examining the in vivo effects of the mitochondria-targeted antioxidant MitoQ. We performed behavioural tests in wild-type and R6/2 HD mice, examined markers of oxidative stress, UPR activation, and the status of key protein degradation pathways in brain and peripheral tissues. We show that R6/2 mice present widespread markers of oxidative stress, with tissue-specific changes in proteostasis that were more pronounced in the brain and muscle than in the liver. R6/2 mice presented increased levels of cytosolic and mitochondrial chaperones, particularly in muscle, indicating UPR activation. Treatment with MitoQ significantly ameliorated fine motor control of R6/2 mice, and reduced markers of oxidative damage in muscle. Additionally, MitoQ attenuated overactive autophagy induction in the R6/2 muscle, which has been associated with muscle wasting. Treatment with MitoQ did not alter autophagy markers in the brain, in agreement with its low brain bioavailability, which limits the risk of impairing neuronal protein clearance mechanisms. This study supports the hypotheses that abnormal redox signalling in muscle contributes to altered proteostasis and motor impairment in HD, and that redox interventions can improve muscle performance, highlighting the importance of peripheral therapeutics in HD.


Assuntos
Doença de Huntington , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Modelos Animais de Doenças , Doença de Huntington/tratamento farmacológico , Doença de Huntington/genética , Doença de Huntington/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Oxirredução , Proteostase
11.
Neurobiol Aging ; 89: 12-23, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32143981

RESUMO

The nigrostriatal dopaminergic system (NDS) controls motor activity, and its impairment during type 2 diabetes (T2D) progression could increase Parkinson's disease risk in diabetics. If so, whether glycemia regulation prevents this impairment needs to be addressed. We investigated whether T2D impairs the NDS and whether dipeptidyl peptidase-4 inhibition (DPP-4i; a clinical strategy against T2D but also neuroprotective in animal models) prevents this effect, in middle-aged mice. Neither T2D (induced by 12 months of high-fat diet) nor aging (14 months) changed striatal dopamine content assessed by high-performance liquid chromatography. However, T2D reduced basal and amphetamine-stimulated striatal extracellular dopamine, assessed by microdialysis. Both the DPP-4i linagliptin and the sulfonylurea glimepiride (an antidiabetic comparator unrelated to DPP-4i) counteracted these effects. The functional T2D-induced effects did not correlate with NDS neuronal/glial alterations. However, aging itself affected striatal neurons/glia, and the glia effects were counteracted mainly by DPP-4i. These findings show NDS functional pathophysiology in T2D and suggest the preventive use of two unrelated anti-T2D drugs. Moreover, DPP-4i counteracted striatal age-related glial alterations suggesting striatal rejuvenation properties.


Assuntos
Envelhecimento/metabolismo , Corpo Estriado/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Dopamina/metabolismo , Linagliptina/farmacologia , Substância Negra/metabolismo , Compostos de Sulfonilureia/farmacologia , Animais , Diabetes Mellitus Tipo 2/complicações , Progressão da Doença , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Doença de Parkinson/etiologia , Doença de Parkinson/prevenção & controle , Risco
12.
Biochim Biophys Acta ; 1783(6): 994-1002, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18348871

RESUMO

Previously we demonstrated that insulin protects against neuronal oxidative stress by restoring antioxidants and energy metabolism. In this study, we analysed how insulin influences insulin-(IR) and insulin growth factor-1 receptor (IGF-1R) intracellular signaling pathways after oxidative stress caused by ascorbate/Fe2+ in rat cortical neurons. Insulin prevented oxidative stress-induced decrease in tyrosine phosphorylation of IR and IGF-1R and Akt inactivation. Insulin also decreased the active form of glycogen synthase kinase-3beta (GSK-3beta) upon oxidation. Since phosphatidylinositol 3-kinase (PI-3K)/Akt-mediated inhibition of GSK-3beta may stimulate protein synthesis and decrease apoptosis, we analysed mRNA and protein expression of "candidate" proteins involved in antioxidant defense, glucose metabolism and apoptosis. Insulin prevented oxidative stress-induced increase in glutathione peroxidase-1 and decrease in hexokinase-II expression, supporting previous findings of changes in glutathione redox cycle and glycolysis. Moreover, insulin precluded Bcl-2 decrease and caspase-3 increased expression. Concordantly, insulin abolished caspase-3 activity and DNA fragmentation caused by oxidative stress. Thus, insulin-mediated activation of IR/IGF-1R stimulates PI-3K/Akt and inhibits GSK-3beta signaling pathways, modifying neuronal antioxidant defense-, glucose metabolism- and anti-apoptotic-associated protein synthesis. These and previous data implicate insulin as a promising neuroprotective agent against oxidative stress associated with neurodegenerative diseases.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Caspase 3/metabolismo , Feminino , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hexoquinase/metabolismo , Neurônios/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Gravidez , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Receptor IGF Tipo 1/metabolismo , Tirosina/metabolismo , Glutationa Peroxidase GPX1
13.
J Alzheimers Dis ; 16(4): 741-61, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19387110

RESUMO

The processes underlying the pathogenesis of Alzheimer's disease involve several factors including impaired glucose/energy metabolism, mitochondrial dysfunction, oxidative stress and altered insulin-signaling pathways. This review is mainly devoted to discuss evidence supporting the notion that mitochondrial dysfunction and oxidative stress are interconnected and intimately associated with the development and progression of Alzheimer's disease. Furthermore, the review explores the role of insulin signaling in the pathophysiology of the disease. Indeed, several studies have begun to find links between insulin and mechanisms with clear pathogenic implications for this disorder. Understanding the key mechanisms involved in the etiopathogenesis of Alzheimer's disease may provide opportunities for the design of efficacious preventive and therapeutic strategies.


Assuntos
Doença de Alzheimer , Insulina/metabolismo , Mitocôndrias/fisiologia , Estresse Oxidativo/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Humanos , Mitocôndrias/patologia
14.
Med Chem ; 4(4): 358-64, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18673148

RESUMO

We aimed to investigate whether metformin protects the brain against the oxidative imbalance promoted by type 2 diabetes. This study analyzed the effect of metformin on oxidative stress markers (thiobarbituric acid reactive substances (TBARS), malondialdehyde (MDA) and carbonyl groups), hydrogen peroxide (H(2)O(2)) levels, non-enzymatic antioxidant defenses [reduced (GSH) and oxidized (GSSG) glutathione and vitamin E] and enzymatic antioxidant defenses [glutathione peroxidase (GPx), glutathione reductase (GRed) and manganese superoxide dismutase (MnSOD)] in brain homogenates of diabetic GK rats, a model of type 2 diabetes. For this purpose we compared brain homogenates obtained from untreated GK rats versus GK rats treated with metformin during a period of 4 weeks. Brain homogenates obtained from Wistar rats were used as control. The MDA levels, GPx and GRed activities are significantly higher in untreated GK rats, while TBARS levels, carbonyl groups, glutathione content and vitamin E levels remain statistically unchanged when compared with control rats. In contrast, MnSOD activity and the levels of H(2)O(2) are significantly decreased in untreated GK rats when compared with control animals. However, metformin treatment normalized the majority of the parameters altered by diabetes. We observed that metformin, besides its antihyperglycemic action, induces a significant decrease in TBARS and MDA levels, GPx and GRed activities and a significant increase in GSH levels and MnSOD activity. These results indicate that metformin protects against diabetes-associated oxidative stress suggesting that metformin could be an effective neuroprotective agent.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metformina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Glutationa/metabolismo , Peróxido de Hidrogênio/metabolismo , Masculino , Oxirredução , Proteínas/metabolismo , Ratos
15.
Neural Regen Res ; 18(7): 1486-1488, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36571346
16.
Sci Rep ; 8(1): 8961, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29895889

RESUMO

Neuronal loss alongside altered energy metabolism, are key features of Huntington's disease (HD) pathology. The orexigenic gut-peptide hormone ghrelin is known to stimulate appetite and affect whole body energy metabolism. Liraglutide is an efficient anti-type 2 diabetes incretin drug, with neuroprotective effects alongside anorectic properties. Combining liraglutide with the orexigenic peptide ghrelin may potentially promote brain/cognitive function in HD. The R6/2 mouse model of HD exhibits progressive central pathology, weight loss, deranged glucose metabolism, skeletal muscle atrophy and altered body composition. In this study, we targeted energy metabolism in R6/2 mice using a co-administration of liraglutide and ghrelin. We investigated their effect on brain cortical hormone-mediated intracellular signalling pathways, metabolic and apoptotic markers, and the impact on motor function in HD. We here demonstrate that liraglutide, alone or together with ghrelin (subcutaneous daily injections of 150 µg/kg (ghrelin) and 0.2 mg/kg (liraglutide), for 2 weeks), normalized glucose homeostatic features in the R6/2 mouse, without substantially affecting body weight or body composition. Liraglutide alone decreased brain cortical active GLP-1 and IGF-1 levels in R6/2 mice, alongside higher ADP levels. Liraglutide plus ghrelin decreased brain insulin, lactate, AMP and cholesterol levels in R6/2 mice. Taken together, our findings encourage further studies targeting energy metabolism in HD.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético/efeitos dos fármacos , Grelina/farmacologia , Doença de Huntington/tratamento farmacológico , Liraglutida/farmacologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Quimioterapia Combinada , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Masculino , Camundongos , Camundongos Transgênicos
17.
Mol Neurobiol ; 55(5): 4030-4050, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28573460

RESUMO

Type 2 diabetes (T2D) is a modern socioeconomic burden, mostly due to its long-term complications affecting nearly all tissues. One of them is the brain, whose dysfunctional intracellular quality control mechanisms (namely autophagy) may upregulate apoptosis, leading to cognitive dysfunction and Alzheimer disease (AD). Since impaired brain insulin signaling may constitute the crosslink between T2D and AD, its restoration may be potentially therapeutic herein. Accordingly, the insulinotropic anti-T2D drugs from glucagon-like peptide-1 (GLP-1) mimetics, namely, exendin-4 (Ex-4), could be a promising therapy. In line with this, we hypothesized that peripherally administered Ex-4 rescues brain intracellular signaling pathways, promoting autophagy and ultimately protecting against chronic T2D-induced apoptosis. Thus, we aimed to explore the effects of chronic, continuous, subcutaneous (s.c.) exposure to Ex-4 in brain cortical GLP-1/insulin/insulin-like growth factor-1 (IGF-1) signaling, and in autophagic and cell death mechanisms in middle-aged (8 months old), male T2D Goto-Kakizaki (GK) rats. We used brain cortical homogenates obtained from middle-aged (8 months old) male Wistar (control) and T2D GK rats. Ex-4 was continuously administered for 28 days, via s.c. implanted micro-osmotic pumps (5 µg/kg/day; infusion rate 2.5 µL/h). Peripheral characterization of the animal models was given by the standard biochemical analyses of blood or plasma, the intraperitoneal glucose tolerance test, and the heart rate. GLP-1, insulin, and IGF-1, their downstream signaling and autophagic markers were evaluated by specific ELISA kits and Western blotting. Caspase-like activities and other apoptotic markers were given by colorimetric methods and Western blotting. Chronic Ex-4 treatment attenuated peripheral features of T2D in GK rats, including hyperglycemia and insulin resistance. Furthermore, s.c. Ex-4 enhanced their brain cortical GLP-1 and IGF-1 levels, and subsequent signaling pathways. Specifically, Ex-4 stimulated protein kinase A (PKA) and phosphoinositide 3-kinase (PI3K)/Akt signaling, increasing cGMP and AMPK levels, and decreasing GSK3ß and JNK activation in T2D rat brains. Moreover, Ex-4 regulated several markers for autophagy in GK rat brains (as mTOR, PI3K class III, LC3 II, Atg7, p62, LAMP-1, and Parkin), ultimately protecting against apoptosis (by decreasing several caspase-like activities and mitochondrial cytochrome c, and increasing Bcl2 levels upon T2D). Altogether, this study demonstrates that peripheral Ex-4 administration may constitute a promising therapy against the chronic complications of T2D affecting the brain.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia , Encéfalo/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/patologia , Exenatida/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Exenatida/administração & dosagem , Masculino , Modelos Biológicos , Ratos Wistar , Transdução de Sinais
18.
Diabetes ; 55(10): 2863-70, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17003354

RESUMO

We previously demonstrated that insulin has a neuroprotective role against oxidative stress, a deleterious condition associated with diabetes, ischemia, and age-related neurodegenerative diseases. In this study, we investigated the effect of insulin on neuronal glucose uptake and metabolism after oxidative stress in rat primary cortical neurons. On oxidative stress, insulin stimulates neuronal glucose uptake and subsequent metabolism into pyruvate, restoring intracellular ATP and phosphocreatine. Insulin also increases intracellular and decreases extracellular adenosine, counteracting the effect of oxidative stress. Insulin effects are apparently mediated by phosphatidylinositol 3-K and extracellular signal-regulated kinase signaling pathways. Extracellular adenosine under oxidative stress is largely inhibited after blockade of ecto-5'-nucleotidase, suggesting that extracellular adenosine results preferentially from ATP release and catabolism. Moreover, insulin appears to interfere with the ATP release induced by oxidative stress, regulating extracellular adenosine levels. In conclusion, insulin neuroprotection against oxidative stress-mediated damage involves 1) stimulation of glucose uptake and metabolism, increasing energy levels and intracellular adenosine and, ultimately, uric acid formation and 2) a decrease in extracellular adenosine, which may reduce the facilitatory activity of adenosine receptors.


Assuntos
Insulina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo , Nucleotídeos de Adenina/metabolismo , Adenosina/metabolismo , Androstadienos/farmacologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Cromonas/farmacologia , Flavonoides/farmacologia , Glucose/metabolismo , Ácido Láctico/metabolismo , Morfolinas/farmacologia , Fosfocreatina/metabolismo , Ácido Pirúvico/metabolismo , Ratos , Ratos Wistar , Tioinosina/análogos & derivados , Tioinosina/farmacologia , Wortmanina
19.
Sci Rep ; 7(1): 13896, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-29066728

RESUMO

Accumulating evidence suggests altered energy metabolism as a key feature in Huntington's disease (HD) pathology. Hyper-catabolism, including weight loss and muscle atrophy, is seen in HD patients and HD mouse models. Metabolic hormones are key players, not only in energy metabolism, but also in neurodegenerative processes. Ghrelin, a gut peptide-hormone, plays an important role in regulating energy metabolism, stimulating appetite, and affects brain function and increases neuronal survival. The R6/2 mouse model of HD has previously been shown to exhibit progressive weight loss, dysregulated glucose metabolism, skeletal muscle atrophy and altered body composition. In this study, we targeted energy metabolism in R6/2 mice using ghrelin administration, with the primary aim to delay weight loss and reduce muscle atrophy. We also evaluated glucose metabolism and behaviour. We here demonstrate that ghrelin administration (subcutaneous 150 µg/kg daily injections) for 4 weeks, reversed the catabolic gene expression profile (increased expression of Caspase 8, Traf-5 and Creb1) seen in R6/2 mouse skeletal muscle. Skeletal muscle morphology was also improved with ghrelin, and importantly, ghrelin administration normalized behavioural deficits in R6/2 mice. Taken together, our findings encourage further studies targeting metabolism in HD.


Assuntos
Grelina/farmacologia , Doença de Huntington/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Biomarcadores/sangue , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Grelina/uso terapêutico , Glucose/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Doença de Huntington/sangue , Doença de Huntington/complicações , Doença de Huntington/tratamento farmacológico , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Músculo Esquelético/patologia , Atrofia Muscular/complicações , Atrofia Muscular/tratamento farmacológico , Comportamento de Nidação/efeitos dos fármacos , Ratos
20.
Neuropeptides ; 58: 73-81, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26876526

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder linked to the expression of mutant huntingtin. Bioenergetic dysfunction has been described to contribute to HD pathogenesis. Thus, treatment paradigms aimed to ameliorate energy deficits appear to be suitable candidates in HD. In previous studies, we observed protective effects of insulin growth factor-1 (IGF-1) in YAC128 and R6/2 mice, two HD mouse models, whereas IGF-1 and/or insulin halted mitochondrial-driven oxidative stress in mutant striatal cells and mitochondrial dysfunction in HD human lymphoblasts. Here, we analyzed the effect of IGF-1 versus insulin on energy metabolic parameters using striatal cells derived from HD knock-in mice and primary cortical cultures from YAC128 mice. STHdh(Q111/Q111) cells exhibited decreased ATP/ADP ratio and increased phosphocreatine levels. Moreover, pyruvate levels were increased in mutant cells, most probably in consequence of a decrease in pyruvate dehydrogenase (PDH) protein expression and increased PDH phosphorylation, reflecting its inactivation. Insulin and IGF-1 treatment significantly decreased phosphocreatine levels, whereas IGF-1 only decreased pyruvate levels in mutant cells. In a different scenario, primary cortical cultures derived from YAC128 mice also displayed energetic abnormalities. We observed a decrease in both ATP/ADP and phosphocreatine levels, which were prevented following exposure to insulin or IGF-1. Furthermore, decreased lactate levels in YAC128 cultures occurred concomitantly with a decline in lactate dehydrogenase activity, which was ameliorated with both insulin and IGF-1. These data demonstrate differential HD-associated metabolic dysfunction in striatal cell lines and primary cortical cultures, both of which being alleviated by insulin and IGF-1.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Proteína Huntingtina/genética , Doença de Huntington/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Insulina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Animais , Linhagem Celular , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Camundongos , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA