Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Appl Physiol ; 119(10): 2301-2312, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31451954

RESUMO

PURPOSE: To compare concentric and eccentric cycling performed by older adults for metabolic demand and post-exercise oxidative stress, inflammation and muscle damage. METHODS: Eight male and two female healthy older adults (60.4 ± 6.8 years) performed 30 min of moderate-intensity concentric (CONC-M: 50% maximum power output; POmax) and eccentric cycling (ECC-M: 50% POmax) and high-intensity eccentric cycling (ECC-H: 100% POmax) in a randomized order. Average power output (PO), oxygen consumption (VO2), heart rate (HR) and rate of perceived exertion were recorded during cycling. Some indirect markers of muscle damage were assessed before, and immediately, 24 and 48 h after cycling. Markers of oxidative stress (malondialdehyde: MDA, protein carbonyl), antioxidant (total antioxidant capacity, glutathione peroxidase activity: GPx) and inflammation (IL-6, TNF-α) were measured before and 5 min after cycling. RESULTS: PO in ECC-H (202.6 ± 78.5 W) was > 50% greater (P < 0.05) than that of CONC-M (98.6 ± 33.1 W) and ECC-M (112.0 ± 42.1 W). VO2 and HR were also greater (P < 0.05) for ECC-H than CONC-M (50% and 17%, respectively) and ECC-M (40% and 23%, respectively). Muscle strength loss at 1 day post-exercise (8-22%), peak soreness (10-62 mm) and creatine kinase activity (30-250 IU/L) after ECC-H were greater (P < 0.05) than those after ECC-M and CONC-M. MDA decreased (P < 0.05) after CONC-M (- 28%) and ECC-M (- 22%), but not after ECC-H. GPx activity increased after all exercises similarly (20-27%). IL-6 increased (P < 0.05) only after ECC-H (18%). CONCLUSION: Oxidative stress was minimal after eccentric cycling, but high-intensity eccentric cycling induced moderate muscle damage and inflammation, which is not desirable for older individuals.


Assuntos
Mialgia/etiologia , Estresse Oxidativo , Condicionamento Físico Humano/métodos , Idoso , Feminino , Glutationa Peroxidase/sangue , Frequência Cardíaca , Humanos , Interleucina-6/sangue , Masculino , Malondialdeído/sangue , Pessoa de Meia-Idade , Contração Muscular , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Mialgia/sangue , Consumo de Oxigênio , Condicionamento Físico Humano/efeitos adversos , Esforço Físico , Carbonilação Proteica , Distribuição Aleatória , Fator de Necrose Tumoral alfa/sangue
2.
Biochim Biophys Acta Mol Basis Dis ; 1864(11): 3685-3695, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30251688

RESUMO

Duchenne muscular dystrophy (DMD) is characterized by a severe and progressive destruction of muscle fibers associated with altered Ca2+ homeostasis. We have previously shown that the IP3 receptor (IP3R) plays a role in elevating basal cytoplasmic Ca2+ and that pharmacological blockade of IP3R restores muscle function. Moreover, we have shown that the IP3R pathway negatively regulates autophagy by controlling mitochondrial Ca2+ levels. Nevertheless, it remains unclear whether IP3R is involved in abnormal mitochondrial Ca2+ levels, mitochondrial dynamics, or autophagy and mitophagy observed in adult DMD skeletal muscle. Here, we show that the elevated basal autophagy and autophagic flux levels were normalized when IP3R was downregulated in mdx fibers. Pharmacological blockade of IP3R in mdx fibers restored both increased mitochondrial Ca2+ levels and mitochondrial membrane potential under resting conditions. Interestingly, mdx mitochondria changed from a fission to an elongated state after IP3R knockdown, and the elevated mitophagy levels in mdx fibers were normalized. To our knowledge, this is the first study associating IP3R1 activity with changes in autophagy, mitochondrial Ca2+ levels, mitochondrial membrane potential, mitochondrial dynamics, and mitophagy in adult mouse skeletal muscle. Moreover, these results suggest that increased IP3R activity in mdx fibers plays an important role in the pathophysiology of DMD. Overall, these results lead us to propose the use of specific IP3R blockers as a new pharmacological treatment for DMD, given their ability to restore both autophagy/mitophagy and mitochondrial function.


Assuntos
Autofagia/efeitos dos fármacos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/patologia , Animais , Cálcio/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Técnicas de Silenciamento de Genes , Humanos , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Receptores de Inositol 1,4,5-Trifosfato/genética , Compostos Macrocíclicos/farmacologia , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos mdx , Mitocôndrias/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Oxazóis/farmacologia , RNA Interferente Pequeno/metabolismo
3.
Front Physiol ; 9: 791, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29988564

RESUMO

Aim: We hypothesize that both type-1 ryanodine receptor (RyR1) and IP3-receptor (IP3R) calcium channels are necessary for the mitochondrial Ca2+ increase caused by membrane depolarization induced by potassium (or by electrical stimulation) of single skeletal muscle fibers; this calcium increase would couple muscle fiber excitation to an increase in metabolic output from mitochondria (excitation-metabolism coupling). Methods: Mitochondria matrix and cytoplasmic Ca2+ levels were evaluated in fibers isolated from flexor digitorium brevis muscle using plasmids for the expression of a mitochondrial Ca2+ sensor (CEPIA3mt) or a cytoplasmic Ca2+ sensor (RCaMP). The role of intracellular Ca2+ channels was evaluated using both specific pharmacological inhibitors (xestospongin B for IP3R and Dantrolene for RyR1) and a genetic approach (shIP3R1-RFP). O2 consumption was detected using Seahorse Extracellular Flux Analyzer. Results: In isolated muscle fibers cell membrane depolarization increased both cytoplasmic and mitochondrial Ca2+ levels. Mitochondrial Ca2+ uptake required functional inositol IP3R and RyR1 channels. Inhibition of either channel decreased basal O2 consumption rate but only RyR1 inhibition decreased ATP-linked O2 consumption. Cell membrane depolarization-induced Ca2+ signals in sub-sarcolemmal mitochondria were accompanied by a reduction in mitochondrial membrane potential; Ca2+ signals propagated toward intermyofibrillar mitochondria, which displayed increased membrane potential. These results are compatible with slow, Ca2+-dependent propagation of mitochondrial membrane potential from the surface toward the center of the fiber. Conclusion: Ca2+-dependent changes in mitochondrial membrane potential have different kinetics in the surface vs. the center of the fiber; these differences are likely to play a critical role in the control of mitochondrial metabolism, both at rest and after membrane depolarization as part of an "excitation-metabolism" coupling process in skeletal muscle fibers.

4.
PLoS One ; 11(12): e0168255, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27977752

RESUMO

Testosterone induces cardiac hypertrophy through a mechanism that involves a concerted crosstalk between cytosolic and nuclear signaling pathways. Nuclear factor of activated T-cells (NFAT) is associated with the promotion of cardiac hypertrophy, glycogen synthase kinase-3ß (GSK-3ß) is considered to function as a negative regulator, mainly by modulating NFAT activity. However, the role played by calcineurin-NFAT and GSK-3ß signaling in testosterone-induced cardiac hypertrophy has remained unknown. Here, we determined that testosterone stimulates cardiac myocyte hypertrophy through NFAT activation and GSK-3ß inhibition. Testosterone increased the activity of NFAT-luciferase (NFAT-Luc) in a time- and dose-dependent manner, with the activity peaking after 24 h of stimulation with 100 nM testosterone. NFAT-Luc activity induced by testosterone was blocked by the calcineurin inhibitors FK506 and cyclosporine A and by 11R-VIVIT, a specific peptide inhibitor of NFAT. Conversely, testosterone inhibited GSK-3ß activity as determined by increased GSK-3ß phosphorylation at Ser9 and ß-catenin protein accumulation, and also by reduction in ß-catenin phosphorylation at residues Ser33, Ser37, and Thr41. GSK-3ß inhibition with 1-azakenpaullone or a GSK-3ß-targeting siRNA increased NFAT-Luc activity, whereas overexpression of a constitutively active GSK-3ß mutant (GSK-3ßS9A) inhibited NFAT-Luc activation mediated by testosterone. Testosterone-induced cardiac myocyte hypertrophy was established by increased cardiac myocyte size and [3H]-leucine incorporation (as a measurement of cellular protein synthesis). Calcineurin-NFAT inhibition abolished and GSK-3ß inhibition promoted the hypertrophy stimulated by testosterone. GSK-3ß activation by GSK-3ßS9A blocked the increase of hypertrophic markers induced by testosterone. Moreover, inhibition of intracellular androgen receptor prevented testosterone-induced NFAT-Luc activation. Collectively, these results suggest that cardiac myocyte hypertrophy induced by testosterone involves a cooperative mechanism that links androgen signaling with the recruitment of NFAT through calcineurin activation and GSK-3ß inhibition.


Assuntos
Cardiomegalia/induzido quimicamente , Glicogênio Sintase Quinase 3 beta/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fatores de Transcrição NFATC/fisiologia , Testosterona/efeitos adversos , Animais , Animais Recém-Nascidos , Cardiomegalia/genética , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/genética , Fatores de Transcrição NFATC/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
5.
Front Physiol ; 7: 282, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27471471

RESUMO

Reactive oxygen species (ROS) participate as signaling molecules in response to exercise in skeletal muscle. However, the source of ROS and the molecular mechanisms involved in these phenomena are still not completely understood. The aim of this work was to study the role of skeletal muscle NADPH oxidase isoform 2 (NOX2) in the molecular response to physical exercise in skeletal muscle. BALB/c mice, pre-treated with a NOX2 inhibitor, apocynin, (3 mg/kg) or vehicle for 3 days, were swim-exercised for 60 min. Phospho-p47(phox) levels were significantly upregulated by exercise in flexor digitorum brevis (FDB). Moreover, exercise significantly increased NOX2 complex assembly (p47(phox)-gp91(phox) interaction) demonstrated by both proximity ligation assay and co-immunoprecipitation. Exercise-induced NOX2 activation was completely inhibited by apocynin treatment. As expected, exercise increased the mRNA levels of manganese superoxide dismutase (MnSOD), glutathione peroxidase (GPx), citrate synthase (CS), mitochondrial transcription factor A (tfam) and interleukin-6 (IL-I6) in FDB muscles. Moreover, the apocynin treatment was associated to a reduced activation of p38 MAP kinase, ERK 1/2, and NF-κB signaling pathways after a single bout of exercise. Additionally, the increase in plasma IL-6 elicited by exercise was decreased in apocynin-treated mice compared with the exercised vehicle-group (p < 0.001). These results were corroborated using gp91-dstat in an in vitro exercise model. In conclusion, NOX2 inhibition by both apocynin and gp91dstat, alters the intracellular signaling to exercise and electrical stimuli in skeletal muscle, suggesting that NOX2 plays a critical role in molecular response to an acute exercise.

6.
Skelet Muscle ; 6: 15, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27069569

RESUMO

BACKGROUND: Electrical activity regulates the expression of skeletal muscle genes by a process known as "excitation-transcription" (E-T) coupling. We have demonstrated that release of adenosine 5'-triphosphate (ATP) during depolarization activates membrane P2X/P2Y receptors, being the fundamental mediators between electrical stimulation, slow intracellular calcium transients, and gene expression. We propose that this signaling pathway would require the proper coordination between the voltage sensor (dihydropyridine receptor, DHPR), pannexin 1 channels (Panx1, ATP release conduit), nucleotide receptors, and other signaling molecules. The goal of this study was to assess protein-protein interactions within the E-T machinery and to look for novel constituents in order to characterize the signaling complex. METHODS: Newborn derived myotubes, adult fibers, or triad fractions from rat or mouse skeletal muscles were used. Co-immunoprecipitation, 2D blue native SDS/PAGE, confocal microscopy z-axis reconstruction, and proximity ligation assays were combined to assess the physical proximity of the putative complex interactors. An L6 cell line overexpressing Panx1 (L6-Panx1) was developed to study the influence of some of the complex interactors in modulation of gene expression. RESULTS: Panx1, DHPR, P2Y2 receptor (P2Y2R), and dystrophin co-immunoprecipitated in the different preparations assessed. 2D blue native SDS/PAGE showed that DHPR, Panx1, P2Y2R and caveolin-3 (Cav3) belong to the same multiprotein complex. We observed co-localization and protein-protein proximity between DHPR, Panx1, P2Y2R, and Cav3 in adult fibers and in the L6-Panx1 cell line. We found a very restricted location of Panx1 and Cav3 in a putative T-tubule zone near the sarcolemma, while DHPR was highly expressed all along the transverse (T)-tubule. By Panx1 overexpression, extracellular ATP levels were increased both at rest and after electrical stimulation. Basal mRNA levels of the early gene cfos and the oxidative metabolism markers citrate synthase and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) were significantly increased by Panx1 overexpression. Interleukin 6 expression evoked by 20-Hz electrical stimulation (270 pulses, 0.3 ms each) was also significantly upregulated in L6-Panx1 cells. CONCLUSIONS: We propose the existence of a relevant multiprotein complex that coordinates events involved in E-T coupling. Unveiling the molecular actors involved in the regulation of gene expression will contribute to the understanding and treatment of skeletal muscle disorders due to wrong-expressed proteins, as well as to improve skeletal muscle performance.


Assuntos
Contração Muscular , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Transcrição Gênica , Trifosfato de Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Caveolina 3/genética , Caveolina 3/metabolismo , Linhagem Celular , Conexinas/genética , Conexinas/metabolismo , Distrofina/genética , Distrofina/metabolismo , Estimulação Elétrica , Regulação da Expressão Gênica , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Complexos Multiproteicos , Proteínas Musculares/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Ratos Wistar , Receptores Purinérgicos P2Y2/genética , Receptores Purinérgicos P2Y2/metabolismo , Ativação Transcricional , Transfecção
7.
Biochim Biophys Acta ; 1852(7): 1410-9, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25857619

RESUMO

Duchenne muscular dystrophy is a fatal X-linked genetic disease, caused by mutations in the dystrophin gene, which cause functional loss of this protein. This pathology is associated with an increased production of reactive oxygen (ROS) and nitrogen species. The aim of this work was to study the alterations in NF-κB activation and interleukin-6 (IL-6) expression induced by membrane depolarization in dystrophic mdx myotubes. Membrane depolarization elicited by electrical stimulation increased p65 phosphorylation, NF-κB transcriptional activity and NF-κB-dependent IL-6 expression in wt myotubes, whereas in mdx myotubes it had the opposite effect. We have previously shown that depolarization-induced intracellular Ca2+ increases and ROS production are necessary for NF-κB activation and stimulation of gene expression in wt myotubes. Dystrophic myotubes showed a reduced amplitude and area under the curve of the Ca2+ transient elicited by electrical stimulation. On the other hand, electrical stimuli induced higher ROS production in mdx than wt myotubes, which were blocked by NOX2 inhibitors. Moreover, mRNA expression and protein levels of the NADPH oxidase subunits: p47phox and gp91phox were increased in mdx myotubes. Looking at ROS-dependence of NF-κB activation we found that in wt myotubes external administration of 50 µM H2O2 increased NF-κB activity; after administration of 100 and 200 µM H2O2 there was no effect. In mdx myotubes there was a dose-dependent reduction in NF-κB activity in response to external administration of H2O2, with a significant effect of 100 µM and 200 µM, suggesting that ROS levels are critical for NF-κB activity. Prior blockage with NOX2 inhibitors blunted the effects of electrical stimuli in both NF-κB activation and IL-6 expression. Finally, to ascertain whether stimulation of NF-κB and IL-6 gene expression by the inflammatory pathway is also impaired in mdx myotubes, we studied the effect of lipopolysaccharide on both NF-κB activation and IL-6 expression. Exposure to lipopolysaccharide induced a dramatic increase in both NF-κB activation and IL-6 expression in both wt and mdx myotubes, suggesting that the altered IL-6 gene expression after electrical stimulation in mdx muscle cells is due to dysregulation of Ca2+ release and ROS production, both of which impinge on NF-κB signaling. IL-6 is a key metabolic modulator that is released by the skeletal muscle to coordinate a multi-systemic response (liver, muscle, and adipocytes) during physical exercise; the alteration of this response in dystrophic muscles may contribute to an abnormal response to contraction and exercise.


Assuntos
Interleucina-6/metabolismo , Potenciais da Membrana , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Estimulação Elétrica , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/fisiologia , NF-kappa B/genética
8.
PLoS One ; 8(12): e81222, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349043

RESUMO

Duchenne Muscular Dystrophy (DMD) is a recessive X-linked genetic disease, caused by mutations in the gene encoding dystrophin. DMD is characterized in humans and in mdx mice by a severe and progressive destruction of muscle fibers, inflammation, oxidative/nitrosative stress, and cell death. In mdx muscle fibers, we have shown that basal ATP release is increased and that extracellular ATP stimulation is pro-apoptotic. In normal fibers, depolarization-induced ATP release is blocked by nifedipine, leading us to study the potential therapeutic effect of nifedipine in mdx muscles and its relation with extracellular ATP signaling. Acute exposure to nifedipine (10 µM) decreased [Ca(2+)]r, NF-κB activity and iNOS expression in mdx myotubes. In addition, 6-week-old mdx mice were treated with daily intraperitoneal injections of nifedipine, 1 mg/Kg for 1 week. This treatment lowered the [Ca(2+)]r measured in vivo in the mdx vastus lateralis. We demonstrated that extracellular ATP levels were higher in adult mdx flexor digitorum brevis (FDB) fibers and can be significantly reduced after 1 week of treatment with nifedipine. Interestingly, acute treatment of mdx FDB fibers with apyrase, an enzyme that completely degrades extracellular ATP to AMP, reduced [Ca(2+)]r to a similar extent as was seen in FDB fibers after 1-week of nifedipine treatment. Moreover, we demonstrated that nifedipine treatment reduced mRNA levels of pro-oxidative/nitrosative (iNOS and gp91(phox)/p47(phox) NOX2 subunits) and pro-apoptotic (Bax) genes in mdx diaphragm muscles and lowered serum creatine kinase (CK) levels. In addition, nifedipine treatment increased muscle strength assessed by the inverted grip-hanging test and exercise tolerance measured with forced swimming test in mdx mice. We hypothesize that nifedipine reduces basal ATP release, thereby decreasing purinergic receptor activation, which in turn reduces [Ca(2+)]r in mdx skeletal muscle cells. The results in this work open new perspectives towards possible targets for pharmacological approaches to treat DMD.


Assuntos
Cálcio/metabolismo , Nifedipino/uso terapêutico , Animais , Células Cultivadas , Creatina Quinase/genética , Creatina Quinase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos mdx , Força Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo
9.
PLoS One ; 8(11): e75340, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24282497

RESUMO

ATP signaling has been shown to regulate gene expression in skeletal muscle and to be altered in models of muscular dystrophy. We have previously shown that in normal muscle fibers, ATP released through Pannexin1 (Panx1) channels after electrical stimulation plays a role in activating some signaling pathways related to gene expression. We searched for a possible role of ATP signaling in the dystrophy phenotype. We used muscle fibers from flexor digitorum brevis isolated from normal and mdx mice. We demonstrated that low frequency electrical stimulation has an anti-apoptotic effect in normal muscle fibers repressing the expression of Bax, Bim and PUMA. Addition of exogenous ATP to the medium has a similar effect. In dystrophic fibers, the basal levels of extracellular ATP were higher compared to normal fibers, but unlike control fibers, they do not present any ATP release after low frequency electrical stimulation, suggesting an uncoupling between electrical stimulation and ATP release in this condition. Elevated levels of Panx1 and decreased levels of Cav1.1 (dihydropyridine receptors) were found in triads fractions prepared from mdx muscles. Moreover, decreased immunoprecipitation of Cav1.1 and Panx1, suggest uncoupling of the signaling machinery. Importantly, in dystrophic fibers, exogenous ATP was pro-apoptotic, inducing the transcription of Bax, Bim and PUMA and increasing the levels of activated Bax and cytosolic cytochrome c. These evidence points to an involvement of the ATP pathway in the activation of mechanisms related with cell death in muscular dystrophy, opening new perspectives towards possible targets for pharmacological therapies.


Assuntos
Trifosfato de Adenosina/farmacologia , Apoptose/efeitos dos fármacos , Músculo Esquelético/patologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Conexinas/metabolismo , Estimulação Elétrica , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Distrofias Musculares/patologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteína X Associada a bcl-2/genética
10.
Curr Vasc Pharmacol ; 7(4): 450-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19485887

RESUMO

Diabetes mellitus is currently considered to be an epidemic disease. Approximately a third of individuals with type 1 and type 2 diabetes develop persistent albuminuria, lose renal function, and are at increased risk of cardiovascular and other microvascular complications. Diabetic nephropathy (DN) is the primary cause of end stage renal disease throughout the world. Microvascular dysfunction in the glomerulus appears as an early pathogenic event in progression of this renal complication. In recent years, studies with animal knockout (KO) models have revealed that uncoupling of the vascular endothelial growth factor/nitric oxide (VEGF/NO) axis leads to the glomerular alterations that characterize diabetic nephropathy. Therefore, new therapeutic alternatives may usefully target VEGF overproduction or endothelial nitric oxide availability. Recent studies also demonstrate a role for adenosine receptors in glomerular physiology and VEGF production that looks promising for therapeutic intervention of the evolution of diabetic nephropathy. However further progress is required in order to understand the dynamics of local adenosine production, in particular the extracellular metabolism of adenine nucleotides by ectoenzymes and the role of nucleoside transporters on external adenosine accumulation in the glomerulus in the pathological state. So far there is no assay that is sufficiently sensitive and accurate for subclinical diagnoses of this renal disease, which is complicated and costly to patients with often devastating effects. Current studies using proteomics offer promising alternatives for the identification of new renal injury markers. It is hoped these will permit evaluation of new therapeutic tools for more opportune intervention of this disease.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Receptores Purinérgicos P1/fisiologia , Adenosina/metabolismo , Animais , Nefropatias Diabéticas/fisiopatologia , Progressão da Doença , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Microvasos/fisiopatologia , Óxido Nítrico/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...