Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
bioRxiv ; 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38585797

ABSTRACT

Mitochondrial stress within the nervous system can trigger non-cell autonomous responses in peripheral tissues. However, the specific neurons involved and their impact on organismal aging and health have remained incompletely understood. Here, we demonstrate that mitochondrial stress in γ-aminobutyric acid-producing (GABAergic) neurons in Caenorhabditis elegans ( C. elegans ) is sufficient to significantly alter organismal lifespan, stress tolerance, and reproductive capabilities. This mitochondrial stress also leads to significant changes in mitochondrial mass, energy production, and levels of reactive oxygen species (ROS). DAF-16/FoxO activity is enhanced by GABAergic neuronal mitochondrial stress and mediates the induction of these non-cell-autonomous effects. Moreover, our findings indicate that GABA signaling operates within the same pathway as mitochondrial stress in GABAergic neurons, resulting in non-cell-autonomous alterations in organismal stress tolerance and longevity. In summary, these data suggest the crucial role of GABAergic neurons in detecting mitochondrial stress and orchestrating non-cell-autonomous changes throughout the organism.

2.
Chronic Illn ; : 17423953241241759, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38532693

ABSTRACT

OBJECTIVE: The clinical aspects of lung cancer patients are well-studied. However, healthcare charge patterns have yet to be explored through a large-scale representative population-based sample investigating differences by socioeconomic factors and comorbidities. AIM: To identify how comorbidities associated with healthcare charges among lung cancer patients. METHODS: We examined the characteristics of the patient sample and the association between comorbidity status (diabetes, hypertension, or both) and healthcare charge. Multivariate survey linear regression models were used to estimate the association. We also investigated sub-group association through various patient and socioeconomic factors. RESULTS: Of 212,745 lung cancer patients, 68.5% had diabetes and/or hypertension. Hospital charges were higher in the population with comorbidities. The results showed that lung cancer patients with comorbidities had 9.4%, 5.1%, and 12.0% (with diabetes, hypertension, and both, respectively) higher hospital charges than those without comorbidities. In sub-group analysis, Black patients also showed a similar trend across socioeconomic (i.e. household income and primary payer) and racial (i.e. White, Black, Hispanic, and Asian/Pacific Islander) factors. DISCUSSION: Black patients may be significantly financially burdened because of the prevalence of comorbidities and low-income status. More work is required to ensure healthcare equality and promote access to care for the uninsured, low-income, and minority populations because comorbidities common in these populations can create more significant financial barriers.

3.
Front Endocrinol (Lausanne) ; 14: 1222532, 2023.
Article in English | MEDLINE | ID: mdl-37583429

ABSTRACT

Background: Physical inactivity increases the risk for metabolic diseases such as obesity and type 2 diabetes. Neuromuscular electrical stimulation (NMES) is an effective method to induce muscle contraction, particularly for populations with physical impairments and/or metabolic diseases. However, its effectiveness to improve glycemic control is unclear. This review aimed to determine the effectiveness of NMES on glycemic control. Methods: Electronic search consisted of MEDLINE (PubMed), EMBASE, Cochrane Library, Google Scholar, and Web of Science to identify studies that investigated the effects of NMES on glycemic control for this systematic review. The meta-analysis consists of the studies designed as randomized controlled trials. Effect sizes were calculated as the standardized mean difference (SMD) and meta-analysis was conducted using a random-effects model. Results: Thirty-five studies met the inclusion criteria for systematic review and of those, nine qualified for the meta-analysis. Existing evidence suggested that NMES effectively improves glycemic control predominantly in middle-aged and elderly population with type 2 diabetes, obesity, and spinal cord injury. The meta-analysis is comprised of 180 participants and reported that NMES intervention lowered fasting blood glucose (SMD: 0.48; 95% CI: 0.17 to 0.78; p=0.002; I²=0%). Additional analysis using the primary measures reported by each study to indicate glycemic control (i.e., OGTT, HOMA-IR, and fasting glucose) also confirmed a significant effect of NMES on improving glycemic control (SMD: 0.41; 95% CI, 0.09 to 0.72; p=0.01; I²=11%). NMES protocol varied across studies and requires standardization. Conclusion: NMES could be considered as a therapeutic strategy to improve glycemic control in populations with physical impairments and/or metabolic disorders. Systematic review registration: https://www.crd.york.ac.uk/prospero/, identifier CRD42020192491.


Subject(s)
Diabetes Mellitus, Type 2 , Electric Stimulation Therapy , Aged , Humans , Middle Aged , Diabetes Mellitus, Type 2/therapy , Electric Stimulation , Electric Stimulation Therapy/methods , Health Services , Obesity
4.
Front Physiol ; 14: 1133423, 2023.
Article in English | MEDLINE | ID: mdl-36969584

ABSTRACT

Doxorubicin is a highly effective chemotherapeutic agent widely used to treat a variety of cancers. However, the clinical application of doxorubicin is limited due to its adverse effects on several tissues. One of the most serious side effects of doxorubicin is cardiotoxicity, which results in life-threatening heart damage, leading to reduced cancer treatment success and survival rate. Doxorubicin-induced cardiotoxicity results from cellular toxicity, including increased oxidative stress, apoptosis, and activated proteolytic systems. Exercise training has emerged as a non-pharmacological intervention to prevent cardiotoxicity during and after chemotherapy. Exercise training stimulates numerous physiological adaptations in the heart that promote cardioprotective effects against doxorubicin-induced cardiotoxicity. Understanding the mechanisms responsible for exercise-induced cardioprotection is important to develop therapeutic approaches for cancer patients and survivors. In this report, we review the cardiotoxic effects of doxorubicin and discuss the current understanding of exercise-induced cardioprotection in hearts from doxorubicin-treated animals.

5.
Nutrients ; 14(12)2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35745205

ABSTRACT

The mechanisms connecting obesity with type 2 diabetes, insulin resistance, nonalcoholic fatty liver disease, and cardiovascular diseases remain incompletely understood. The function of MAPK phosphatase-2 (MKP-2), a type 1 dual-specific phosphatase (DUSP) in whole-body metabolism, and how this contributes to the development of diet-induced obesity, type 2 diabetes (T2D), and insulin resistance is largely unknown. We investigated the physiological contribution of MKP-2 in whole-body metabolism and whether MKP-2 is altered in obesity and human fatty liver disease using MKP-2 knockout mice models and human liver tissue derived from fatty liver disease patients. We demonstrate that, for the first time, MKP-2 expression was upregulated in liver tissue in humans with obesity and fatty liver disease and in insulin-responsive tissues in mice with obesity. MKP-2-deficient mice have enhanced p38 MAPK, JNK, and ERK activities in insulin-responsive tissues compared with wild-type mice. MKP-2 deficiency in mice protects against diet-induced obesity and hepatic steatosis and was accompanied by improved glucose homeostasis and insulin sensitivity. Mkp-2-/- mice are resistant to diet-induced obesity owing to reduced food intake and associated lower respiratory exchange ratio. This was associated with enhanced circulating insulin-like growth factor-1 (IGF-1) and stromal cell-derived factor 1 (SDF-1) levels in Mkp-2-/- mice. PTEN, a negative regulator of Akt, was downregulated in livers of Mkp-2-/- mice, resulting in enhanced Akt activity consistent with increased insulin sensitivity. These studies identify a novel role for MKP-2 in the regulation of systemic metabolism and pathophysiology of obesity-induced insulin resistance and fatty liver disease.


Subject(s)
Diabetes Mellitus, Type 2 , Fatty Liver , Insulin Resistance , Animals , Diabetes Mellitus, Type 2/metabolism , Dual Specificity Phosphatase 1/metabolism , Dual-Specificity Phosphatases , Fatty Liver/metabolism , Humans , Insulin/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Phosphatases , Obesity/metabolism , Protein Tyrosine Phosphatases , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation
6.
Membranes (Basel) ; 12(4)2022 Apr 09.
Article in English | MEDLINE | ID: mdl-35448380

ABSTRACT

Obesity has reached global epidemic proportions and it affects the development of insulin resistance, type 2 diabetes, fatty liver disease and other metabolic diseases. Membrane lipids are important structural and signaling components of the cell membrane. Recent studies highlight their importance in lipid homeostasis and are implicated in the pathogenesis of fatty liver disease. Here, we discuss the numerous membrane lipid species and their metabolites including, phospholipids, sphingolipids and cholesterol, and how dysregulation of their composition and physiology contribute to the development of fatty liver disease. The development of new genetic and pharmacological mouse models has shed light on the role of lipid species on various mechanisms/pathways; these lipids impact many aspects of the pathophysiology of fatty liver disease and could potentially be targeted for the treatment of fatty liver disease.

7.
Nutrients ; 13(11)2021 Nov 04.
Article in English | MEDLINE | ID: mdl-34836195

ABSTRACT

The liver plays a key role in whole-body, glucose and lipid homeostasis. Nutritional signals in response to fasting and refeeding regulate hepatic lipid synthesis. It is established that activation of mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP-1) in response to overnutrition regulates MAPK-dependent pathways that control lipid metabolism in the liver. However, the regulatory mechanisms and the impact of the actions of MKP-1 in hepatic response to fasting remains unclear. We investigated the effect of fasting on the expression of MKP-1 and the impact on hepatic response to feeding. In this study, we demonstrate that fasting stress induced upregulation of hepatic MKP-1 protein levels with a corresponding downregulation of p38 MAPK and JNK phosphorylation in mouse livers. We found that MKP-1-deficient livers are resistant to fasting-induced hepatic steatosis. Hepatic MKP-1 deficiency impaired fasting-induced changes in the levels of key transcription factors involved in the regulation of fatty acid and cholesterol metabolism including Srebf2 and Srebf1c. Mechanistically, MKP-1 negatively regulates Srebf2 expression by attenuating p38 MAPK pathway, suggesting its contribution to the metabolic effects of MKP-1 deficiency in the fasting liver. These findings support the hypothesis that upregulation of MKP-1 is a physiological relevant response and might be beneficial in hepatic lipid utilization during fasting in the liver. Collectively, these data unravel some of the complexity and tissue specific interaction of MKP-1 action in response to changes in nutritional cues, including fasting and excess nutrients.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Eating/physiology , Fasting/metabolism , Liver/metabolism , Up-Regulation/physiology , Animal Nutritional Physiological Phenomena , Animals , Fasting/adverse effects , Fatty Liver/etiology , Fatty Liver/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipid Metabolism/physiology , Lipogenesis/physiology , Mice , Models, Animal , Phosphorylation/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Cell Signal ; 2(3): 172-180, 2021.
Article in English | MEDLINE | ID: mdl-34557866

ABSTRACT

The western diet and overuse of anti-inflammatory medication have caused a great deal of stress on the liver. Obesity and the associated inflammatory state in insulin-responsive tissues result in the release of pro-inflammatory cytokine that activates the stress-responsive MAPKs, p38 MAPK, and JNK. These MAPKs have figured prominently as critical effectors in physiological and pathophysiological hepatic inflammation. In contrast, evidence for a role for ERK1/2 in hepatic inflammation has been less well developed. In this review article, we describe recent insights into the physiology and pathophysiology of the role of stress-responsive MAPKs in hepatic inflammation during obesity and liver injury with a focus on macrophages, hepatocytes and hepatic stellate cells. In response to metabolic stress and liver injury, JNK activation in macrophages and hepatocytes promotes the secretion of inflammatory cytokines and macrophage and neutrophil infiltration. p38 MAPK plays an important role in contributing to the progression of hepatic inflammation in response to various hepatic cellular stresses, although the precise substrates mediating these effects in hepatocytes and hepatic stellate cells remain to be identified. Both JNK and p38 MAPK promotes profibrotic behavior in hepatic stellate cells.

9.
Article in English | MEDLINE | ID: mdl-33669288

ABSTRACT

Although low socioeconomic status (SES) and decreased muscle strength have been found to be associated with the risk factors of non-alcoholic fatty liver disease (NAFLD), including insulin resistance, obesity, and metabolic syndrome, the associations among SES, muscle strength, and NAFLD are still unclear. We aimed to investigate the combined effect of SES and relative handgrip strength (HGS) on the risk of NAFLD in middle-aged adults. Data from 5272 middle-aged adults who participated in the Korea National Health and Nutrition Examination Surveys (KNHANES) from 2014-2018 were analyzed. NAFLD was defined using the hepatic steatosis index (HSI) > 36 and the comprehensive NAFLD score (CNS) ≥ 40 in the absence of other causes of liver disease. SES was based on a self-reported questionnaire. Overall, individuals with low SES (odds ratio (OR) = 1.703, 95% confidence interval (CI): 1.424-2.037, p < 0.001) or low HGS (OR = 12.161, 95% CI: 9.548-15.488, p < 0.001) had a significantly higher risk of NAFLD. The joint association analysis showed that a low SES combined with a low HGS (OR = 2.479, 95% CI: 1.351-4.549, p = 0.003) further significantly increased the risk of NAFLD when adjusted for all the covariates, compared with individuals with a high SES and a high HGS (OR = 1). The current findings suggest that both low SES and low HGS were independently and synergistically associated with an increased risk of NAFLD in middle-aged Korean adults.


Subject(s)
Non-alcoholic Fatty Liver Disease , Adult , Hand Strength , Humans , Middle Aged , Non-alcoholic Fatty Liver Disease/epidemiology , Nutrition Surveys , Republic of Korea/epidemiology , Risk Factors , Social Class
10.
Front Immunol ; 12: 790511, 2021.
Article in English | MEDLINE | ID: mdl-34992607

ABSTRACT

Cardiac fibrosis, a pathological condition due to excessive extracellular matrix (ECM) deposition in the myocardium, is associated with nearly all forms of heart disease. The processes and mechanisms that regulate cardiac fibrosis are not fully understood. In response to cardiac injury, macrophages undergo marked phenotypic and functional changes and act as crucial regulators of myocardial fibrotic remodeling. Here we show that the mitogen-activated protein kinase (MAPK) phosphatase-5 (MKP-5) in macrophages is involved in pressure overload-induced cardiac fibrosis. Cardiac pressure overload resulting from transverse aortic constriction (TAC) leads to the upregulation of Mkp-5 gene expression in the heart. In mice lacking MKP-5, p38 MAPK and JNK were hyperactivated in the heart, and TAC-induced cardiac hypertrophy and myocardial fibrosis were attenuated. MKP-5 deficiency upregulated the expression of the ECM-degrading matrix metalloproteinase-9 (Mmp-9) in the Ly6Clow (M2-type) cardiac macrophage subset. Consistent with in vivo findings, MKP-5 deficiency promoted MMP-9 expression and activity of pro-fibrotic macrophages in response to IL-4 stimulation. Furthermore, using pharmacological inhibitors against p38 MAPK, JNK, and ERK, we demonstrated that MKP-5 suppresses MMP-9 expression through a combined effect of p38 MAPK/JNK/ERK, which subsequently contributes to the inhibition of ECM-degrading activity. Taken together, our study indicates that pressure overload induces MKP-5 expression and facilitates cardiac hypertrophy and fibrosis. MKP-5 deficiency attenuates cardiac fibrosis through MAPK-mediated regulation of MMP-9 expression in Ly6Clow cardiac macrophages.


Subject(s)
Cardiomegaly/immunology , Dual-Specificity Phosphatases/deficiency , Heart Failure/immunology , MAP Kinase Signaling System/immunology , Myocardium/pathology , Animals , Blood Pressure , Cardiomegaly/diagnosis , Cardiomegaly/pathology , Cells, Cultured , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Echocardiography , Fibrosis , Heart/diagnostic imaging , Heart Failure/pathology , Humans , Interleukin-4/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Phosphorylation/immunology , Primary Cell Culture , Ventricular Remodeling/immunology
11.
Sci Signal ; 13(646)2020 08 25.
Article in English | MEDLINE | ID: mdl-32843541

ABSTRACT

The mitogen-activated protein kinase (MAPK) phosphatases (MKPs) have been considered "undruggable," but their position as regulators of the MAPKs makes them promising therapeutic targets. MKP5 has been suggested as a potential target for the treatment of dystrophic muscle disease. Here, we identified an inhibitor of MKP5 using a p38α MAPK-derived, phosphopeptide-based small-molecule screen. We solved the structure of MKP5 in complex with this inhibitor, which revealed a previously undescribed allosteric binding pocket. Binding of the inhibitor to this pocket collapsed the MKP5 active site and was predicted to limit MAPK binding. Treatment with the inhibitor recapitulated the phenotype of MKP5 deficiency, resulting in activation of p38 MAPK and JNK. We demonstrated that MKP5 was required for TGF-ß1 signaling in muscle and that the inhibitor blocked TGF-ß1-mediated Smad2 phosphorylation. TGF-ß1 pathway antagonism has been proposed for the treatment of dystrophic muscle disease. Thus, allosteric inhibition of MKP5 represents a therapeutic strategy against dystrophic muscle disease.


Subject(s)
Dual-Specificity Phosphatases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Mitogen-Activated Protein Kinase Phosphatases/antagonists & inhibitors , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Allosteric Site/genetics , Amino Acid Sequence , Animals , Cell Differentiation/drug effects , Cell Line , Dual-Specificity Phosphatases/chemistry , Dual-Specificity Phosphatases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Female , Humans , Kinetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Phosphatases/chemistry , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Protein Binding/drug effects , Sequence Homology, Amino Acid , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
12.
Med Hypotheses ; 134: 109423, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31627121

ABSTRACT

Exercise intolerance is a hallmark of symptoms in patients with heart failure. In addition to reduced cardiac output, a series of impairments in pulmonary and vascular systems leads to decreases in oxygen delivery and availability in locomotor muscles. This contributes to exercise intolerance in heart failure. The oxy-hemoglobin dissociation curve is essentially a graph illustrating the relationship between the partial pressure of oxygen (PO2, X-axis) and oxygen saturation (SaO2, Y-axis) of hemoglobin. The rightward shift of the curve indicates that hemoglobin's affinity for oxygen decreases and in turn, it may allow the release of more oxygen to tissues. In the present study, we discuss the pathophysiological impairment, which causes exercise intolerance in heart failure patients and suggest a strategy to improve exercise capacity without altering cardiac output via modulating the oxy-hemoglobin dissociation curve.


Subject(s)
Exercise Tolerance , Heart Failure/physiopathology , Hemoglobins/metabolism , Oxygen/blood , Oxyhemoglobins/metabolism , Allosteric Regulation , Cardiac Output , Female , Heart Failure/blood , Humans , Hypoxia/etiology , Hypoxia/physiopathology , Male , Models, Cardiovascular , Muscle, Skeletal/metabolism , Ventricular Dysfunction, Left/physiopathology
13.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R227-R233, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31774307

ABSTRACT

Doxorubicin (DOX) is a highly effective antineoplastic agent used in cancer treatment. Unfortunately, clinical use of DOX is limited due to the development of dose-dependent toxicity to cardiac and respiratory (i.e., diaphragm) muscles. After administration, DOX preferentially localizes to the inner mitochondrial membrane, where it promotes cellular toxicity via enhanced mitochondrial reactive oxygen species (ROS) production. Although recent evidence suggests that amelioration of mitochondrial ROS emission preserves cardiorespiratory muscle function following DOX treatment, the mechanisms responsible for this protection remain unknown. Therefore, we tested the hypothesis that DOX-induced mitochondrial ROS production is required to stimulate pathological signaling by the autophagy/lysosomal system (ALS), the ubiquitin-proteasome pathway (UPP), and the unfolded protein response (UPR). Cause and effect were determined by administration of the mitochondria-targeted peptide SS-31 to DOX-treated animals. Interestingly, while SS-31 abrogated aberrant ROS emission in cardiorespiratory muscles of DOX-treated animals, our results revealed muscle-specific regulation of effector pathways. In the heart, SS-31 prevented DOX-induced proteolytic signaling through the ALS and UPP. In contrast, ALS signaling was inhibited by SS-31 in the diaphragm, but the UPP was not affected. UPR signaling was activated in both muscles at eukaryotic translation initiation factor 2α (eIF2α) S51 in the heart and diaphragm of DOX-treated animals and was attenuated with SS-31 treatment in both tissues. However, downstream signaling of eIF2α (activating transcription factor 4 and CCAAT/enhancer-binding protein homologous protein) was diminished in the heart but upregulated in the diaphragm with DOX. Collectively, these results show that DOX-induced ROS production plays distinct roles in the regulation of cardiac and diaphragm muscle proteolysis.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Diaphragm/drug effects , Doxorubicin/toxicity , Heart Diseases/chemically induced , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Proteolysis/drug effects , Activating Transcription Factor 4/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cardiotoxicity , Diaphragm/metabolism , Eukaryotic Initiation Factor-2/metabolism , Female , Heart Diseases/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Myocytes, Cardiac/metabolism , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction , Unfolded Protein Response/drug effects
14.
Am J Physiol Lung Cell Mol Physiol ; 317(5): L678-L689, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31483681

ABSTRACT

Mitogen-activated protein kinase (MAPK) phosphatase 5 (MKP-5) is a member of the dual-specificity family of protein tyrosine phosphatases that negatively regulates p38 MAPK and the JNK. MKP-5-deficient mice exhibit improved muscle repair and reduced fibrosis in an animal model of muscular dystrophy. Here, we asked whether the effects of MKP-5 on muscle fibrosis extend to other tissues. Using a bleomycin-induced model of pulmonary fibrosis, we found that MKP-5-deficient mice were protected from the development of lung fibrosis, expressed reduced levels of hydroxyproline and fibrogenic genes, and displayed marked polarization towards an M1-macrophage phenotype. We showed that the profibrogenic effects of the transforming growth factor-ß1 (TGF-ß1) were inhibited in MKP-5-deficient lung fibroblasts. MKP-5-deficient fibroblasts exhibited enhanced p38 MAPK activity, impaired Smad3 phosphorylation, increased Smad7 levels, and decreased expression of fibrogenic genes. Myofibroblast differentiation was attenuated in MKP-5-deficient fibroblasts. Finally, we found that MKP-5 expression was increased in idiopathic pulmonary fibrosis (IPF)-derived lung fibroblasts but not in whole IPF lungs. These data suggest that MKP-5 plays an essential role in promoting lung fibrosis. Our results couple MKP-5 with the TGF-ß1 signaling machinery and imply that MKP-5 inhibition may serve as a therapeutic target for human lung fibrosis.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/physiology , Fibroblasts/pathology , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Pulmonary Fibrosis/pathology , Transforming Growth Factor beta1/pharmacology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Dual-Specificity Phosphatases/genetics , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phosphorylation , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Signal Transduction
15.
J Cachexia Sarcopenia Muscle ; 10(4): 767-781, 2019 08.
Article in English | MEDLINE | ID: mdl-30972953

ABSTRACT

BACKGROUND: Mechanical ventilation (MV) is a life-saving measure for patients in respiratory failure. However, prolonged MV results in significant diaphragm atrophy and contractile dysfunction, a condition referred to as ventilator-induced diaphragm dysfunction (VIDD). While there are currently no clinically approved countermeasures to prevent VIDD, increased expression of heat shock protein 72 (HSP72) has been demonstrated to attenuate inactivity-induced muscle wasting. HSP72 elicits cytoprotection via inhibition of NF-κB and FoxO transcriptional activity, which contribute to VIDD. In addition, exercise-induced prevention of VIDD is characterized by an increase in the concentration of HSP72 in the diaphragm. Therefore, we tested the hypothesis that increased HSP72 expression is required for the exercise-induced prevention of VIDD. We also determined whether increasing the abundance of HSP72 in the diaphragm, independent of exercise, is sufficient to prevent VIDD. METHODS: Cause and effect was determined by inhibiting the endurance exercise-induced increase in HSP72 in the diaphragm of exercise trained animals exposed to prolonged MV via administration of an antisense oligonucleotide targeting HSP72. Additional experiments were performed to determine if increasing HSP72 in the diaphragm via genetic (rAAV-HSP72) or pharmacological (BGP-15) overexpression is sufficient to prevent VIDD. RESULTS: Our results demonstrate that the exercise-induced increase in HSP72 protein abundance is required for the protective effects of exercise against VIDD. Moreover, both rAAV-HSP72 and BGP-15-induced overexpression of HSP72 were sufficient to prevent VIDD. In addition, modification of HSP72 in the diaphragm is inversely related to the expression of NF-κB and FoxO target genes. CONCLUSIONS: HSP72 overexpression in the diaphragm is an effective intervention to prevent MV-induced oxidative stress and the transcriptional activity of NF-κB and FoxO. Therefore, overexpression of HSP72 in the diaphragm is a potential therapeutic target to protect against VIDD.


Subject(s)
Exercise/physiology , HSP72 Heat-Shock Proteins/metabolism , Respiration, Artificial/methods , Animals , Diaphragm/physiopathology , Female , Humans , Rats
16.
Diabetes ; 67(4): 624-635, 2018 04.
Article in English | MEDLINE | ID: mdl-29317435

ABSTRACT

Stress responses promote obesity and insulin resistance, in part, by activating the stress-responsive mitogen-activated protein kinases (MAPKs), p38 MAPK, and c-Jun NH2-terminal kinase (JNK). Stress also induces expression of MAPK phosphatase-1 (MKP-1), which inactivates both JNK and p38 MAPK. However, the equilibrium between JNK/p38 MAPK and MKP-1 signaling in the development of obesity and insulin resistance is unclear. Skeletal muscle is a major tissue involved in energy expenditure and glucose metabolism. In skeletal muscle, MKP-1 is upregulated in high-fat diet-fed mice and in skeletal muscle of obese humans. Mice lacking skeletal muscle expression of MKP-1 (MKP1-MKO) showed increased skeletal muscle p38 MAPK and JNK activities and were resistant to the development of diet-induced obesity. MKP1-MKO mice exhibited increased whole-body energy expenditure that was associated with elevated levels of myofiber-associated mitochondrial oxygen consumption. miR-21, a negative regulator of PTEN expression, was upregulated in skeletal muscle of MKP1-MKO mice, resulting in increased Akt activity consistent with enhanced insulin sensitivity. Our results demonstrate that skeletal muscle MKP-1 represents a critical signaling node through which inactivation of the p38 MAPK/JNK module promotes obesity and insulin resistance.


Subject(s)
Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Insulin Resistance , MAP Kinase Kinase 4/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Diet, High-Fat , Energy Metabolism , Humans , Mice , Mice, Knockout , MicroRNAs/metabolism , Mitochondria, Muscle/metabolism , Oxygen Consumption , Signal Transduction
17.
Free Radic Biol Med ; 115: 179-190, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29197632

ABSTRACT

Mechanical ventilation (MV) results in the rapid development of ventilator-induced diaphragm dysfunction (VIDD). While the mechanisms responsible for VIDD are not fully understood, recent data reveal that prolonged MV activates autophagy in the diaphragm, which may occur as a result of increased cellular reactive oxygen species (ROS) production. Therefore, we tested the hypothesis that (1) accelerated autophagy is a key contributor to VIDD; and that (2) oxidative stress is required to increase the expression of autophagy genes in the diaphragm. Our findings reveal that targeted inhibition of autophagy in the rat diaphragm prevented MV-induced muscle atrophy and contractile dysfunction. Attenuation of VIDD in these animals occurred as a result of increased diaphragm concentration of the antioxidant catalase and reduced mitochondrial ROS emission, which corresponded to reductions in the activity of calpain and caspase-3. To determine if increased ROS production is required for the upregulation of autophagy biomarkers in the diaphragm, rats that were administered the mitochondrial-targeted peptide SS-31 during MV. Results from this study demonstrated that mitochondrial ROS production in the diaphragm during MV is required for the increased expression of key autophagy genes (i.e. LC3, Atg7, Atg12, Beclin1 and p62), as well as for increased activity of cathepsin L. Together, these data reveal that autophagy is required for VIDD, and that autophagy inhibition reduces MV-induced diaphragm ROS production and prevents a positive feedback loop whereby increased autophagy is stimulated by oxidative stress, resulting in further increases in ROS and autophagy.


Subject(s)
Diaphragm/physiology , Mitochondria/metabolism , Muscular Atrophy/metabolism , Respiration, Artificial/adverse effects , Animals , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Cells, Cultured , Disease Models, Animal , Female , Humans , Muscle Contraction , Muscular Atrophy/etiology , Oxidative Stress/genetics , Proteolysis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
18.
Skelet Muscle ; 7(1): 21, 2017 10 18.
Article in English | MEDLINE | ID: mdl-29047406

ABSTRACT

BACKGROUND: The mitogen-activated protein kinases (MAPKs) have been shown to be involved in regulating myofiber survival. In skeletal muscle, p38 MAPK and JNK are negatively regulated by MAPK phosphatase-5 (MKP-5). During muscle regeneration, MKP-5 is downregulated, thereby promoting p38 MAPK/JNK signaling, and subsequent repair of damaged muscle. Mice lacking MKP-5 expression exhibit enhanced regenerative myogenesis. However, the effect of MKP-5 on myofiber survival during regeneration is unclear. METHODS: To investigate whether MKP-5 is involved in myofiber survival, skeletal muscle injury was induced by cardiotoxin injection, and the effects on apoptosis were assessed by TUNEL assay in wild type and MKP-5-deficient mice. The contribution of MKP-5 to apoptotic signaling and its link to this pathway through mitochondrial function were determined in regenerating skeletal muscle of MKP-5-deficient mice. RESULTS: We found that loss of MKP-5 in skeletal muscle resulted in improved myofiber survival. In response to skeletal muscle injury, loss of MKP-5 decreased activation of the mitochondrial apoptotic pathway involving the signal transducer and activator of transcription 3 (STAT3) and increased expression of the anti-apoptotic transcription factor Bcl-2. Skeletal muscle of MKP-5-deficient mice also exhibited an improved anti-oxidant capacity as a result of increased expression of catalase further contributing to myofiber survival by attenuating oxidative damage. CONCLUSIONS: Taken together, these findings suggest that MKP-5 coordinates skeletal muscle regeneration by regulating mitochondria-mediated apoptosis. MKP-5 negatively regulates apoptotic signaling, and during regeneration, MKP-5 downregulation contributes to the restoration of myofiber survival. Finally, these results suggest that MKP-5 inhibition may serve as an important therapeutic target for the preservation of skeletal muscle survival in degenerative muscle diseases.


Subject(s)
Apoptosis , Dual-Specificity Phosphatases/genetics , Muscle Development , Muscle Fibers, Skeletal/metabolism , Signal Transduction , Animals , Cell Line , Dual-Specificity Phosphatases/metabolism , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Regeneration , STAT3 Transcription Factor/metabolism
19.
J Biol Chem ; 292(9): 3581-3590, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28096466

ABSTRACT

The mitogen-activated protein kinases (MAPKs) have been shown to regulate skeletal muscle function. Previously, we showed that MAPK phosphatase-5 (MKP-5) negatively regulates myogenesis and regeneration of skeletal muscle through inhibition of p38 MAPK and c-Jun N-terminal kinase (JNK). However, the identity and contribution of MKP-5-regulated MAPK targets in the control of skeletal muscle function and regenerative myogenesis have not been established. To identify MKP-5-regulated MAPK substrates in skeletal muscle, we performed a global differential phospho-MAPK substrate screen in regenerating skeletal muscles of wild type and MKP-5-deficient mice. We discovered a novel MKP-5-regulated MAPK substrate called guanine nucleotide exchange factor for Rab3A (GRAB) that was hyperphosphorylated on a phospho-MAPK motif in skeletal muscle of MKP-5-deficient mice. GRAB was found to be phosphorylated by JNK on serine 169. Myoblasts overexpressing a phosphorylation-defective mutant of GRAB containing a mutation at Ser-169 to Ala-169 (GRAB-S169A) inhibited the ability of C2C12 myoblasts to differentiate. We found that GRAB phosphorylation at Ser-169 was required for the secretion of the promyogenic cytokine interleukin 6 (IL-6). Consistent with this observation, MKP-5-deficient mice exhibited increased circulating IL-6 expression as compared with wild type mice. Collectively, these data demonstrate a novel mechanism whereby MKP-5-mediated regulation of JNK negatively regulates phosphorylation of GRAB, which subsequently controls secretion of IL-6. These data support the notion that MKP-5 serves as a negative regulator of MAPK-dependent signaling of critical skeletal muscle signaling pathways.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Gene Expression Regulation, Enzymologic , Guanine Nucleotide Exchange Factors/metabolism , Interleukin-6/metabolism , Muscle Development , rab3A GTP-Binding Protein/metabolism , Amino Acid Motifs , Animals , Cell Movement , Cell Proliferation , MAP Kinase Signaling System , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Mutation , Myoblasts/metabolism , Phosphorylation , Proteomics , Regeneration , Serine/chemistry
20.
Crit Care Med ; 43(5): e133-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25746508

ABSTRACT

OBJECTIVES: Mechanical ventilation is a lifesaving measure for patients with respiratory failure. However, prolonged mechanical ventilation results in diaphragm weakness, which contributes to problems in weaning from the ventilator. Therefore, identifying the signaling pathways responsible for mechanical ventilation-induced diaphragm weakness is essential to developing effective countermeasures to combat this important problem. In this regard, the forkhead boxO family of transcription factors is activated in the diaphragm during mechanical ventilation, and forkhead boxO-specific transcription can lead to enhanced proteolysis and muscle protein breakdown. Currently, the role that forkhead boxO activation plays in the development of mechanical ventilation-induced diaphragm weakness remains unknown. DESIGN: This study tested the hypothesis that mechanical ventilation-induced increases in forkhead boxO signaling contribute to ventilator-induced diaphragm weakness. SETTING: University research laboratory. SUBJECTS: Young adult female Sprague-Dawley rats. INTERVENTIONS: Cause and effect was determined by inhibiting the activation of forkhead boxO in the rat diaphragm through the use of a dominant-negative forkhead boxO adeno-associated virus vector delivered directly to the diaphragm. MEASUREMENTS AND MAIN RESULTS: Our results demonstrate that prolonged (12 hr) mechanical ventilation results in a significant decrease in both diaphragm muscle fiber size and diaphragm-specific force production. However, mechanically ventilated animals treated with dominant-negative forkhead boxO showed a significant attenuation of both diaphragm atrophy and contractile dysfunction. In addition, inhibiting forkhead boxO transcription attenuated the mechanical ventilation-induced activation of the ubiquitin-proteasome system, the autophagy/lysosomal system, and caspase-3. CONCLUSIONS: Forkhead boxO is necessary for the activation of key proteolytic systems essential for mechanical ventilation-induced diaphragm atrophy and contractile dysfunction. Collectively, these results suggest that targeting forkhead boxO transcription could be a key therapeutic target to combat ventilator-induced diaphragm dysfunction.


Subject(s)
Diaphragm/physiopathology , Forkhead Transcription Factors/antagonists & inhibitors , Respiration, Artificial/adverse effects , Animals , Diaphragm/pathology , Female , Hemodynamics , Muscle Contraction , Muscular Atrophy , Nerve Tissue Proteins , Rats , Rats, Sprague-Dawley , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...