Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
3.
Aging Cell ; 21(9): e13674, 2022 09.
Article in English | MEDLINE | ID: mdl-35934931

ABSTRACT

Mitochondrial dysfunction has been associated with age-related diseases, including idiopathic pulmonary fibrosis (IPF). We provide evidence that implicates chronic elevation of the mitochondrial anion carrier protein, uncoupling protein-2 (UCP2), in increased generation of reactive oxygen species, altered redox state and cellular bioenergetics, impaired fatty acid oxidation, and induction of myofibroblast senescence. This pro-oxidant senescence reprogramming occurs in concert with conventional actions of UCP2 as an uncoupler of oxidative phosphorylation with dissipation of the mitochondrial membrane potential. UCP2 is highly expressed in human IPF lung myofibroblasts and in aged fibroblasts. In an aging murine model of lung fibrosis, the in vivo silencing of UCP2 induces fibrosis regression. These studies indicate a pro-fibrotic function of UCP2 in chronic lung disease and support its therapeutic targeting in age-related diseases associated with impaired tissue regeneration and organ fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Myofibroblasts , Uncoupling Protein 2 , Aged , Animals , Fibroblasts/metabolism , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Mice , Myofibroblasts/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism
4.
Nat Aging ; 1(2): 205-217, 2021 02.
Article in English | MEDLINE | ID: mdl-34386777

ABSTRACT

Aging is a risk factor for progressive fibrotic disorders involving diverse organ systems, including the lung. Idiopathic pulmonary fibrosis, an age-associated degenerative lung disorder, is characterized by persistence of apoptosis-resistant myofibroblasts. In this report, we demonstrate that sirtuin-3 (SIRT3), a mitochondrial deacetylase, is downregulated in lungs of IPF human subjects and in mice subjected to lung injury. Over-expression of the SIRT3 cDNA via airway delivery restored capacity for fibrosis resolution in aged mice, in association with activation of the forkhead box transcription factor, FoxO3a, in fibroblasts, upregulation of pro-apoptotic members of the Bcl-2 family, and recovery of apoptosis susceptibility. While transforming growth factor-ß1 reduced levels of SIRT3 and FoxO3a in lung fibroblasts, cell non-autonomous effects involving macrophage secreted products were necessary for SIRT3-mediated activation of FoxO3a. Together, these findings reveal a novel role of SIRT3 in pro-resolution macrophage functions that restore susceptibility to apoptosis in fibroblasts via a FoxO3a-dependent mechanism.


Subject(s)
Idiopathic Pulmonary Fibrosis , Sirtuin 3 , Humans , Animals , Mice , Sirtuin 3/genetics , Lung/metabolism , Fibrosis , Idiopathic Pulmonary Fibrosis/metabolism , Gene Expression
5.
Sci Signal ; 13(644)2020 08 11.
Article in English | MEDLINE | ID: mdl-32788339

ABSTRACT

The oxidation of tyrosine residues to generate o,o'-dityrosine cross-links in extracellular proteins is necessary for the proper function of the extracellular matrix (ECM) in various contexts in invertebrates. Tyrosine oxidation is also required for the biosynthesis of thyroid hormone in vertebrates, and there is evidence for oxidative cross-linking reactions occurring in extracellular proteins secreted by myofibroblasts. The ECM protein fibronectin circulates in the blood as a globular protein that dimerizes through disulfide bridges generated by cysteine oxidation. We found that cellular (fibrillar) fibronectin on the surface of transforming growth factor-ß1 (TGF-ß1)-activated human myofibroblasts underwent multimerization by o,o'-dityrosine cross-linking under reducing conditions that disrupt disulfide bridges, but soluble fibronectin did not. This reaction on tyrosine residues required both the TGF-ß1-dependent production of hydrogen peroxide and the presence of myeloperoxidase (MPO) derived from inflammatory cells, which are active participants in wound healing and fibrogenic processes. Oxidative cross-linking of matrix fibronectin attenuated both epithelial and fibroblast migration and conferred resistance to proteolysis by multiple proteases. The abundance of circulating o,o'-dityrosine-modified fibronectin was increased in a murine model of lung fibrosis and in human subjects with interstitial lung disease compared to that in control healthy subjects. These studies indicate that tyrosine can undergo stable, covalent linkages in fibrillar fibronectin under inflammatory conditions and that this modification affects the migratory behavior of cells on such modified matrices, suggesting that this modification may play a role in both physiologic and pathophysiologic tissue repair.


Subject(s)
Cell Movement/physiology , Fibronectins/metabolism , Myofibroblasts/metabolism , Oxidative Stress/physiology , Peptide Hydrolases/metabolism , A549 Cells , Animals , Cell Line , Cells, Cultured , Cross-Linking Reagents/chemistry , Extracellular Matrix/metabolism , Female , Fibronectins/chemistry , Humans , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/cytology , Neutrophils/cytology , Neutrophils/metabolism , Oxidation-Reduction , Peroxidase/genetics , Peroxidase/metabolism , Transforming Growth Factor beta1/metabolism , Tyrosine/analogs & derivatives , Tyrosine/chemistry , Tyrosine/metabolism
7.
Nat Med ; 24(10): 1627, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30104770

ABSTRACT

In the version of this article originally published, a grant was omitted from the Acknowledgements section. The following sentence should have been included: "R.B.M. was supported by a Department of Veterans Affairs Merit Award (5I01BX003272)." The error has been corrected in the HTML and PDF versions of this article.

8.
Nat Med ; 24(8): 1121-1127, 2018 08.
Article in English | MEDLINE | ID: mdl-29967351

ABSTRACT

Fibrosis is a pathological result of a dysfunctional repair response to tissue injury and occurs in a number of organs, including the lungs1. Cellular metabolism regulates tissue repair and remodelling responses to injury2-4. AMPK is a critical sensor of cellular bioenergetics and controls the switch from anabolic to catabolic metabolism5. However, the role of AMPK in fibrosis is not well understood. Here, we demonstrate that in humans with idiopathic pulmonary fibrosis (IPF) and in an experimental mouse model of lung fibrosis, AMPK activity is lower in fibrotic regions associated with metabolically active and apoptosis-resistant myofibroblasts. Pharmacological activation of AMPK in myofibroblasts from lungs of humans with IPF display lower fibrotic activity, along with enhanced mitochondrial biogenesis and normalization of sensitivity to apoptosis. In a bleomycin model of lung fibrosis in mice, metformin therapeutically accelerates the resolution of well-established fibrosis in an AMPK-dependent manner. These studies implicate deficient AMPK activation in non-resolving, pathologic fibrotic processes, and support a role for metformin (or other AMPK activators) to reverse established fibrosis by facilitating deactivation and apoptosis of myofibroblasts.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Lung/pathology , Metformin/therapeutic use , Adenylate Kinase/metabolism , Animals , Bleomycin , Disease Models, Animal , Enzyme Activation/drug effects , Extracellular Matrix Proteins/metabolism , Humans , Male , Metformin/pharmacology , Mice, Inbred C57BL , Mitochondria/metabolism , Myofibroblasts/drug effects , Myofibroblasts/pathology
10.
Ann Am Thorac Soc ; 14(Supplement_5): S383-S388, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29161077

ABSTRACT

The aging of the human population has resulted in an unprecedented increase in the incidence and prevalence of age-related diseases, including those of the lung. Idiopathic pulmonary fibrosis is a disease of aging, and is characterized by a progressive decline in lung function and high mortality. Recent studies suggest that mitochondrial dysfunction, which can accompany aging phenotypes, may contribute to the pathogenesis of idiopathic pulmonary fibrosis. In this review, we explore current evidence for mitochondrial dysfunction in alveolar epithelial cells, fibroblasts, and immune cells that participate in the fibrotic process. Further, the fates of these cell populations and the potential to target mitochondrial dysfunction as a therapeutic strategy are discussed.


Subject(s)
Aging/metabolism , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/metabolism , Mitochondria/pathology , Aging/immunology , Alveolar Epithelial Cells/metabolism , Fibroblasts/metabolism , Humans , Lung/physiopathology
11.
Article in English | MEDLINE | ID: mdl-28756121

ABSTRACT

BACKGROUND: Long-term survival of lung transplant recipients (LTRs) is limited by the occurrence of bronchiolitis obliterans syndrome (BOS). Recent evidence suggests a role for microbiome alterations in the occurrence of BOS, although the precise mechanisms are unclear. In this study we evaluated the relationship between the airway microbiome and distinct subsets of immunoregulatory myeloid-derived suppressor cells (MDSCs) in LTRs. METHODS: Bronchoalveolar lavage (BAL) and simultaneous oral wash and nasal swab samples were collected from adult LTRs. Microbial genomic DNA was isolated, 16S rRNA genes amplified using V4 primers, and polymerase chain reaction (PCR) products sequenced and analyzed. BAL MDSC subsets were enumerated using flow cytometry. RESULTS: The oral microbiome signature differs from that of the nasal, proximal and distal airway microbiomes, whereas the nasal microbiome is closer to the airway microbiome. Proximal and distal airway microbiome signatures of individual subjects are distinct. We identified phenotypic subsets of MDSCs in BAL, with a higher proportion of immunosuppressive MDSCs in the proximal airways, in contrast to a preponderance of pro-inflammatory MDSCs in distal airways. Relative abundance of distinct bacterial phyla in proximal and distal airways correlated with particular airway MDSCs. Expression of CCAAT/enhancer binding protein (C/EBP)-homologous protein (CHOP), an endoplasmic (ER) stress sensor, was increased in immunosuppressive MDSCs when compared with pro-inflammatory MDSCs. CONCLUSIONS: The nasal microbiome closely resembles the microbiome of the proximal and distal airways in LTRs. The association of distinct microbial communities with airway MDSCs suggests a functional relationship between the local microbiome and MDSC phenotype, which may contribute to the pathogenesis of BOS.

13.
JCI Insight ; 2(2): e91377, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28138565

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal progressive fibrotic lung disease characterized by the presence of invasive myofibroblasts in the lung. Currently, there are only two FDA-approved drugs (pirfenidone and nintedanib) for the treatment of IPF. There are no defined criteria to guide specific drug therapy. New methodologies are needed not only to predict personalized drug therapy, but also to screen novel molecules that are on the horizon for treatment of IPF. We have developed a model system that exploits the invasive phenotype of IPF lung tissue. This ex vivo 3D model uses lung tissue from patients to develop pulmospheres. Pulmospheres are 3D spheroids composed of cells derived exclusively from primary lung biopsies and inclusive of lung cell types reflective of those in situ, in the patient. We tested the pulmospheres of 20 subjects with IPF and 9 control subjects to evaluate the responsiveness of individual patients to antifibrotic drugs. Clinical parameters and outcomes were also followed in the same patients. Our results suggest that pulmospheres simulate the microenvironment in the lung and serve as a personalized and predictive model for assessing responsiveness to antifibrotic drugs in patients with IPF.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Enzyme Inhibitors/pharmacology , Idiopathic Pulmonary Fibrosis/drug therapy , Indoles/pharmacology , Lung/drug effects , Myofibroblasts/drug effects , Pyridones/pharmacology , Spheroids, Cellular/drug effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biopsy , Case-Control Studies , Disease Progression , Enzyme Inhibitors/therapeutic use , Humans , Indoles/therapeutic use , Lung/pathology , Models, Biological , Precision Medicine , Pyridones/therapeutic use , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transforming Growth Factor beta1/pharmacology
14.
Sci Rep ; 6: 37445, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27869174

ABSTRACT

Cellular plasticity and de-differentiation are hallmarks of tissue/organ regenerative capacity in diverse species. Despite a more restricted capacity for regeneration, humans with age-related chronic diseases, such as cancer and fibrosis, show evidence of a recapitulation of developmental gene programs. We have previously identified a resident population of mesenchymal stromal cells (MSCs) in the terminal airways-alveoli by bronchoalveolar lavage (BAL) of human adult lungs. In this study, we characterized MSCs from BAL of patients with stable and progressive idiopathic pulmonary fibrosis (IPF), defined as <5% and ≥10% decline, respectively, in forced vital capacity over the preceding 6-month period. Gene expression profiles of MSCs from IPF subjects with progressive disease were enriched for genes regulating lung development. Most notably, genes regulating early tissue patterning and branching morphogenesis were differentially regulated. Network interactive modeling of a set of these genes indicated central roles for TGF-ß and SHH signaling. Importantly, fibroblast growth factor-10 (FGF-10) was markedly suppressed in IPF subjects with progressive disease, and both TGF-ß1 and SHH signaling were identified as critical mediators of this effect in MSCs. These findings support the concept of developmental gene re-activation in IPF, and FGF-10 deficiency as a potentially critical factor in disease progression.


Subject(s)
Cellular Reprogramming , Idiopathic Pulmonary Fibrosis/pathology , Mesenchymal Stem Cells/pathology , Bronchoalveolar Lavage Fluid/cytology , Disease Progression , Down-Regulation/genetics , Fibroblast Growth Factor 10/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Genes, Developmental , Hedgehog Proteins/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Immunohistochemistry , Lung/pathology , Mesenchymal Stem Cells/metabolism , Reproducibility of Results , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation/genetics
15.
FASEB J ; 30(6): 2135-50, 2016 06.
Article in English | MEDLINE | ID: mdl-26884454

ABSTRACT

Matricellular proteins mediate pleiotropic effects during tissue injury and repair. CCN1 is a matricellular protein that has been implicated in angiogenesis, inflammation, and wound repair. In this study, we identified CCN1 as a gene that is differentially up-regulated in alveolar mesenchymal cells of human subjects with rapidly progressive idiopathic pulmonary fibrosis (IPF). Elevated levels of CCN1 mRNA were confirmed in lung tissues of IPF subjects undergoing lung transplantation, and CCN1 protein was predominantly localized to fibroblastic foci. CCN1 expression in ex vivo IPF lung fibroblasts correlated with gene expression of the extracellular matrix proteins, collagen (Col)1a1, Col1a2, and fibronectin as well as the myofibroblast marker, α-smooth muscle actin. RNA interference (RNAi)-mediated knockdown of CCN1 down-regulated the constitutive expression of these profibrotic genes in IPF fibroblasts. TGF-ß1, a known mediator of tissue fibrogenesis, induces gene and protein expression of CCN1 via a mothers against decapentaplegic homolog 3 (SMAD3)-dependent mechanism. Importantly, endogenous CCN1 potentiates TGF-ß1-induced SMAD3 activation and induction of profibrotic genes, supporting a positive feedback loop leading to myofibroblast activation. In vivo RNAi-mediated silencing of CCN1 attenuates fibrogenic responses to bleomycin-induced lung injury. These studies support previously unrecognized, cooperative interaction between the CCN1 matricellular protein and canonical TGF-ß1/SMAD3 signaling that promotes lung fibrosis.-Kurundkar, A. R., Kurundkar, D., Rangarajan, S., Locy, M. L., Zhou, Y., Liu, R.-M., Zmijewski, J., Thannickal, V. J. The matricellular protein CCN1 enhances TGF-ß1/SMAD3-dependent profibrotic signaling in fibroblasts and contributes to fibrogenic responses to lung injury.


Subject(s)
Cysteine-Rich Protein 61/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/physiology , Lung Injury/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cells, Cultured , Cysteine-Rich Protein 61/genetics , Gene Knockdown Techniques , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pulmonary Fibrosis/metabolism , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Smad3 Protein/genetics , Transforming Growth Factor beta1/genetics , Up-Regulation
16.
Drugs ; 76(3): 291-300, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26729185

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an aging-associated, recalcitrant lung disease with historically limited therapeutic options. The recent approval of two drugs, pirfenidone and nintedanib, by the US Food and Drug Administration in 2014 has heralded a new era in its management. Both drugs have demonstrated efficacy in phase III clinical trials by retarding the rate of progression of IPF; neither drug appears to be able to completely arrest disease progression. Advances in the understanding of IPF pathobiology have led to an unprecedented expansion in the number of potential therapeutic targets. Drugs targeting several of these are under investigation in various stages of clinical development. Here, we provide a brief overview of the drugs that are currently approved and others in phase II clinical trials. Future therapeutic opportunities that target novel pathways, including some that are associated with the biology of aging, are examined. A multi-targeted approach, potentially with combination therapies, and identification of individual patients (or subsets of patients) who may respond more favourably to specific agents are likely to be more effective.


Subject(s)
Aging/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Indoles/therapeutic use , Molecular Targeted Therapy , Pyridones/therapeutic use , Aging/metabolism , Aging/pathology , Clinical Trials, Phase II as Topic , Drug Approval , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Indoles/administration & dosage , Indoles/adverse effects , Indoles/pharmacokinetics , Pyridones/administration & dosage , Pyridones/adverse effects , Pyridones/pharmacokinetics
17.
Am J Respir Cell Mol Biol ; 54(1): 51-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26072676

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a disease with relentless course and limited therapeutic options. Nintedanib (BIBF-1120) is a multiple tyrosine kinase inhibitor recently approved by the U.S. Food and Drug Administration for the treatment of IPF. The precise antifibrotic mechanism(s) of action of nintedanib, however, is not known. Therefore, we studied the effects of nintedanib on fibroblasts isolated from the lungs of patients with IPF. Protein and gene expression of profibrotic markers were assessed by Western immunoblotting and real-time PCR. Autophagy markers and signaling events were monitored by biochemical assays, Western immunoblotting, microscopy, and immunofluorescence staining. Silencing of autophagy effector proteins was achieved with small interfering RNAs. Nintedanib down-regulated protein and mRNA expression of extracellular matrix (ECM) proteins, fibronectin, and collagen 1a1 while inhibiting transforming growth factor (TGF)-ß1-induced myofibroblast differentiation. Nintedanib also induced beclin-1-dependent, ATG7-independent autophagy. Nintedanib's ECM-suppressive actions were not mediated by canonical autophagy. Nintedanib inhibited early events in TGF-ß signaling, specifically tyrosine phosphorylation of the type II TGF-ß receptor, activation of SMAD3, and p38 mitogen-activated protein kinase. Nintedanib down-regulates ECM production and induces noncanonical autophagy in IPF fibroblasts while inhibiting TGF-ß signaling. These mechanisms appear to be uncoupled and function independently to mediate its putative antifibrotic effects.


Subject(s)
Idiopathic Pulmonary Fibrosis/prevention & control , Indoles/pharmacology , Lung/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 7 , Beclin-1 , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Dose-Response Relationship, Drug , Fibronectins/genetics , Fibronectins/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad3 Protein/metabolism , Time Factors , Transfection , Transforming Growth Factor beta1/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Biol Chem ; 290(42): 25427-38, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26318453

ABSTRACT

Contraction is crucial in maintaining the differentiated phenotype of myofibroblasts. Contraction is an energy-dependent mechanism that relies on the production of ATP by mitochondria and/or glycolysis. Although the role of mitochondrial biogenesis in the adaptive responses of skeletal muscle to exercise is well appreciated, mechanisms governing energetic adaptation of myofibroblasts are not well understood. Our study demonstrates induction of mitochondrial biogenesis and aerobic glycolysis in response to the differentiation-inducing factor transforming growth factor ß1 (TGF-ß1). This metabolic reprogramming is linked to the activation of the p38 mitogen-activated protein kinase (MAPK) pathway. Inhibition of p38 MAPK decreased accumulation of active peroxisome proliferator-activated receptor γ coactivator 1α in the nucleus and altered the translocation of mitochondrial transcription factor A to the mitochondria. Genetic or pharmacologic approaches that block mitochondrial biogenesis or glycolysis resulted in decreased contraction and reduced expression of TGF-ß1-induced α-smooth muscle actin and collagen α-2(I) but not of fibronectin or collagen α-1(I). These data indicate a critical role for TGF-ß1-induced metabolic reprogramming in regulating myofibroblast-specific contractile signaling and support the concept of integrating bioenergetics with cellular differentiation.


Subject(s)
Cell Differentiation , Energy Metabolism , Myofibroblasts/metabolism , Cell Line , Electron Transport , Glycolysis , Humans , Lung/cytology , Lung/metabolism , Mitochondria/metabolism , Myofibroblasts/cytology , Oxygen Consumption , Transforming Growth Factor beta1/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
19.
PLoS One ; 10(2): e0116995, 2015.
Article in English | MEDLINE | ID: mdl-25658089

ABSTRACT

Transforming growth factor-ß (TGF-ß) mediates growth-inhibitory effects on most target cells via activation of the canonical SMAD signaling pathway. This growth-inhibitory activity may be coupled with cellular differentiation. Our studies demonstrate that TGF-ß1 inhibits proliferation of primary, non-transformed human lung fibroblasts in association with the induction of myofibroblast differentiation. Differentiated myofibroblasts maintain the capacity to proliferate in response to exogenous mitogenic stimuli and are resistant to serum deprivation-induced apoptosis. These proliferative and anti-apoptotic properties of myofibroblasts are related, in part, to the down-regulation of caveolin-1 (Cav-1) by TGF-ß1. Cav-1 down-regulation is mediated by early activation of p38 MAPK and does not require SMAD signaling. In contrast, myofibroblast differentiation is dependent on activation of the SMAD pathway, but not on p38 MAPK. Thus, combinatorial signaling by TGF-ß1 of myofibroblast differentiation and down-regulation of Cav-1 by SMAD and p38 MAPK pathways, respectively, confer proliferative and apoptosis-resistant properties to myofibroblasts. Selective targeting of this SMAD-independent, p38-MAPK/Cav-1-dependent pathway is likely to be effective in the treatment of pathological conditions characterized by TGF-ß signaling and myofibroblast activation.


Subject(s)
Caveolin 1/metabolism , Cell Proliferation , MAP Kinase Signaling System , Myofibroblasts/metabolism , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Survival , Humans , Myofibroblasts/pathology , Smad Proteins/metabolism
20.
Sleep Med ; 9(1): 88-93, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17825618

ABSTRACT

BACKGROUND: The prevalence of restless legs syndrome (RLS) in India is unknown. OBJECTIVES: The primary objective was to assess the occurrence of RLS in residents of Bangalore. The secondary objective was to correlate demographic and socioeconomic factors with RLS occurrence and severity. METHODS: This was a cross-sectional, questionnaire-based survey conducted during August 2005 among adult residents of Bangalore, who participated in a face-to-face interview. Diagnosis of RLS was based on fulfillment of all National Institutes of Health/International Restless Legs Syndrome Study Group (NIH/IRLSSG) essential criteria. Severity of RLS was assessed using the IRLSSG scale. RESULTS: RLS occurred in 27 (2.1%) of 1266 respondents. Predominant symptoms included "pulling," "tingling" and "pain". RLS was associated with delayed sleep onset and RLS severity correlated with the duration of delay in sleep onset. RLS was associated with per-capita income less than the equivalent of US$1/day, education less than high school level, chronic daily alcohol consumption and chronic blood loss. CONCLUSION: This is the first Indian population study on RLS which reveals prevalence of the disorder in a South Indian urban population at 2.1%. Larger studies are warranted to better characterize RLS in India.


Subject(s)
Restless Legs Syndrome/epidemiology , Severity of Illness Index , Urban Population/statistics & numerical data , Adult , Cross-Sectional Studies , Female , Humans , India/epidemiology , Male , Middle Aged , Prevalence , Research Design , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL
...