Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Am J Hum Genet ; 111(4): 778-790, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38531365

RESUMO

Selenophosphate synthetase (SEPHS) plays an essential role in selenium metabolism. Two mammalian SEPHS paralogues, SEPHS1 and SEPHS2, share high sequence identity and structural homology with SEPHS. Here, we report nine individuals from eight families with developmental delay, growth and feeding problems, hypotonia, and dysmorphic features, all with heterozygous missense variants in SEPHS1. Eight of these individuals had a recurrent variant at amino acid position 371 of SEPHS1 (p.Arg371Trp, p.Arg371Gln, and p.Arg371Gly); seven of these variants were known to be de novo. Structural modeling and biochemical assays were used to understand the effect of these variants on SEPHS1 function. We found that a variant at residue Trp352 results in local structural changes of the C-terminal region of SEPHS1 that decrease the overall thermal stability of the enzyme. In contrast, variants of a solvent-exposed residue Arg371 do not impact enzyme stability and folding but could modulate direct protein-protein interactions of SEPSH1 with cellular factors in promoting cell proliferation and development. In neuronal SH-SY5Y cells, we assessed the impact of SEPHS1 variants on cell proliferation and ROS production and investigated the mRNA expression levels of genes encoding stress-related selenoproteins. Our findings provided evidence that the identified SEPHS1 variants enhance cell proliferation by modulating ROS homeostasis. Our study supports the hypothesis that SEPHS1 plays a critical role during human development and provides a basis for further investigation into the molecular mechanisms employed by SEPHS1. Furthermore, our data suggest that variants in SEPHS1 are associated with a neurodevelopmental disorder.


Assuntos
Deficiência Intelectual , Anormalidades Musculoesqueléticas , Transtornos do Neurodesenvolvimento , Animais , Criança , Humanos , Deficiências do Desenvolvimento/genética , Éxons , Deficiência Intelectual/genética , Mamíferos/genética , Hipotonia Muscular/genética , Anormalidades Musculoesqueléticas/genética , Neuroblastoma/genética , Transtornos do Neurodesenvolvimento/genética , Espécies Reativas de Oxigênio
2.
J Family Med Prim Care ; 11(9): 5593-5598, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36505561

RESUMO

Context: Gender-based discrimination is more predominant in India. In spite of various laws, gender inequality is an evil that plagues society even today. This is an important challenge for meeting our Sustainable Development Goals. Methods: This cross-sectional study was carried out in an urban field practice area. Study subjects were married women and their husbands in the age-group of 15-49 years along with their under-five children. Gender egalitarianism was assessed for factors like education, employment and media exposure. Factors which were studied for revealing gender egalitarianism among children included sex ratio, immunization status, nutritional status and health care expenditure. Completed family size and preference for the sex of the child were enquired about to assess the inclination towards male gender of the baby. Anthro software was used for statistical analysis. Results: Gender egalitarianism was found with regards to education. However, significant difference was noted in the employment status of men and women. Overall, sex ratio was in favor of girls. Though gender inequality was evident from the results, it was more in favor of girls. There was no evidence of gender bias for immunization of children. It was observed that more boys were stunted than girls and almost equal proportion of boys and girls were wasted. Conclusions: Factors like high literacy, control over income, access to financial resources made women more empowered and such empowered women were less likely to show son preference. Hence, there was no gender inequality among children in the present study.

4.
Elife ; 82019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30994454

RESUMO

How huntingtin (HTT) triggers neurotoxicity in Huntington's disease (HD) remains unclear. We report that HTT forms a transcription-coupled DNA repair (TCR) complex with RNA polymerase II subunit A (POLR2A), ataxin-3, the DNA repair enzyme polynucleotide-kinase-3'-phosphatase (PNKP), and cyclic AMP-response element-binding (CREB) protein (CBP). This complex senses and facilitates DNA damage repair during transcriptional elongation, but its functional integrity is impaired by mutant HTT. Abrogated PNKP activity results in persistent DNA break accumulation, preferentially in actively transcribed genes, and aberrant activation of DNA damage-response ataxia telangiectasia-mutated (ATM) signaling in HD transgenic mouse and cell models. A concomitant decrease in Ataxin-3 activity facilitates CBP ubiquitination and degradation, adversely impacting transcription and DNA repair. Increasing PNKP activity in mutant cells improves genome integrity and cell survival. These findings suggest a potential molecular mechanism of how mutant HTT activates DNA damage-response pro-degenerative pathways and impairs transcription, triggering neurotoxicity and functional decline in HD.


Assuntos
Ataxina-3/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Proteína Huntingtina/metabolismo , Proteínas Mutantes/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Animais , Linhagem Celular , RNA Polimerases Dirigidas por DNA/metabolismo , Humanos , Proteína Huntingtina/genética , Camundongos Transgênicos , Proteínas Mutantes/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Multimerização Proteica , Sialoglicoproteínas/metabolismo
5.
Expert Opin Ther Targets ; 22(8): 703-714, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30015535

RESUMO

INTRODUCTION: HIV-1-infected smokers are at risk of oxidative damage to neuronal cells in the central nervous system by both HIV-1 and cigarette smoke. Since neurons have a weak antioxidant defense system, they mostly depend on glial cells, particularly astrocytes, for protection against oxidative damage and neurotoxicity. Astrocytes augment the neuronal antioxidant system by supplying cysteine-containing products for glutathione synthesis, antioxidant enzymes such as SOD and catalase, glucose for antioxidant regeneration via the pentose-phosphate pathway, and by recycling of ascorbic acid. Areas covered: The transport of antioxidants and energy substrates from astrocytes to neurons could possibly occur via extracellular nanovesicles called exosomes. This review highlights the neuroprotective potential of exosomes derived from astrocytes against smoking-induced oxidative stress, HIV-1 replication, and subsequent neurotoxicity observed in HIV-1-positive smokers. Expert opinion: During stress conditions, the antioxidants released from astrocytes either via extracellular fluid or exosomes to neurons may not be sufficient to provide neuroprotection. Therefore, we put forward a novel strategy to combat oxidative stress in the central nervous system, using synthetically developed exosomes loaded with antioxidants such as glutathione and the anti-aging protein Klotho.


Assuntos
Astrócitos/metabolismo , Infecções por HIV/virologia , Fumar/efeitos adversos , Animais , Antioxidantes/metabolismo , Sistema Nervoso Central/fisiopatologia , Sistema Nervoso Central/virologia , Exossomos/metabolismo , HIV-1/fisiologia , Humanos , Neuroproteção/fisiologia , Estresse Oxidativo/fisiologia , Replicação Viral/fisiologia
6.
Physiol Rep ; 4(21)2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27905294

RESUMO

Glucocorticoids (GC) are a frontline therapy for numerous acute and chronic diseases because of their demonstrated efficacy at reducing systemic inflammation. An unintended side effect of GC therapy is the stimulation of skeletal muscle atrophy. Pathophysiological mechanisms responsible for GC-induced skeletal muscle atrophy have been extensively investigated, and the ability to treat patients with GC without unintended muscle atrophy has yet to be realized. We have reported that a single, standard-of-care dose of Methylprednisolone increases in vivo expression of NF-κB-inducing kinase (NIK), an important upstream regulatory kinase controlling NF-κB activation, along with other key muscle catabolic regulators such as Atrogin-1 and MuRF1 that induce skeletal muscle proteolysis. Here, we provide experimental evidence that overexpressing NIK by intramuscular injection of recombinant human NIK via adenoviral vector in mouse tibialis anterior muscle induces a 30% decrease in the average fiber cross-sectional area that is associated with increases in mRNA expression of skeletal muscle atrophy biomarkers MuRF1, Atrogin-1, myostatin and Gadd45. A single injection of GC induced NIK mRNA and protein within 2 h, with the increased NIK localized to nuclear and sarcolemmal locations within muscle fibers. Daily GC injections induced skeletal muscle fore limb weakness as early as 3 days with similar atrophy of muscle fibers as observed with NIK overexpression. NIK overexpression in primary human skeletal muscle myotubes increased skeletal muscle atrophy biomarkers, while NIK knockdown significantly attenuated GC-induced increases in NIK and Atrogin-1. These results suggest that NIK may be a novel, previously unrecognized mediator of GC-induced skeletal muscle atrophy.


Assuntos
Glucocorticoides/farmacologia , Músculo Esquelético/enzimologia , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Glucocorticoides/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/efeitos dos fármacos , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/patologia , Proteínas Serina-Treonina Quinases/administração & dosagem , RNA Mensageiro/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
7.
World J Gastrointest Pathophysiol ; 7(1): 38-47, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26909227

RESUMO

The gastrointestinal tract (GIT) is a complex system, which changes in response to requirements of the body. GIT represents a barrier to the external environment. To achieve this, epithelial cells must renew rapidly. This renewal of epithelial cells starts in the fetal life under the influence of many GIT peptides by swallowing amniotic fluid (AF). Development and maturation of GIT is a very complex cascade that begins long before birth and continues during infancy and childhood by breast-feeding. Many factors like genetic preprogramming, local and systemic endocrine secretions and many trophic factors (TF) from swallowed AF contribute and modulate the development and growth of the GIT. GIT morphogenesis, differentiation and functional development depend on the activity of various TF in the AF. This manuscript will review the role of AF borne TF in the development of GIT.

8.
J Innate Immun ; 8(2): 143-55, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26756522

RESUMO

The airway mucosa is responsible for mounting a robust innate immune response (IIR) upon encountering pathogen-associated molecular patterns. The IIR produces protective gene networks that stimulate neighboring epithelia and components of the immune system to trigger adaptive immunity. Little is currently known about how cellular reactive oxygen species (ROS) signaling is produced and cooperates in the IIR. We discuss recent discoveries about 2 nuclear ROS signaling pathways controlling innate immunity. Nuclear ROS oxidize guanine bases to produce mutagenic 8-oxoguanine, a lesion excised by 8-oxoguanine DNA glycosylase1/AP-lyase (OGG1). OGG1 forms a complex with the excised base, inducing its nuclear export. The cytoplasmic OGG1:8-oxoG complex functions as a guanine nucleotide exchange factor, triggering small GTPase signaling and activating phosphorylation of the nuclear factor (NF)x03BA;B/RelA transcription factor to induce immediate early gene expression. In parallel, nuclear ROS are detected by ataxia telangiectasia mutated (ATM), a PI3 kinase activated by ROS, triggering its nuclear export. ATM forms a scaffold with ribosomal S6 kinases, inducing RelA phosphorylation and resulting in transcription-coupled synthesis of type I and type III interferons and CC and CXC chemokines. We propose that ATM and OGG1 are endogenous nuclear ROS sensors that transmit nuclear signals that coordinate with outside-in pattern recognition receptor signaling, regulating the IIR.


Assuntos
Núcleo Celular/imunologia , Imunidade Inata , Pulmão/imunologia , Espécies Reativas de Oxigênio/imunologia , Transdução de Sinais/imunologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/imunologia , Núcleo Celular/patologia , DNA Glicosilases/imunologia , Guanina/análogos & derivados , Guanina/imunologia , Humanos , Pulmão/patologia , Oxirredução , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Quinases S6 Ribossômicas/imunologia , Fator de Transcrição RelA/imunologia
9.
PLoS Genet ; 11(1): e1004834, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25590633

RESUMO

Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD), is an untreatable autosomal dominant neurodegenerative disease, and the most common such inherited ataxia worldwide. The mutation in SCA3 is the expansion of a polymorphic CAG tri-nucleotide repeat sequence in the C-terminal coding region of the ATXN3 gene at chromosomal locus 14q32.1. The mutant ATXN3 protein encoding expanded glutamine (polyQ) sequences interacts with multiple proteins in vivo, and is deposited as aggregates in the SCA3 brain. A large body of literature suggests that the loss of function of the native ATNX3-interacting proteins that are deposited in the polyQ aggregates contributes to cellular toxicity, systemic neurodegeneration and the pathogenic mechanism in SCA3. Nonetheless, a significant understanding of the disease etiology of SCA3, the molecular mechanism by which the polyQ expansions in the mutant ATXN3 induce neurodegeneration in SCA3 has remained elusive. In the present study, we show that the essential DNA strand break repair enzyme PNKP (polynucleotide kinase 3'-phosphatase) interacts with, and is inactivated by, the mutant ATXN3, resulting in inefficient DNA repair, persistent accumulation of DNA damage/strand breaks, and subsequent chronic activation of the DNA damage-response ataxia telangiectasia-mutated (ATM) signaling pathway in SCA3. We report that persistent accumulation of DNA damage/strand breaks and chronic activation of the serine/threonine kinase ATM and the downstream p53 and protein kinase C-δ pro-apoptotic pathways trigger neuronal dysfunction and eventually neuronal death in SCA3. Either PNKP overexpression or pharmacological inhibition of ATM dramatically blocked mutant ATXN3-mediated cell death. Discovery of the mechanism by which mutant ATXN3 induces DNA damage and amplifies the pro-death signaling pathways provides a molecular basis for neurodegeneration due to PNKP inactivation in SCA3, and for the first time offers a possible approach to treatment.


Assuntos
Dano ao DNA/genética , Enzimas Reparadoras do DNA/genética , Doença de Machado-Joseph/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteínas Repressoras/genética , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/genética , Ataxina-3 , Reparo do DNA/genética , Enzimas Reparadoras do DNA/biossíntese , Humanos , Doença de Machado-Joseph/patologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Agregados Proteicos/genética , Proteína Quinase C-delta/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Expansão das Repetições de Trinucleotídeos/genética
10.
J Virol ; 89(5): 2628-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25520509

RESUMO

UNLABELLED: Respiratory syncytial virus (RSV) is a primary etiological agent of childhood lower respiratory tract disease. Molecular patterns induced by active infection trigger a coordinated retinoic acid-inducible gene I (RIG-I)-Toll-like receptor (TLR) signaling response to induce inflammatory cytokines and antiviral mucosal interferons. Recently, we discovered a nuclear oxidative stress-sensitive pathway mediated by the DNA damage response protein, ataxia telangiectasia mutated (ATM), in cytokine-induced NF-κB/RelA Ser 276 phosphorylation. Here we observe that ATM silencing results in enhanced single-strand RNA (ssRNA) replication of RSVand Sendai virus, due to decreased expression and secretion of type I and III interferons (IFNs), despite maintenance of IFN regulatory factor 3 (IRF3)-dependent IFN-stimulated genes (ISGs). In addition to enhanced oxidative stress, RSV replication enhances foci of phosphorylated histone 2AX variant (γH2AX), Ser 1981 phosphorylation of ATM, and IKKγ/NEMO-dependent ATM nuclear export, indicating activation of the DNA damage response. ATM-deficient cells show defective RSV-induced mitogen and stress-activated kinase 1 (MSK-1) Ser 376 phosphorylation and reduced RelA Ser 276 phosphorylation, whose formation is required for IRF7 expression. We observe that RelA inducibly binds the native IFN regulatory factor 7 (IRF7) promoter in an ATM-dependent manner, and IRF7 inducibly binds to the endogenous retinoic acid-inducible gene I (RIG-I) promoter. Ectopic IRF7 expression restores RIG-I expression and type I/III IFN expression in ATM-silenced cells. We conclude that paramyxoviruses trigger the DNA damage response, a pathway required for MSK1 activation of phospho Ser 276 RelA formation to trigger the IRF7-RIG-I amplification loop necessary for mucosal IFN production. These data provide the molecular pathogenesis for defects in the cellular innate immunity of patients with homozygous ATM mutations. IMPORTANCE: RNA virus infections trigger cellular response pathways to limit spread to adjacent tissues. This "innate immune response" is mediated by germ line-encoded pattern recognition receptors that trigger activation of two, largely independent, intracellular NF-κB and IRF3 transcription factors. Downstream, expression of protective antiviral interferons is amplified by positive-feedback loops mediated by inducible interferon regulatory factors (IRFs) and retinoic acid inducible gene (RIG-I). Our results indicate that a nuclear oxidative stress- and DNA damage-sensing factor, ATM, is required to mediate a cross talk pathway between NF-κB and IRF7 through mediating phosphorylation of NF-κB. Our studies provide further information about the defects in cellular and innate immunity in patients with inherited ATM mutations.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , RNA Helicases DEAD-box/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferons/biossíntese , NF-kappa B/metabolismo , Vírus Sinciciais Respiratórios/imunologia , Vírus Sendai/imunologia , Linhagem Celular , Proteína DEAD-box 58 , Células Epiteliais/imunologia , Células Epiteliais/virologia , Inativação Gênica , Humanos , Fosforilação , Processamento de Proteína Pós-Traducional , Receptores Imunológicos , Vírus Sinciciais Respiratórios/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Vírus Sendai/fisiologia , Replicação Viral
11.
Indian Pediatr ; 51(11): 934-5, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25432234

RESUMO

We describe profile of 60 children [mean (SD) age, 9.5 (3.8) y] presenting to the department of Pediatrics with snake envenomation. Neurotoxic bites were predominant (53%) and required mean (SD) 21.5 (9.29) antisnake venom vials, while children with neurohemotoxic features required mean (SD) 31.2 (10.8) vials to improve. Duration of hospital stay was median (SD) 4.0 (2.71) days. The commonest complication was respiratory dysfunction; mortality rate was 13.3%.


Assuntos
Mordeduras de Serpentes/epidemiologia , Adolescente , Antivenenos/uso terapêutico , Criança , Pré-Escolar , Feminino , Humanos , Índia/epidemiologia , Masculino , Estudos Retrospectivos , Mordeduras de Serpentes/tratamento farmacológico , Resultado do Tratamento
12.
World Allergy Organ J ; 7(1): 13, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24982697

RESUMO

A pathological hallmark of asthma is chronic injury and repair, producing dysfunction of the epithelial barrier function. In this setting, increased oxidative stress, growth factor- and cytokine stimulation, together with extracellular matrix contact produces transcriptional reprogramming of the epithelial cell. This process results in epithelial-mesenchymal transition (EMT), a cellular state associated with loss of epithelial polarity, expression of mesenchymal markers, enhanced mobility and extracellular matrix remodeling. As a result, the cellular biology of the EMT state produces characteristic changes seen in severe, refractory asthma: myofibroblast expansion, epithelial trans-differentiation and subepithelial fibrosis. EMT also induces profound changes in epithelial responsiveness that affects innate immune signaling that may have impact on the adaptive immune response and effectiveness of glucocorticoid therapy in severe asthma. We discuss how this complex phenotype is beginning to be understood using systems biology-level approaches through perturbations coupled with high throughput profiling and computational modeling. Understanding the distinct changes induced by EMT at the systems level may provide translational strategies to reverse the altered signaling and physiology of refractory asthma.

13.
Nucleic Acids Res ; 42(13): 8416-32, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24957606

RESUMO

Ataxia-telangiectasia mutated (ATM), a member of the phosphatidylinositol 3 kinase-like kinase family, is a master regulator of the double strand DNA break-repair pathway after genotoxic stress. Here, we found ATM serves as an essential regulator of TNF-induced NF-kB pathway. We observed that TNF exposure of cells rapidly induced DNA double strand breaks and activates ATM. TNF-induced ROS promote nuclear IKKγ association with ubiquitin and its complex formation with ATM for nuclear export. Activated cytoplasmic ATM is involved in the selective recruitment of the E3-ubiquitin ligase ß-TrCP to phospho-IκBα proteosomal degradation. Importantly, ATM binds and activates the catalytic subunit of protein kinase A (PKAc), ribosmal S6 kinase that controls RelA Ser 276 phosphorylation. In ATM knockdown cells, TNF-induced RelA Ser 276 phosphorylation is significantly decreased. We further observed decreased binding and recruitment of the transcriptional elongation complex containing cyclin dependent kinase-9 (CDK9; a kinase necessary for triggering transcriptional elongation) to promoters of NF-κB-dependent immediate-early cytokine genes, in ATM knockdown cells. We conclude that ATM is a nuclear damage-response signal modulator of TNF-induced NF-κB activation that plays a key scaffolding role in IκBα degradation and RelA Ser 276 phosphorylation. Our study provides a mechanistic explanation of decreased innate immune response associated with A-T mutation.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Quinase 9 Dependente de Ciclina/genética , Regulação da Expressão Gênica , Genes Precoces , NF-kappa B/metabolismo , Fator de Transcrição RelA/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Citocinas/genética , Citocinas/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Inibidor de NF-kappaB alfa , Fosforilação , Regiões Promotoras Genéticas , Serina/metabolismo , Fator de Transcrição RelA/química , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitinação , Proteínas Contendo Repetições de beta-Transducina/metabolismo
14.
Biomed Res Int ; 2013: 505864, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24228254

RESUMO

The respiratory mucosa is a major coordinator of the inflammatory response in chronic airway diseases, including asthma and chronic obstructive pulmonary disease (COPD). Signals produced by the chronic inflammatory process induce epithelial mesenchymal transition (EMT) that dramatically alters the epithelial cell phenotype. The effects of EMT on epigenetic reprogramming and the activation of transcriptional networks are known, its effects on the innate inflammatory response are underexplored. We used a multiplex gene expression profiling platform to investigate the perturbations of the innate pathways induced by TGF ß in a primary airway epithelial cell model of EMT. EMT had dramatic effects on the induction of the innate pathway and the coupling interval of the canonical and noncanonical NF- κ B pathways. Simulation experiments demonstrate that rapid, coordinated cap-independent translation of TRAF-1 and NF- κ B2 is required to reduce the noncanonical pathway coupling interval. Experiments using amantadine confirmed the prediction that TRAF-1 and NF- κ B2/p100 production is mediated by an IRES-dependent mechanism. These data indicate that the epigenetic changes produced by EMT induce dynamic state changes of the innate signaling pathway. Further applications of systems approaches will provide understanding of this complex phenotype through deterministic modeling and multidimensional (genomic and proteomic) profiling.


Assuntos
Asma/genética , Inflamação/genética , Doença Pulmonar Obstrutiva Crônica/genética , Mucosa Respiratória/metabolismo , Fator de Crescimento Transformador beta/genética , Asma/metabolismo , Asma/patologia , Transição Epitelial-Mesenquimal/genética , Perfilação da Expressão Gênica , Humanos , Imunidade Inata/genética , Inflamação/metabolismo , Inflamação/patologia , NF-kappa B/genética , Proteômica , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Mucosa Respiratória/patologia , Transdução de Sinais/genética , Fator 1 Associado a Receptor de TNF/biossíntese , Fator 1 Associado a Receptor de TNF/genética , Fator de Crescimento Transformador beta/biossíntese
15.
Cell Physiol Biochem ; 31(4-5): 659-72, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23711492

RESUMO

BACKGROUND: Both endoplasmic reticulum (ER) stress, a fundamental cell response associated with stress-initiated unfolded protein response (UPR), and loss of Klotho, an anti-aging hormone linked to NF-κB-induced inflammation, occur in chronic metabolic diseases such as obesity and type 2 diabetes. We investigated if the loss of Klotho is causally linked to increased ER stress. METHODS: We treated human renal epithelial HK-2, alveolar epithelial A549, HEK293, and SH-SH-SY5Y neuroblastoma cells with ER stress-inducing agents, thapsigargin and/or tunicamycin. Effects of overexpression or siRNA-mediated knockdown of Klotho on UPR signaling was investigated by immunoblotting and Real-time PCR. RESULTS: Elevated Klotho levels in HK-2 cells decreased expression of ER stress markers phospho--IRE1, XBP-1s, BiP, CHOP, pJNK, and phospho-p38, all of which were elevated in response to tunicamycin and/or thapsigargin. Similar results were observed using A549 cells for XBP-1s, BiP, and CHOP in response to thapsigargin. Conversely, knockdown of Klotho in HEK 293 cells using siRNA caused further thapsigargin-induced increases in pIRE-1, XBP-1s, and BiP. Klotho overexpression in A549 cells blocked thapsigargin-induced caspase and PARP cleavage and improved cell viability. CONCLUSION: Our data indicate that Klotho has an important role in regulating ER stress and that loss of Klotho is causally linked to ER stress-induced apoptosis.


Assuntos
Glucuronidase/metabolismo , Apoptose , Caspases/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Glucuronidase/antagonistas & inibidores , Glucuronidase/genética , Células HEK293 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas Klotho , Fosforilação , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Tapsigargina/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tunicamicina/farmacologia , Resposta a Proteínas não Dobradas , Regulação para Cima/efeitos dos fármacos
16.
J Biol Chem ; 288(20): 14612-14623, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23543740

RESUMO

The NF-κB transcription factor mediates the inflammatory response through distinct (canonical and non-canonical) signaling pathways. The mechanisms controlling utilization of either of these pathways are largely unknown. Here we observe that TNF stimulation induces delayed NF-κB2/p100 processing and investigate the coupling mechanism. TNF stimulation induces TNF-associated factor-1 (TRAF-1) that directly binds NF-κB-inducing kinase (NIK) and stabilizes it from degradation by disrupting its interaction with TRAF2·cIAP2 ubiquitin ligase complex. We show that TRAF1 depletion prevents TNF-induced NIK stabilization and reduces p52 production. To further examine the interactions of TRAF1 and NIK with NF-κB2/p100 processing, we mathematically modeled TRAF1·NIK as a coupling signaling complex and validated computational inference by siRNA knockdown to show non-canonical pathway activation is dependent not only on TRAF1 induction but also NIK stabilization by forming TRAF1·NIK complex. Thus, these integrated computational-experimental studies of TNF-induced TRAF1 expression identified TRAF1·NIK as a central complex linking canonical and non-canonical pathways by disrupting the TRAF2-cIAP2 ubiquitin ligase complex. This feed-forward kinase pathway is essential for the activation of non-canonical pathway.


Assuntos
Subunidade p52 de NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fator 1 Associado a Receptor de TNF/metabolismo , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Espectrometria de Massas , Microscopia Confocal , Modelos Teóricos , RNA Interferente Pequeno/metabolismo , Frações Subcelulares/metabolismo
18.
J Biol Chem ; 286(43): 37187-95, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21900239

RESUMO

Nuclear factor-κB (NF-κB) is an inducible cytoplasmic transcription factor that plays a role as a master regulator of airway mucosal inflammation. The prototypical ("canonical") NF-κB pathway controls cytoplasmic to nuclear translocation in response to stimulation by the mononuclear cytokine, TNF. Despite intensive investigation, the spectrum of kinases involved in the canonical NF-κB pathway has not yet been systematically determined. Here we have applied a high throughput siRNA-mediated loss-of-function screening assay to identify novel kinases important in TNF-induced NF-κB signaling. Type II A549 epithelial cells stably expressing an IL-8/luciferase reporter gene optimized for high throughput siRNA format (Z' score of 0.65) and siRNAs for 636 human kinases were reverse-transfected and screened in the assay. 36 candidate genes were identified that inhibited TNF signaling with a Z score deviation of <-1.3 in replicate plates. From this group, 11 kinases were selected for independent validation, of which eight were successfully silenced. Six kinases were validated, including ATM, CDK2, -5, and -7, CALM3, MAPAKP5, and MAP3K/MEKK3. The surprising function of ATM in TNF signaling was confirmed where reduced NF-κB/RelA translocation and Ser-276 phosphorylation were seen in ATM(-/-) mouse embryo fibroblasts. These data indicate that ATM is a key regulatory kinase that may control global NF-κB activation in the TNF-induced canonical pathway.


Assuntos
Proteínas Quinases/metabolismo , RNA Interferente Pequeno/biossíntese , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/metabolismo , Animais , Linhagem Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Quinases/genética , RNA Interferente Pequeno/genética , Fator de Transcrição RelA/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
19.
Endocrinology ; 152(10): 3622-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21846802

RESUMO

Enhanced levels of nuclear factor (NF)-κB-inducing kinase (NIK), an upstream kinase in the NF-κB pathway, have been implicated in the pathogenesis of chronic inflammation in diabetes. We investigated whether increased levels of NIK could induce skeletal muscle insulin resistance. Six obese subjects with metabolic syndrome underwent skeletal muscle biopsies before and six months after gastric bypass surgery to quantitate NIK protein levels. L6 skeletal myotubes, transfected with NIK wild-type or NIK kinase-dead dominant negative plasmids, were treated with insulin alone or with adiponectin and insulin. Effects of NIK overexpression on insulin-stimulated glucose uptake were estimated using tritiated 2-deoxyglucose uptake. NF-κB activation (EMSA), phosphatidylinositol 3 (PI3) kinase activity, and phosphorylation of inhibitor κB kinase ß and serine-threonine kinase (Akt) were measured. After weight loss, skeletal muscle NIK protein was significantly reduced in association with increased plasma adiponectin and enhanced AMP kinase phosphorylation and insulin sensitivity in obese subjects. Enhanced NIK expression in cultured L6 myotubes induced a dose-dependent decrease in insulin-stimulated glucose uptake. The decrease in insulin-stimulated glucose uptake was associated with a significant decrease in PI3 kinase activity and protein kinase B/Akt phosphorylation. Overexpression of NIK kinase-dead dominant negative did not affect insulin-stimulated glucose uptake. Adiponectin treatment inhibited NIK-induced NF-κB activation and restored insulin sensitivity by restoring PI3 kinase activation and subsequent Akt phosphorylation. These results indicate that NIK induces insulin resistance and further indicate that adiponectin exerts its insulin-sensitizing effect by suppressing NIK-induced skeletal muscle inflammation. These observations suggest that NIK could be an important therapeutic target for the treatment of insulin resistance associated with inflammation in obesity and type 2 diabetes.


Assuntos
Adiponectina/farmacologia , Resistência à Insulina , Músculo Esquelético/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Adulto , Células Cultivadas , Glucose/metabolismo , Humanos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Serina-Treonina Quinases/análise , Proteínas Proto-Oncogênicas c-akt/metabolismo
20.
Exp Diabetes Res ; 2011: 192564, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21869881

RESUMO

The impact of increased NF-κB-inducing kinase (NIK), a key component of the NF-κB activation pathways, on diabetes-induced renal inflammation remains unknown. We overexpressed NIK wild type (NIKwt) or kinase-dead dominant negative mutants (NIKdn) in HK-2 cells and demonstrated that RelB and p52, but not RelA, abundance and DNA binding increased in nuclei of NIKwt but not NIKdn overexpressed cells, and this corresponded with increases in multiple proinflammatory cytokines. Since TRAF3 negatively regulates NIK expression, we silenced TRAF3 by >50%; this increased nuclear levels of p52 and RelB, and transcript levels of proinflammatory cytokines and transcription factors. In HK-2 cells and mouse primary proximal tubule epithelial cells treated with methylglyoxal-modified albumin, multiple proinflammatory cytokines and NIK were increased in association with increased nuclear RelB and p52. These observations indicate that NIK regulates proinflammatory responses of renal proximal tubular epithelial cells via mechanisms involving TRAF3 and suggest a role for NF-κB noncanonical pathway activation in modulating diabetes-induced inflammation in renal tubular epithelium.


Assuntos
Nefropatias Diabéticas/genética , Túbulos Renais/patologia , Nefrite/genética , Proteínas Serina-Treonina Quinases/fisiologia , Urotélio/patologia , Animais , Células Cultivadas , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Túbulos Renais/metabolismo , Camundongos , Nefrite/etiologia , Nefrite/metabolismo , Nefrite/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Aldeído Pirúvico/química , Aldeído Pirúvico/farmacologia , RNA Interferente Pequeno/farmacologia , Albumina Sérica/química , Albumina Sérica/farmacologia , Transfecção , Urotélio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...