Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Comput Chem ; 44(8): 887-901, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36478400

RESUMO

The COVID-19 pandemic has been a public health emergency, with deadly forms constantly emerging around the world, highlighting the dire need for highly effective antiviral therapeutics. Peptide therapeutics show significant potential for this viral disease due to their efficiency, safety, and specificity. Here, two thousand seven hundred eight antibacterial peptides were screened computationally targeting the Main protease (Mpro) of SARS CoV-2. Six top-ranked peptides according to their binding scores, binding pose were investigated by molecular dynamics to explore the interaction and binding behavior of peptide-Mpro complexes. The structural and energetic characteristics of Mpro-DRAMP01760 and Mpro-DRAMP01808 complexes fluctuated less during a 250 ns MD simulation. In addition, three peptides (DRAMP01760, DRAMP01808, and DRAMP01342) bind strongly to Mpro protein, according to the free energy landscape and principal component analysis. Peptide helicity and secondary structure analysis are in agreement with our findings. Interaction analysis of protein-peptide complexes demonstrated that Mpro's residue CYS145, HIS41, PRO168, GLU166, GLN189, ASN142, MET49, and THR26 play significant contributions in peptide-protein attachment. Binding free energy analysis (MM-PBSA) demonstrated the energy profile of interacting residues of Mpro in peptide-Mpro complexes. To summarize, the peptides DRAMP01808 and DRAMP01760 may be highly Mpro specific, resulting disruption in a viral replication and transcription. The results of this research are expected to assist future research toward the development of antiviral peptide-based therapeutics for Covid-19 treatment.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Tratamento Farmacológico da COVID-19 , Pandemias , Peptídeos/farmacologia , Antivirais/farmacologia , Peptídeo Hidrolases , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular
2.
Inhal Toxicol ; 23(8): 486-94, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21689010

RESUMO

Ozone (O3), a commonly encountered environmental pollutant, has been shown to induce pulmonary fibrosis in different animal models; the underlying mechanism, however, remains elusive. To investigate the molecular mechanism underlying O3-induced pulmonary fibrosis, 6- to 8-week-old C57BL/6 male mice were exposed to a cyclic O3 exposure protocol consisting of 2 days of filtered air and 5 days of O3 exposure (0.5 ppm, 8 h/day) for 5 and 10 cycles with or without intraperitoneal injection of IN-1233, a specific inhibitor of the type 1 receptor of transforming growth factor beta (TGF-ß), the most potent profibrogenic cytokine. The results showed that O3 exposure for 5 or 10 cycles increased the TGF-ß protein level in the epithelial lining fluid (ELF), associated with an increase in the expression of plasminogen activator inhibitor 1 (PAI-1), a TGF-ß-responsive gene that plays a critical role in the development of fibrosis under various pathological conditions. Cyclic O3 exposure also increased the deposition of collagens and alpha smooth muscle actin (α-SMA) in airway walls. However, these fibrotic changes were not overt until after 10 cycles of O3 exposure. Importantly, blockage of the TGF-ß signaling pathway with IN-1233 suppressed O3-induced Smad2/3 phosphorylation, PAI-1 expression, as well as collagens and α-SMA deposition in the lung. Our data demonstrate for the first time that O3 exposure increases TGF-ß expression and activates TGF-ß signaling pathways, which mediates O3-induced lung fibrotic responses in vivo.


Assuntos
Brônquios/efeitos dos fármacos , Oxidantes Fotoquímicos/toxicidade , Ozônio/toxicidade , Fibrose Pulmonar/induzido quimicamente , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Actinas/metabolismo , Animais , Benzamidas/farmacologia , Brônquios/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Exposição por Inalação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Quinolinas/farmacologia , Serpina E2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/análise , Fator de Crescimento Transformador beta1/metabolismo
3.
Exp Cell Res ; 314(19): 3427-43, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18718466

RESUMO

Yip1p/Yif1p family proteins are five-span transmembrane proteins localized in the Golgi apparatus and the ER. There are nine family members in humans, and YIPF5 and YIF1A are the human orthologs of budding yeast Yip1p and Yif1p, respectively. We raised antisera against YIPF5 and YIF1A and examined the localization of endogenous proteins in HeLa cells. Immunofluorescence, immunoelectron microscopy and subcellular fractionation analysis suggested that YIPF5 and YIF1A are not restricted to ER exit sites but also localized in the ER-Golgi intermediate compartment (ERGIC) and some in the cis-Golgi at steady state. Along with ERGIC53, YIPF5 and YIF1A remained in the cytoplasmic punctate structures after brefeldin A treatment, accumulated in the ERGIC and the cis-Golgi after treatment with AlF4- and accumulated in the ER when ER to Golgi transport was inhibited by Sar1(H79G). These results supported the localization of YIPF5 and YIF1A in the ERGIC and the cis-Golgi, and strongly suggested that they are recycling between the ER and the Golgi apparatus. Analysis by blue native PAGE and co-immunoprecipitation showed that YIPF5 and YIF1A form stable complexes of three different sizes. Interestingly, the knockdown of YIPF5 or YIF1A caused partial disassembly of the Golgi apparatus suggesting that YIPF5 and YIF1A are involved in the maintenance of the Golgi structure.


Assuntos
Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Retículo Endoplasmático/ultraestrutura , Imunofluorescência , Complexo de Golgi/ultraestrutura , Células HeLa , Humanos , Proteínas de Membrana/análise , Proteínas de Membrana/genética , Microscopia Imunoeletrônica , Transporte Proteico , Proteínas de Transporte Vesicular/análise , Proteínas de Transporte Vesicular/genética
4.
J Alzheimers Dis ; 64(2): 447-457, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29914038

RESUMO

Alzheimer's disease (AD) is a major cause of dementia in the elderly with no effective treatment. Accumulation of amyloid-ß peptide (Aß) in the brain is a pathological hallmark of AD and is believed to be a central disease-causing and disease-promoting event. In a previous study, we showed that deletion of plasminogen activator inhibitor 1 (PAI-1), a primary inhibitor of tissue type and urokinase type plasminogen activators (tPA and uPA), significantly reduced brain Aß load in APP/PS1 mice, an animal model of familial AD. In this study, we further show that oral administration of TM5275, a small molecule inhibitor of PAI-1, for a period of 6 weeks, inhibits the activity of PAI-1 and increases the activities of tPA and uPA as well as plasmin, which is associated with a reduction of Aß load in the hippocampus and cortex and improvement of learning/memory function in APP/PS1 mice. Protein abundance of low density lipoprotein related protein-1 (LRP-1), a multi ligand endocytotic receptor involved in transporting Aß out of the brain, as well as plasma Aß42 are increased, whereas the expression and processing of full-length amyloid-ß protein precursor is not affected by TM5275 treatment in APP/PS1 mice. In vitro studies further show that PAI-1 increases, whereas TM5275 reduces, Aß40 level in the culture medium of SHSY5Y-APP neuroblastoma cells. Collectively, our data suggest that TM5275 improves memory function of APP/PS1 mice, probably by reducing brain Aß accumulation through increasing plasmin-mediated degradation and LRP-1-mediated efflux of Aß in the brain.


Assuntos
Doença de Alzheimer/complicações , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Transtornos da Memória , Piperazinas/uso terapêutico , para-Aminobenzoatos/uso terapêutico , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Mutação/genética , Neuroblastoma/patologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Presenilina-1/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
5.
Sci Rep ; 7(1): 14346, 2017 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-29085012

RESUMO

Chronic inflammation plays a key role in development of many liver diseases. Stimulation of Toll-like receptor 4 (TLR4) by bacterial lipopolysaccharide (LPS) initiates inflammation and promotes development of hepatocellular carcinoma and other liver diseases. NADPH oxidases contribute to LPS-induced reactive oxygen species (ROS) production and modulate TLR responses, but whether these enzymes function in TLR4 responses of hepatocytes is unknown. In the present work, we examined the role of NADPH oxidase 4 (Nox4) in LPS-induced TLR4 responses in human hepatoma cells and wildtype and Nox4-deficient mice. We found that LPS increased expression of Nox4, TNF-α, and proliferating cell nuclear antigen (PCNA). Nox4 silencing suppressed LPS-induced TNF-α and PCNA increases in human cells. The LPS-induced TNF-α increases were MyD88-dependent, and were attenuated in primary hepatocytes isolated from Nox4-deficient mice. We found that Nox4 mediated LPS-TLR4 signaling in hepatocytes via NF-ĸB and AP-1 pathways. Moreover, the effect of Nox4 depletion was time-dependent; following six weeks of repeated LPS stimulation in vivo, hepatic TNF-α and PCNA responses subsided in Nox4-deficient mice compared with wildtype mice. Therefore, our data suggest that Nox4 mediates LPS-TLR4 signaling in human hepatoma cells and murine hepatocytes and may contribute to the ability of LPS to stimulate liver pathology.


Assuntos
Carcinoma Hepatocelular/metabolismo , NADPH Oxidase 4/metabolismo , NADPH Oxidase 4/fisiologia , Animais , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Hepatócitos/metabolismo , Humanos , Lipopolissacarídeos/farmacologia , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidase 4/genética , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/fisiologia
6.
BMJ Open ; 7(8): e017768, 2017 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-28801442

RESUMO

INTRODUCTION: Environmental enteric dysfunction (EED) is a subacute inflammatory condition of the small intestinal mucosa with unclear aetiology that may account for more than 40% of all cases of stunting. Currently, there are no universally accepted protocols for the diagnosis, treatment and ultimately prevention of EED. The Bangladesh Environmental Enteric Dysfunction (BEED) study is designed to validate non-invasive biomarkers of EED with small intestinal biopsy, better understand disease pathogenesis and identify potential therapeutic targets for interventions designed to control EED and stunting. METHODS AND ANALYSIS: The BEED study is a community-based intervention where participants are recruited from three cohorts: stunted children aged 12-18 months (length for age Z-score (LAZ) <-2), at risk of stunting children aged 12-18 months (LAZ <-1 to -2) and malnourished adults aged 18-45 years (body mass index <18.5 kg/m2). After screening, participants eligible for study provide faecal, urine and plasma specimens to quantify the levels of candidate EED biomarkers before and after receiving a nutritional intervention. Participants who fail to respond to nutritional therapy are considered as the candidates for upper gastrointestinal endoscopy with biopsy. Histopathological scoring for EED will be performed on biopsies obtained from several locations within the proximal small intestine. Candidate EED biomarkers will be correlated with nutritional status, the results of histochemical and immunohistochemical analyses of epithelial and lamina propria cell populations, plus assessments of microbial community structure. ETHICS AND DISSEMINATION: Ethics approval was obtained in all participating institutes. Results of this study will be submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov ID: NCT02812615. Registered on 21 June 2016.


Assuntos
Transtornos do Crescimento , Doenças Inflamatórias Intestinais/diagnóstico , Mucosa Intestinal/patologia , Intestino Delgado/patologia , Desnutrição , Adolescente , Adulto , Bangladesh , Biomarcadores/metabolismo , Criança , Transtornos da Nutrição Infantil/dietoterapia , Transtornos da Nutrição Infantil/etiologia , Estudos de Coortes , Endoscopia Gastrointestinal , Feminino , Microbioma Gastrointestinal , Transtornos do Crescimento/dietoterapia , Transtornos do Crescimento/etiologia , Transtornos do Crescimento/metabolismo , Humanos , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Masculino , Desnutrição/dietoterapia , Desnutrição/etiologia , Desnutrição/metabolismo , Pessoa de Meia-Idade , Estado Nutricional , Projetos de Pesquisa , Adulto Jovem
7.
Toxicol Sci ; 147(1): 222-34, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26116027

RESUMO

Alzheimer's disease (AD) is a major cause of dementia in the elderly. Although early-onset (familial) AD is attributed to gene mutations, the cause for late-onset (sporadic) AD, which accounts for 95% of AD cases, is unknown. In this study, we show that exposure of 6-week-old amyloid beta precursor protein (APP)/presenilin (PS1) overexpressing mice, a well-established animal model of AD, and nontransgenic littermates to a cyclic O3 exposure protocol, which mimics environmental exposure episodes, accelerated learning/memory function loss in male APP/PS1 mice but not in female APP/PS1 mice or nontransgenic littermates. Female APP/PS1 mice had higher brain levels of amyloid beta peptide (Aß42) and Aß40, compared with male APP/PS1 mice; O3 exposure, however, had no significant effect on brain Aß load in either male or female mice. Our results further show that male APP/PS1 mice had lower levels of antioxidants (glutathione and ascorbate) and experienced augmented induction of NADPH oxidases, lipid peroxidation, and neuronal apoptosis upon O3 exposure, compared with female APP/PS1 mice. No significant effect of O3 on any of these parameters was detected in nontransgenic littermates. In vitro studies further show that 4-hydroxynonenal, a lipid peroxidation product which was increased in the plasma and cortex/hippocampus of O3-exposed male APP/PS1 mice, induced neuroblastoma cell apoptosis. Together, the results suggest that O3 exposure per se may not cause AD but can synergize with genetic risk factors to accelerate the pathophysiology of AD in genetically predisposed populations. The results also suggest that males may be more sensitive to O3-induced neuropathophysiology than females due to lower levels of antioxidants.


Assuntos
Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/psicologia , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/psicologia , Doenças do Sistema Nervoso/induzido quimicamente , Doenças do Sistema Nervoso/psicologia , Ozônio/toxicidade , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/genética , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Feminino , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , NADPH Oxidases/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Presenilinas/biossíntese , Presenilinas/genética , Caracteres Sexuais
8.
Exp Gerontol ; 61: 62-75, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25451236

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a fatal lung disorder with unknown cause and no effective treatment. The incidence of and mortality from IPF increase with age, suggesting that advanced age is a major risk factor for IPF. The mechanism underlying the increased susceptibility of the elderly to IPF, however, is unknown. In this study, we show for the first time that the protein level of plasminogen activator inhibitor 1 (PAI-1), a protease inhibitor which plays an essential role in the control of fibrinolysis, was significantly increased with age in mouse lung homogenate and lung fibroblasts. Upon bleomycin challenge, old mice experienced augmented PAI-1 induction and lung fibrosis as compared to young mice. Most interestingly, we show that fewer (myo)fibroblasts underwent apoptosis and more (myo)fibroblasts with increased level of PAI-1 accumulated in the lung of old than in young mice after bleomycin challenge. In vitro studies further demonstrate that fibroblasts isolated from lungs of old mice were resistant to H2O2 and tumor necrosis factor alpha-induced apoptosis and had augmented fibrotic responses to TGF-ß1, compared to fibroblasts isolated from young mice. Inhibition of PAI-1 activity with a PAI-1 inhibitor, on the other hand, eliminated the aging-related apoptosis resistance and TGF-ß1 sensitivity in isolated fibroblasts. Moreover, we show that knocking down PAI-1 in human lung fibroblasts with PAI-1 siRNA significantly increased their sensitivity to apoptosis and inhibited their responses to TGF-ß1. Together, the results suggest that increased PAI-1 expression may underlie the aging-related sensitivity to lung fibrosis in part by protecting fibroblasts from apoptosis.


Assuntos
Envelhecimento , Apoptose , Fibroblastos/fisiologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Fibrose Pulmonar/etiologia , Animais , Bleomicina/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta1/fisiologia
9.
J Alzheimers Dis ; 26(4): 767-78, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21860091

RESUMO

Alzheimer's disease (AD) is a major cause of dementia in the elderly with no effective treatment. Accumulation of amyloid-ß peptide (Aß) in the brain, one of the pathological features of AD, is considered to be a central disease-causing and disease-promoting event in AD. In this study, we showed that feeding male AßPP/PS1 transgenic mice, a well established mouse model of AD, with a diet containing phenolic antioxidant tert-butylhydroquinone (TBHQ) dramatically reduced brain Aß load with no significant effect on the amounts of alpha- and beta-C-terminal fragments or full-length AßPP. Further studies showed that TBHQ diet inhibited the expression of plasminogen activator inhibitor-1 (PAI-1), a protease inhibitor which plays a critical role in brain Aß accumulation in AD, accompanied by increases in the activities of tissue type and urokinase type plasminogen activators (tPA and uPA) as well as plasmin. Moreover, we showed that TBHQ diet increased the expression of low density lipoprotein related protein-1, a multi ligand endocytotic receptor involved in transporting Aß out of the brain, and plasma Aß(40) and Aß(42) levels. We also showed that TBHQ diet increased the concentration of glutathione, an important antioxidant, and suppressed the expression of NADPH oxidase 2 as well as lipid peroxidation. Collectively, our data suggest that TBHQ may have therapeutic potential for AD by increasing brain antioxidant capacity/reducing oxidative stress level and by stimulating Aß degradation/clearance pathways.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Amiloidose/prevenção & controle , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Hidroquinonas/farmacologia , Hidroquinonas/uso terapêutico , Peptídeos beta-Amiloides/metabolismo , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Antioxidantes/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Western Blotting , Química Encefálica/efeitos dos fármacos , Química Encefálica/genética , Dieta , Fibrinolisina/metabolismo , Glutationa/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estresse Oxidativo/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Presenilina-1/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA