Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(39): e2306288120, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37729198

RESUMO

Nonsmall cell lung cancer (NSCLC) is highly malignant with limited treatment options, platinum-based chemotherapy is a standard treatment for NSCLC with resistance commonly seen. NSCLC cells exploit enhanced antioxidant defense system to counteract excessive reactive oxygen species (ROS), which contributes largely to tumor progression and resistance to chemotherapy, yet the mechanisms are not fully understood. Recent studies have suggested the involvement of histones in tumor progression and cellular antioxidant response; however, whether a major histone variant H1.2 (H1C) plays roles in the development of NSCLC remains unclear. Herein, we demonstrated that H1.2 was increasingly expressed in NSCLC tumors, and its expression was correlated with worse survival. When crossing the H1c knockout allele with a mouse NSCLC model (KrasLSL-G12D/+), H1.2 deletion suppressed NSCLC progression and enhanced oxidative stress and significantly decreased the levels of key antioxidant glutathione (GSH) and GCLC, the catalytic subunit of rate-limiting enzyme for GSH synthesis. Moreover, high H1.2 was correlated with the IC50 of multiple chemotherapeutic drugs and with worse prognosis in NSCLC patients receiving chemotherapy; H1.2-deficient NSCLC cells presented reduced survival and increased ROS levels upon cisplatin treatment, while ROS scavenger eliminated the survival inhibition. Mechanistically, H1.2 interacted with NRF2, a master regulator of antioxidative response; H1.2 enhanced the nuclear level and stability of NRF2 and, thus, promoted NRF2 binding to GCLC promoter and the consequent transcription; while NRF2 also transcriptionally up-regulated H1.2. Collectively, these results uncovered a tumor-driving role of H1.2 in NSCLC and indicate an "H1.2-NRF2" antioxidant feedforward cycle that promotes tumor progression and chemoresistance.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Camundongos , Humanos , Histonas/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Antioxidantes , Fator 2 Relacionado a NF-E2/genética , Espécies Reativas de Oxigênio , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Glutationa , Modelos Animais de Doenças
2.
BMC Infect Dis ; 19(1): 547, 2019 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-31226950

RESUMO

BACKGROUND: A nationwide investigation on the carriage proportion of H. influenzae among healthy populations is lacking in China. The purpose of the study was to review the prevalence of pharyngeal carriage of H. influenzae among healthy populations in China, and explore its influencing factors. The serotypes distribution of H. influenzae was also analyzed. METHODS: A systematic search was conducted with key words "Haemophilus influenzae", "Carriage", and "China" or "Chinese" from inception to March 2018. After careful screening, the data of included articles were extracted with a pre-designed excel form. Then, the pooled carriage proportion of H. influenzae was calculated using the random effect model. RESULTS: A total of 42 studies with 17,388 participants were included. The overall pooled carriage proportion of H. influenzae was 0.17 (95% CI: 0.13-0.21), and the carriage proportion largely varied by province. Subgroup analysis indicated that the pooled carriage proportion was 0.17 (0.13-0.21) for children, and 0.14 (0.7-0.23) for adults. There were no statistically significant heterogeneity between subgroups by age (p = 0.65), sex (p = 0.88), and season (p = 0.10). The pooled carriage proportion of Hib was 0.01 (0-0.02), while the carriage proportion of NTHi was 0.22 (0.13-0.31). CONCLUSION: In China, the carriage proportion of H. influenzae among healthy population was low, but it largely varied by provinces.


Assuntos
Portador Sadio/epidemiologia , Infecções por Haemophilus/epidemiologia , Haemophilus influenzae/isolamento & purificação , Faringe/microbiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Infecções Assintomáticas/epidemiologia , Criança , Pré-Escolar , China/epidemiologia , Feminino , Infecções por Haemophilus/microbiologia , Voluntários Saudáveis , Humanos , Lactente , Recém-Nascido , Masculino , Programas de Rastreamento , Pessoa de Meia-Idade , Prevalência , Estações do Ano , Adulto Jovem
3.
Cancer Sci ; 109(11): 3450-3460, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30191636

RESUMO

Kinesin family member 20B (KIF20B, also known as MPHOSPH1) is a kinesin protein that plays a critical role in cytokinesis. Previously, we and others have demonstrated the oncogenic role of KIF20B in several cancers; however, the exact mechanisms underlying its tumorigenic effects remain unclear. Herein, we showed overexpression of KIF20B in human hepatocellular carcinoma (HCC) and reported a negative correlation between KIF20B level and prognosis of patients. Mechanistically, reducing KIF20B blockades mitotic exit of HCC cells at telophase in a spindle assembly checkpoint independent way. Importantly, reducing KIF20B acts synergistically with three microtubule-associated agents (MTA) to p53- or p14ARF-dependently suppress p53-wt or p53-null HCC cells. In addition to taxol, reducing KIF20B also enhanced the toxicity of two chemotherapeutic drugs, hydroxycamptothecin and mitomycin C. In conclusion, we found a novel mechanism in that blocking cytokinesis by KIF20B inhibition increases the efficacy of MTA; our results thus suggested a dual-mitotic suppression approach against HCC by combining MTA with KIF20B inhibition, which simultaneously blocks mitosis at both metaphase and telophase.


Assuntos
Carcinoma Hepatocelular/metabolismo , Cinesinas/antagonistas & inibidores , Neoplasias Hepáticas/metabolismo , RNA Interferente Pequeno/farmacologia , Moduladores de Tubulina/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinese/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Mitose/efeitos dos fármacos , Prognóstico , Regulação para Cima/efeitos dos fármacos
4.
Biochim Biophys Acta ; 1852(7): 1278-87, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25748499

RESUMO

Renal ischemia/reperfusion (I/R) injury is the most common cause of acute kidney injury, having a high rate of mortality and no effective therapy currently available. Apelin-13, a bioactive peptide, has been shown to inhibit the early lesions of diabetic nephropathy in several mouse models by us and others. To test whether apelin-13 protects against renal I/R induced injury, male rats were exposed to renal I/R injury with or without apelin-13 treatment for 3 days. Apelin-13 treatment markedly reduced the injury-induced tubular lesions, renal cell apoptosis, and normalized the injury induced renal dysfunction. Apelin-13 treatment inhibited the injury-induced elevation of inflammatory factors and Tgf-ß1, as well as apoptosis. Apelin-13 treatment also inhibited the injury-induced elevation of histone methylation and Kmt2d, a histone methyltransferase of H3K4me2, following renal I/R injury. Furthermore, in cultured renal mesangial and tubular cells, apelin-13 suppressed the injury-induced elevation of Tgf-ß1, apoptosis, H3K4me2 and Kmt2d under the in vitro hypoxia/reperfusion (H/R) conditions. Consistently, over-expression of apelin significantly inhibited H/R-induced elevation of TGF-ß1, apoptosis, H3K4me2 and Kmt2d. The present study therefore suggests apelin-13 may be a therapeutic candidate for treating acute kidney injury.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Rim/irrigação sanguínea , Traumatismo por Reperfusão/prevenção & controle , Fator de Crescimento Transformador beta1/metabolismo , Animais , Apoptose , Linhagem Celular , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Fator de Crescimento Transformador beta1/genética
5.
J Physiol ; 592(3): 505-21, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24247978

RESUMO

Diabetic nephropathy is the primary cause of end-stage renal disease. Increasing numbers of patients are suffering from this disease and therefore novel medications and therapeutic approaches are urgently needed. Here, we investigated whether apelin-13, the most active member of the adipokine apelin group, could effectively suppress the development of nephropathy in Akita mouse, a spontaneous type 1 diabetic model. Apelin-13 treatment decreased diabetes-induced glomerular filtration rate, proteinuria, glomerular hypertrophy, mesangial expansion and renal inflammation. The inflammatory factors, activation of NF-κB, histone acetylation and the enzymes involved in histone acetylation were further examined in diabetic kidneys and high glucose- or sodium butyrate-treated mesangial cells in the presence or absence of apelin-13. Apelin-13 treatment inhibited diabetes-, high glucose- and NaB-induced elevation of inflammatory factors, and histone hyperacetylation by upregulation of histone deacetylase 1. Furthermore, overexpression of apelin in mesangial cells induced histone deacetylation under high glucose condition. Thus, apelin-13 may be a novel therapeutic candidate for treatment of diabetic nephropathy via regulation of histone acetylation.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Acetilação , Animais , Nefropatias Diabéticas/metabolismo , Taxa de Filtração Glomerular , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , NF-kappa B/metabolismo
6.
Biochem Biophys Res Commun ; 441(1): 133-8, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24129189

RESUMO

Endoplasmic reticulum (ER) stress is associated with the development of diabetes. The present study sought to investigate the effect of Liraglutide, a glucagon like peptide 1 analogue, on ER stress in ß-cells. We found that Liraglutide protected the pancreatic INS-1 cells from thapsigargin-induced ER stress and the ER stress associated cell apoptosis, mainly by suppressing the PERK and IRE1 pathways. We further tested the effects of Liraglutide in the Akita mouse, an ER-stress induced type 1 diabetes model. After administration of Liraglutide for 8weeks, p-eIF2α and p-JNK were significantly decreased in the pancreas of the Akita mouse, while the treatment showed no significant impact on the levels of insulin of INS-cells. Taken together, our findings suggest that Liraglutide may protect pancreatic cells from ER stress and its related cell death.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Animais , Morte Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Fator de Iniciação 2 em Eucariotos/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Liraglutida , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia
7.
Biophys Chem ; 296: 106984, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36889133

RESUMO

Protein misfolding diseases (PMDs) in humans are characterized by the deposition of protein aggregates in tissues, including Alzheimer's disease, Parkinson's disease, type 2 diabetes, and amyotrophic lateral sclerosis. Misfolding and aggregation of amyloidogenic proteins play a central role in the onset and progression of PMDs, and these processes are regulated by multiple factors, especially the interaction between proteins and bio-membranes. Bio-membranes induce conformational changes in amyloidogenic proteins and affect their aggregation; on the other hand, the aggregates of amyloidogenic proteins may cause membrane damage or dysfunction leading to cytotoxicity. In this review, we summarize the factors that affect the binding of amyloidogenic proteins and membranes, the effects of bio-membranes on the aggregation of amyloidogenic proteins, mechanisms of membrane disruption by amyloidogenic aggregates, technical approaches for detecting these interactions, and finally therapeutic strategies targeting membrane damage caused by amyloidogenic proteins.


Assuntos
Doença de Alzheimer , Diabetes Mellitus Tipo 2 , Doença de Parkinson , Humanos , Proteínas Amiloidogênicas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Doença de Alzheimer/metabolismo
8.
Theranostics ; 13(15): 5348-5364, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37908721

RESUMO

Rationale: Increased methylation of key genes has been observed in kidney diseases, suggesting that the ten-eleven translocation (Tet) methyl-cytosine dioxygenase family as well as 5mC oxidation may play important roles. As a member of the Tet family, the role of Tet1 in acute kidney injury (AKI) remains unclear. Methods: Tet1 knockout mice, with or without tempol treatment, a scavenger of reactive oxygen species (ROS), were challenged with ischemia and reperfusion (I/R) injury or unilateral ureteral obstruction (UUO) injury. RNA-sequencing, Western blotting, qRT-PCR, bisulfite sequencing, chromatin immunoprecipitation, immunohistochemical staining, and dot blot assays were performed. Results: Tet1 expression was rapidly upregulated following I/R or UUO injury. Moreover, Tet1 knockout mice showed increased renal injury and renal cell death, increased ROS accumulation, G2/M cell cycle arrest, inflammation, and fibrosis. Severe renal damage in injured Tet1 knockout mice was alleviated by tempol treatment. Mechanistically, Tet1 reduced the 5mC levels in an enzymatic activity-dependent manner on the promoters of Sod1 and Sod2 to promote their expression, thus lowering injury-induced excessive ROS and reducing I/R or UUO injury. Conclusions: Tet1 plays an important role in the development of AKI by promoting SOD expression through a DNA demethylase-dependent mechanism.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Obstrução Ureteral , Animais , Camundongos , Injúria Renal Aguda/metabolismo , Rim/metabolismo , Camundongos Knockout , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Superóxido Dismutase/metabolismo , Obstrução Ureteral/metabolismo
9.
Nat Commun ; 14(1): 4261, 2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37460623

RESUMO

Acute kidney injury (AKI) exhibits high morbidity and mortality. Kidney injury molecule-1 (KIM1) is dramatically upregulated in renal tubules upon injury, and acts as a biomarker for various renal diseases. However, the exact role and underlying mechanism of KIM1 in the progression of AKI remain elusive. Herein, we report that renal tubular specific knockout of Kim1 attenuates cisplatin- or ischemia/reperfusion-induced AKI in male mice. Mechanistically, transcription factor Yin Yang 1 (YY1), which is downregulated upon AKI, binds to the promoter of KIM1 and represses its expression. Injury-induced KIM1 binds to the ECD domain of death receptor 5 (DR5), which activates DR5 and the following caspase cascade by promoting its multimerization, thus induces renal cell apoptosis and exacerbates AKI. Blocking the KIM1-DR5 interaction with rationally designed peptides exhibit reno-protective effects against AKI. Here, we reveal a YY1-KIM1-DR5 axis in the progression of AKI, which warrants future exploration as therapeutic targets.


Assuntos
Injúria Renal Aguda , Rim , Animais , Masculino , Camundongos , Injúria Renal Aguda/metabolismo , Apoptose , Cisplatino/efeitos adversos , Rim/metabolismo , Túbulos Renais/metabolismo , Camundongos Endogâmicos C57BL , Receptores do Ligante Indutor de Apoptose Relacionado a TNF
10.
Theranostics ; 13(10): 3387-3401, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37351176

RESUMO

Rationale: Ischemia-reperfusion injury (I/R) is a common cause of acute kidney injury (AKI). Post-ischemic recovery of renal blood supply plays an important role in attenuating injury. Exogenous application of elabela (ELA) peptides has been demonstrated by us and others to alleviate AKI, partly through its receptor APJ. However, the endogenous role of ELA in renal I/R remains unclear. Methods: Renal tubule specific ELA knockout (ApelaKsp KO) mice challenged with bilateral or unilateral I/R were used to investigate the role of endogenous ELA in renal I/R. RNA-sequencing analysis was performed to unbiasedly investigate altered genes in kidneys of ApelaKsp KO mice. Injured mice were treated with ELA32 peptide, Nω-hydroxy-nor-L-arginine (nor-NOHA), prostaglandin E2 (PGE2), Paricalcitol, ML221 or respective vehicles, individually or in combination. Results: ELA is mostly expressed in renal tubules. Aggravated pathological injury and further reduction of renal microvascular blood flow were observed in ApelaKsp KO mice during AKI and the following transition to chronic kidney disease (AKI-CKD). RNA-seq analysis suggested that two blood flow regulators, arginine metabolizing enzyme arginase 2 (ARG2) and PGE2 metabolizing enzyme carbonyl reductases 1 and 3 (CBR1/3), were altered in injured ApelaKsp KO mice. Notably, combination application of an ARG2 inhibitor nor-NOHA, and Paricalcitol, a clinically used activator for PGE2 synthesis, alleviated injury-induced AKI/AKI-CKD stages and eliminated the worst outcomes observed in ApelaKsp KO mice. Moreover, while the APJ inhibitor ML221 blocked the beneficial effects of ELA32 peptide on AKI, it showed no effect on combination treatment of nor-NOHA and Paricalcitol. Conclusions: An endogenous tubular ELA-APJ axis regulates renal microvascular blood flow that plays a pivotal role in I/R-induced AKI. Furthermore, improving renal blood flow by inhibiting ARG2 and activating PGE2 is an effective treatment for AKI and prevents the subsequent AKI-CKD transition.


Assuntos
Injúria Renal Aguda , Hormônios Peptídicos , Insuficiência Renal Crônica , Traumatismo por Reperfusão , Camundongos , Animais , Microcirculação , Dinoprostona/farmacologia , Rim/patologia , Injúria Renal Aguda/patologia , Insuficiência Renal Crônica/etiologia , Traumatismo por Reperfusão/patologia , Isquemia/patologia , Hormônios Peptídicos/efeitos adversos , Hormônios Peptídicos/genética , Reperfusão/efeitos adversos
11.
Front Nutr ; 9: 927092, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35811982

RESUMO

The Coronavirus Disease 2019 (COVID-19) showed worse prognosis and higher mortality in individuals with obesity. Dyslipidemia is a major link between obesity and COVID-19 severity. Statins as the most common lipid regulating drugs have shown favorable effects in various pathophysiological states. Importantly, accumulating observational studies have suggested that statin use is associated with reduced risk of progressing to severe illness and in-hospital death in COVID-19 patients. Possible explanations underlie these protective impacts include their abilities of reducing cholesterol, suppressing viral entry and replication, anti-inflammation and immunomodulatory effects, as well as anti-thrombosis and anti-oxidative properties. Despite these benefits, statin therapies have side effects that should be considered, such as elevated creatinine kinase, liver enzyme and serum glucose levels, which are already elevated in severe COVID-19. Concerns are also raised whether statins interfere with the efficacy of COVID-19 vaccines. Randomized controlled trials are being conducted worldwide to confirm the values of statin use for COVID-19 treatment. Generally, the results suggest no necessity to discontinue statin use, and no evidence suggesting interference between statins and COVID-19 vaccines. However, concomitant administration of statins and COVID-19 antiviral drug Paxlovid may increase statin exposure and the risk of adverse effects, because most statins are metabolized mainly through CYP3A4 which is potently inhibited by ritonavir, a major component of Paxlovid. Therefore, more clinical/preclinical studies are still warranted to understand the benefits, harms and mechanisms of statin use in the context of COVID-19.

12.
Curr Pharm Des ; 28(37): 3033-3048, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36111767

RESUMO

Parkinson's disease (PD) is a common neurodegenerative disease that affects the motor system and progressively worsens with age. Current treatment options for PD mainly target symptoms, due to our limited understanding of the etiology and pathophysiology of PD. A variety of preclinical models have been developed to study different aspects of the disease. The models have been used to elucidate the pathogenesis and for testing new treatments. These models include cell models, non-mammalian models, rodent models, and non-human primate models. Over the past few decades, Caenorhabditis elegans (C. elegans) has been widely adopted as a model system due to its small size, transparent body, short generation time and life cycle, fully sequenced genome, the tractability of genetic manipulation and suitability for large scale screening for disease modifiers. Here, we review studies using C. elegans as a model for PD and highlight the strengths and limitations of the C. elegans model. Various C. elegans PD models, including neurotoxin-induced models and genetic models, are described in detail. Moreover, met.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Animais , Caenorhabditis elegans/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Modelos Animais de Doenças , Neurotoxinas , Roedores
13.
Diabetes Care ; 43(7): 1399-1407, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32409498

RESUMO

OBJECTIVE: Diabetes is one of the most distinct comorbidities of COVID-19. Here, we describe the clinical characteristics of and outcomes in patients with diabetes in whom COVID-19 was confirmed or clinically diagnosed (with typical features on lung imaging and symptoms) and their association with glucose-lowering or blood pressure-lowering medications. RESEARCH DESIGN AND METHODS: In this retrospective study involving 904 patients with COVID-19 (136 with diabetes, mostly type 2 diabetes), clinical and laboratory characteristics were collected and compared between the group with diabetes and the group without diabetes, and between groups taking different medications. Logistic regression was used to explore risk factors associated with mortality or poor prognosis. RESULTS: The proportion of comorbid diabetes is similar between cases of confirmed and of clinically diagnosed COVID-19. Risk factors for higher mortality of patients with diabetes and COVID-19 were older age (adjusted odds ratio [aOR] 1.09 [95% CI 1.04, 1.15] per year increase; P = 0.001) and elevated C-reactive protein (aOR 1.12 [95% CI 1.00, 1.24]; P = 0.043). Insulin usage (aOR 3.58 [95% CI 1.37, 9.35]; P = 0.009) was associated with poor prognosis. Clinical outcomes of those who use an ACE inhibitor (ACEI) or angiotensin II type-I receptor blocker (ARB) were comparable with those of patients who do not use ACEI/ARB among COVID-19 patients with diabetes and hypertension. CONCLUSIONS: C-reactive protein may help to identify patients with diabetes who are at greater risk of dying during hospitalization. Older patients with diabetes were prone to death related to COVID-19. Attention needs to be paid to patients with diabetes and COVID-19 who use insulin. ACEI/ARB use showed no significant impact on patients with diabetes and hypertension who have COVID-19.


Assuntos
Betacoronavirus , Infecções por Coronavirus/epidemiologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/epidemiologia , Pneumonia Viral/epidemiologia , Fatores Etários , Idoso , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , COVID-19 , Comorbidade , Feminino , Glucose/uso terapêutico , Humanos , Hipertensão/epidemiologia , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Pandemias , Estudos Retrospectivos , SARS-CoV-2 , Fatores de Tempo
14.
DNA Repair (Amst) ; 7(3): 452-63, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18206427

RESUMO

Cytotoxic action (tumor cell killing) and carcinogenic side effect (therapy-related secondary leukemia) of etoposide are closely related to its ability in stabilizing topoisomerase II cleavable complex (TOP2cc), a unique form of protein-linked DNA break. How cells process and detect TOP2-concealed DNA damage for the activation of downstream cellular responses remains unclear. Here, we showed proteasomal degradation of both TOP2 isozymes in a transcription-dependent manner upon etoposide treatment. Downregulation of TOP2 was preferentially associated with proteasomal removal of TOP2 in TOP2cc rather than proteolysis of free TOP2. Interestingly, blockage of TOP2 downregulation in TOP2cc also caused reduction in etoposide-induced activation of DNA damage molecules, an observation suggesting that the processing pathways of TOP2cc are involved in activation of etoposide-induced cellular responses. In this regard, we observed two TOP2cc processing pathways, replication- and transcription-initiated processing (RIP and TIP) with proteasome involved in the latter. Importantly, two processing pathways contributed to differential activation of various DNA damage signaling and downstream cellular responses. Etoposide-induced phosphorylation of p53 relied mainly on RIP, whereas activation of Chk1, Chk2 depended largely on TIP. Both RIP and TIP played roles in activating non-homologous end joining pathway, while only RIP modulated etoposide-induced cell killing in a p53-dependent manner. Collectively, our results are consistent with the notion that protein-linked DNA breakage (e.g., TOP2cc) requires processing pathways for initiating downstream DNA damage detection, repair as well as cell death programs.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Dano ao DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Etoposídeo/farmacologia , Transcrição Gênica/efeitos dos fármacos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Reparo do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/genética , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Glutaminase/metabolismo , Células HCT116 , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Complexo de Endopeptidases do Proteassoma , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína de Replicação A/metabolismo , Inibidores da Topoisomerase II , Proteína Supressora de Tumor p53/metabolismo
15.
Eur J Pharmacol ; 819: 169-180, 2018 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-29208474

RESUMO

Over the past decades, Caenorhabditis elegans (C. elegans) has been widely used as a model system because of its small size, transparent body, short generation time and lifespan (~3 days and 3 weeks, respectively), completely sequenced genome and tractability to genetic manipulation. Protein misfolding and aggregation are key pathological features in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Animal models, including C. elegans, have been extensively used to discover and validate new drugs against neurodegenerative diseases. The well-defined and genetically tractable nervous system of C. elegans offers an effective model to explore basic mechanistic pathways of neurodegenerative diseases. Recent progress in high-throughput drug screening also provides a powerful approach for identifying chemical modulators of biological processes. Here, we summarize the latest progress of using C. elegans as a model system for target identification and drug screening in neurodegenerative diseases.


Assuntos
Caenorhabditis elegans , Avaliação Pré-Clínica de Medicamentos/métodos , Doenças Neurodegenerativas/tratamento farmacológico , Animais , Modelos Animais de Doenças , Humanos , Terapia de Alvo Molecular
16.
Cell Death Dis ; 9(5): 534, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29748662

RESUMO

Proteins that bind to microtubule are important for cell cycle, and some of these proteins show oncogenic characteristics with mechanisms not fully understood. Herein we demonstrate overexpression of protein regulator of cytokinesis 1 (PRC1), a microtubule-associated regulator of mitosis, in human hepatocellular carcinoma (HCC). Moreover, upregulated PRC1 is associated with lower survival rates of HCC patients. Mechanistically, reducing PRC1 blocks mitotic exit of HCC cells at telophase in a spindle assembly checkpoint independent manner, and acts synergistically with microtubule-associated agents (MTAs) to suppress p53-wt or p53-null HCC cells in a p53- or p14ARF-dependent manner; while overexpressing PRC1 increases the resistance of HCC to taxol. A combined treatment of taxol/shPRC1 results in 90% suppression of tumor growth in subcutaneous HCC xenograft models. In orthotopic xenograft mice, reducing PRC1 significantly alleviates HCC development and hepatic injury. Together, our results suggest a dual-mitotic suppression approach against HCC by combining MTAs with cytokinesis inhibition, which blocks mitosis at both metaphase and telophase.


Assuntos
Carcinoma Hepatocelular , Proteínas de Ciclo Celular/biossíntese , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Hepáticas , Mitose/efeitos dos fármacos , Paclitaxel/farmacologia , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Células HEK293 , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitose/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Food Funct ; 7(6): 2624-34, 2016 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-27171848

RESUMO

The aim of our study was to investigate the protective effects and underlying mechanisms of myricetin, a bioactive food compound, on brain injury and neurological deficits after ischemic stroke. Treatment of myricetin significantly attenuated oxygen-glucose deprivation (OGD)-induced cell death in SHSY5Y cells in vitro. In a rat model of cerebral ischemia, myricetin was administered intragastrically at 2 h before and every day after middle cerebral artery occlusion (MCAO). The effects of myricetin were evaluated by various biochemical assays and neurobehavioral tests. Treatment with myricetin resulted in decreased infarction volume, reduced neuronal loss as well as lessened production of reactive oxygen species (ROS) and malondialdehyde following MCAO. We also found evidence that myricetin treatment could enhance the activity of antioxidant enzymes and mitochondrial function. Meanwhile, myricetin treatment reversed the suppression of Nrf2 nuclear translocation, and increased HO-1 expression in the ipsilateral ischemic brain and in the normal brain. Moreover, our results suggested that myricetin treatment resulted in significant improvement in neurological function. In conclusion, treatment with myricetin attenuates brain injury and neurological deficits in a rat model of cerebral ischemia via improvement of mitochondrial function and activation of the Nrf2 pathway.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Flavonoides/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase (Desciclizante)/metabolismo , Masculino , Malondialdeído/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fator 2 Relacionado a NF-E2/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
18.
Sci Rep ; 5: 13556, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26324190

RESUMO

Amyloid formation is associated with multiple amyloidosis diseases. Human calcitonin (hCT) is a typical amyloidogenic peptide, its aggregation is associated with medullary carcinoma of the thyroid (MTC), and also limits its clinical application. Magnolia officinalis is a traditional Chinese herbal medicine; its two major polyphenol components, magnolol (Mag) and honokiol (Hon), have displayed multiple functions. Polyphenols like flavonoids and their derivatives have been extensively studied as amyloid inhibitors. However, the anti-amyloidogenic property of a biphenyl backbone containing polyphenols such as Mag and Hon has not been reported. In this study, these two compounds were tested for their effects on hCT aggregation. We found that Mag and Hon both inhibited the amyloid formation of hCT, whereas Mag showed a stronger inhibitory effect; moreover, they both dose-dependently disassembled preformed hCT aggregates. Further immuno-dot blot and dynamic light scattering studies suggested Mag and Hon suppressed the aggregation of hCT both at the oligomerization and the fibrillation stages, while MTT-based and dye-leakage assays demonstrated that Mag and Hon effectively reduced cytotoxicity caused by hCT aggregates. Furthermore, isothermal titration calorimetry indicated Mag and Hon both interact with hCT. Together, our study suggested a potential anti-amyloidogenic property of these two compounds and their structure related derivatives.


Assuntos
Compostos de Bifenilo/metabolismo , Calcitonina/metabolismo , Lignanas/metabolismo , Compostos de Bifenilo/química , Calcitonina/química , Calorimetria , Linhagem Celular Tumoral , Difusão Dinâmica da Luz , Humanos , Lignanas/química , Magnolia/química , Magnolia/metabolismo , Medicina Tradicional Chinesa , Microscopia Eletrônica de Transmissão , Polifenóis/química , Ligação Proteica
19.
Food Funct ; 6(2): 584-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25518982

RESUMO

Long-term hyperglycemia is a typical symptom of diabetes mellitus (DM) which can cause a high level of protein glycation and lead to the formation of advanced glycation end products (AGEs). The accumulation of AGEs in turn deteriorates DM and its complications. Insulin, the only hormone that directly decreases blood sugar in vivo, is vulnerable to glycation which causes the loss of its biological activity. In this study, we used a porcine insulin (PI)-methylglyoxal (MGO) model to investigate the inhibitory effect of leonurine (LN), a natural alkaloid extracted from Herba leonuri, on AGE formation. Assays including AGE-specific fluorescence, and fructosamine level and carbonyl group content determination showed that LN can dose-dependently suppress PI glycation. A significantly decreased cross-linking level on the glycated PI was also proven by SDS-PAGE electrophoresis. A further liquid chromatography-mass spectrometry study suggested that LN may inhibit PI glycation through trapping MGO and keeping it from reacting with PI. Our results thus indicate that LN is a promising anti-glycation agent for the prevention of diabetes and its complications via inhibiting AGE formation.


Assuntos
Ácido Gálico/análogos & derivados , Produtos Finais de Glicação Avançada/metabolismo , Animais , Glicemia/metabolismo , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Ácido Gálico/farmacologia , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Hiperglicemia/sangue , Hiperglicemia/tratamento farmacológico , Insulina/análogos & derivados , Insulina/metabolismo , Leonurus/química , Espectrometria de Massas , Extratos Vegetais/farmacologia , Aldeído Pirúvico/metabolismo , Suínos
20.
Open Biochem J ; 8: 107-11, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25598850

RESUMO

Glucagon-like peptide-1 (GLP-1), which has been extensively applied for treating type 2 diabetes mellitus (T2DM), is an incretin hormone that regulates glucose homeostasis. GLP-1(28-36)amide, a C-terminal nonapeptide (FIAWLVKGRamide) of GLP-1, is a major product derived from the cleavage of GLP-1 by the neutral endopeptidase (NEP). GLP-1(28-36)amide has long been regarded as a metabolically inactive byproduct, however, recent findings reveal that GLP-1(28-36)amide plays multiple novel roles in ameliorating hepatic metabolism, protecting ß cells, improving glucose disposal and inhibiting weight gain. Here, we summarize the latest progress on the effects of GLP-1(28-36)amide with a focus on its roles in regulating the Wnt and mitochondrial-mediated signaling pathways.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA