Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Ind Med ; 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38980251

RESUMO

OBJECTIVES: Prior analyses of the Occupational Disease Surveillance System (ODSS) have compared cancer rates using internal referent groups. As an exploratory analysis, we sought to estimate cancer risk using general population reference rates to evaluate the impact that the comparison population has on findings from our surveillance program. METHODS: A cohort of approximately 2.3 million workers in Ontario, Canada with an accepted lost-time workers' compensation claim were followed for all cancer diagnoses between 1983 and 2018. Standardized incidence ratios (SIRs) and 95% confidence intervals were calculated for workers in specific occupational groups using (1) all other workers in the ODSS cohort, and (2) the general population of Ontario. RESULTS: SIRs using the general population reference group were generally equal to or modestly lower compared to SIRs using the internal reference group. Within occupation groups, SIRs had a discordant direction of association (increased rate in the internal comparison and decreased in the external comparison) for some cancer sites including urinary, prostate, and colorectal. CONCLUSIONS: Findings emphasize the importance of the choice of reference group when evaluating cancer risks in large occupational surveillance cohorts. Importantly, the magnitude of confounding and the healthy worker hire bias may depend on the occupation group and cancer site of interest.

2.
Biochim Biophys Acta Gen Subj ; 1868(8): 130651, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38825256

RESUMO

Cannabidiol (CBD) has antioxidant and anti-inflammatory activities. However, the anti-tumor effect of CBD on hepatocellular carcinoma (HCC) remains unclear. Here, we investigated whether CBD displays anti-tumorigenic effects in HCC cells and whether it could reduce tumorigenesis and metastases in vivo. First, this study treated HCC cells with different concentrations of CBD, followed by analyzing the changes in the proliferative, apoptotic, migratory and invasive abilities. The effects of CBD on the growth and metastasis of HCC cells in vivo were verified by tumorigenesis and metastasis assays. Subsequently, the target genes of CBD were predicted through the SwissTarget website and the genes differentially expressed in cells after CBD treatment were analyzed by microarray for intersection. The enrichment of the pathways after CBD treatment was analyzed by KEGG enrichment analysis, followed by western blot validation. Finally, rescue assays were used to validate the functions of genes as well as pathways in the growth and metastasis of HCC cells. A significant weakening of the ability of HCC cells to grow and metastasize in vitro and in vivo was observed upon CBD treatment. Mechanistically, CBD reduced GRP55 expression in HCC cells, along with increased TP53 expression and blocked MAPK signaling activation. In CBD-treated cells, the anti-tumor of HCC cells was restored after overexpression of GRP55 or deletion of TP53. CBD inhibits the MAPK signaling activation and increases the TP53 expression by downregulating GRP55 in HCC cells, thereby suppressing the growth and metastasis of HCC cells.


Assuntos
Canabidiol , Carcinoma Hepatocelular , Neoplasias Hepáticas , Receptores de Canabinoides , Proteína Supressora de Tumor p53 , Canabidiol/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Humanos , Proteína Supressora de Tumor p53/metabolismo , Receptores de Canabinoides/metabolismo , Receptores de Canabinoides/genética , Animais , Proliferação de Células/efeitos dos fármacos , Linhagem Celular Tumoral , Camundongos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Fenótipo , Camundongos Nus
3.
Phytother Res ; 37(7): 2965-2978, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36879546

RESUMO

Acute lung injury (ALI) caused by acute bacterial infection remains a common life-threatening lung disease. An increased inflammatory response is the basis for the occurrence and development of ALI. Most antibiotics can only reduce the bacterial load but do not protect from lung damage because of an excessive immune response. Chrysophanol (chrysophanic acid, Chr), as a natural anthraquinone extracted from Rheum palmatum L., has various biological functions, including anti-inflammatory, anti-cancer activities, and ameliorative effects on cardiovascular diseases. Considering these properties, we investigated the effect of Chr in Klebsiella pneumoniae (KP)-induced ALI mice and its potential mechanism. Our results showed that Chr had protective effects against KP-infected mice, including increased survival rate, decreased bacterial burden, reduced recruitment of immune cells, and reduced reactive oxygen species level of lung macrophages. Chr reduced the expression of inflammatory cytokines by inhibiting the toll-like receptor 4/nuclear factor kappa-B (TLR4/NF-κB) signaling pathway and inflammasome activation and strengthening autophagy. Overactivation of the TLR4/NF-κB signaling pathway by the activator Neoseptin 3 led to Chr losing control of inflammatory cytokines in cells, resulting in increased cell death. Similarly, overactivation of the c-Jun N-terminal kinase signaling pathway using the activator anisomycin resulted in Chr losing its inhibitory effect on NOD-like receptor thermal protein domain associated protein 3 (NFRP3) inflammasome activation, and cell viability was reduced. In addition, autophagy was blocked by siBeclin1, so Chr could not reduce inflammatory factors, and cell viability was markedly inhibited. Collectively, this work unravels the molecular mechanism underpinning Chr-alleviated ALI via inhibiting pro-inflammatory cytokines. Thus, Chr is a potential therapeutic agent for KP-induced ALI.


Assuntos
Lesão Pulmonar Aguda , NF-kappa B , Camundongos , Animais , NF-kappa B/metabolismo , Receptor 4 Toll-Like/metabolismo , Klebsiella pneumoniae/metabolismo , Inflamassomos , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/induzido quimicamente , Antraquinonas/farmacologia , Antraquinonas/uso terapêutico , Pulmão , Citocinas/metabolismo , Lipopolissacarídeos/farmacologia
4.
Phytomedicine ; 93: 153742, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34624808

RESUMO

BACKGROUND: Lung cancer is the leading cause of cancer death worldwide, yet no effective medication for this disease is available. Cochlioquinone B derivative (CoB1), purified from Salvia miltiorrhiza endophytic Bipolaris sorokiniana, affects the defense against pulmonary pathogens by regulating inflammatory responses. However, the effect of CoB1 on lung cancer and the underlying molecular mechanisms remain unknown. In the present study, we investigate the protective effects of CoB1 on lung cancer and explore its underlying mechanism. METHOD: We examined the inhibitory effect of CoB1 on lung cancer cells (A549 cells) by MTT and colony formation assay. The effect of CoB1 on cytostatic autophagy in lung cancer cells was verified by Western blot, transmission electron microscopy, and confocal microscopy. The differentially expressed miRNAs were identified using quantitative RT-PCR. Luciferase assay and Northern blot were performed to verify the correlation between miRNA-125b and Foxp3. Protein expression in autophagy-related pathways was detected by Western blot. Xenograft tumor models were constructed to explore the inhibitory effect of CoB1 and the role of miRNA-125b as a suppressor in lung cancer in vivo. RESULT: CoB1 inhibited lung cancer cell proliferation by inducing cytostatic autophagy both in vitro and in vivo. CoB1-induced autophagy was related to blocking of the PI3K/Akt1/mTOR signaling pathway. In addition, CoB1 induced miR-125b expression via activating the TAK1/MKK4/JNK/Smad axis, thereby reducing Foxp3 expression and further inducing autophagy. CONCLUSION: This study is the first to report the specific inhibitory function of CoB1 purified from Salvia miltiorrhiza endophytic Bipolaris sorokiniana in lung cancer, which may be due to the induction of autophagy. This study provides evidence and novel insights into the anticancer efficacy of CoB1.


Assuntos
Citostáticos , Neoplasias Pulmonares , MicroRNAs , Autofagia , Linhagem Celular Tumoral , Proliferação de Células , Fatores de Transcrição Forkhead , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
5.
Mol Ther Oncolytics ; 20: 82-93, 2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33575473

RESUMO

Lung cancer is the most frequent and fatal malignancy in humans worldwide, yet novel successful drugs for control of this disease are still lacking. Ipomoea batatas polysaccharides (IBPs) have been implicated in inhibiting diverse cancer types, but their functions in mitigating lung cancer are largely unknown. In this study, we identify a role of IBP in inhibiting lung cancer proliferation. We found that IBP significantly impedes the proliferation of lung cancer cells by inducing cytostatic macroautophagy both in vitro and in vivo. Mechanistically, IBP specifically promotes ubiquitination-mediated degradation of PAK1 (p21-activated kinase 1) and blocks its downstream Akt1/mTOR signaling pathway, leading to increased autophagic flux. In lung cancer xenografts in mice, IBP-induced cytostatic autophagy suppresses tumor development. Through site-directed mutational analysis, the underlying signaling augments ubiquitination via PAK1-ubiquitin interaction. Collectively, this work unravels the molecular mechanism underpinning IBP-induced cytostatic autophagy in lung cancer and characterizes IBP as a potential therapeutic agent for lung cancer treatment.

6.
Sci Rep ; 10(1): 20013, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33203903

RESUMO

The study aimed to investigate the antibacterial effect and potential mechanisms of chlorogenic acid (CA) in Klebsiella pneumonia (KPN) induced infection in vitro and in vivo. 62 KPN strains were collected from the First People's Hospital of Yunnan Province. CA and CA combined Levofloxacin (LFX) were detected for KPN biofilm (BF) formation in vitro. The lung infection mice model were established by KPN. The effect of CA (500 mg/kg), LFX (50 mg/kg) and CA combined LFX (250 mg/kg + 25 mg/kg) were evaluated through the survival of mice, the changes of inflammation factors of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1ß and IL-6 in serum, the histopathological analysis of lung and the protein expression of NLRP3 signaling pathway in vivo. A total of 62 KPNs were isolated and identified, of which 13 (21%) strains were BF positive. 8 (13%) strains were extended spectrum ß-lactamase strains (ESBLs), and 20 (32%) strains are ESBLs biofilm positive. In vitro study, CA and LFX showed a synergistic effect on KPN biofilm formation. In vivo mice experiment, CA, especially CA + LFX treated group significantly decreased the serum levels of TNF-α, IL-1ß and IL-6, improved the survival ratio and lung pathology changes, and also reduced the protein expression of ASC, caspase 1 p20, IL-1ß and phosphor NF-κB p65. CA could effectively alleviate lung infection of KPN infected mice, and the antibacterial effection is strengthened by combined with LFX. The study provide a theroy basis for making rational and scientific antibacterial therapy strategy in clinic.


Assuntos
Antibacterianos/farmacologia , Ácido Clorogênico/farmacologia , Infecções por Klebsiella/tratamento farmacológico , Klebsiella pneumoniae/efeitos dos fármacos , Levofloxacino/farmacologia , Animais , Antibacterianos/uso terapêutico , Biofilmes/efeitos dos fármacos , Ácido Clorogênico/uso terapêutico , Farmacorresistência Bacteriana , Sinergismo Farmacológico , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Infecções por Klebsiella/microbiologia , Infecções por Klebsiella/mortalidade , Levofloxacino/uso terapêutico , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/metabolismo
7.
Front Cell Dev Biol ; 8: 552020, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33240872

RESUMO

BACKGROUND: Salmonella typhimurium (ST) causes several intestinal diseases. Polyphenols including chlorogenic acid (CGA) inhibit pathogenesis. OBJECTIVE: This study aimed to investigate the mechanisms of CGA in ST infection. METHODS: The intestinal pathological changes and survival rate of ST-infected mice were measured to verify the protection of CGA on ST infection. The antibacterial effects of CGA in vitro on the invasion to intestinal epithelial cells and autophagy was evaluated. The relationships among GAS5, miR-23a, and PTEN were verified. Expression of inflammation- and autophagy-related proteins was detected. RESULTS: CGA treatment alleviated pathological damage, improved the secretion disturbance of intestinal cytokines caused by ST infection, and reduced the mortality of mice. Intestinal GAS5 was upregulated after CGA treatment. LncRNA GAS5 competitively bound to miR-23a to upregulate PTEN and inhibit the p38 MAPK pathway. CGA regulated the p38 MAPK pathway through lncRNA GAS5/miR-23a/PTEN axis to promote autophagy in ST infection. The functional rescue experiments of miR-23a and PTEN further identified these effects. CONCLUSION: CGA promotes autophagy and inhibits ST infection through the GAS5/miR-23a/PTEN axis and the p38 MAPK pathway.

8.
J Immunol ; 205(5): 1293-1305, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32747503

RESUMO

Owing to multiple antibiotic resistance, Pseudomonas aeruginosa causes the most intractable infections to human beings worldwide, thus exploring novel drugs to defend against this bacterium remains of great importance. In this study, we purified a novel cochlioquinone B derivative (CoB1) from Salvia miltiorrhiza endophytic Bipolaris sorokiniana and reveal its role in host defense against P. aeruginosa infection by activating cytoprotective autophagy in alveolar macrophages (AMs) both in vivo and in vitro. Using a P. aeruginosa infection model, we observed that CoB1-treated mice manifest weakened lung injury, reduced bacterial systemic dissemination, decreased mortality, and dampened inflammatory responses, compared with the wild type littermates. We demonstrate that CoB1-induced autophagy in mouse AMs is associated with decreased PAK1 expression via the ubiquitination-mediated degradation pathway. The inhibition of PAK1 decreases the phosphorylation level of Akt, blocks the Akt/mTOR signaling pathway, and promotes the release of ULK1/2-Atg13-FIP200 complex from mTOR to initiate autophagosome formation, resulting in increased bacterial clearance capacity. Together, our results provide a molecular basis for the use of CoB1 to regulate host immune responses against P. aeruginosa infection and indicate that CoB1 is a potential option for the treatment of infection diseases.


Assuntos
Autofagia/efeitos dos fármacos , Benzoquinonas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Células Cultivadas , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Pseudomonas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos
9.
Colloids Surf B Biointerfaces ; 185: 110616, 2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-31740323

RESUMO

Multidrug-resistant (MDR) bacterial strains have led to notable heathy threats to human beings. The demand for the development of effective antibacterial materials is increasing. Silver nanoparticles (AgNPs) and graphene-based nanomaterials are two major types of nanomaterials that are studied to inhibit and/or kill bacteria. In this study, by combining the excellent photothermal effect of graphene and antibacterial activity of AgNPs, we have applied reduced graphene oxide/silver (RGO/Ag) nanocomposite to destroy the MDR bacteria. The antibacterial activity of the RGO/Ag nanocomposite was systematically investigated using a regular bacterium of Escherichia coli (E. coli) and an MDR bacterium of Klebsiella pneumoniae (Kp). Compared with AgNPs, graphene oxide (GO) and RGO, the RGO/Ag nanocomposite showed significant higher antibacterial efficiency for both regular bacteria and MDR bacteria. Under a near-infrared (NIR) irradiation (0.30 W/cm2 for 10 min), the RGO/Ag nanocomposite demonstrated an enhanced synergetic antibacterial activity through the photothermal effect. Nearly 100 % of E. coli and Kp were killed by the treatment of 15 µg/mL and 30 µg/mL of RGO/Ag nanocomposite, respectively. Moreover, a membrane integrity assay and a reactive oxygen species (ROS) assay demonstrated that the RGO/Ag nanocomposite under NIR irradiation induced the cell membrane disruption and generation of ROS, providing possible mechanisms for their high antibacterial activity besides the photothermal effect. Finally, the cytotoxicity of the RGO/Ag nanocomposites toward different mammalian cells was studied, the cell viabilities retained above 60 % at higher concentrations of RGO/Ag, indicating that the RGO/Ag nanocomposites may be a low cytotoxic, efficient antibacterial agent with the irradiation.


Assuntos
Antibacterianos/farmacologia , Grafite/farmacologia , Hipertermia Induzida , Nanocompostos/química , Fototerapia , Prata/farmacologia , Bactérias/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
10.
Breast Cancer ; 26(6): 766-775, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31172425

RESUMO

BACKGROUND: Icariin is a major component isolated from Epimedium brevicornum Maxim and has been reported to exhibit anti-tumor activity. However, whether icariin could reverse the acquired drug resistance in breast cancer remains largely unclear. Therefore, this study was designed to explore the antitumor effects of icariin and its underlying mechanisms in a tamoxifen-resistant breast cancer cell line MCF-7/TAM. METHODS: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and Lactate dehydrogenase (LDH) assay were performed to determine the effects of icariin on cell viability and cell death. Cell cycle progression and apoptosis were detected by flow cytometry analysis. Transmission electron microscopy (TEM) assay was utilized to observe cell autophagy. The downstream protein levels were measured using western blotting. RESULTS: Here, we observed that icariin treatment not only inhibited the growth of MCF-7 but also has a potential function to overcome tamoxifen resistance in MCF-7/TAM. Moreover, icariin significantly induced cell cycle G0/G1 phase arrest and apoptosis, as well as suppressed autophagy. At molecular levels, icariin treatment remarkably down-regulated the expression levels of CDK2, CDK4, Cyclin D1, Bcl-2, LC3-1, LC3-II, AGT5, Beclin-1, but upregulated the expression levels of caspase-3, PARP and p62. Most importantly, we found inhibition of autophagy via 3-MA treatment could significantly enhance the effects of icariin on cell viability and apoptosis. Enhanced autophagy via autophagy related 5 (ATG5) overexpression could partially reverse the effects of icariin on cell viability and apoptosis. CONCLUSION: These results revealed that icariin might potentially be useful as an adjuvant agent in cancer chemotherapy to enhance the effect of tamoxifen through suppression of autophagy in vitro and provide insight into the therapeutic potential of icariin for the treatment of chemo-resistant breast cancer.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Flavonoides/farmacologia , Extratos Vegetais/farmacologia , Tamoxifeno/efeitos adversos , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Neoplasias da Mama/tratamento farmacológico , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Epimedium/química , Feminino , Humanos , Células MCF-7 , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/uso terapêutico , Transfecção
11.
Mol Med Rep ; 19(3): 2057-2064, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30664158

RESUMO

The present study aimed to investigate the anti­arthritic effects of curculigoside isolated from the rhizome of Curculigo orchioides Gaertn in vivo and in vitro, as well as to determine the potential underlying mechanisms. A rat model of arthritis was induced with type II collagen. Arthritic rats were treated with curculigoside (50 mg/kg) and blood samples were collected to determine serum levels of tumor necrosis factor (TNF)­α, interleukin (IL)­1ß, IL­6, IL­10, IL­12 and IL­17A. Furthermore, indices of the thymus and spleen were determined. The anti­proliferative effects of curculigoside were detected with Cell Counting kit­8 assays in rheumatoid arthritis­derived fibroblast­like synoviocyte MH7A cells. In addition, expression levels of Janus kinase (JAK)1, JAK3, signal transducer and activator of transcription (STAT)3, nuclear factor (NF)­κB p65 and its inhibitor (IκB) were determined by western blotting. The results revealed that curculigoside inhibited paw swelling and arthritis scores in type II collagen­induced arthritic (CIA) rats. Additionally, curculigoside decreased serum levels of TNF­α, IL­1ß, IL­6, IL­10, IL­12 and IL­17A in CIA rats. Curculigoside also significantly inhibited MH7A cell proliferation in a time and concentration­dependent manner. Furthermore, treatment downregulated the expression of JAK1, JAK3 and STAT3, and upregulated cytosolic nuclear factor (NF)­κB p65 and IκB. In conclusion, the results of the present study indicated that curculigoside exhibited significant anti­arthritic effects in vivo and in vitro, and the molecular mechanism may be associated with the JAK/STAT/NF­κB signaling pathway.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Benzoatos/administração & dosagem , Glucosídeos/administração & dosagem , Janus Quinase 1/genética , Janus Quinase 3/genética , Fator de Transcrição STAT3/genética , Animais , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo II/toxicidade , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas I-kappa B/genética , Interleucinas/genética , Ratos , Transdução de Sinais , Sinoviócitos/efeitos dos fármacos , Fator de Transcrição RelA/genética , Fator de Necrose Tumoral alfa/genética
12.
Oncol Rep ; 38(2): 611-624, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627697

RESUMO

Nanomaterials are increasingly used as drug carriers for cancer therapy. Nanomaterials also appeal to researchers in the areas of cancer diagnosis and biomarker discovery. Several antitumor nanodrugs are currently being tested in preclinical and clinical trials and show promise in therapeutic and other settings. We review the development of nanomaterial drug carriers, including liposomes, polymer nanoparticles, dendritic polymers, and nanomicelles, for the diagnosis and treatment of various cancers. The prospects of nanomaterials as drug carriers for future clinical applications are also discussed.


Assuntos
Sistemas de Liberação de Medicamentos/tendências , Nanotecnologia/tendências , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Dendrímeros/uso terapêutico , Humanos , Lipossomos/uso terapêutico , Nanopartículas/uso terapêutico
13.
Cell Res ; 26(12): 1273-1287, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27857054

RESUMO

Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated (Cas) systems in bacteria and archaea provide adaptive immunity against invading foreign nucleic acids. Previous studies suggest that certain bacteria employ their Type II CRISPR-Cas systems to target their own genes, thus evading host immunity. However, whether other CRISPR-Cas systems have similar functions during bacterial invasion of host cells remains unknown. Here we identify a novel role for Type I CRISPR-Cas systems in evading host defenses in Pseudomonas aeruginosa strain UCBPP-PA14. The Type I CRISPR-Cas system of PA14 targets the mRNA of the bacterial quorum-sensing regulator LasR to dampen the recognition by toll-like receptor 4, thus diminishing the pro-inflammatory responses of the host in cell and mouse models. Mechanistically, this nuclease-mediated RNA degradation requires a "5'-GGN-3'" recognition motif in the target mRNA, and HD and DExD/H domains in Cas3 of the Type I CRISPR-Cas system. As LasR and Type I CRISPR-Cas systems are ubiquitously present in bacteria, our findings elucidate an important common mechanism underlying bacterial virulence.


Assuntos
Sistemas CRISPR-Cas/genética , Marcação de Genes , Pseudomonas aeruginosa/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Proteínas Associadas a CRISPR/química , Proteínas Associadas a CRISPR/genética , Células Cultivadas , Feminino , Imunidade Inata , Inflamação/patologia , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Motivos de Nucleotídeos , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/mortalidade , Infecções por Pseudomonas/patologia , Infecções por Pseudomonas/veterinária , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , RNA Mensageiro/metabolismo , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Virulência/genética
14.
Tumour Biol ; 37(5): 6661-71, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26646563

RESUMO

Although the Warburg effect is a dominant metabolic phenotype observed in cancers, the metabolic changes and adaptation occurring in tumors have been demonstrated to extend beyond the Warburg effect and thus considered a secondary effect to the transformation process of carcinogenesis, including nutritional deficiencies. However, the role of nutritional deficiencies in this metabolic reprogramming (e. g., oxidative phosphorylation (OXPHOS)/glycolysis interconversion) is not completely known yet. Here, we showed that under regular culture condition, the proliferation of U251 cells, but not other tumor cell lines, preferentially performed the Warburg effect and was remarkably inhibited by oxamic acid which can inhibit the activity of lactate dehydrogenase (LDH); whereas under serum starvation, glycolysis was depressed, tricarboxylic acid cycle (TCA) was enhanced, and the activity of OXPHOS was reinforced to maintain cellular ATP content in a high level, but interestingly, we observed a decreased expression of reactive oxygen species (ROS). Moreover, the upregulated activity of mitochondrial complex I was confirmed by Western blots and showed that the mitochondrial-related protein, NDUFA9, NDUFB8, ND1, and VDAC1 were remarkably increased after serum starved. Mechanistically, nutritional deficiencies could reduce hypoxia-inducible factor α (HIF-1α) protein expression to increase C-MYC protein level, which in turn increased nuclear respiratory factor 1 (NRF1) and mitochondrial transcription factor A (TFAM) transcription to enhance the activity of OXPHOS, suggesting that metabolic reprogramming by the changes of microenvironment during the carcinogenesis can provide some novel therapeutic clues to traditional cancer treatments.


Assuntos
Metabolismo Energético , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Trifosfato de Adenosina/biossíntese , Linhagem Celular Tumoral , Células Cultivadas , Ciclo do Ácido Cítrico , Glicólise , Humanos , Redes e Vias Metabólicas , Mitocôndrias/genética , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Fenótipo , Inanição/metabolismo
15.
J Immunol ; 195(8): 3901-11, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26371245

RESUMO

Earlier studies reported that a cell membrane protein, Annexin A2 (AnxA2), plays multiple roles in the development, invasion, and metastasis of cancer. Recent studies demonstrated that AnxA2 also functions in immunity against infection, but the underlying mechanism remains largely elusive. Using a mouse infection model, we reveal a crucial role for AnxA2 in host defense against Pseudomonas aeruginosa, as anxa2(-/-) mice manifested severe lung injury, systemic dissemination, and increased mortality compared with wild-type littermates. In addition, anxa2(-/-) mice exhibited elevated inflammatory cytokines (TNF-α, IL-6, IL-1ß, and IFN-γ), decreased bacterial clearance by macrophages, and increased superoxide release in the lung. We further identified an unexpected molecular interaction between AnxA2 and Fam13A, which activated Rho GTPase. P. aeruginosa infection induced autophagosome formation by inhibiting Akt1 and mTOR. Our results indicate that AnxA2 regulates autophagy, thereby contributing to host immunity against bacteria through the Akt1-mTOR-ULK1/2 signaling pathway.


Assuntos
Anexina A2/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/imunologia , Animais , Anexina A2/genética , Autofagia/genética , Autofagia/imunologia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Citocinas/genética , Citocinas/imunologia , Feminino , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Infecções por Pseudomonas/genética , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética
16.
Mol Cell Biol ; 35(16): 2729-39, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26031335

RESUMO

Transient receptor potential channel 1 (TRPC1) is a nonselective cation channel that is required for Ca(2+) homeostasis necessary for cellular functions. However, whether TRPC1 is involved in infectious disease remains unknown. Here, we report a novel function for TRPC1 in host defense against Gram-negative bacteria. TRPC1(-/-) mice exhibited decreased survival, severe lung injury, and systemic bacterial dissemination upon infection. Furthermore, silencing of TRPC1 showed decreased Ca(2+) entry, reduced proinflammatory cytokines, and lowered bacterial clearance. Importantly, TRPC1 functioned as an endogenous Ca(2+) entry channel critical for proinflammatory cytokine production in both alveolar macrophages and epithelial cells. We further identified that bacterium-mediated activation of TRPC1 was dependent on Toll-like receptor 4 (TLR4), which induced endoplasmic reticulum (ER) store depletion. After activation of phospholipase Cγ (PLC-γ), TRPC1 mediated Ca(2+) entry and triggered protein kinase Cα (PKCα) activity to facilitate nuclear translocation of NF-κB/Jun N-terminal protein kinase (JNK) and augment the proinflammatory response, leading to tissue damage and eventually mortality. These findings reveal that TRPC1 is required for host defense against bacterial infections through the TLR4-TRPC1-PKCα signaling circuit.


Assuntos
Lesão Pulmonar/genética , Lesão Pulmonar/microbiologia , Proteína Quinase C-alfa/imunologia , Infecções por Pseudomonas/complicações , Infecções por Pseudomonas/genética , Pseudomonas aeruginosa/imunologia , Canais de Cátion TRPC/genética , Animais , Cálcio/imunologia , Células Cultivadas , Técnicas de Inativação de Genes , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/imunologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , NF-kappa B/imunologia , Infecções por Pseudomonas/imunologia , Transdução de Sinais , Canais de Cátion TRPC/imunologia , Receptor 4 Toll-Like/imunologia
17.
J Infect Dis ; 212(11): 1816-26, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26022442

RESUMO

BACKGROUND: Klebsiella pneumoniae causes serious infections and healthcare burdens in humans. We have previously reported that the deficiency of autophagy-related gene (Atg) 7 in macrophages (murine alveolar macrophage cell line [MH-S]) induced irregular host immunity against K. pneumoniae and worsened pathologic effects in the lung. In the current study, we investigated the molecular mechanism by which Atg7 influenced K. pneumoniae-induced inflammatory responses. METHODS: Expression levels of Atg7, ubiquitin (Ub), and tumor necrosis factor (TNF) α and phosphorylation of IκBα (p-IκBα) were determined with immunoblotting. Ubiquitylation of p-IκBα was determined with immunoprecipitation. RESULTS: We noted an interaction between Atg7 and p-IκBα, which was decreased in MH-S after K. pneumoniae infection, whereas the interaction between Ub and p-IκBα was increased. Knock-down of Atg7 with small interfering RNA increased p-IκBα ubiquitylation, promoted nuclear factor κB translocation into the nucleus, and increased the production of TNF-α. Moreover, knock-down of Ub with lentivirus-short hairpin RNA Ub particles decreased binding of p-IκBα to Ub and inhibited TNF-α expression in the primary alveolar macrophages and lung tissue of atg7-knockout mice on K. pneumoniae infection. CONCLUSIONS: Loss of Atg7 switched binding of p-IκBα from Atg7 to Ub, resulting in increased ubiquitylation of p-IκBα and intensified inflammatory responses against K. pneumoniae. Our findings not only reveal a regulatory role of Atg7 in ubiquitylation of p-IκBα but also indicate potential therapeutic targets for K. pneumoniae control.


Assuntos
Proteínas I-kappa B/metabolismo , Infecções por Klebsiella/imunologia , Proteínas Associadas aos Microtúbulos/metabolismo , Ubiquitinação/imunologia , Animais , Autofagia/imunologia , Proteína 7 Relacionada à Autofagia , Linhagem Celular , Inflamação/imunologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Transgênicos , Transdução de Sinais/imunologia , Receptor 4 Toll-Like
18.
Int J Nanomedicine ; 10: 1045-59, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25709431

RESUMO

Klebsiella pneumoniae (Kp) is one of the most common pathogens in nosocomial infections and is increasingly becoming multiple drug resistant. However, the molecular pathogenesis of Kp in causing tissue injury and dysregulated host defense remains elusive, further dampening the development of novel therapeutic measures. We have previously screened a series of synthetic antimicrobial beta-sheet forming peptides and identified a peptide (IRIKIRIK; ie, IK8L) with a broad range of bactericidal activity and low cytotoxicity in vitro. Here, employing an animal model, we investigated the antibacterial effects of IK8L in acute infection and demonstrated that peritoneal injection of IK8L to mice down-regulated inflammatory cytokines, alleviated lung injury, and importantly, decreased mortality compared to sham-injected controls. In addition, a math model was used to evaluate in vivo imaging data and predict infection progression in infected live animals. Mechanistically, IK8L can kill Kp by inhibiting biofilm formation and modulating production of inflammatory cytokines through the STAT3/JAK signaling both in vitro and in vivo. Collectively, these findings reveal that IK8L may have potential for preventing or treating Kp infection.


Assuntos
Antibacterianos , Infecções por Klebsiella , Klebsiella pneumoniae/efeitos dos fármacos , Peptídeos , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Modelos Animais de Doenças , Infecções por Klebsiella/tratamento farmacológico , Infecções por Klebsiella/microbiologia , Camundongos , Peptídeos/química , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Estrutura Secundária de Proteína
19.
Acta Biochim Biophys Sin (Shanghai) ; 46(6): 512-21, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24777141

RESUMO

The mitochondrial transcription termination factor (MTERF) family is a group of highly conserved DNA-binding proteins composed of four key members, MTERF1-4. To date, several studies have investigated the binding sites of MTERF1 on mitochondrial genome and the regulation of mitochondrial gene transcription, but the more intricate connection between mitochondrial genes transcription regulation, mitochondrial oxidative phosphorylation (OXPHOS), and cell proliferation is still poorly understood. In this study, we constructed over-expression and knockdown vectors of MTERF1 that were transfected into HeLa cells to investigate the functions of MTERF1. Results showed that although MTERF1 is a positive regulatory factor of mitochondrial genes transcription, it had no significant effect on the replication of mitochondrial DNA. Over-expression of MTERF1 increased mitochondrial oxidative phosphorylation activity and promoted ATP synthesis, cyclin D1 expression, and cell proliferation, while its knockdown inhibited ATP synthesis, decreased cyclin D1 expression, and slowed the cell growth. These results suggested that MTERF1 may promote cell proliferation by regulating oxidative phosphorylation activity in HeLa cells. Ultimately, these findings create a foundation for further and more conclusive studies on the physiological functions of MTERF family by providing novel insights into the potential mechanisms underlying cell proliferation regulation.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Proliferação de Células , Proteínas Mitocondriais/fisiologia , Trifosfato de Adenosina/metabolismo , Sequência de Bases , Primers do DNA , Replicação do DNA , DNA Mitocondrial/metabolismo , DNA Mitocondrial/fisiologia , Citometria de Fluxo , Células HeLa , Humanos , Fosforilação Oxidativa , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA