Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
BMC Microbiol ; 24(1): 37, 2024 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-38279108

RESUMO

BACKGROUND: Vibrio vulnificus exists as one of the most serious foodborne pathogens for humans, and rapid and sensitive detection methods are needed to control its infections. As an emerging method, The Loop-Mediated Isothermal Amplification (LAMP) assay has been applied to the early detection of various foodborne pathogens due to its high efficiency, but sample preprocessing still prolongs the complete detection. To optimize the detection process, our study established a novel sample preprocessing method that was more efficient compared to common methods. RESULT: Using V. vulnificus as the detecting pathogen, the water-lysis-based detecting LAMP method shortened the preprocessing time to ≤ 1 min with 100% LAMP specificity; the detection limits of the LAMP assay were decreased to 1.20 × 102 CFU/mL and 1.47 × 103 CFU/g in pure culture and in oyster, respectively. Furthermore, the 100% LAMP specificity and high sensitivity of the water-lysis method were also obtained on detecting V. parahaemolyticus, V. alginolyticus, and P. mirabilis, revealing its excellent LAMP adaption with improvement in sensitivity and efficiency. CONCLUSION: Our study provided a novel LAMP preprocessing method that was more efficient compared to common methods and possessed the practical potential for LAMP application in the future.


Assuntos
Técnicas de Diagnóstico Molecular , Vibrio vulnificus , Humanos , Vibrio vulnificus/genética , Técnicas de Amplificação de Ácido Nucleico/métodos , Água , Manejo de Espécimes , Sensibilidade e Especificidade
2.
Nat Commun ; 14(1): 6473, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37833415

RESUMO

Tumor growth requires elevated ribosome biogenesis. Targeting ribosomes is an important strategy for cancer therapy. The ribosome inhibitor, homoharringtonine (HHT), is used for the clinical treatment of leukemia, yet it is ineffective for the treatment of solid tumors, the reasons for which remain unclear. Here we show that Snail1, a key factor in the regulation of epithelial-to-mesenchymal transition, plays a pivotal role in cellular surveillance response upon ribotoxic stress. Mechanistically, ribotoxic stress activates the JNK-USP36 signaling to stabilize Snail1 in the nucleolus, which facilitates ribosome biogenesis and tumor cell survival. Furthermore, we show that HHT activates the JNK-USP36-Snail1 axis in solid tumor cells, but not in leukemia cells, resulting in solid tumor cell resistance to HHT. Importantly, a combination of HHT with the inhibition of the JNK-USP36-Snail1 axis synergistically inhibits solid tumor growth. Together, this study provides a rationale for targeting the JNK-USP36-Snail1 axis in ribosome inhibition-based solid tumor therapy.


Assuntos
Leucemia , Neoplasias , Humanos , Sobrevivência Celular , Ribossomos , Nucléolo Celular , Ubiquitina Tiolesterase
3.
Oncogene ; 42(5): 339-350, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36460773

RESUMO

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with a high risk of metastasis and recurrence. Although chemotherapy has greatly improved the clinical outcome of TNBC patients, acquired drug resistance remains a huge challenge for TNBC treatment. Breast cancer stem cells (BCSCs) play a critical role in breast cancer development, metastasis, recurrence, and chemotherapy resistance. Thus, it is of great importance to decipher the underlying molecular mechanism of BCSCs regulation for TNBC drug resistance. In this study, we demonstrate that the F-box protein FBXL2 is a critical negative regulator of BCSCs stemness and that downregulation of FBXL2 plays a causal role in TNBC drug resistance. We show that expression levels of FBXL2 significantly influence CD44high/CD24low subpopulation and the mammosphere formation ability of TNBC cells. Ectopic expression of FBXL2 inhibits initiation of TNBC and overcomes paclitaxel resistance in vivo. In addition, activation of FBXL2 by nebivolol, a clinically used small-molecule inhibitor of the beta-1 receptor, markedly overcomes BCSCs-induced paclitaxel resistance. Mechanistically, we show that FBXL2 targets transcriptional factor E47 for polyubiquitin- and proteasome-mediated degradation, resulting in inhibition of BCSC stemness. Clinical analyses indicate that low expression of FBXL2 correlates with high expression of E47 as well as with high stemness features, and is associated with poor clinical outcomes of breast cancer patients. Taken together, these results highlight that the FBXL2-E47 axis plays a critical role in the regulation of BCSC stemness and paclitaxel resistance. Thus, targeting FBXL2 might be a potential therapeutic strategy for drug-resistant TNBC.


Assuntos
Proteínas F-Box , Neoplasias de Mama Triplo Negativas , Humanos , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Linhagem Celular Tumoral , Mama/patologia , Células-Tronco Neoplásicas/patologia , Proteínas F-Box/genética , Proteínas F-Box/metabolismo
4.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-929614

RESUMO

@#[Abstract] Objective: The co-culture model of LAG3+Jurkat cells and tumor cells was constructed to investigate the anti-tumor effect and mechanism of a novel fully human anti-LAG3 monoclonal antibody in vitro. Methods: Jurkat cells were stimulated with PHA to simulate TIL, and the secretion of IL-2 was detected by ELISA to evaluate the degree of Jurkat cell activation. Meanwhile, FCM, Immunofluorescence and WB assays were employed to detect the expression of LAG3 in activated Jurkat cells and MHC classⅡmolecule(MHC-Ⅱ), a LAG3 ligand, in HGC-27, MGC-803 and A549 tumor cells. The co-culture model of activated LAG3+Jurkat cells and tumor cells was constructed, and CCK-8 assays were employed to detect the killing efficiency of LAG3+Jurkat cells against tumor cells at different effector-target ratios and the effect of the anti-LAG3 antibody . The secretion levels of cytokines IL-2, IL-10 and TNF-α in supernatant of co-culture system were detected by ELISA. Results: After 48 h treatment, 2 μg/mL PHA exhibited no obvious cytotoxicity to Jurkat cells (P>0.05), but could significantly induce IL-2 secretion (P<0.01) and LAG3 expression (P<0.01), indicating activated LAG3+Jurkat cells were acquired. MGC-803 and A549 cells significantly expressed MHC-Ⅱ (P<0.01), but HGC-27 cells did not express MHC-Ⅱ (P>0.05). The co-culture model of LAG3+Jurkat cells and tumor cells was constructed at a effector-target ratio of 10∶1. The anti-LAG3 antibody could effectively enhance the killing efficiency of Jurkat cells against MHC-Ⅱ+ tumor cells (P<0.05). Further analysis revealed that the secretion levels of cytokines IL-2, IL-10 and TNF-α were increased in the co-culture supernatant of MHC-Ⅱ+ target cell group (all P<0.01). Conclusion: A co-culture model of LAG3+Jurkat cells and tumor cells was successfully constructed in vitro. The anti-LAG3 antibody might increase the killing effect of Jurkat cells against MGC-803 and A549 tumor cells through blocking LAG3/MHC-Ⅱ interaction, which may be related to the increased secretion levels of cytokines IL-2, IL-10 and TNF-α in the supernatant of co-culture system.

5.
Nat Commun ; 12(1): 5919, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34635651

RESUMO

Abnormal activation of epidermal growth factor receptor (EGFR) drives non-small cell lung cancer (NSCLC) development. EGFR mutations-mediated resistance to tyrosine-kinase inhibitors (TKIs) is a major hurdle for NSCLC treatment. Here, we show that F-box protein FBXL2 targets EGFR and EGFR TKI-resistant mutants for proteasome-mediated degradation, resulting in suppression of EGFR-driven NSCLC growth. Reduced FBXL2 expression is associated with poor clinical outcomes of NSCLC patients. Furthermore, we show that glucose-regulated protein 94 (Grp94) protects EGFR from degradation via blockage of FBXL2 binding to EGFR. Moreover, we have identified nebivolol, a clinically used small molecule inhibitor, that can upregulate FBXL2 expression to inhibit EGFR-driven NSCLC growth. Nebivolol in combination with osimertinib or Grp94-inhibitor-1 exhibits strong inhibitory effects on osimertinib-resistant NSCLC. Together, this study demonstrates that the FBXL2-Grp94-EGFR axis plays a critical role in NSCLC development and suggests that targeting FBXL2-Grp94 to destabilize EGFR may represent a putative therapeutic strategy for TKI-resistant NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Proteínas F-Box/genética , Neoplasias Pulmonares/genética , Glicoproteínas de Membrana/genética , Acrilamidas/farmacologia , Compostos de Anilina/farmacologia , Animais , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Proteínas F-Box/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Nebivolol/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Análise de Sobrevida , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Theranostics ; 11(15): 7235-7246, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34158847

RESUMO

Rationale: Hosts defend against viral infection by sensing viral pathogen-associated molecular patterns and activating antiviral innate immunity through TBK1-IRF3 signaling. However, the underlying molecular mechanism remains unclear. Methods: SiRNAs targeting Sirt1-7 were transfected into macrophages to screen the antiviral function. Sirt5 deficient mice or macrophages were subjected to viral infection to assess in vivo and in vitro function of Sirt5 by detecting cytokines, viral replicates and survival rate. Immunoprecipitation, WesternBlot and luciferase reporter assay were used to reveal molecular mechanism. Results: In this study, we functionally screened seven Sirtuin family members, and found that Sirtuin5 (Sirt5) promotes antiviral signaling and responses. Sirt5 deficiency leads to attenuated antiviral innate immunity in vivo and in vitro upon viral infection by decreasing TBK1-IRF3 activation and type I IFN production. Sirt5 overexpression increased antiviral innate immunity. Mechanism investigation revealed that Sirt5 interacts with DDX3 and demalonylates DDX3, which is critical for TBK1-IRF3 activation. Mutation of the demalonylation lysine sites (K66, K130, and K162) of DDX3 increased ifnß transcription. Furthermore, the acetylation on lysine 118 of DDX3 positively regulated ifnß transcription, whereas Sirt5 could not deacetylate this site. Conclusion: Sirt5 promotes anti- RNA and DNA virus innate immune responses by increasing TBK1 signaling through demalonylating DDX3, which identifies a novel regulatory pathway of antiviral innate immune response.


Assuntos
RNA Helicases DEAD-box/imunologia , Imunidade Inata , Macrófagos/imunologia , Sirtuínas/imunologia , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , RNA Helicases DEAD-box/genética , Células HEK293 , Humanos , Lipoilação/genética , Lipoilação/imunologia , Macrófagos/virologia , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Células RAW 264.7 , Sirtuínas/genética , Estomatite Vesicular/genética , Vírus da Estomatite Vesicular Indiana/genética
7.
Front Cell Infect Microbiol ; 11: 652957, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34055666

RESUMO

Vibrio parahaemolyticus is a common pathogenic marine bacterium that causes gastrointestinal infections and other health complications, which could be life-threatening to immunocompromised patients. For the past two decades, the pathogenicity of environmental V. parahaemolyticus has increased greatly, and the genomic change behind this phenomenon still needs an in-depth exploration. To investigate the difference in pathogenicity at the genomic level, three strains with different hemolysin expression and biofilm formation capacity were screened out of 69 environmental V. parahaemolyticus strains. Subsequently, 16S rDNA analysis, de novo sequencing, pathogenicity test, and antibiotic resistance assays were performed. Comparative genome-scale interpretation showed that various functional region differences in pathogenicity of the selected V. parahaemolyticus strains were due to dissimilarities in the distribution of key genetic elements and in the secretory system compositions. Furthermore, the genomic analysis-based hypothesis of distinct pathogenic effects was verified by the survival rate of mouse models infected with different V. parahaemolyticus strains. Antibiotic resistance results also presented the multi-directional evolutionary potential in environmental V. parahaemolyticus, in agreement with the phylogenetic analysis results. Our study provides a theoretical basis for better understanding of the increasing pathogenicity of environmental V. parahaemolyticus at the genome level. Further, it has a key referential value for the exploration of pathogenicity and prevention of environmental V. parahaemolyticus in the future.


Assuntos
Vibrioses , Vibrio parahaemolyticus , Animais , Proteínas Hemolisinas/genética , Humanos , Camundongos , Filogenia , Virulência , Fatores de Virulência
8.
Sci Rep ; 10(1): 19885, 2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33173121

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 36(9): 776-781, 2020 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-32967760

RESUMO

Objective To explore the regulatory effect of quorum sensing molecule N-3-oxodecanoyl-L-homoserine lactone (3-oxo-C10-HSL) on lipopolysaccharide (LPS)-induced inflammation in RAW264.7 macrophages. Methods RAW264.7 macrophages were divided into experimental group, control group and blank group. The experimental group was treated with different concentrations of 3-oxo-C10-HSL and LPS; the control group was treated with DMSO and LPS; and the blank group was treated with DMSO and PBS. Cells and supernatants were collected after 12 hours of stimulation. The mRNA expression of interleukin-6 (IL-6), interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) were detected by real-time quantitative PCR, and the protein levels of IL-6 and TNF-α in supernatant were detected by ELSIA. Further, 25 µmol/L 3-oxo-C10-HSL and 100 ng/mL LPS were used to stimulate the cells for 15, 30 and 60 minutes, and the phosphorylation of nuclear factor κBp65 (NF-κBp65) was detected by Western blot analysis. Results The 3-oxo-C10-HSL could decrease the mRNA levels of IL-6, IL-1ß, TNF-α, MCP-1 and the protein levels of IL-6 and TNF-α in LPS-treated RAW264.7 macrophages in a dose-dependent manner. In addition, 3-oxo-C10-HSL could inhibit the phosphorylation of NF-κBp65 induced by LPS. Conclusion 3-oxo-C10-HSL can alleviate LPS-induced inflammatory responses in RAW264.7 macrophages by inhibiting activation of NF-κB signaling pathway.


Assuntos
Percepção de Quorum , 4-Butirolactona/análogos & derivados , Lipopolissacarídeos , Macrófagos , NF-kappa B , Fator de Necrose Tumoral alfa/genética
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 36(5): 451-456, 2020 May.
Artigo em Chinês | MEDLINE | ID: mdl-32696758

RESUMO

Objective Lymphocyte activation gene 3 (LAG-3) is a potential therapeutic target of tumor. This work aims to screen for anti-LAG-3 human scFvs from a human phage antibody library, and transform them into fully human antibodies. Methods The human phage antibody library was screened for anti-LAG-3 human scFvs. The specificity and affinity of phage scFvs were determined by ELISA and surface plasmon resonance (SPR), respectively. The anti-LAG-3 full human antibodies were obtained using eukaryotic expression system. Results Through 3 rounds of screening, we obtained 4 phage scFv clones that could bind to human LAG-3 recombinant protein. Affinity determination showed that the highest affinity reached 3.48×10-10. Then through the construction of whole human antibody expression vector, expression in eukaryotic cells, and purification, 3 clones of anti-LAG-3 full human antibodies were harvested finally. Conclusion Using a large-capacity human antibody phage display library, we obtained three anti-LAG-3 full human antibodies with high affinity.


Assuntos
Bacteriófagos , Anticorpos de Cadeia Única , Antígenos CD , Bacteriófagos/genética , Ensaio de Imunoadsorção Enzimática , Humanos , Biblioteca de Peptídeos , Anticorpos de Cadeia Única/genética , Proteína do Gene 3 de Ativação de Linfócitos
11.
Front Immunol ; 10: 2575, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31781099

RESUMO

Hyper-inflammation during acute phase and sequential hypo-inflammation during immunosuppressive phase in macrophages/monocytes lead to multiorgan failure syndrome and immune collapse of sepsis, in which toll-like receptor (TLR)-triggered inflammatory responses play a major role. Here, we reported that Siglecg deficiency attenuated TLR4-triggered pro-inflammatory cytokine production and increased anti-inflammatory cytokine [interleukin-10 [IL-10]] production in vivo and in vitro at both acute and immunosuppressive phases. Siglecg deficiency also protected mice from lipopolysaccharide (LPS)-induced sepsis with less inflammation in the lung and less tissue destruction in the spleen. Siglec-G inhibited proto-oncogene tyrosine-protein kinase Src (Src) activation via recruiting and activating tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP1) through immunoreceptor tyrosine-based inhibitory motif (ITIM) domain. Src could inhibit TLR4-induced inflammatory cytokines and promote anti-inflammatory cytokine IL-10. Mechanical investigation showed that Src could interact with and phosphorylate STAT3. Src could also promote HIF1α degradation through activating GSK3ß. Our study reveals that Siglec-G orchestrates TLR-induced inflammation, which outlines that blocking Siglec-G or activating Src may be a promising strategy for both acute and chronic inflammatory diseases.


Assuntos
Inflamação/imunologia , Lectinas/deficiência , Receptores de Antígenos de Linfócitos B/deficiência , Sepse/imunologia , Quinases da Família src/metabolismo , Animais , Citocinas/metabolismo , Ativação Enzimática , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-10/metabolismo , Lectinas/fisiologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Receptores de Antígenos de Linfócitos B/fisiologia , Proteínas Tirosina Fosfatases Contendo o Domínio SH2/metabolismo , Fator de Transcrição STAT3/metabolismo , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico , Transdução de Sinais , Receptores Toll-Like/metabolismo
12.
Microb Pathog ; 137: 103789, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31605759

RESUMO

Vibrio vulnificus is a food-borne marine pathogen that causes both life-threatening primary septicemia and necrotizing wound infections which accompany severe inflammation. Cytolysin is a very powerful virulence factor of V. vulnificus and is one of the likely candidates in the pathogenesis of V. vulnificus infections. However, the pathogenetic roles of cytolysin in V. vulnificus-induced inflammation are not well understood. In this study, we used the recombinant protein Vibrio vulnificus cytolysin (VVC) to demonstrate that VVC can induce inflammatory responses in RAW264.7 macrophages. Low dose (<5 µg/ml) VVC had no impact on cell viability and induced pro-inflammatory cytokines production in RAW264.7 macrophages such as IL-6 and TNF-α. Moreover, VVC induced p65, p38, ERK1/2, and AKT phosphorylation in RAW264.7 macrophages. We further demonstrated that BAPTA-AM, a specific intracellular calcium chelator, inhibited VVC-induced inflammatory responses including pro-inflammatory cytokines production and inflammatory signaling activation in RAW264.7 macrophages. In addition, VVC primed rather than actived NLRP3 inflammasome in RAW264.7 macrophages. To determine whether VVC have a direct inflammatory effect on the host, we examined the effects of VVC injected into the skin of mice. VVC stimulated a significant induction of mRNAs for the pro-inflammatory cytokine IL-6 and inflammatory chemokines such as MCP-1 and IP-10. Histology data also showed that VVC caused inflammatory responses in the skin of mice. Collectively, our findings indicated that VVC induced inflammatory responses in RAW264.7 macrophages and in vivo and suggested the possibility of targeting VVC as a strategy for the clinical management of V. vulnificus-induced inflammatory responses.


Assuntos
Sinalização do Cálcio , Macrófagos/imunologia , Perforina/imunologia , Vibrioses/imunologia , Vibrio vulnificus/imunologia , Animais , Cálcio/imunologia , Feminino , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Células RAW 264.7 , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Vibrioses/genética , Vibrioses/microbiologia , Vibrio vulnificus/genética
13.
Proc Natl Acad Sci U S A ; 116(26): 12958-12963, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31189603

RESUMO

Macrophages can internalize the invading pathogens by raft/caveolae and/or clathrin-dependent endocytosis and elicit an immune response against infection. However, the molecular mechanism for macrophage endocytosis remains elusive. Here we report that LAPF (lysosome-associated and apoptosis-inducing protein containing PH and FYVE domains) is required for caveolae-mediated endocytosis. Lapf-deficient macrophages have impaired capacity to endocytose and eliminate bacteria. Macrophage-specific Lapf-deficient mice are more susceptible to Escherichia coli (E. coli) infection with higher bacterial loads. Moreover, Lapf deficiency impairs TLR4 endocytosis, resulting in attenuated production of TLR-triggered proinflammatory cytokines. LAPF is localized to early endosomes and interacts with caveolin-1. Phosphorylation of LAPF by the tyrosine kinase Src is required for LAPF-Src-Caveolin complex formation and endocytosis and elimination of bacteria. Collectively, our work demonstrates that LAPF is critical for endocytosis of bacteria and induction of inflammatory responses, suggesting that LAPF and Src could be potential targets for the control of infectious diseases.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Caveolina 1/metabolismo , Endocitose/imunologia , Infecções por Escherichia coli/imunologia , Macrófagos/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Caveolina 1/imunologia , Linhagem Celular , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Endossomos/imunologia , Endossomos/metabolismo , Endossomos/microbiologia , Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Imunidade Inata , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Knockout , Cultura Primária de Células , Quinases da Família src/imunologia , Quinases da Família src/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-29868495

RESUMO

N-Acyl Homoserine Lactones (N-AHLs) are an important group of small quorum-sensing molecules generated and released into the surroundings by Gram-negative bacteria. N-AHLs play a crucial role in various infection-related biological processes of marine Vibrio species, including survival, colonization, invasion, and pathogenesis. With the increasing problem of antibiotic abuse and subsequently the emergence of drug-resistant bacteria, studies on AHLs are therefore expected to bring potential new breakthroughs for the prevention and treatment of Vibrio infections. This article starts from AHLs generation in marine Vibrio, and then discusses the advantages, disadvantages, and trends in the future development of various detection methods for AHLs characterization. In addition to a detailed classification of the various marine Vibrio-derived AHL types that have been reported over the years, the regulatory mechanisms of AHLs and their roles in marine Vibrio biofilms, pathogenicity and interaction with host cells are also highlighted. Intervention measures for AHLs in different stages are systematically reviewed, and the prospects of their future development and application are examined.


Assuntos
Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/metabolismo , Percepção de Quorum/fisiologia , Vibrio/fisiologia , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Biofilmes , Hidrolases de Éster Carboxílico/biossíntese , Interações Hospedeiro-Patógeno , Humanos , Oceanos e Mares , Vibrio/patogenicidade , Vibrioses/microbiologia
15.
J Autoimmun ; 81: 120-129, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28461090

RESUMO

The induction and persistence of a hypo-inflammatory and immunosuppressive state in severe sepsis is commonly associated with increased risks of secondary infections and mortality. Toll-like receptor (TLR)-triggered inflammatory response of macrophages/monocytes plays an important role in determining the outcome of hyper-inflammation during the acute phase and the hypo-inflammation during immunosuppressive phase of sepsis. However, the mechanisms for controlling hypo-inflammatory response in endotoxin tolerant macrophages remain to be fully understood. Considering that metabolic control of inflammation is an emerging field and the balance between AMP/ATP and oxidized NAD+/reduced NADH is associated with inflammation and metabolism, we analyzed the level of NAD+ in TLR-triggered innate inflammatory response, and found that the decreased level of NAD+ was significantly related to the increased inflammatory cytokine production both in vivo and in vitro. By screening the expression and function of NAD+ dependent type III deacetylase Sirtuin family members, we found that SIRT5 and SIRT1/2 had opposite expression patterns and functions in macrophages. SIRT5 deficiency decreased TLR-triggered inflammation in both acute and immunosuppressive phases of sepsis. Interestingly, cytoplasmic SIRT5 counteracted the inhibitory effects of SIRT2 and enhanced the innate inflammatory responses in macrophages and even in endotoxin-tolerant macrophages by promoting acetylation of p65 and activation of NF-κB pathway. Mechanistically, SIRT5 competed with SIRT2 to interact with NF-κB p65, in a deacetylase activity-independent way, to block the deacetylation of p65 by SIRT2, which consequently led to increased acetylation of p65 and the activation of NF-κB pathway and its downstream cytokines. Our study discovered the new functions of different Sirtuin members in sepsis, indicating that targeting of Sirtuin family members at different sepsis phases can be helpful to precisely control the progression of sepsis.


Assuntos
Endotoxinas/imunologia , Tolerância Imunológica , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , NAD/metabolismo , Sirtuínas/metabolismo , Fator de Transcrição RelA/metabolismo , Acetilação , Animais , Linhagem Celular , Citocinas/metabolismo , Expressão Gênica , Genótipo , Humanos , Imunidade Inata , Imunomodulação , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout
16.
Sci Rep ; 6: 26252, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27188220

RESUMO

Interleukin-10 (IL-10) plays a central role in regulation of intestinal mucosal homeostasis and prevention of inflammatory bowel disease (IBD). We previously reported that CD11b(hi) regulatory dendritic cells (DCs) can produce more IL-10, and CD11b can negatively regulate Toll-like receptors (TLRs)-induced inflammatory responses in macrophages. However whether CD11b and its signaling can control autoimmunity via IL-10 production remains unclear. Here we found that CD11b deficient (Itgam(-/-)) mice were more susceptible to dextran sulfate sodium (DSS)-induced colitis, with more tumor necrosis factor α (TNF-α) while less IL-10 production. CD11b inhibited nuclear factor-kappa B (NF-κB) while promoted activator protein 1 (AP-1) activation through activating sarcoma oncogene (Src), leading to decreased TNF-α while increased IL-10 production. Src interacted with and promoted c-casitas B lineage lymphoma proto-oncogene (c-Cbl)-mediated degradation of the inhibitory subunit p85 of phosphatidylinositol 3-kinase (PI3K). Importantly, Src inhibitor dasatinib aggravated DSS-induced colitis by decreasing IL-10 while increasing TNF-α in vivo. Therefore, CD11b promotes IL-10 production by activating Src-Akt signal pathway. An axis of CD11b-Src pathway is important in balancing homeostasis of TLR-induced pro-inflammatory and anti-inflammatory responses.


Assuntos
Antígeno CD11b/genética , Colite/genética , Interleucina-10/biossíntese , Animais , Colite/induzido quimicamente , Sulfato de Dextrana , Feminino , Interleucina-10/genética , Masculino , Camundongos Mutantes , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...