Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-37302095

RESUMO

BACKGROUND: Toll-like receptor 9 (TLR9), located in the endosomal compartment, is known to play a role in inflammation by recognizing oligonucleotides that contain CpG motive (CpG-ODN). Signaling by TLR9 leads to the production of proinflammatory cytokines and can trigger cell death. OBJECTIVE: This study aims to investigate the molecular mechanism of pyroptosis induced by ODN1826 in the mouse macrophage cell line (Raw264.7). METHODS: The protein expression and the amount of lactate dehydrogenase (LDH) of ODN1826-treated cells were determined by immunoblotting and LDH assay, respectively. In addition, the level of cytokine production was observed by ELISA assay and the ROS production was determined by flow cytometry. RESULTS: Our results showed that ODN1826 induced pyroptosis as judged by LDH releases. Furthermore, caspase-11 and gasdermin D activation, which are the key molecules in pyroptosis, were also observed in ODN1826-activated cells. Moreover, we also demonstrated that Reactive Oxygen Species (ROS) production by ODN1826 is essential for caspase-11 activation and gasdermin D release, which leads to pyroptosis. CONCLUSIONS: ODN1826 induces pyroptosis in Raw264.7 cells via caspase-11 and GSDMD activation. Moreover, the production of ROS by this ligand plays an essential role in the regulation of caspase-11 and GSDMD activation, which then controls pyroptosis in TLR9 activation.

2.
Artigo em Inglês | MEDLINE | ID: mdl-36278779

RESUMO

BACKGROUND: NLRP12 has been shown to play an essential role as a negative regulator in several bacterial infection. OBJECTIVE: The purpose of this study is to elucidate the role of NLRP12 in B. pseudomallei-infected RAW264.7 macrophages. METHODS: The protein expression and the level of TNF-α production were determined by immunoblotting and ELISA assay, respectively. RESULTS: The results demonstrated that unlike the LPS-mutant strain which lacks O antigenic polysaccharide, the wild-type B. pseudomallei was able to upregulate NLRP12 protein expression in RAW264.7 macrophages. NLRP12 expression also correlated with the suppression of TNF-α production as demonstrated in wild-type B. pseudomallei-infected Nlrp12-depleted macrophages when compared to that of the control siRNA-transfected cells. The expression of NLRP12 was also inhibited in cytochalasin D treated cells. CONCLUSIONS: Our findings showed that wild-type B. pseudomallei can activate NLRP12 expression leading to the suppression of TNF-α production. It is possible that the regulation of NLRP12 may contribute to the pathogenesis of B. pseudomallei infection in melioidosis patients.

3.
Infect Immun ; 88(3)2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31818963

RESUMO

Melioidosis is an infectious disease with a high mortality rate responsible for community-acquired sepsis in Southeast Asia and Northern Australia. The causative agent of this disease is Burkholderia pseudomallei, a Gram-negative bacterium that resides in soil and contaminated natural water. After entering into host cells, the bacteria escape into the cytoplasm, which has numerous cytosolic sensors, including the noncanonical inflammatory caspases. Although the noncanonical inflammasome (caspase-11) has been investigated in a murine model of B. pseudomallei infection, its role in humans, particularly in lung epithelial cells, remains unknown. We, therefore, investigated the function of caspase-4 (ortholog of murine caspase-11) in intracellular killing of B. pseudomallei The results showed that B. pseudomallei induced caspase-4 activation at 12 h postinfection in human alveolar epithelial A549 cells. The number of intracellular B. pseudomallei bacteria was increased in the absence of caspase-4, suggesting its function in intracellular bacterial restriction. In contrast, a high level of caspase-4 processing was observed when cells were infected with lipopolysaccharide (LPS) mutant B. pseudomallei The enhanced bacterial clearance in LPS-mutant-infected cells is also correlated with a higher degree of caspase-4 activation. These results highlight the susceptibility of the LPS mutant to caspase-4-mediated intracellular bacterial killing.


Assuntos
Células Epiteliais Alveolares/fisiologia , Burkholderia pseudomallei/patogenicidade , Caspases Iniciadoras/fisiologia , Melioidose/imunologia , Animais , Burkholderia pseudomallei/fisiologia , Melioidose/microbiologia , Camundongos
4.
Inflamm Res ; 68(10): 841-844, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31292668

RESUMO

OBJECTIVE: The aim of the present study is to investigate the participation of NLRP12 in Porphyromonas gingivalis LPS-activated mouse macrophages. METHODS: NLRP12-depleted mouse macrophages were stimulated with P. gingivalis LPS (1 µg/ml.). At indicated time points, the treated cells were lysed and the supernatant from treated cells was collected. Gene and protein expression of NLRP12 and iNOS were determined by RT-PCR and immunoblotting, respectively. The level of TNF-α production in the supernatant of the activated cells was determined by ELISA. RESULTS AND CONCLUSION: NLRP12 was upregulated in response to stimulation with P. gingivalis LPS. In addition, when NLRP12 was depleted in P. gingivalis LPS-treated macrophages, an increase in TNF-α production and iNOS expression were observed when compared to those of the control cells, indicating that NLRP12 downregulates the inflammatory cytokine and antimicrobial molecule production in the macrophages.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Porphyromonas gingivalis , Células RAW 264.7
5.
Proc Natl Acad Sci U S A ; 113(25): E3519-28, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27185909

RESUMO

IFN-γ is a major regulator of immune functions and has been shown to induce liver-stage Plasmodium elimination both in vitro and in vivo. The molecular mechanism responsible for the restriction of liver-stage Plasmodium downstream of IFN-γ remains uncertain, however. Autophagy, a newly described immune defense mechanism, was recently identified as a downstream pathway activated in response to IFN-γ in the control of intracellular infections. We thus hypothesized that the killing of liver-stage malarial parasites by IFN-γ involves autophagy induction. Our results show that whereas IFN-γ treatment of human hepatocytes activates autophagy, the IFN-γ-mediated restriction of liver-stage Plasmodium vivax depends only on the downstream autophagy-related proteins Beclin 1, PI3K, and ATG5, but not on the upstream autophagy-initiating protein ULK1. In addition, IFN-γ enhanced the recruitment of LC3 onto the parasitophorous vacuole membrane (PVM) and increased the colocalization of lysosomal vesicles with P. vivax compartments. Taken together, these data indicate that IFN-γ mediates the control of liver-stage P. vivax by inducing a noncanonical autophagy pathway resembling that of LC3-associated phagocytosis, in which direct decoration of the PVM with LC3 promotes the fusion of P. vivax compartments with lysosomes and subsequent killing of the pathogen. Understanding the hepatocyte response to IFN-γ during Plasmodium infection and the roles of autophagy-related proteins may provide an urgently needed alternative strategy for the elimination of this human malaria.


Assuntos
Fosfatidilinositol 3-Quinases , Plasmodium vivax , Humanos , Fígado/parasitologia , Malária/imunologia , Malária Vivax
6.
Inflamm Res ; 67(9): 723-726, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29980803

RESUMO

OBJECTIVE: The aim of this study is to investigate the involvement of TLR9 in the regulation of iNOS expression and nitric oxide (NO) production in Porphyromonas gingivalis LPS-treated mouse macrophages. METHODS: Mouse macrophage cell line (RAW264.7) was transfected with siRNAs against TLR9 and then stimulated with P. gingivalis LPS. At indicated time points, the activated cells were lysed. Gene and protein expression of iNOS were determined by RT-PCR and immunoblotting, respectively. The level of nitric oxide (NO) production in the supernatant of the activated cells was determined by Griess reaction assay. RESULTS AND CONCLUSION: Depletion of TLR9 in mouse macrophages demonstrated the markedly decreased iNOS gene and protein expression by P. gingivalis LPS compared to those of the wild-type or control siRNA transfected cells. In consistent with these results, the level of NO secretion was also significantly diminished in TLR9-depleted cells after challenged with P. gingivalis LPS. These results indicate that TLR9 involves in the regulation of the iNOS expression and the NO secretion in P. gingivalis LPS-treated macrophages.


Assuntos
Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Receptor Toll-Like 9/metabolismo , Animais , Indução Enzimática/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Porphyromonas gingivalis , Células RAW 264.7 , RNA Mensageiro/metabolismo
7.
Inflamm Res ; 66(12): 1099-1105, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28889202

RESUMO

INTRODUCTION: We aimed to investigate the involvement of surface TLRs and endosomal TLRs in the regulation of SARM expression by TLR2 ligands (Pam2CSK4 and Pam3CSK4). MATERIALS AND METHODS: Mouse macrophage cell line (RAW264.7) was treated with either Pam2CSK4 or Pam3CSK4 (TLR2 ligands) at a concentration of 100 ng/ml. At indicated time points, the treated cells were lysed. The gene and protein expression of SARM were determined by RT-PCR and immunoblotting, respectively. For silencing of TLR9 function, the cells were transfected with TLR9 siRNAs before stimulation by these two TLR2 ligands RESULTS: The SARM expression was upregulated at both transcriptional and translational levels in time-dependent manner during activation of Pam2CSK4 and Pam3CSK4 in mouse macrophages. Blocking of ligand internalization by cytochalasin D showed interference effect with SARM expression. Moreover, our results also demonstrated that endosomal acidification and TLR9 were required for SARM expression suggesting the essential role of endosomal compartment acidification and TLR9 in regulating SARM expression. CONCLUSION: Our findings suggested the collaboration of TLR2-TLR9 at least in the regulation of SARM expression. However, the underlying mechanism that participated in these two TLRs cooperation is underinvestigated.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Proteínas do Domínio Armadillo/genética , Proteínas do Citoesqueleto/genética , Ligantes , Lipopeptídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , RNA Interferente Pequeno/genética , Receptor 2 Toll-Like/imunologia , Receptor Toll-Like 9/genética
8.
Inflamm Res ; 66(10): 843-853, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28593434

RESUMO

OBJECTIVE: The aim of this study was to investigate the involvement of TLR adaptor molecules, such as TRIF, MyD88, and TBK1 in the induction of iNOS and nitric oxide (NO) production in Pam2CSK4 and Pam3CSK4-treated mouse macrophages. METHOD: Mouse macrophage cell line (RAW264.7) was transfected with trif, myd88, and tbk1 siRNAs before stimulated with Pam2CSK4 and Pam3CSK4. The iNOS gene and protein expression were determined by RT-PCR and immunoblotting, respectively. The NO production was determined by Griess reaction assay. RESULTS: The results showed that the induction of iNOS expression and NO production by Pam2CSK4 and Pam3CSK4 were diminished in tbk1 and myd88-depleted mouse macrophages but not trif-depleted cells. CONCLUSION: These results suggested that the TBK1 and MyD88 molecules were essential for the induction of iNOS expression and NO production by both Pam2CSK4 and Pam3CSK4 via TLR2 signaling.


Assuntos
Lipopeptídeos/imunologia , Fator 88 de Diferenciação Mieloide/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Adaptadoras de Transporte Vesicular/efeitos dos fármacos , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Expressão Gênica/efeitos dos fármacos , Ligantes , Lipopeptídeos/farmacologia , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Proteínas Serina-Treonina Quinases/genética , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Receptor 2 Toll-Like/efeitos dos fármacos , Receptor 2 Toll-Like/metabolismo
9.
Med Microbiol Immunol ; 205(3): 255-60, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26563410

RESUMO

Burkholderia pseudomallei, a gram-negative intracellular bacterium, is a causative agent of melioidosis. The bacterium has been shown to induce the innate immune response, particularly pro-inflammatory cytokine production in several of both mouse and human cell types. In the present study, we investigate host immune response in B. pseudomallei-infected primary human monocytes. We discover that wild-type B. pseudomallei is able to survive and multiply inside the primary human monocytes. In contrast, B. pseudomallei LPS mutant, a less virulent strain, is susceptible to host killing during bacterial infection. Moreover, microarray result showed that wild-type B. pseudomallei but not B. pseudomallei LPS mutant is able to activate gene expression of IL-23 as demonstrated by the up-regulation of p19 and p40 subunit expression. Consistent with gene expression analysis, the secretion of IL-23 analyzed by ELISA also showed that wild-type B. pseudomallei induces a significantly higher level of IL-23 secretion than that of B. pseudomallei LPS mutant. These results implied that IL-23 may be an important cytokine for the innate immune response during B. pseudomallei infection. The regulation of IL-23 production may drive the different host innate immune responses between patients and may relate to the severity of melioidosis.


Assuntos
Burkholderia pseudomallei/imunologia , Imunidade Inata , Subunidade p19 da Interleucina-23/biossíntese , Monócitos/imunologia , Monócitos/microbiologia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Humanos , Subunidade p19 da Interleucina-23/metabolismo , Análise em Microsséries , Viabilidade Microbiana
10.
J Hazard Mater ; 471: 134270, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38640676

RESUMO

Alachlor, a widely used chloroacetanilide herbicide for controlling annual grasses in crops, has been reported to rapidly trigger protein denaturation and aggregation in the eukaryotic model organism Saccharomyces cerevisiae. Therefore, this study aimed to uncover cellular mechanisms involved in preventing alachlor-induced proteotoxicity. The findings reveal that the ubiquitin-proteasome system (UPS) plays a crucial role in eliminating alachlor-denatured proteins by tagging them with polyubiquitin for subsequent proteasomal degradation. Exposure to alachlor rapidly induced an inhibition of proteasome activity by 90 % within 30 min. The molecular docking analysis suggests that this inhibition likely results from the binding of alachlor to ß subunits within the catalytic core of the proteasome. Notably, our data suggest that nascent proteins in the endoplasmic reticulum (ER) are the primary targets of alachlor. Consequently, the unfolded protein response (UPR), responsible for coping with aberrant proteins in the ER, becomes activated within 1 h of alachlor treatment, leading to the splicing of HAC1 mRNA into the active transcription activator Hac1p and the upregulation of UPR gene expression. These findings underscore the critical roles of the protein quality control systems UPS and UPR in mitigating alachlor-induced proteotoxicity by degrading alachlor-denatured proteins and enhancing the protein folding capacity of the ER.


Assuntos
Acetamidas , Retículo Endoplasmático , Herbicidas , Complexo de Endopeptidases do Proteassoma , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Resposta a Proteínas não Dobradas , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Acetamidas/farmacologia , Acetamidas/toxicidade , Herbicidas/toxicidade , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Citosol/metabolismo , Citosol/efeitos dos fármacos , Simulação de Acoplamento Molecular , Estresse Proteotóxico
11.
Infect Immun ; 81(9): 3463-71, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23836818

RESUMO

Burkholderia pseudomallei, the causative agent of melioidosis, evades macrophage killing by suppressing the TRIF-dependent pathway, leading to inhibition of inducible nitric oxide synthase (iNOS) expression. We previously demonstrated that virulent wild-type B. pseudomallei inhibits the TRIF-dependent pathway by upregulating sterile-α- and armadillo motif-containing protein (SARM) and by inhibiting downregulation of signal regulatory protein α (SIRPα); both molecules are negative regulators of Toll-like receptor signaling. In contrast, the less virulent lipopolysaccharide (LPS) mutant of B. pseudomallei is unable to exhibit these features and is susceptible to macrophage killing. However, the functional relationship of these two negative regulators in the evasion of macrophage defense has not been elucidated. We demonstrated here that SIRPα downregulation was observed after inhibition of SARM expression by small interfering RNA in wild-type-infected macrophages, indicating that SIRPα downregulation is regulated by SARM. Furthermore, this downregulation requires activation of the TRIF signaling pathway, as we observed abrogation of SIRPα downregulation as well as restricted bacterial growth in LPS mutant-infected TRIF-depleted macrophages. Although inhibition of SARM expression is correlated to SIRPα downregulation and iNOS upregulation in gamma interferon-activated wild-type-infected macrophages, these phenomena appear to bypass the TRIF-dependent pathway. Similar to live bacteria, the wild-type LPS is able to upregulate SARM and to prevent SIRPα downregulation, implying that the LPS of B. pseudomallei may play a crucial role in regulating the expression of these two negative regulators. Altogether, our findings show a previously unrecognized role of B. pseudomallei-induced SARM in inhibiting SIRPα downregulation-mediated iNOS upregulation, facilitating the ability of the bacterium to multiply in macrophages.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas do Domínio Armadillo/genética , Burkholderia pseudomallei/genética , Proteínas do Citoesqueleto/genética , Macrófagos/metabolismo , Melioidose/genética , Receptores Imunológicos/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Proteínas do Domínio Armadillo/imunologia , Proteínas do Domínio Armadillo/metabolismo , Burkholderia pseudomallei/imunologia , Burkholderia pseudomallei/metabolismo , Proteínas do Citoesqueleto/imunologia , Proteínas do Citoesqueleto/metabolismo , Regulação para Baixo/imunologia , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Melioidose/imunologia , Melioidose/metabolismo , Melioidose/microbiologia , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Regulação para Cima/genética , Regulação para Cima/imunologia
12.
PLoS One ; 18(11): e0292340, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38011122

RESUMO

BACKGROUND: Cleistanthin A (CA), extracted from Phyllanthus taxodiifolius Beille, was previously reported as a potential V-ATPase inhibitor relevant to cancer cell survival. In the present study, ECDD-S16, a derivative of cleistanthin A, was investigated and found to interfere with pyroptosis induction via V-ATPase inhibition. OBJECTIVE: This study examined the ability of ECDD-S16 to inhibit endolysosome acidification leading to the attenuation of pyroptosis in Raw264.7 macrophages activated by both surface and endosomal TLR ligands. METHODS: To elucidate the activity of ECDD-S16 on pyroptosis-induced inflammation, Raw264.7 cells were pretreated with the compound before stimulation with surface and endosomal TLR ligands. The release of lactate dehydrogenase (LDH) was determined by LDH assay. Additionally, the production of cytokines and the expression of pyroptosis markers were examined by ELISA and immunoblotting. Moreover, molecular docking was performed to demonstrate the binding of ECDD-S16 to the vacuolar (V-)ATPase. RESULTS: This study showed that ECDD-S16 could inhibit pyroptosis in Raw264.7 cells activated with surface and endosomal TLR ligands. The attenuation of pyroptosis by ECDD-S16 was due to the impairment of endosome acidification, which also led to decreased Reactive Oxygen Species (ROS) production. Furthermore, molecular docking also showed the possibility of inhibiting endosome acidification by the binding of ECDD-S16 to the vacuolar (V-)ATPase in the region of V0. CONCLUSION: Our findings indicate the potential of ECDD-S16 for inhibiting pyroptosis and prove that vacuolar H+ ATPase is essential for pyroptosis induced by TLR ligands.


Assuntos
ATPases Vacuolares Próton-Translocadoras , Humanos , ATPases Vacuolares Próton-Translocadoras/metabolismo , Piroptose , Simulação de Acoplamento Molecular , Inflamação
13.
Infect Immun ; 80(12): 4223-31, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22988019

RESUMO

The facultative intracellular gram-negative bacterium Burkholderia pseudomallei is the causative agent of melioidosis and is known for its ability to evade the Toll-like receptor (TLR)-mediated innate immune response. Previously it has been demonstrated that this bacterium was able to suppress the MyD88-independent pathway and can survive macrophage intracellular killing. However, the underlying mechanisms responsible for the suppression of this pathway are not fully understood. In the present study, we showed that both living and heat-killed B. pseudomallei bacteria restrict the TLR signaling response, particularly macrophage inducible nitric oxide synthase (iNOS) expression, by preventing downregulation of constitutively expressed signal regulatory protein α (SIRPα) molecule, a known negative regulator of TLR signaling. In contrast, a lipopolysaccharide (LPS) mutant of B. pseudomallei, a less virulent strain, was able to downregulate SIRPα expression in mouse macrophages. However, depletion of constitutively expressed SIRPα was able to induce the gene expression downstream of TLR signaling pathways (particularly the MyD88-independent pathway), such as that of the iNOS gene, leading to enhanced macrophage intracellular killing of B. pseudomallei. Induction of gene expression was consistent with the enhanced degradation pattern of IκBα with SIRPα depletion. Additionally, the downregulation of SIRPα expression with upregulation of iNOS was observed when the macrophages were pretreated with gamma interferon (IFN-γ) prior to the infection, suggesting that the enhanced intracellular killing of bacteria by IFN-γ is associated with the decreased SIRPα expression. Altogether our findings demonstrate that B. pseudomallei evades macrophage intracellular killing by preventing the downregulation of SIRPα that results in the inhibition of gene expression downstream of the MyD88-independent pathway.


Assuntos
Burkholderia pseudomallei/patogenicidade , Regulação da Expressão Gênica/imunologia , Macrófagos , Melioidose/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Receptores Imunológicos/metabolismo , Animais , Burkholderia pseudomallei/crescimento & desenvolvimento , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Evasão da Resposta Imune , Macrófagos/imunologia , Macrófagos/microbiologia , Melioidose/microbiologia , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Receptores Imunológicos/genética , Transdução de Sinais , Receptores Toll-Like/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-36232220

RESUMO

Microglial activation has been found to play a crucial role in various neurological disorders. Proinflammatory substances overproduced by activated microglia, such as cytokines, chemokines, reactive oxygen species, and nitric oxide (NO), can result in neuroinflammation that further exacerbates the course of the diseases. This study aimed to explore the anti-inflammatory effect of the ethyl acetate extract of Pueraria mirifica on microglial activation. Lipopolysaccharide (LPS)-induced inflammation was used as a model to investigate the effects of P. mirifica on HAPI (highly aggressive proliferating immortalized), a rat microglial cell line. Administration of ethyl acetate extract from the tuberous roots of P. mirifica to HAPI cells dose-dependently reduced NO production and iNOS expression induced by LPS. Attenuation of IRF-1 (interferon regulatory factor-1) induction, one of the transcription factors governing iNOS expression, suggested that the inhibitory effect on NO production by the plant extract was at least partially mediated through this transcription factor. In addition, LPS-stimulated mRNA expression of MCP-1 (monocyte chemoattractant protein-1), IL-6 (interleukin-6), and TNF-α (tumor necrosis factor-α) was also suppressed with P. mirifica extract pretreatment. This study indicates that the ethyl acetate extract of P. mirifica could potentially serve as an anti-inflammatory mediator and may be useful in relieving the severity of neurological diseases where microglia play a role.


Assuntos
Lipopolissacarídeos , Pueraria , Acetatos , Animais , Anti-Inflamatórios/farmacologia , Quimiocina CCL2 , Quimiocinas/metabolismo , Citocinas/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Microglia , Óxido Nítrico/metabolismo , Extratos Vegetais/metabolismo , Extratos Vegetais/farmacologia , Pueraria/genética , Pueraria/metabolismo , RNA Mensageiro/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
15.
Microbiol Spectr ; 10(5): e0348822, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36194127

RESUMO

Melioidosis is a serious infectious disease caused by Burkholderia pseudomallei. This bacterium is able to survive and multiply inside the immune cells such as macrophages. It is well established that Toll-like receptors (TLRs), particularly surface TLRs such as TLR2, TLR4, and TLR5, play an essential role in defending against this bacterial infection. However, the involvement of endosomal TLRs in the infection has not been elucidated. In this study, we demonstrated that the number of intracellular bacteria is reduced in TLR9-depleted RAW264.7 cells infected with B. pseudomallei, suggesting that TLR9 is involved in intracellular bacterial killing in macrophages. As several reports have previously demonstrated that pyroptosis is essential for restricting intracellular bacterial killing, particularly in B. pseudomallei infection, we also observed an increased release of cytosolic enzyme lactate dehydrogenase (LDH) in TLR9-depleted cells infected with B. pseudomallei, suggesting TLR9 involvement in pyroptosis in this context. Consistently, the increases in caspase-11 and gasdermind D (GSDMD) activations, which are responsible for the LDH release, were also detected. Moreover, we demonstrated that the increases in pyroptosis and bacterial killing in B. pseudomallei-infected TLR9-depleted cells were due to the augmentation of the IFN-ß, one of the key cytokines known to regulate caspase-11. Altogether, this finding showed that TLR9 suppresses macrophage killing of B. pseudomallei by regulating pyroptosis. This information provides a novel mechanism of TLR9 in the regulation of intracellular bacterial killing by macrophages, which could potentially be leveraged for therapeutic intervention. IMPORTANCE Surface TLRs have been well established to play an essential role in Burkholderia pseudomallei infection. However, the role of endosomal TLRs has not been elucidated. In the present study, we demonstrated that TLR9 plays a crucial role by negatively regulating cytokine production, particularly IFN-ß, a vital cytokine to control pyroptosis via caspase-11 activation. By depletion of TLR9, the percentage of pyroptosis was significantly increased, leading to suppression of intracellular survival in B. pseudomallei-infected macrophages. These findings provide a new role of TLR9 in macrophages.


Assuntos
Burkholderia pseudomallei , Melioidose , Camundongos , Animais , Burkholderia pseudomallei/metabolismo , Receptor Toll-Like 9/metabolismo , Receptor 2 Toll-Like/metabolismo , Piroptose , Receptor 4 Toll-Like/metabolismo , Receptor 5 Toll-Like/metabolismo , Melioidose/metabolismo , Melioidose/microbiologia , Macrófagos , Linhagem Celular , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Citocinas/metabolismo , Caspases/metabolismo , Lactato Desidrogenases/metabolismo
16.
Pathog Dis ; 80(1)2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35038342

RESUMO

Mycobacterium tuberculosis utilizes several mechanisms to block phagosome-lysosome fusion to evade host cell restriction. However, induction of host cell autophagy by starvation was shown to overcome this block, resulting in enhanced lysosomal delivery to mycobacterial phagosomes and the killing of the M. tuberculosis reference strain H37Rv. Nevertheless, our previous studies found that strains belonging to the M. tuberculosis Beijing genotype can resist starvation-induced autophagic elimination, though the mycobacterial factors involved remain unclear. In this study, we showed that KatG expression is upregulated in the autophagy-resistant M. tuberculosis Beijing strain (BJN) during autophagy induction by the starvation of host macrophages, while such increase was not observed in the H37Rv. KatG depletion using the CRISPR-dCas9 interference system in the BJN resulted in increased lysosomal delivery to its phagosome and decreased its survival upon autophagy induction by starvation. As KatG functions by catabolizing ROS, we determined the source of ROS contributing to the starvation-induced autophagic elimination of mycobacteria. Using siRNA-mediated knockdown, we found that Superoxide dismutase 2, which generates mitochondrial ROS but not NADPH oxidase 2, is important for the starvation-induced lysosomal delivery to mycobacterial phagosomes. Taken together, these findings showed that KatG is vital for the BJN to evade starvation-induced autophagic restriction.


Assuntos
Mycobacterium tuberculosis , Autofagia/genética , Pequim , Lisossomos/microbiologia , Mycobacterium tuberculosis/genética , Fagossomos/metabolismo
18.
Sci Rep ; 11(1): 4342, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33619301

RESUMO

Induction of host cell autophagy by starvation was shown to enhance lysosomal delivery to mycobacterial phagosomes, resulting in the restriction of Mycobacterium tuberculosis reference strain H37Rv. Our previous study showed that strains belonging to M. tuberculosis Beijing genotype resisted starvation-induced autophagic elimination but the factors involved remained unclear. Here, we conducted RNA-Seq of macrophages infected with the autophagy-resistant Beijing strain (BJN) compared to macrophages infected with H37Rv upon autophagy induction by starvation. Results identified several genes uniquely upregulated in BJN-infected macrophages but not in H37Rv-infected cells, including those encoding Kxd1 and Plekhm2, which function in lysosome positioning towards the cell periphery. Unlike H37Rv, BJN suppressed enhanced lysosome positioning towards the perinuclear region and lysosomal delivery to its phagosome upon autophagy induction by starvation, while depletion of Kxd1 and Plekhm2 reverted such effects, resulting in restriction of BJN intracellular survival upon autophagy induction by starvation. Taken together, these data indicated that Kxd1 and Plekhm2 are important for the BJN strain to suppress lysosome positioning towards the perinuclear region and lysosomal delivery into its phagosome during autophagy induction by starvation to evade starvation-induced autophagic restriction.


Assuntos
Autofagia , Interações Hospedeiro-Patógeno , Lisossomos/metabolismo , Lisossomos/microbiologia , Mycobacterium tuberculosis/fisiologia , Tuberculose/metabolismo , Tuberculose/microbiologia , Autofagia/genética , Proteínas de Transporte/genética , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Ontologia Genética , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Anotação de Sequência Molecular , Transcriptoma , Tuberculose/genética , Tuberculose/imunologia
19.
Biochem Biophys Res Commun ; 398(4): 752-8, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20627090

RESUMO

Information on the immune response against H5N1 within the lung is lacking. Here we describe the sustained antiviral immune responses, as indicated by the expression of MxA protein and IFN-alpha mRNA, in autopsy lung tissue from an H5N1-infected patient. H5N1 infection of primary bronchial/tracheal epithelial cells and lung microvascular endothelial cells induced IP-10, and also up-regulated the retinoic acid-inducible gene-I (RIG-I). Down-regulation of RIG-I gene expression decreased IP-10 response. Co-culturing of H5N1-infected pulmonary cells with TNF-alpha led to synergistically enhanced production of IP-10. In the absence of viral infection, TNF-alpha and IFN-alpha also synergistically enhanced IP-10 response. Methylprednisolone showed only a partial inhibitory effect on this chemokine response. Our findings strongly suggest that both the H5N1 virus and the locally produced antiviral cytokines; IFN-alpha and TNF-alpha may have an important role in inducing IP-10 hyperresponse, leading to inflammatory damage in infected lung.


Assuntos
Quimiocina CXCL10/biossíntese , Virus da Influenza A Subtipo H5N1 , Influenza Humana/imunologia , Pulmão/imunologia , Pulmão/virologia , Pneumonia Viral/imunologia , Células Cultivadas , Quimiocina CXCL10/antagonistas & inibidores , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Proteínas de Ligação ao GTP/biossíntese , Humanos , Interferon-alfa/biossíntese , Interferon-alfa/farmacologia , Metilprednisolona/farmacologia , Proteínas de Resistência a Myxovirus , Receptores Imunológicos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
20.
BMC Immunol ; 10: 20, 2009 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-19397822

RESUMO

BACKGROUND: Burkholderia pseudomallei (Bp) is a category B biothreat organism that causes a potentially fatal disease in humans and animals, namely melioidosis. Burkholderia thailandensis (Bt) is another naturally occurring species that is very closely related to Bp. However, despite this closely related genotype, Bt is considered avirulent as it does not cause the disease. In the present study, we compared the growth kinetics of B. pseudomallei strain 844 (Bp-844) in human monocyte-derived dendritic cells (MoDCs) and macrophages (Mphis), as well as its ability to stimulate host cell responses with those of B. thailandensis strain UE5 (Bt-UE5). RESULTS: Primary human MoDCs and Mphis were infected with Bp-844 and its intracellular growth kinetics and ability to induce host cell responses were evaluated. The results were compared with those obtained using the Bt-UE5. In human MoDCs, both bacteria were similar in respect to their ability to survive and replicate intracellularly, induce upregulation of costimulatory molecules and cytokines and bias T helper cell differentiation toward a Th1 phenotype. By contrast, the two bacteria exhibited different growth kinetics in human Mphis, where the intracellular growth of Bt-UE5, but not Bp-844, was significantly suppressed. Moreover, the ability of Mphis to kill Bp-844 was markedly enhanced following stimulation with IFN-gamma. CONCLUSION: The data presented showed that while both strains were similar in their ability to survive and replicate in human MoDCs, only Bp-844 could readily replicate in human Mphis. Both bacteria induced similar host cellular responses, particularly with regard to their ability to bias T cell differentiation toward a Th1 phenotype.


Assuntos
Infecções por Burkholderia/microbiologia , Burkholderia pseudomallei/fisiologia , Citotoxicidade Imunológica , Células Dendríticas/microbiologia , Macrófagos/microbiologia , Infecções por Burkholderia/imunologia , Burkholderia pseudomallei/patogenicidade , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Citoplasma , Células Dendríticas/imunologia , Células Dendríticas/patologia , Humanos , Interferon gama/metabolismo , Ativação Linfocitária , Macrófagos/imunologia , Macrófagos/patologia , Especificidade da Espécie , Células Th1/imunologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA