Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Transl Med ; 21(1): 898, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38082290

RESUMO

BACKGROUND: Early microbial exposure is associate with protective allergic asthma. We have previously demonstrated that Streptococcus pneumoniae aminopeptidase N (PepN), one of the pneumococcal components, inhibits ovalbumin (OVA) -induced airway inflammation in murine models of allergic asthma, but the underlying mechanism was incompletely determined. METHODS: BALB/c mice were pretreated with the PepN protein and exposed intranasally to HDM allergen. The anti-inflammatory mechanisms were investigated using depletion and adoptive transfer experiments as well as transcriptome analysis and isolated lung CD11chigh macrophages. RESULTS: We found pretreatment of mice with PepN promoted the proliferation of lung-resident F4/80+CD11chigh macrophages in situ but also mobilized bone marrow monocytes to infiltrate lung tissue that were then transformed into CD11high macrophages. PepN pre-programmed the macrophages during maturation to an anti-inflammatory phenotype by shaping the metabolic preference for oxidative phosphorylation (OXPHOS) and also inhibited the inflammatory response of macrophages by activating AMP-activated protein kinase. Furthermore, PepN treated macrophages also exhibited high-level costimulatory signaling molecules which directed the differentiation into Treg. CONCLUSION: Our results demonstrated that the expansion of CD11chigh macrophages in lungs and the OXPHOS metabolic bias of macrophages are associated with reduced allergic airway inflammation after PepN exposure, which paves the way for its application in preventing allergic asthma.


Assuntos
Asma , Pneumonia , Camundongos , Animais , Streptococcus pneumoniae/metabolismo , Antígenos CD13 , Citocinas/metabolismo , Asma/metabolismo , Pulmão/metabolismo , Inflamação/prevenção & controle , Macrófagos/metabolismo , Anti-Inflamatórios , Fenótipo , Ovalbumina , Modelos Animais de Doenças , Camundongos Endogâmicos BALB C
2.
Virulence ; : 2283898, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37964595

RESUMO

The detoxified pneumolysin derivative ΔA146Ply has been proven to have a direct anti-triple negative breast cancer effect by our group, but its work model remains unclear. In this study, we focused on its ability to inhibit triple-negative breast cancer metastasis. We found that ΔA146Ply suppressed the migration and invasion of triple-negative breast cancer cells by activating mannose receptor and toll-like receptor 4. Their activation triggers the activation of the mammalian target of rapamycin signaling, sequentially leading to autophagy, transforming growth factor-ß1, and epithelial-mesenchymal transition inhibition. Furthermore, the combination of doxorubicin and ΔA146Ply significantly inhibited triple-negative breast cancer progression and prolonged survival in tumor-bearing mice. Taken together, our study provides an alternative microbiome-based mannose receptor-targeted therapy for triple-negative breast cancer and a novel theoretical and experimental basis for the downstream signaling pathway of the mannose receptor.

3.
Am J Pathol ; 193(5): 567-578, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37080661

RESUMO

Protein kinase CK2 is a constitutively active and ubiquitously expressed serine/threonine kinase that is closely associated with various types of cancers, autoimmune disorders, and inflammation. However, the role of CK2 in psoriasis remains unknown. Herein, the study indicated elevated expression of CK2 in skin lesions from patients with psoriasis and from psoriasis-like mice. In the psoriasis-like mouse model, the CK2-specific inhibitor CX-4945 ameliorated imiquimod-induced psoriasis symptoms with reduced proliferation, abnormal differentiation, inflammatory cytokine production (especially IL-17A) of keratinocytes, and infiltration of γδ T cells. In in vitro studies, exogenous CK2 promoted hyperproliferation and abnormal differentiation of human keratinocytes, which were reversed by the suppression of CK2 with CX-4945 or siRNA. Furthermore, knockdown of CK2 reduced IL-17A expression and abolished IL-17A-induced proliferation and inflammatory cytokine expression in keratinocytes. Interestingly, IL-17A increased the expression of CK2 in keratinocytes, thereby establishing a positive feedback loop. In addition, suppression of CK2 inhibited the activation of STAT3 and Akt signaling pathways in human keratinocytes and imiquimod-induced psoriatic lesions of mice. These findings indicate that a highly expressed CK2 level in the skin lesions is required in the development of psoriasis by promoting epidermal hyperplasia, abnormal differentiation, and inflammatory response via regulation of the STAT3 and Akt signaling pathways. CK2 may be a target for the treatment of psoriasis.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Psoríase , Animais , Humanos , Camundongos , Caseína Quinase II/metabolismo , Diferenciação Celular , Proliferação de Células , Citocinas/metabolismo , Imiquimode/efeitos adversos , Interleucina-17/metabolismo , Queratinócitos/patologia , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-akt/metabolismo , Psoríase/induzido quimicamente , Pele/metabolismo , Fator de Transcrição STAT3/metabolismo
4.
Immun Inflamm Dis ; 11(2): e779, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36840485

RESUMO

Efferocytosis can resolve airway inflammation and enhance airway tolerance in allergic asthma. While previous work has reported that progranulin (PGRN) regulated macrophage efferocytosis, but it is unclear whether PGRN-mediated efferocytosis is associated with asthma. Here, we found that in an ovalbumin (OVA)-induced allergic asthma model, the airway inflammation was suppressed and the apoptosis in lung tissues was ameliorated in PGRN-deficient mice. In contrast, PGRN knockdown in human bronchial epithelial cells increased apoptosis in vitro. Furthermore, PGRN-deficient macrophages had significantly stronger efferocytosis ability than wild type (WT) macrophages both in vitro and in vivo. PGRN-deficient peritoneal macrophages (PMs) exhibited increased expression of genes associated with efferocytosis including milk fat globule-epidermal growth factor 8 (MFG-E8), peroxisome proliferator-activated receptor gamma (PPAR-γ) and sirtuin1 (SIRT1) and increased capacity to produce the anti-inflammatory mediator interleukin (IL)-10 during efferocytosis. GW9662, the inhibitor of PPAR-γ, abolished increased efferocytosis and MFG-E8 expression in PGRN-deficient PMs suggesting that PGRN deficiency enhanced MFG-E8-mediated efferocytosis through PPAR-γ. Correspondingly, efferocytosis genes were increased in the lungs of OVA-induced PGRN-deficient mice. GW9662 treatment reduced MFG-E8 expression but did not significantly affect airway inflammation. Our results demonstrated that PGRN deficiency enhanced efferocytosis via the PPAR-γ/MFG-E8 pathway and this may be one of the reasons PGRN deficiency results in inhibition of airway inflammation in allergic asthma.


Assuntos
Asma , PPAR gama , Camundongos , Animais , Humanos , PPAR gama/metabolismo , Progranulinas , Fator VIII/metabolismo , Macrófagos/metabolismo , Asma/metabolismo , Inflamação/metabolismo
5.
Microbiol Spectr ; 10(2): e0204921, 2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35297653

RESUMO

Streptococcus pneumoniae is a leading bacterial cause of a wide range of infections, and pneumococcal pneumosepsis causes high mortality in hosts infected with antibiotic-resistant strains and those who cannot resolve ongoing inflammation. The factors which influence the development and outcome of pneumosepsis are currently unclear. IL-6 is critical for maintaining immune homeostasis, and we determined that this cytokine is also essential for resisting pneumosepsis, as it inhibits macrophage pyroptosis and pyroptosis-related inflammation injury in the lung. IL-6 affected infection outcomes in mice and exerted a protective role, primarily via macrophages. We further found that IL-6 deficiency led to increased lung macrophage death and aggravated lung inflammation, and that exogenous administration of IL-6 protein could decrease macrophage death and alleviate lung tissue inflammation. IL-6 also protected Streptococcus pneumoniae-induced lung macrophage death and lung inflammation injury by inhibiting gasdermin E (GSDME)- and gasdermin D (GSDMD)-mediated pyroptosis. Together, these data reveal a novel mechanism for the development of pneumosepsis and the critical protective role of IL-6. These findings may assist in the early identification and treatment of pneumococcal pneumosepsis. IMPORTANCE Pneumococcal pneumonia has been a significant cause of morbidity and mortality throughout human history. Failing to control pneumococcal pneumonia and resolve ongoing inflammation in a host can cause sepsis, namely pneumococcal pneumosepsis, and death ensues. Few theories have suggested an optimally therapeutic option for this infectious disease. The interleukin-6 (IL-6, a cytokine featuring pleiotropic activity) theory, proposed here, implies that IL-6 acts as a protector against pneumococcal pneumosepsis. IL-6 prevents lung macrophage death and lung inflammation injury by inhibiting a caspase-3-GSDME-mediated switch from apoptosis to pyroptosis and inhibiting caspase-1-GSDMD-mediated classic pyroptosis during pneumococcal pneumosepsis. Thus, IL-6 is an important determinant for controlling bacterial invasion and a homeostatic coordinator of pneumococcal pneumosepsis. This study clarifies a novel mechanism of occurrence and development of pneumonia and secondary sepsis following a Streptococcus pneumoniae infection. It is important for the early identification and treatment of pneumococcal pneumosepsis.


Assuntos
Pneumonia Pneumocócica , Sepse , Animais , Citocinas/metabolismo , Inflamação/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão , Macrófagos/metabolismo , Camundongos , Pneumonia Pneumocócica/metabolismo , Piroptose , Streptococcus pneumoniae
6.
EBioMedicine ; 72: 103607, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34628355

RESUMO

BACKGROUND: The initial clinical manifestations and abdominal imaging findings of neonates with necrotising enterocolitis (NEC) and food protein-induced enterocolitis syndrome (FPIES) are sometimes similar; however, their prognosis and therapies are different. We aimed to evaluate the utility of interleukin (IL)-27 as a differentiation marker between NEC and highly suspected early onset (HSEO)-FPIES. METHODS: All samples used in this study were obtained from the neonatal diagnosis centre of Children's Hospital of Chongqing Medical University. In the case-control study, neonates with NEC (n = 13), HSEO-FPIES (n = 9), and jaundice (control, n = 8) were enroled to determine the serum IL-27 levels using commercial enzyme-linked immunosorbent assay (ELISA) kits. In the validation cohort study, the NEC (n = 87), HSEO-FPIES (n = 62), and jaundice (control, n = 54) groups were included to analyse the diagnostic efficiency of IL-27 for discriminating between NEC and HSEO-FPIES using a receiver operating characteristic (ROC) curve. FINDINGS: In the case-control study, IL-27 levels were higher in the NEC group than in the HSEO-FPIES group (p = 0·005). In the cohort study, the area under the ROC curve (AUC) of IL-27 for differentiating NEC from HSEO-FPIES was 0·878, which was higher than the AUCs of IL-6 (0·761), C-reactive protein (0·800), white blood cell count (0·637), neutrophils (0·765), lymphocytes (0·782), neutrophil to lymphocyte ratio (0·781), and platelet count (0·729). INTERPRETATION: Serum IL-27 is a novel biomarker that may potentially discriminate NEC from HSEO-FPIES in neonates. FUNDING: None.


Assuntos
Biomarcadores/metabolismo , Enterocolite Necrosante/metabolismo , Hipersensibilidade Alimentar/metabolismo , Interleucinas/metabolismo , Contagem de Células Sanguíneas/métodos , Proteína C-Reativa/metabolismo , Estudos de Casos e Controles , Feminino , Humanos , Recém-Nascido , Doenças do Recém-Nascido/metabolismo , Interleucina-6/metabolismo , Masculino , Prognóstico , Estudos Prospectivos , Curva ROC , Síndrome
7.
Mol Ther Oncolytics ; 18: 247-261, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32728613

RESUMO

Increasing evidence demonstrates that microorganisms and their products can modulate host responses to cancer therapies and contribute to tumor shrinkage via various mechanisms, including intracellular signaling pathways modulation and immunomodulation. Detoxified pneumolysin derivative ΔA146Ply is a pneumolysin mutant lacking hemolytic activity. To determine the antitumor activity of ΔA146Ply, the combination of ΔA146Ply and berbamine, a well-established antitumor agent, was used for breast cancer therapy, especially for triple-negative breast cancer. The efficacy of the combination therapy was evaluated in vitro using four breast cancer cell lines and in vivo using a synergistic mouse tumor model. We demonstrated that in vitro, the combination therapy significantly inhibited cancer cell proliferation, promoted cancer cell apoptosis, caused cancer cell-cycle arrest, and suppressed cancer cell migration and invasion. In vivo, the combination therapy significantly suppressed tumor growth and prolonged the median survival time of tumor-bearing mice partially through inhibiting tumor cell proliferation, promoting tumor cell apoptosis, and activating systemic antitumor immune responses. The safety analysis demonstrated that the combination therapy showed no obvious liver and kidney toxicity to tumor-bearing mice. Our study provides a new treatment option for breast cancer and lays the experimental basis for the development of ΔA146Ply as an antitumor agent.

8.
Can J Microbiol ; 66(7): 435-445, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32191844

RESUMO

Pneumolysin (Ply) is a major virulence factor of Streptococcus pneumoniae. Ply-induced interferon-ß (IFN-ß) expression in host macrophages has been shown to be due to the accumulation of mitochondrial deoxyribonucleic acid (mtDNA) in the cytoplasm during S. pneumoniae infection. Our findings extend this work to show human bronchial epithelial cells that reside at the interface of inflammatory injury, BEAS-2B, adapt to local cues by altering mitochondrial states and releasing excess mtDNA. The results in this research showed that purified Ply induced the expression of IFN-ß in human epithelial cells, which was accompanied by mitochondrial damage both in vivo and in vitro. The observations also were supported by the increased mtDNA concentrations in the bronchial lavage fluid of mice infected with S. pneumoniae. In summary, our study demonstrated that Ply triggered the production of IFN-ß in epithelial cells, and this response was mediated by mtDNA released from Ply-damaged mitochondria. It displayed an impressive modulation of IFN-ß response to S. pneumoniae in epithelial cells.


Assuntos
Citosol/metabolismo , DNA Mitocondrial/metabolismo , Interferon beta/metabolismo , Mitocôndrias/efeitos dos fármacos , Estreptolisinas/toxicidade , Animais , Proteínas de Bactérias/toxicidade , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/microbiologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Streptococcus pneumoniae/patogenicidade
9.
J Microbiol ; 58(4): 330-339, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32103444

RESUMO

Streptococcus pneumoniae is a Gram-positive pathogen with high morbidity and mortality globally but some of its pathogenesis remains unknown. Previous research has provided evidence that aminopeptidase N (PepN) is most likely a virulence factor of S. pneumoniae. However, its role in S. pneumoniae virulence and its interaction with the host remains to be confirmed. We generated a pepN gene deficient mutant strain and found that its virulence for mice was significantly attenuated as were in vitro adhesion and invasion of host cells. The PepN protein could induce a strong innate immune response in vivo and in vitro and induced secretion of IL-6 and TNF-α by primary peritoneal macrophages via the rapid phosphorylation of MAPK and PI3K/AKT signaling pathways and this was confirmed using specific pathway inhibitors. In conclusion, PepN is a novel virulence factor that is essential for the virulence of S. pneumoniae and induces host innate immunity via MAPK and PI3K/AKT signaling.


Assuntos
Aminopeptidases/fisiologia , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/patogenicidade , Fatores de Virulência/fisiologia , Células A549 , Aminopeptidases/genética , Animais , Proteínas de Bactérias/fisiologia , Adesão Celular , Feminino , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata , Sistema de Sinalização das MAP Quinases , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Virulência
10.
FEBS J ; 286(23): 4754-4768, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31315154

RESUMO

Pneumolysin (Ply), a major virulence factor of Streptococcus pneumoniae (S. pn), affects the immunity of host cells during infection. It has been reported that Ply is involved in S. pn standard strain D39-induced interferon-ß (IFN-ß) expression; however, other findings suggest that recombinant Ply protein is incapable of triggering IFN-ß expression. Here, we demonstrated that purified Ply was capable of initiating oxidative damage to mitochondria, resulting in the subsequent release of mitochondrial deoxyribonucleic acid (mtDNA), which mediated IFN-ß expression in macrophages. Importantly, we determined that IFN-ß expression was regulated by stimulator of interferon genes (STING) signaling in response to Ply. In conclusion, our study identified that IFN-ß production was triggered by Ply in macrophages and mtDNA released from Ply-damaged mitochondria mediated this process, through the STING pathway. This is a novel mechanism by which S. pn modulates type I IFN response in macrophages.


Assuntos
Citosol/metabolismo , DNA Mitocondrial/metabolismo , Interferon Tipo I/metabolismo , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Estreptolisinas/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Transporte Biológico/fisiologia , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Streptococcus pneumoniae/metabolismo
11.
Front Immunol ; 9: 2952, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619312

RESUMO

Acute otitis media (AOM) is one of the most common infectious diseases primarily caused by Streptococcus pneumoniae (S.pn) among children. Progranulin (PGRN) is a multifunctional growth factor widely expressed in various tissues and cells. Studies have confirmed that PGRN is involved in the development of a variety of inflammatory diseases. We found that the expression of PGRN increased significantly in the middle ear of wild mice with AOM. However, its physiological functions in AOM still remain unknown. To examine the role of PGRN during AOM, we established an acute otitis media model in both C57BL/6 wild mice and PGRN-deficient (PGRN-/-) mice via transbullar injection with S.pn clinical strain serotype 19F. Interestingly, we observed dual results: on one hand, macrophage recruitment notably increased in PGRN-/- mice compared with WT mice; on the other hand, the overall bacterial clearance was surprisingly dampened in PGRN-/- mice. The enhanced recruitment of macrophages was associated with increased production of chemokine (C-C motif) ligand 2 (CCL2), while the decreased bacterial clearance was associated with impaired endocytosis capacity of macrophages. The scavenging ability of bacteria in PGRN-/- mice was recovered with administration of recombinant PGRN. These results suggested a novel dual role of PGRN in affecting the activities of macrophages.


Assuntos
Quimiotaxia/imunologia , Imunidade Inata/imunologia , Otite Média/imunologia , Progranulinas/imunologia , Streptococcus pneumoniae/imunologia , Doença Aguda , Animais , Células Cultivadas , Quimiocina CCL2/imunologia , Quimiocina CCL2/metabolismo , Quimiotaxia/genética , Modelos Animais de Doenças , Endocitose/genética , Endocitose/imunologia , Humanos , Imunidade Inata/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Otite Média/genética , Otite Média/microbiologia , Progranulinas/genética , Progranulinas/metabolismo , Streptococcus pneumoniae/fisiologia
12.
Immunobiology ; 222(2): 384-393, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27594384

RESUMO

Pneumococcal DnaJ was recently shown to be a potential protein vaccine antigen that induces strong Th1 and Th17 immune response against streptococcus pneumoniae infection in mice. However, how DnaJ mediates T cell immune response against S. pneumoniae infection has not been addressed. Here, we investigate whether DnaJ contributes to the development of T cell immunity through the activation of bone marrow-derived dendritic cells (BMDCs). We found that endotoxin-free recombinant DnaJ (rDnaJ) induced activation and maturation of BMDCs via recognition of Toll-like receptor 4 (TLR4) and activation of MAPKs, NF-κB and PI3K-Akt pathways. rDnaJ-treated BMDCs effectively stimulated naïve CD4+ T cells to secrete IFN-γ and IL-17A. Splenocytes from mice that were adoptively transferred with rDnaJ-pulsed BMDCs secreted higher levels of IFN-γ and IL-17A compared with those that received PBS-activated BMDCs. Splenocytes from TLR4-/- mice immunized with rDnaJ produced lower levels of IFN-γ and IL-17A compared with those from wild type mice. Our findings indicate that DnaJ can induce Th1 and Th17 immune responses against S. pneumoniae through activation of BMDCs in a TLR4-dependent manner.


Assuntos
Proteínas de Bactérias/imunologia , Células Dendríticas/fisiologia , Proteínas de Choque Térmico HSP40/imunologia , Transdução de Sinais , Streptococcus pneumoniae/imunologia , Células Th1/fisiologia , Células Th17/fisiologia , Receptor 4 Toll-Like/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Biomarcadores , Diferenciação Celular/imunologia , Citocinas/metabolismo , Células Dendríticas/citologia , Proteínas de Choque Térmico HSP40/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunomodulação , Imunofenotipagem , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Infecções Pneumocócicas/prevenção & controle , Vacinas Pneumocócicas/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Streptococcus pneumoniae/metabolismo
13.
Prep Biochem Biotechnol ; 47(2): 123-128, 2017 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-27143404

RESUMO

To obtain recombinant cystatin C (CysC) protein, which can be used in immunological diagnostic kits, we focused on the preparation of tag-free CysC. The 6 × His-TF-CysC fusion protein was found to overexpress in soluble form in cells of BL21-Gold (DE3)/pCold TF-CysC, which had been induced with isopropyl-D-1-thiogalactopyranoside. Subsequently, we established a protein purification method for tag-free CysC using immobilized metal-affinity chromatography and size-exclusion chromatography. In this method, glutathione-S-transferase-human rhinovirus 3C proteases were used to remove the protein tags. High homogeneity of the purified CysC was determined by SDS-PAGE, while the purity of the tag-free CysC was ascertained to be above 95%. With a yield of 25 mg/L from bacterial culture, the biological activity of the tag-free CysC was evaluated as inhibitors like natural CysC. The performance of this purification method was successfully evaluated in the preparation of other low molecular weight heterologous proteins in Escherichia coli.


Assuntos
Cistatina C/isolamento & purificação , Escherichia coli/genética , Cromatografia de Afinidade , Cromatografia em Gel , Cistatina C/genética , Eletroforese em Gel de Poliacrilamida , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação
14.
Pediatr Res ; 80(6): 886-893, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27463737

RESUMO

BACKGROUND: The natural course of otitis media (OM) in most children is acute and self-limiting; however, approximately 10-20% of children can experience persistent or recurrent OM. Determining the host factors that influence outcome of OM will help us design better therapies. This study focused on the role of Toll-like receptor 2 (TLR2) in a pneumococcal OM mouse model. METHODS: The middle ears (MEs) of wild-type (WT) and TLR2-/- mice were inoculated with Streptococcus pneumoniae (Spn) serotype 19F via transbullar injection. ME TLR2 expression in WT mice was determined by qRT-PCR and immunofluorescence. ME pathological manifestations, inflammatory response, and pneumococcal clearance between WT and TLR2-/- mice were compared after Spn inoculation. RESULTS: TLR2 expression in ME mucosa was markedly enhanced following infection with Spn in WT mice. In contrast to WT mice, TLR2-/- mice exhibited unaffected early ME inflammatory response. During late stage of ME infection, however, the absence of TLR2 can lead to reduced macrophage recruitment, impaired Spn clearance, and prolonged ME inflammation. CONCLUSION: Our results demonstrate that TLR2 signaling is critical for bacterial clearance and timely resolution of inflammation in OM induced by Spn.


Assuntos
Macrófagos/imunologia , Otite Média/imunologia , Otite Média/microbiologia , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/microbiologia , Receptor 2 Toll-Like/imunologia , Animais , Modelos Animais de Doenças , Orelha Média/imunologia , Orelha Média/microbiologia , Orelha Média/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Otite Média/patologia , Infecções Pneumocócicas/patologia , Streptococcus pneumoniae/classificação , Streptococcus pneumoniae/isolamento & purificação , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Pesquisa Translacional Biomédica , Regulação para Cima
15.
Artigo em Inglês | MEDLINE | ID: mdl-26973817

RESUMO

Interaction between virulence factors of Streptococcus pneumoniae and innate immune receptors elicits host responses through specific signaling pathways during infection. Insights into the signaling events may provide a better knowledge of the starting events for host-pathogen interaction. Here we demonstrated a significant induction of innate immune response elicited by recombinant S. pneumoniae endopeptidase O (rPepO), a newer pneumococcal virulence protein, both in vivo and in vitro. Intratracheal instillation of rPepO protein resulted in significant increase of cytokines production and neutrophils infiltration in mouse lungs. TLR2 or TLR4 deficient mice subjected to rPepO treatment showed decreased cytokines production, reduced neutrophils infiltration and intensified tissue injury as compared with WT mice. Upon stimulation, cytokines TNF-α, IL-6, CXCL1, and CXCL10 were produced by peritoneal exudate macrophages (PEMs) in a TLR2 and TLR4 dependent manner. rPepO-induced cytokines production was markedly decreased in TLR2 or TLR4 deficient PEMs. Further study revealed that cytokines induction relied on the rapid phosphorylation of p38, Akt and p65, not the activation of ERK or JNK. While in TLR2 or TLR4 deficient PEMs the activation of p65 was undetectable. Taken together, these results indicate for the first time that the newer pneumococcal virulence protein PepO activates host innate immune response partially through TLR2 and TLR4 signaling pathways.


Assuntos
Proteínas de Bactérias/imunologia , Endopeptidases/imunologia , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/patogenicidade , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Quimiocina CXCL1/metabolismo , Quimiocina CXCL10/metabolismo , Imunidade Inata/imunologia , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Virulência/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(6): 730-5, 2015 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-26062411

RESUMO

OBJECTIVE: To investigate the mechanism of immune response in mouse macrophage induced by Pneumococcal heat shock protein 40 (HSP40). METHODS: After recombinant HSP40 (rHSP40) underwent expression detection and purification, lipopolysaccharide (LPS) was removed from it. Then rHSP40 was used to stimulate bone marrow derived macrophages (BMDMs) derived from C57BL/6 wild-type mice. The mRNA levels of tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), IL-1ß, IL-23p19, IL-12p40, IL-12p35 and IL-10 in BMDMs were determined by reverse transcription PCR; the expressions of TNF-α, IL-6 and IL-12p40 were measured by ELISA. After stimulated by rHSP40, the levels of TNF-α and IL-6 expressed by wide-type, TLR2-/- and TLR4-/- BMDMs were detected by ELISA. The effects of the pretreatment of mitogen-activated protein kinases (MAPK) inhibitors on the secretion of TNF-α and IL-6 induced by rHSP40 were also evaluated by ELISA in BMDMs. The phosphorylation levels of p38MAPK and c-Jun N-terminal kinase (JNK) were determined by Western blotting. RESULTS: The rHSP40 protein reached a purity of more than 90%. It significantly enhanced the phosphorylation levels of p38MAPK and JNK as well as the expressions of TNF-α and IL-6. The p38MAPK and JNK inhibitors significantly suppressed the expressions of TNF-α and IL-6. The expressions of TNF-α and IL-6 in TLR4-/- BMDMs significantly decreased compared with wide-type BMDMs. CONCLUSION: HSP40-induced immune response of mouse macrophages is regulated by p38MAPK and JNK signaling pathways, and this induction process depends on TLR4.


Assuntos
Proteínas de Choque Térmico HSP40/imunologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Streptococcus pneumoniae/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Proteínas de Bactérias/imunologia , Células da Medula Óssea/citologia , Citocinas/genética , Regulação da Expressão Gênica/imunologia , Proteínas de Choque Térmico HSP40/química , Lipopolissacarídeos/isolamento & purificação , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/imunologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
17.
Immunology ; 144(2): 282-90, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25157456

RESUMO

Airway inflammatory diseases such as chronic obstructive pulmonary disease (COPD) and asthma are associated with elevated expression of interleukin-32 (IL-32), a recently described cytokine that appears to play a critical role in inflammation. However, so far, the regulation of pulmonary IL-32 production has not been fully established. We examined the expression of IL-32 by tumour necrosis factor-α (TNF-α) in primary human lung fibroblasts. Human lung fibroblasts were cultured in the presence or absence of TNF-α and/or other cytokines/Toll-like receptor (TLR) ligands or various signalling molecule inhibitors to analyse the expression of IL-32 by quantitative RT-PCR and ELISA. Next, activation of Akt and c-Jun N-terminal kinase (JNK) signalling pathways was investigated by Western blot. Interleukin-32 mRNA of four spliced isoforms (α, ß, γ and δ) was up-regulated upon TNF-α stimulation, which was associated with a significant IL-32 protein release from TNF-α-activated human lung fibroblasts. The combination of interferon-γ and TNF-α induced enhanced IL-32 release in human lung fibroblasts, whereas IL-4, IL-17A, IL-27 and TLR ligands did not alter IL-32 release in human lung fibroblasts either alone, or in combination with TNF-α. Furthermore, the activation of Akt and JNK pathways regulated TNF-α-induced IL-32 expression in human lung fibroblasts, and inhibition of the Akt and JNK pathways was able to suppress the increased release of IL-32 to nearly the basal level. These data suggest that TNF-α may be involved in airway inflammation via the induction of IL-32 by activating Akt and JNK signalling pathways. Therefore, the TNF-α/IL-32 axis may be a potential therapeutic target for airway inflammatory diseases.


Assuntos
Inflamação/imunologia , Interleucinas/genética , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Asma/imunologia , Células Cultivadas , Fibroblastos/citologia , Humanos , Interferon gama/farmacologia , Interleucina-17/farmacologia , Interleucina-4/farmacologia , Interleucinas/biossíntese , Interleucinas/farmacologia , Pulmão/citologia , Isoformas de Proteínas/genética , Doença Pulmonar Obstrutiva Crônica/imunologia , RNA Mensageiro/biossíntese , Proteínas Recombinantes/farmacologia , Receptores Toll-Like/metabolismo , Regulação para Cima
18.
J Infect Dis ; 211(10): 1616-27, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25489003

RESUMO

BACKGROUND: Interleukin 4 (IL-4) is an important cytokine that may modulate development of secondary bacterial pneumonia during sepsis-induced immunosuppression. METHODS: We established an experimental model of cecal ligation and puncture (CLP)-induced sublethal polymicrobial sepsis followed by secondary Pseudomonas aeruginosa pulmonary infection, RESULTS: IL-4-deficient mice that underwent CLP were resistant to secondary pulmonary P. aeruginosa infection. As compared to wild-type mice, IL-4 knockout (KO) mice displayed improved survival and better bacterial clearance. Furthermore, IL-4 KO mice exhibited enhanced lung inflammation, neutrophil recruitment to airspaces, and elevated pulmonary cytokine production, with significantly increased tumor necrosis factor α (TNF-α) production. Neutralization of TNF-α could reverse the enhanced protection against secondary P. aeruginosa pneumonia in septic IL-4 KO mice, indicating that the resistance of septic IL-4 KO mice to secondary bacterial pneumonia was partially mediated by TNF-α. In addition, IL-4 priming displayed marked impairment of the ability of alveolar macrophages to phagocytose and kill P. aeruginosa in vitro, and this defect was associated with decreased activation of Akt, JNK, p38MAPK, and ERK intracellular signaling pathways by IL-4. Finally, neutralization of IL-4 in septic mice could improve survival and clearance of bacteria from the lungs of septic mice infected with P. aeruginosa. CONCLUSIONS: Our findings provide new insight for immunopathologic mechanisms of sepsis-induced secondary bacterial pneumonia.


Assuntos
Tolerância Imunológica , Interleucina-4/deficiência , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/isolamento & purificação , Sepse/complicações , Animais , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/microbiologia , Fator de Necrose Tumoral alfa/imunologia
19.
FEBS J ; 281(17): 3803-15, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24989111

RESUMO

Interaction between pneumococcal virulence factors and innate immune receptors triggers host responses via specific signaling pathways after infection. By generating a deficient mutant, we show here that, compared with the wild-type parent strain, glycosyl hydrolase 25 relating to invasion protein (GHIP) mutant strain was impaired in rapid dissemination into vessels and caused less severe inflammation in mice lungs. Further study demonstrated that the lack of this protein in Streptococcus pneumoniae caused an increased susceptibility to whole blood or neutrophils, while this impairment could be recovered by supplementing recombinant GHIP (rGHIP). Additionally, secreted GHIP could be detected in culture medium, and purified protein was able to induce the release of tumor necrosis factor α and interleukin 6 from peritoneal macrophages. Further investigations revealed that the induction of interleukin 6 by this virulence factor depended on the phosphorylation of c-Jun N-terminal kinase and p38 mitogen activated protein kinase and Toll-like receptor 2. Taken together, GHIP, a novel pneumococcal virulence factor, appeared to play a critical role in bacterial survival and the induction of host innate immune response during pneumococcal infection.


Assuntos
Proteínas de Bactérias/imunologia , Imunidade Inata , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/genética , Fatores de Virulência/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Interleucina-6/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Pulmão/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Fosforilação , Infecções Pneumocócicas/imunologia , Proteínas Recombinantes/farmacologia , Streptococcus pneumoniae/patogenicidade , Receptor 2 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
20.
J Microbiol ; 51(4): 453-60, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23990296

RESUMO

Hsp100/Clp proteins have crucial functions in the protein quality control, stress tolerance, and virulence of many pathogenic bacteria. ClpE is an important virulence factor involved in adherence and invasion in Streptococcus pneumoniae. To explore the underlying mechanism, we screened ClpE interaction proteins using a bacterial two-hybrid system and co-immunoprecipitation. We used ClpE as bait and constructed the pBT-ClpE bait plasmid for two-hybrid screening. Then, we constructed ClpE::GFP fusion for co-immunoprecipitation screening using anti-GFP monoclonal antibody. We obtained eight potential ClpE interaction proteins, including carbamoyl-phosphate synthase, pyruvate oxidase (SpxB), phosphoenolpyruvate-protein phosphotransferase, aminopeptidase N (pepN), L-lactate dehydrogenase, ribosomal protein S4, sensor histidine kinase (SPD_2019), and FtsW (a cell division protein). FtsW, SpxB, pepN, and SPD_2019 were confirmed to interact with ClpE using Bacterial Two-hybrid or Co-immunoprecipitation. Morphologic observations found that ΔclpE strain existed in abnormal division. ß-Galactosidase activity assay suggested that ClpE contributed to the degradation of FtsW. Furthermore, FtsW could be induced by heat shock. The results suggested that ClpE might affect cell division by regulating the level of FtsW. These data may provide new insights in studying the role of ClpE in S. pneumoniae.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Choque Térmico/metabolismo , Mapeamento de Interação de Proteínas , Streptococcus pneumoniae/metabolismo , Adenosina Trifosfatases/genética , Proteínas de Bactérias/genética , Deleção de Genes , Proteínas de Choque Térmico/genética , Temperatura Alta , Imunoprecipitação , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas/métodos , Proteólise , Streptococcus pneumoniae/genética , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA