RESUMO
The accumulation of self-renewed polarized microglia in the penumbra is a critical neuroinflammatory process after ischemic stroke, leading to secondary demyelination and neuronal loss. Although known to regulate tumor cell proliferation and neuroinflammation, HDAC3's role in microgliosis and microglial polarization remains unclear. We demonstrated that microglial HDAC3 knockout (HDAC3-miKO) ameliorated poststroke long-term functional and histological outcomes. RNA-seq analysis revealed mitosis as the primary process affected in HDAC3-deficent microglia following stroke. Notably, HDAC3-miKO specifically inhibited proliferation of proinflammatory microglia without affecting anti-inflammatory microglia, preventing microglial transition to a proinflammatory state. Moreover, ATAC-seq showed that HDAC3-miKO induced closing of accessible regions enriched with PU.1 motifs. Overexpressing microglial PU.1 via an AAV approach reversed HDAC3-miKO-induced proliferation inhibition and protective effects on ischemic stroke, indicating PU.1 as a downstream molecule that mediates HDAC3's effects on stroke. These findings uncovered that HDAC3/PU.1 axis, which mediated differential proliferation-related reprogramming in different microglia populations, drove poststroke inflammatory state transition, and contributed to pathophysiology of ischemic stroke.
Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Microglia , Acidente Vascular Cerebral/genética , Proliferação de Células , SementesRESUMO
BACKGROUND: Hemifacial macrosomia (HFM, OMIM 164210) is a complex and highly heterogeneous disease. FORKHEAD BOX I3 (FOXI3) is a susceptibility gene for HFM, and mice with loss of function of Foxi3 did exhibit a phenotype similar to craniofacial dysmorphism. However, the specific pathogenesis of HFM caused by FOXI3 deficiency remains unclear till now. METHOD: In this study, we first constructed a Foxi3 deficiency (Foxi3-/- ) mouse model to verify the craniofacial phenotype of Foxi3-/- mice, and then used RNAseq data for gene differential expression analysis to screen candidate pathogenic genes, and conducted gene expression verification analysis using quantitative real-time PCR. RESULTS: By observing the phenotype of Foxi3-/- mice, we found that craniofacial dysmorphism was present. The results of comprehensive bioinformatics analysis suggested that the craniofacial dysmorphism caused by Foxi3 deficiency may be involved in the PI3K-Akt signaling pathway. Quantitative real-time PCR results showed that the expression of PI3K-Akt signaling pathway-related gene Akt2 was significantly increased in Foxi3-/- mice. CONCLUSION: The craniofacial dysmorphism caused by the deficiency of Foxi3 may be related to the expression of Akt2 and PI3K-Akt signaling pathway. This study laid a foundation for understanding the function of FOXI3 and the pathogenesis and treatment of related craniofacial dysmorphism caused by FOXI3 dysfunction.
Assuntos
Anormalidades Craniofaciais , Anormalidades Musculoesqueléticas , Animais , Camundongos , Biologia Computacional , Anormalidades Craniofaciais/genética , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt/genéticaRESUMO
Introduction: The Notch signaling pathway is involved in the development of many diseases; it regulates the development of dendritic cells (DCs), and affects the immune response of DC-mediated T cells. We previously found that ferritin and malate dehydrogenase (mMDH) in Echinococcus granulosus (E.granulosus) induced different immune responses through sensitized DCs. Therefore, in the study we explored whether the Notch signaling pathway affects the development and differentiation of DCs, causing changes in the immune response of DCs sensitized with E. granulosus antigens, and clarified whether it is involved in E.granulosus infection. Methods: We used the Notch signaling pathway inhibitor [N-[3,5-difluorophenace-tyl] -L-alanyl]-S-phenylglycinet-butyl ester (DAPT) or activator Jagged1 to construct in vitro cell models with blocked or activated Notch signaling respectively. We analyzed the effect of Notch signaling on the development and differentiation of DCs by detecting their morphology, migration function, capacity to promote T cell proliferation, and cytokine secretion. We observed the changes in DC response to E. granulosus antigens and the mediated immune response. Results: DAPT inhibited the development and maturation of DCs, which were in a non-responsive or incompetent state, reduced the sensitization of DCs to Eg.ferritin, weakened the migration ability of DCs, disrupted their ability to mediate T-cell proliferation, reduced DC expression of MHCII, CD80, CD60, and CD40 co-stimulatory molecules, prevented the secretion of cytokines and attenuated the expression of Notch1, Notch2, Notch3 receptors, Jagged1, Delta-like 4 (Delta4), and Hes1. Following Jagged1 addition, the function of DCs was restored to some extent, and the expression of Notch1, Delta4 and Hes1 was activated in response to the stimulation of Eg.ferritin. However, Eg.mMDH stimulated DCs to produce an immune response showing weak interference by DAPT and Jagged1. Discussion: The study suggests that the Notc h signaling pathway is involved in the Eg.ferritin-sensitized DC-mediated immune response, which may become a new target for treating E.granulosus infection.
Assuntos
Equinococose , Inibidores da Agregação Plaquetária , Humanos , Inibidores da Agregação Plaquetária/farmacologia , Diferenciação Celular , Transdução de Sinais , Células Dendríticas , FerritinasRESUMO
Although both are myeloid cells located surrounding cerebral vasculature, vessel-associated microglia (VAM) and perivascular macrophages (PVMs) can be distinguished by their distinct morphologies, signatures and microscopic location. As key component of neuro-glia-vascular unit (NGVU), they play prominent roles in neurovasculature development and pathological process of various central nervous system (CNS) diseases, including phagocytosis, angiogenesis, vessel damage/protection and blood flow regulation, therefore serving as potential targets for therapeutics of a broad array of CNS diseases. Herein, we will provide a comprehensive overview of heterogeneity of VAM/PVMs, highlight limitations of current understanding in this field, and discuss possible directions of future investigations.
Assuntos
Doenças do Sistema Nervoso Central , Microglia , Humanos , Microglia/fisiologia , Encéfalo/patologia , Macrófagos , Fagocitose , Doenças do Sistema Nervoso Central/patologiaRESUMO
Echinococcosis is a common human and animal parasitic disease that seriously endangers human health and animal husbandry. Although studies have been conducted on vaccines for echinococcosis, to date, there is no human vaccine available for use. One of the main reasons for this is the lack of in-depth research on basic immunization with vaccines. Our previous results confirmed that recombinant antigen P29 (rEg.P29) induced more than 90% immune protection in both mice and sheep, but data on its induction of sheep-associated cellular immune responses are lacking. In this study, we investigated the changes in CD4+ T cells, CD8+ T cells, and antigen-specific cytokines IFN-γ, IL-4, and IL-17A after rEg.P29 immunization using enzyme-linked immunospot assay (ELISPOT), enzyme-linked immunosorbent assay (ELISA), and flow cytometry to investigate the cellular immune response induced by rEg.P29 in sheep. It was found that rEg.P29 immunization did not affect the percentage of CD4+ and CD8+ T cells in peripheral blood mononuclear cells (PBMCs), and was able to stimulate the proliferation of CD4+ and CD8+ T cells after immunization in vitro. Importantly, the results of both ELISPOT and ELISA showed that rEg.P29 can induce the production of the specific cytokines IFN-γ and IL-17A, and flow cytometry verified that rEg.P29 can induce the expression of IFN-γ in CD4+ and CD8+ T cells and IL-17A in CD4+ T cells; however, no IL-4 expression was observed. These results indicate that rEg.P29 can induce Th1, Th17, and Tc1 cellular immune responses in sheep against echinococcosis infection, providing theoretical support for the translation of rEg.P29 vaccine applications.
Assuntos
Equinococose , Echinococcus granulosus , Vacinas , Humanos , Animais , Camundongos , Ovinos , Interleucina-17 , Linfócitos T CD8-Positivos , Leucócitos Mononucleares , Células Th17 , Mieloblastina , Equinococose/prevenção & controle , Citocinas , ELISPOT , ImunidadeRESUMO
The spread of coronavirus disease 2019 posed a public health crisis beginning in January 2020, affecting hospitals and health care personnel worldwide and disrupting perioperative services. Organization leaders at Xijing Hospital, Xi'an, China, developed a mitigation system for the OR that involved creating a pandemic response team to identify and implement appropriate infection control practices to prevent virus transmission. The leaders addressed managing the daily surgery schedule through patient screening and a focus on the urgency and volume of procedures. They required increased use of personal protective equipment and more stringent cleaning and disinfection protocols and ensured that the physical and mental health of staff members were monitored and prioritized. This article describes how leaders implemented these enhanced processes to protect personnel from infection as they continued to provide patient care. It also describes how high-risk procedures involving patients with confirmed or suspected infections were managed and discusses lessons learned.
Assuntos
COVID-19 , Humanos , Controle de Infecções/métodos , Pandemias/prevenção & controle , Equipamento de Proteção Individual , SARS-CoV-2RESUMO
Echinococcus granulosus causes echinococcosis, an important zoonotic disease worldwide and a major public health issue. Vaccination is an economical and practical approach for controlling E. granulosus. We have previously revealed that a recombinant protein P29 (rEg.P29) is a good vaccine candidate against E. granulosus. However, T cell immunogenic epitopes have not been identified. In the present study, we use rEg.P29-immunized mice as models to screen immunogenic epitopes for the construction of a novel multi-epitope vaccine. We search for immunodominant epitopes from an overlapping peptide library to screen the peptides of rEg.P29. Our results confirm that rEg.P29 immunization in mice elicits the activation of T cells and induces cellular immune responses. Further analyses show that a T cell epitope within amino acids 86100 of rEg.P29 elicits significant antigen-specific IFN-γ production in CD4+ and CD8+ T cells and promotes specific T-cell activation and proliferation. Collectively, these results provide a reference for the construction of a novel vaccine against broad E. granulosus genotypes based on epitopes of rEg.P29.
Assuntos
Equinococose , Epitopos de Linfócito T , Animais , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Epitopos de Linfócito T/genética , Camundongos , Proteínas Recombinantes/genética , ZoonosesRESUMO
The α thalassemia/mental retardation syndrome X-linked (ATRX) mutation impairs DNA damage repair in glioblastoma (GBM), making these cells more susceptible to treatment, which may contribute to the survival advantage in patients with GBM containing ATRX mutations. To better understand the role of ATRX in GBM, genes correlated with ATRX expression were screened in the Cancer Genome Atlas (702 cases) and Chinese Glioma Genome Atlas (325 cases) databases. Sodium-vitamin C cotransporter 2 (SVCT2) was the most positively correlated gene with ATRX expression. ATRX (about 1.99-fold) and SVCT2 (about 2.25-fold) were upregulated in GBM tissues from 40 patients compared with normal brain tissues from 23 subjects. ShSVCT2 transfection did not alter the in vitro viability of GL261 cells. At the same time, it could inhibit the proliferation of GL261 cells in the orthotopic transplantation model with diminished infiltrating macrophages (CD45highCD11b+), downregulated chemokine (C-C motif) ligand 2 (Ccl2), Ccl4, C-X-C motif chemokine ligand 1 (Cxcl1), and Cxcl15 expression, and decreased p-IκBα and p-c-Jun expression. Effect of ShSVCT2 transfection could be reversed by overexpression of SVCT2. siRNA interference of ATRX-dependent SVCT2 signal with shSVCT2 could inhibit tumor cell proliferation in Glu261-LuNeo xenograft tumor model with more survival advantage, probably by the inhibited macrophage chemotaxis. These results indicate that ATRX-dependent SVCT2-mediated chemokine-induced macrophage infiltration is regulated by the NF-κB pathway, which could be considered as treatment targets.NEW & NOTEWORTHY This study demonstrates that interference of ATRX-dependent SVCT2-mediated chemokine-induced macrophage infiltration could inhibit tumor cell proliferation in the GBM cell line-derived xenograft model. ATRX and SVCT2 are potential treatment targets identified in this study.
Assuntos
Neoplasias Encefálicas , Glioblastoma , Simportadores , Talassemia alfa , Animais , Ácido Ascórbico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Xenoenxertos , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Deficiência Intelectual Ligada ao Cromossomo X , Sódio/metabolismo , Transportadores de Sódio Acoplados à Vitamina C , Proteína Nuclear Ligada ao X/genética , Proteína Nuclear Ligada ao X/metabolismoRESUMO
High circulating levels of lactate and high mobility group box-1 (HMGB1) are associated with the severity and mortality of sepsis. However, it is unclear whether lactate could promote HMGB1 release during sepsis. The present study demonstrated a novel role of lactate in HMGB1 lactylation and acetylation in macrophages during polymicrobial sepsis. We found that macrophages can uptake extracellular lactate via monocarboxylate transporters (MCTs) to promote HMGB1 lactylation via a p300/CBP-dependent mechanism. We also observed that lactate stimulates HMGB1 acetylation by Hippo/YAP-mediated suppression of deacetylase SIRT1 and ß-arrestin2-mediated recruitment of acetylases p300/CBP to the nucleus via G protein-coupled receptor 81 (GPR81). The lactylated/acetylated HMGB1 is released from macrophages via exosome secretion which increases endothelium permeability. In vivo reduction of lactate production and/or inhibition of GPR81-mediated signaling decreases circulating exosomal HMGB1 levels and improves survival outcome in polymicrobial sepsis. Our results provide the basis for targeting lactate/lactate-associated signaling to combat sepsis.
Assuntos
Proteína HMGB1 , Sepse , Acetilação , Proteína HMGB1/metabolismo , Humanos , Ácido Láctico , Macrófagos/metabolismoRESUMO
BACKGROUND: Cystic echinococcosis (CE) is a parasitic disease that is caused by Echinococcus granulosus (Eg). The recombinant Echinococcus granulosus antigen P29 (rEg.P29) was shown to confer effective immunity to sheep and mice during E. granulosus secondary infection in our previous study. In this study, we sought to investigate the ability of long noncoding RNA 028466 (lncRNA028466) as a regulator for the protective immunity mediated by rEg.P29 vaccination and to study the effects of lncRNA028466 on CD4+T cell differentiation in mice spleen. METHODS: Female BALB/c mice were divided into two groups and were vaccinated subcutaneously with rEg.P29 antigen and PBS as a control (12 mice each group). Following prime-boost vaccination, CD4+T, CD8+T, and B cells from the spleen were isolated by flow cytometry. Quantitative real-time PCR (qRT-PCR) was performed to measure the expression of lncRNA028466 in these three kinds of cells. Then, lncRNA028466 was overexpressed and knocked down in naive CD4+T cells, and Th1 and Th2 cytokine expression was detected. qRT-PCR, western blot, and ELISA were performed to evaluate the production of IFN-γ, IL-2, IL-4, and IL-10, and flow cytometry was performed to detect the differentiation of Th1 and Th2 subgroups. RESULTS: lncRNA028466 was significantly decreased after the second week of immunization with rEg.P29 antigen. The proportion of CD4+ T cells was increased after rEg.P29 immunization. Overexpression of lncRNA028466 facilitated the production of IL-4, IL-10 and suppressed the production of IFN-γ, IL-2. Furthermore, after transfection with siRNA028466, IL-2 production was facilitated and IL-10 production was suppressed in naive CD4+ T cells. CONCLUSIONS: Immunization with rEg.P29 downregulated the expression of lncRNA028466, which was related to a higher Th1 immune response and a lower Th2 immune response. Our results suggest that lncRNA028466 may be involved in rEg.P29-mediated immune response by regulating cytokine expression of Th1 and Th2.
Assuntos
Antígenos de Helmintos/imunologia , Citocinas/genética , Echinococcus granulosus/imunologia , Regulação da Expressão Gênica , RNA Longo não Codificante/genética , Células Th1/imunologia , Células Th2/imunologia , Animais , Anticorpos Anti-Helmínticos/sangue , Antígenos de Helmintos/administração & dosagem , Antígenos de Helmintos/genética , Citocinas/imunologia , Feminino , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Imunização , Camundongos , Camundongos Endogâmicos BALB C , RNA Longo não Codificante/imunologiaRESUMO
SUMOylation is a reversible post-translational modification that regulates protein function through covalent attachment of small ubiquitin-like modifier (SUMO) proteins. The process of SUMOylating proteins involves an enzymatic cascade, the first step of which entails the activation of a SUMO protein through an ATP-dependent process catalyzed by SUMO-activating enzyme (SAE). Here, we describe the identification of TAK-981, a mechanism-based inhibitor of SAE which forms a SUMO-TAK-981 adduct as the inhibitory species within the enzyme catalytic site. Optimization of selectivity against related enzymes as well as enhancement of mean residence time of the adduct were critical to the identification of compounds with potent cellular pathway inhibition and ultimately a prolonged pharmacodynamic effect and efficacy in preclinical tumor models, culminating in the identification of the clinical molecule TAK-981.
Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Neoplasias/tratamento farmacológico , Ácidos Sulfônicos/uso terapêutico , Sumoilação/efeitos dos fármacos , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Humanos , Camundongos , Estrutura Molecular , Ligação Proteica , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Relação Estrutura-Atividade , Ácidos Sulfônicos/síntese química , Ácidos Sulfônicos/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
Angiogenesis is essential for cardiac functional recovery after myocardial infarction (MI). HSPA12B is predominately expressed in endothelial cells and required for angiogenesis. Yes-associated protein (YAP) plays an important role in tumor angiogenesis. This study investigated the cooperative role of HSPA12B and YAP in angiogenesis after MI. Silencing of either HSPA12B or YAP impaired hypoxia-promoted endothelial cell proliferation and angiogenesis. Deficiency of HSPA12B suppressed YAP expression and nuclear translocation after hypoxia. Knockdown of YAP attenuated hypoxia-stimulated HSPA12B nuclear translocation and abrogated HSPA12B-promoted endothelial cell angiogenesis. Mechanistically, hypoxia induced an interaction between endothelial HSPA12B and YAP. ChIP assay showed that HSPA12B is a target gene of YAP/transcriptional enhanced associated domain 4 (TEAD4) and a coactivator in YAP-associated angiogenesis. In vivo studies using the MI model showed that endothelial cell-specific deficiency of HSPA12B (eHspa12b-/-) or YAP (eYap-/-) impaired angiogenesis and exacerbated cardiac dysfunction compared with WT mice. MI increased YAP expression and nuclear translocation in WT hearts but not eHspa12b-/- hearts. HSPA12B expression and nuclear translocation were upregulated in WT MI hearts but not eYap-/- MI myocardium. Our data demonstrate that endothelial HSPA12B is a target and coactivator for YAP/TEAD4 and cooperates with YAP to regulate endothelial angiogenesis after MI.
Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Endotélio Vascular/patologia , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP70/fisiologia , Infarto do Miocárdio/fisiopatologia , Neovascularização Patológica/patologia , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Endotélio Vascular/metabolismo , Proteínas de Choque Térmico HSP70/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/metabolismo , Transporte Proteico , Fatores de Transcrição/genética , Proteínas de Sinalização YAPRESUMO
Endothelial cell dysfunction contributes to sepsis induced initiate immune response and the infiltration of immune cells into organs, resulting in organ injury. Heat shock protein A12B (HSPA12B) is predominantly expressed in endothelial cells. The present study investigated whether endothelial HSPA12B could regulate macrophage pro-inflammatory response during sepsis. Wild type (WT) and endothelial cell-specific HSPA12B deficient (HSPA12B-/-) mice were subjected to CLP sepsis. Mortality and cardiac function were monitored. Higher mortality, worsened cardiac dysfunction, and greater infiltrated macrophages in the myocardium and spleen were observed in HSPA12B-/- septic mice compared with the WT septic mice. The serum levels of TNF-α and IL-1ß were higher and the levels of IL-10 were lower in HSPA12B-/- septic mice than in WT septic mice. Importantly, endothelial exosomes contain HSPA12B which can be uptaken by macrophages. Interestingly, endothelial exosomal HSPA12B significantly increases IL-10 levels and decreases TNF-α and IL-1ß production in LPS-stimulated macrophages. Mechanistic studies show that endothelial exosomal HSPA12B downregulates NF-κB activation and nuclear translocation in LPS stimulated macrophages. These data suggest that endothelial HSPA12B plays a novel role in the regulation of macrophage pro-inflammatory response via exosomes during sepsis and that sepsis induced cardiomyopathy and mortality are associated with endothelial cell deficiency of HSPA12B.
Assuntos
Coinfecção/imunologia , Exossomos/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Células Endoteliais da Veia Umbilical Humana/imunologia , Macrófagos/imunologia , Sepse/imunologia , Sepse/microbiologia , Animais , Células Cultivadas , Coinfecção/sangue , Citocinas/sangue , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Proteínas de Choque Térmico HSP70/genética , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sepse/sangue , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , TransfecçãoRESUMO
Aims: Inadequate healing after myocardial infarction (MI) leads to heart failure and fatal ventricular rupture, while optimal healing requires timely induction and resolution of inflammation. This study tested the hypothesis that heat shock protein B1 (HSPB1), which limits myocardial inflammation during endotoxemia, modulates wound healing after MI. Methods and results: To test this hypothesis, cardiomyocyte-specific HSPB1 knockout (Hspb1-/-) mice were generated using the Cre-LoxP recombination system. MI was induced by ligation of the left anterior descending coronary artery in Hspb1-/- and wild-type (WT) littermates. HSPB1 was up-regulated in cardiomyocytes of WT animals in response to MI, and deficiency of cardiomyocyte HSPB1 increased MI-induced cardiac rupture and mortality within 21 days after MI. Serial echocardiography showed more aggravated remodelling and cardiac dysfunction in Hspb1-/- mice than in WT mice at 1, 3, and 7 days after MI. Decreased collagen deposition and angiogenesis, as well as increased MMP2 and MMP9 activity, were also observed in Hspb1-/- mice compared with WT controls after MI, using immunofluorescence, polarized light microscopy, and zymographic analyses. Notably, Hspb1-/- hearts exhibited enhanced and prolonged leucocyte infiltration, enhanced expression of inflammatory cytokines, and enhanced TLR4/MyD88/NFκB activation compared with WT controls after MI. In-depth molecular analyses in both mice and primary cardiomyocytes demonstrated that cardiomyocyte-specific knockout of HSPB1 increased nuclear factor-κB (NFκB) activation, which promoted the expression of proinflammatory mediators. This led to increased leucocyte recruitment, thereby to excessive inflammation, ultimately resulting in adverse remodelling, cardiac dysfunction, and cardiac rupture following MI. Conclusion: These data suggest that HSPB1 acts as a negative regulator of NFκB-mediated leucocyte recruitment and the subsequent inflammation in cardiomyocytes. Cardiomyocyte HSPB1 is required for wound healing after MI and could be a target for myocardial repair in MI patients.
Assuntos
Quimiotaxia de Leucócito , Ruptura Cardíaca Pós-Infarto/metabolismo , Proteínas de Choque Térmico/deficiência , Leucócitos/metabolismo , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Remodelação Ventricular , Animais , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Proteínas de Choque Térmico HSP27/deficiência , Proteínas de Choque Térmico HSP27/genética , Ruptura Cardíaca Pós-Infarto/imunologia , Ruptura Cardíaca Pós-Infarto/patologia , Ruptura Cardíaca Pós-Infarto/fisiopatologia , Proteínas de Choque Térmico/genética , Leucócitos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Chaperonas Moleculares/genética , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Neovascularização Fisiológica , Ratos Sprague-Dawley , Transdução de Sinais , CicatrizaçãoRESUMO
Mice immunized with recombinant Echinococcus granulosus antigens Eg10 and Eg mMDH do not show elevated resistance to E. granulosus infection but show aggravated infection instead. To gain a deeper insight in the immune tolerance mechanisms in mice immunized with Eg10 and Eg mMDH, this study simulated the immune tolerance process in vitro by culturing bone marrow-derived dendritic cells (BMDCs) in the presence of Eg10 or Eg mMDH. Scanning electron microscopy revealed that Eg10- and Eg mMDH-treated DCs exhibited immature cell morphology, while addition of LPS to the cells induced changes in cell morphology and an increase in the number of cell-surface protrusions. This observation was consistent with the increased expression of the cell-surface molecules MHCII and CD80 in Eg10- and Eg mMDH-treated DCs pretreated with LPS. DCs exposed to the two antigens had a very weak ability to induce T-cell proliferation, but could promote the formation of Treg cells. Introduction of the indoleamine 2,3-dioxygenase (IDO) inhibitor, 1-methyl tryptopha (1-MT) enhanced the ability of the antigens to induce T cells and inhibited the induction of Treg cells. Eg mMDH-treated DCs showed a strong response to 1-MT: the DCs had high mRNA levels of IDO, IL-6, and IL-10, while 1-MT decreased the expression. In contrast, DCs treated with Eg10 did not show significant changes after 1-MT treatment. Eg mMDH inhibited DC maturation and promoted IDO expression, which, on the one hand, decreased the ability of DCs to induce T-cell proliferation, resulting in T-cell anergy, and on the other hand, induced the formation of Tregs, resulting in an immunosuppressive effect. In contrast, the escape mechanisms induced by Eg10 did not primarily depend on the IDO pathway and might involve other mechanisms that need to be further explored.
Assuntos
Antígenos de Helmintos/imunologia , Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Equinococose/imunologia , Tolerância Imunológica , Linfócitos T Reguladores/imunologia , Animais , Antígenos de Helmintos/genética , Antígenos de Helmintos/farmacologia , Células da Medula Óssea/patologia , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/patologia , Equinococose/patologia , Echinococcus granulosus , Masculino , Camundongos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Linfócitos T Reguladores/patologiaRESUMO
A new on-off-on fluorescent probe, CMOS, based on coumarin was developed to detect the process of hypochlorous acid (HOCl) oxidative stress and cysteine/homocysteine (Cys/Hcy) reduction. The probe exhibited a fast response, good sensitivity and selectivity. Moreover, it was applied for monitoring the redox process in living cells.
RESUMO
OBJECTIVE: To explore the immunoreaction against protoscolex infection by the recombinant ferritin of echinococcus granulosus (rEg.ferritin) immunized mice. METHODS: Bone marrow deriedstem cells (BMDCs) were collected and exposed in different antigens by cell culture and included 6 groups,there were hydatid fluid (HF) group (HF 30 µg/mL),rEg.ferritin group (rEg.ferritin 1 µg/mL), rEg.ferritin+LPS group (rEg.ferritin 1 µg/mL and LPS 100 ng/mL),HF+LPS group (HF 30 µg/mL and LPS 100 ng/mL),LPS group (LPS 100 ng/mL) and control (not add antigen). Morphology of BMDCs in different groups were detected by scan electron microscope (SEM). Phagocytic-ability,T cell multiplication capability and surface marker expression of BMDCs in different groups were detected by flow cytometry.Cytokines of Th1 [interleukin (IL)-12p70,tumor necrosis factor-α (TNF-α)] and Th2 (IL-6 and IL-10) in surpernatant of BMDCs in different groups were detected by enzyme-linked immunosorbent assay (ELISA). RESULTS: rEg.ferritin or rEg.ferritin+LPS stimulated BMDCs maturation, which the dendrites number of BMDCs in these 2 groups were more and longer than control ,HF and HF+LPS groups ( P<0.05), and induced higher levels of surface molecules major histocompatibility complexâ ¡ (MHCâ ¡),CD80,CD86 and CD40,also had strong ability to induce T cell multiplication than control,HF,HF+LPS groups ( P<0.05),but had weak phagocytosis than HF group,and secreted higher levels of IL-6,IL-12p70,TNF-α and IL-10 than control group. CONCLUSION: BMDCs are stimulated with rEg.ferritin and became maturation. By activating T cell and releasing more cytokines of Th1 and Th2,rEg.ferrtin can induce BMDCs to produce Th1 and Th2 immunoreaction.
Assuntos
Células Dendríticas/imunologia , Ferritinas/imunologia , Proteínas de Helminto/imunologia , Linfócitos T/imunologia , Animais , Células da Medula Óssea , Diferenciação Celular , Citocinas/imunologia , Echinococcus granulosus , Camundongos , Fagocitose , Proteínas Recombinantes/imunologia , Células-TroncoRESUMO
Endothelial damage is a critical mediator of myocardial ischemia/reperfusion (I/R) injury. HSPA12B is an endothelial-cell-specifically expressed heat shock protein. However, the roles of HSPA12B in acute myocardial I/R injury is unknown. Here we reported that myocardial I/R upregulated HSPA12B expression in ventricular tissues, and endothelial overexpression of HSPA12B in transgenic mice (Tg) limited infarct size, attenuated cardiac dysfunction and improved cardiomyocyte survival compared with their wild type littermates. These improvements were accompanied with the diminished myocardial no-reflow phenomenon, decreased microvascular leakage, and better maintained endothelial tight junctions. The I/R-evoked neutrophil infiltration was also suppressed in Tg hearts compared with its wild type (WT) littermates. Moreover, Tg hearts exhibited the enhanced activation of PI3K/Akt//mTOR signaling following I/R challenge. However, pharmacological inhibition of PI3K abolished the HSPA12B-induced cardioprotection against myocardial I/R injury. The data demonstrate for the first time that the endothelial HSPA12B protected hearts against myocardial I/R injury. This cardioprotective action of HSPA12B was mediated, at least in part, by improving endothelial integrity in a PI3K/Akt/mTOR-dependent mechanism. Our study suggests that targeting endothelial HSPA12B could be an alternative approach for the management of patients with myocardial I/R injury.
Assuntos
Endotélio/metabolismo , Proteínas de Choque Térmico HSP70/genética , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Apoptose/genética , Sobrevivência Celular/genética , Modelos Animais de Doenças , Endotélio/patologia , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/metabolismo , Infiltração de Neutrófilos , Transdução de Sinais , Junções Íntimas/metabolismo , Junções Íntimas/ultraestrutura , Molécula 1 de Adesão de Célula Vascular/metabolismo , Disfunção VentricularRESUMO
Aging-induced cardiac dysfunction is a prominent feature of cardiac aging. Heat shock protein 27 (HSP27) protects cardiac function against ischemia or chemical challenge. We hypothesized that HSP27 attenuates cardiac aging. Transgenic (Tg) mice with cardiac-specific expression of the HSP27 gene and wild-type (WT) littermates were employed in the experiments. Echocardiography revealed a significant decline in the cardiac function of old WT mice compared with young WT mice. In striking contrast, the aging-induced impairment of cardiac function was attenuated in old Tg mice compared with old WT mice. Levels of cardiac aging markers were lower in old Tg mouse hearts than in old WT mouse hearts. Less interstitial fibrosis and lower contents of reactive oxygen species and ubiquitin-conjugated proteins were detected in old Tg hearts than in old WT hearts. Furthermore, old Tg hearts demonstrated lower accumulation of LC3-II and p62 than old WT hearts. Levels of Atg13, Vps34, and Rab7 were also higher in old Tg hearts than in old WT hearts. Additionally, old Tg hearts had higher levels of PINK1 and Parkin than old WT hearts, suggesting that mitophagy was activated in old Tg hearts. Taken together, HSP27 alleviated cardiac aging and this action involved antioxidation and mitophagy activation.
Assuntos
Envelhecimento , Antioxidantes/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Mitofagia/fisiologia , Miocárdio/metabolismo , Animais , Proteína 7 Relacionada à Autofagia/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Ecocardiografia , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Chaperonas Moleculares , Carbonilação Proteica , Proteínas Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7RESUMO
The aim of this study was to investigate the immunoprotective effects of recombinant Echinococcus granulosus glutathione S-transferase (rEgGST) against the development of protoscolices (PSCs), and to determine the mechanisms underlying this protection. ICR mice were subcutaneously immunized three times with rEgGST at weeks 0, 2 and 4, followed by the intraperitoneal administration of E. granulosus PSCs at week 10. Six mice in each group were sacrificed at 0, 2, 4, 6, 10, 18 and 30 weeks following the initial vaccination in order to observe the macroscopic and microscopic effects of parasite development. Various analyses were subsequently conducted, including determination of the levels of immunoglobulins (Igs) and cytokines. Significant differences were observed a number of indices of immune response following immunization with rEgGST. These included reduced cyst formation and elevated levels of IgG1, IgG2a, IgG3, IL-2, IL-4, IL-10 and IFN-γ, which indicated an increased percentage of immune helper cells. The results of the present study suggest that immunization with rEgGST in mice is able to successfully reduce the PSC-induced formation of cysts and to stimulate an immune response, suggesting that rEgGST possesses potential value as a candidate vaccine for PSC infection.