Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Inflamm Res ; 73(6): 897-913, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38625657

RESUMO

OBJECTIVES AND DESIGN: As an interferon-inducible protein, Viperin has broad-spectrum antiviral effects and regulation of host immune responses. We aim to investigate how Viperin regulates interferon-γ (IFN-γ) production in macrophages to control Mycobacterium tuberculosis (Mtb) infection. METHODS: We use Viperin deficient bone-marrow-derived macrophage (BMDM) to investigate the effects and machines of Viperin on Mtb infection. RESULTS: Viperin inhibited IFN-γ production in macrophages and in the lung of mice to promote Mtb survival. Further insight into the mechanisms of Viperin-mediated regulation of IFN-γ production revealed the role of TANK-binding kinase 1 (TBK1), the TAK1-dependent inhibition of NF-kappa B kinase-epsilon (IKKε), and interferon regulatory factor 3 (IRF3). Inhibition of the TBK1-IKKε-IRF3 axis restored IFN-γ production reduced by Viperin knockout in BMDM and suppressed intracellular Mtb survival. Moreover, Viperin deficiency activated the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway, which promoted IFN-γ production and inhibited Mtb infection in BMDM. Additionally, a combination of the anti-TB drug INH treatment in the absence of Viperin resulted in further IFN-γ production and anti-TB effect. CONCLUSIONS: This study highlights the involvement of TBK1-IKKε-IRF3 axis and JAK-STAT signaling pathways in Viperin-suppressed IFN-γ production in Mtb infected macrophages, and identifies a novel mechanism of Viperin on negatively regulating host immune response to Mtb infection.


Assuntos
Fator Regulador 3 de Interferon , Interferon gama , Macrófagos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis , Proteínas Serina-Treonina Quinases , Proteínas , Transdução de Sinais , Animais , Interferon gama/metabolismo , Interferon gama/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Mycobacterium tuberculosis/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Camundongos , Proteínas/genética , Proteínas/metabolismo , Quinase I-kappa B/metabolismo , Janus Quinases/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Camundongos Knockout , Tuberculose/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Proteína Viperina
2.
Int J Mol Sci ; 25(4)2024 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-38397085

RESUMO

Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), remains a global health crisis with substantial morbidity and mortality rates. Type II alveolar epithelial cells (AEC-II) play a critical role in the pulmonary immune response against Mtb infection by secreting effector molecules such as antimicrobial peptides (AMPs). Here, human ß-defensin 1 (hBD1), an important AMP produced by AEC-II, has been demonstrated to exert potent anti-tuberculosis activity. HBD1 overexpression effectively inhibited Mtb proliferation in AEC-II, while mice lacking hBD1 exhibited susceptibility to Mtb and increased lung tissue inflammation. Mechanistically, in A549 cells infected with Mtb, STAT1 negatively regulated hBD1 transcription, while CEBPB was the primary transcription factor upregulating hBD1 expression. Furthermore, we revealed that the ERK1/2 signaling pathway activated by Mtb infection led to CEBPB phosphorylation and nuclear translocation, which subsequently promoted hBD1 expression. Our findings suggest that the ERK1/2-CEBPB-hBD1 regulatory axis can be a potential therapeutic target for anti-tuberculosis therapy aimed at enhancing the immune response of AEC-II cells.


Assuntos
Mycobacterium tuberculosis , Tuberculose , beta-Defensinas , Animais , Humanos , Camundongos , Células Epiteliais Alveolares , beta-Defensinas/genética , beta-Defensinas/farmacologia , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células Epiteliais , Sistema de Sinalização das MAP Quinases , Tuberculose/metabolismo
3.
Inflamm Res ; 72(1): 27-41, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36315280

RESUMO

OBJECTIVES AND DESIGN: Dendritic cells (DCs) are one of the key immune cells in bridging innate and adaptive immune response against Mycobacterium tuberculosis (Mtb) infection. Interferons (IFNs) play important roles in regulating DC activation and function. Virus-inhibitory protein, endoplasmic reticulum-associated, interferon-inducible (Viperin) is one of the important IFN-stimulated genes (ISGs), and elicits host defense against infection. METHODS: We investigated the effects and mechanisms of Viperin on DC activation and function using Viperin deficient bone marrow-derived dendritic cells (BMDCs) during Mtb infection. RESULTS: Viperin deficiency enhanced phagocytic activity and increased clearance of Mtb in DCs, produced higher abundance of NO, cytokine including interleukin-12 (IL-12), Tumor necrosis factor-α (TNF-α), IL-1ß, IL-6 and chemokine including CXCL1, CXCL2 and CXCL10, elevated MHC I, MHC II and co-stimulatory molecules expression, and enhanced CD4+ and CD8+ T cell responses. Mechanistically, Viperin deficiency promoted DC activation and function through NF-κB p65 activation. NF-κB p65 inhibitor prevented cytokine and chemokine production, and co-stimulatory molecules expression promoted by Viperin deficiency. CONCLUSIONS: These results suggest that Mtb induced Viperin expression could impair the activation of host defense function of DCs and DC-T cell cross talk during Mtb infection. This research may provide a potential target for future HDT in TB therapy.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Proteína Viperina , Quimiocinas/metabolismo , Citocinas , Células Dendríticas , Mycobacterium tuberculosis/metabolismo , NF-kappa B/metabolismo , Proteína Viperina/metabolismo , Animais
4.
J Immunol ; 206(11): 2623-2637, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34001657

RESUMO

Mycobacterium tuberculosis, the pathogen that causes tuberculosis, exhibits complex host-pathogen interactions. Pattern recognition receptors and their downstream signaling pathways play crucial roles in determining the outcome of infection. In particular, the scaffold protein ß-arrestin 2 mediates downstream signaling of G protein-coupled receptors. However, the role of ß-arrestin 2 in conferring immunity against M. tuberculosis has not yet been explored. We found that ß-arrestin 2 was upregulated in the lesioned regions of lung tissues in patients with tuberculosis. M. tuberculosis infection upregulated ß-arrestin 2 expression in human macrophages, and silencing of ß-arrestin 2 significantly enhanced bactericidal activity by enhancing the expression of proinflammatory cytokines such as TNF-α. ß-Arrestin 2 was shown to inhibit the activation of the TLR2/ERK1/2 pathway and its transcriptional regulation activity upon M. tuberculosis infection. Furthermore, ß-arrestin 2 transcriptionally regulates TNF-α by binding to CREB1. These observations revealed that the upregulation of ß-arrestin 2 is critical for M. tuberculosis to escape immune surveillance through an unknown mechanism. Our research offers a novel interference modality to enhance the immune response against tuberculosis by targeting ß-arrestin 2 to modulate the TLR2-ß-arrestin 2-ERK1/2-CREB1-TNF-α regulatory axis.


Assuntos
Inflamação/imunologia , Tuberculose/imunologia , beta-Arrestina 2/imunologia , Adolescente , Células Cultivadas , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Pessoa de Meia-Idade
5.
J Immunol ; 203(4): 922-935, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31235551

RESUMO

Mycobacterium tuberculosis, which primarily infects mononuclear phagocytes, remains the leading bacterial cause of enormous morbidity and mortality because of bacterial infections in humans throughout the world. The IL-1 family of cytokines is critical for host resistance to M. tuberculosis As a newly discovered subgroup of the IL-1 family, although IL-36 cytokines have been proven to play roles in protection against M. tuberculosis infection, the antibacterial mechanisms are poorly understood. In this study, we demonstrated that IL-36γ conferred to human monocyte-derived macrophages bacterial resistance through activation of autophagy as well as induction of WNT5A, a reported downstream effector of IL-1 involved in several inflammatory diseases. Further studies showed that WNT5A could enhance autophagy of monocyte-derived macrophages by inducing cyclooxygenase-2 (COX-2) expression and in turn decrease phosphorylation of AKT/mTOR via noncanonical WNT signaling. Consistently, the underlying molecular mechanisms of IL-36γ function are also mediated by the COX-2/AKT/mTOR signaling axis. Altogether, our findings reveal a novel activity for IL-36γ as an inducer of autophagy, which represents a critical inflammatory cytokine that control the outcome of M. tuberculosis infection in human macrophages.


Assuntos
Interleucina-1/imunologia , Macrófagos/imunologia , Tuberculose Pulmonar/imunologia , Proteína Wnt-5a/imunologia , Autofagia/imunologia , Humanos , Interleucina-1/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Transdução de Sinais/imunologia , Tuberculose Pulmonar/metabolismo , Proteína Wnt-5a/metabolismo
6.
J Cell Mol Med ; 24(22): 13129-13138, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32967056

RESUMO

Vitamin B6 is necessary to maintain normal metabolism and immune response, especially the anti-inflammatory immune response. However, the exact mechanism by which vitamin B6 plays the anti-inflammatory role is still unclear. Here, we report a novel mechanism of preventing excessive inflammation by vitamin B6 via reduction in the accumulation of sphingosine-1-phosphate (S1P) in a S1P lyase (SPL)-dependent manner in macrophages. Vitamin B6 supplementation decreased the expression of pro-inflammatory cytokines by suppressing nuclear factor-κB and mitogen-activated protein kinases signalling pathways. Furthermore, vitamin B6-reduced accumulation of S1P by promoting SPL activity. The anti-inflammatory effects of vitamin B6 were inhibited by S1P supplementation or SPL deficiency. Importantly, vitamin B6 supplementation protected mice from lethal endotoxic shock and attenuated experimental autoimmune encephalomyelitis progression. Collectively, these findings revealed a novel anti-inflammatory mechanism of vitamin B6 and provided guidance on its clinical use.


Assuntos
Aldeído Liases/metabolismo , Inflamação/metabolismo , Lisofosfolipídeos/metabolismo , Macrófagos/metabolismo , Esfingosina/análogos & derivados , Vitamina B 6/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Progressão da Doença , Encefalomielite Autoimune Experimental/metabolismo , Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Choque/metabolismo , Transdução de Sinais , Esfingosina/metabolismo
7.
PLoS Pathog ; 14(8): e1007266, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30133544

RESUMO

NLRC3, a member of the NLR family, has been reported as a negative regulator of inflammatory signaling pathways in innate immune cells. However, the direct role of NLRC3 in modulation of CD4+ T-cell responses in infectious diseases has not been studied. In the present study, we showed that NLRC3 plays an intrinsic role by suppressing the CD4+ T cell phenotype in lung and spleen, including differentiation, activation, and proliferation. NLRC3 deficiency in CD4+ T cells enhanced the protective immune response against Mycobacterium tuberculosis infection. Finally, we demonstrated that NLRC3 deficiency promoted the activation, proliferation, and cytokine production of CD4+ T cells via negatively regulating the NF-κB and MEK-ERK signaling pathways. This study reveals a critical role of NLRC3 as a direct regulator of the adaptive immune response and its protective effects on immunity during M. tuberculosis infection. Our findings also suggested that NLRC3 serves as a potential target for therapeutic intervention against tuberculosis.


Assuntos
Linfócitos T CD4-Positivos/patologia , Imunidade/genética , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/fisiologia , Células Cultivadas , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tuberculose/genética , Tuberculose/patologia
8.
Int J Mol Sci ; 20(3)2019 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-30717477

RESUMO

Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) represents one of the greatest threats to human health., Interferons (IFNs) in combination with the first-line of anti-TB drugs have been used for treating TB for decades in the clinic, but how Mtb infection regulates interferon-stimulated genes (ISGs) in human macrophages (Mϕs) remains unknown. In this study, we investigated the expression-signature and associated innate signaling mechanisms of ISGs in Mtb-infected human monocyte-derived Mϕs (hMDMs) and THP-1-derived Mϕs (THP-1-Mϕs). Among 28 of the detected ISGs, 90% of them exerted a significant increase in Mtb-infected Mϕs. Additionally, we found that cytosolic cyclic (GMP-AMP) synthase (cGAS), toll-like receptor-2 (TLR-2) and TLR-4 signaling pathways participated in ISG induction. Their downstream elements of TANK-binding kinase 1 (TBK1), nuclear factor-kappa B (NF-κB), mitogen-activated protein kinase (MAPK), and Janus kinase-signal transducer and activator of transcription (JAK-STAT) were selectively involved in Mtb-mediated ISG production. Finally, the numerous types of ISG expression in hMDMs of TB patients were more susceptible to restimulation of Mtb infection or/and IFN treatment than that of healthy people. Hence, different signaling pathways define different ISG expression during Mtb infection and this helps to illustrate how ISGs are elucidated and to better understand the host immune responses to Mtb infection in Mϕs.


Assuntos
Interferon gama/farmacologia , Macrófagos/metabolismo , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Tuberculose Pulmonar/tratamento farmacológico , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Interferon gama/metabolismo , Interferon gama/uso terapêutico , Janus Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Mycobacterium tuberculosis , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição STAT1/metabolismo , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/metabolismo
9.
J Immunol ; 197(2): 580-9, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27296666

RESUMO

Tuberculosis is still the widest spread infectious disease in the world, and more in-depth studies are needed on the interaction between the pathogen and the host. Due to the highest lipid components in Mycobacterium tuberculosis, the CD1 family that specifically presents antigenic lipids plays important roles in the antituberculosis immunity, especially CD1c, which functions as the intracellular Ag inspector at the full intracellular range. However, downregulation of the CD1c mRNA level has been observed in M. tuberculosis-infected cells, which is consistent with the regulatory mechanism of miRNA on gene expression. In this study, through combinatory analysis of previous miRNA transcriptomic assays and bioinformatic predictions by web-based algorithms, miR-381-3p was predicted to bind the 3'-untranslated region of CD1c gene. In vivo expression of miR-381-3p in dendritic cells (DCs) of TB patients is higher than in DCs of healthy individuals, inversely related to CD1c. Suppression of CD1c expression in bacillus Calmette-Guérin (BCG)-infected DCs was accompanied with upregulation of miR-381-3p, whereas inhibition of miR-381-3p could reverse suppression of CD1c expression and promote T cell responses against BCG infection. Further study indicated that miR-381-3p is also one of the mediators of the immune suppressor IL-10. Collectively, these results demonstrated the mechanism that suppression of CD1c by BCG infection is mediated by miR-381-3p. This finding may provide a novel approach to boost immune responses to M. tuberculosis.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos CD1/imunologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica/imunologia , Glicoproteínas/imunologia , MicroRNAs/imunologia , Tuberculose/imunologia , Algoritmos , Apresentação de Antígeno/genética , Vacina BCG , Western Blotting , Separação Celular , Biologia Computacional , Humanos , Imunidade Celular/genética , Imunidade Celular/imunologia , Mycobacterium tuberculosis , Reação em Cadeia da Polimerase em Tempo Real
10.
Int J Mol Sci ; 19(3)2018 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-29510550

RESUMO

Absence of effective therapeutic methods for avascular necrosis of femoral head (ANFH) is still perplexing the world's medical community. Bone marrow mesenchymal stem cells (BMSCs) adoptive cell therapy combined with core decompression is a promising modality, which is highly dependent on the cellular activities of BMSCs. Hepatocyte growth factor (HGF) is a survival factor for BMSCs, yet the underlying mechanism is not fully elucidated. In this study, the effects of multiplicity of infections (MOIs) of recombinant adenovirus carrying HGF gene (rAd-HGF) on human BMSC proliferation and osteogenic differentiation were systemically examined. Infection of rAd-HGF produced secretory HGF and promoted hBMSC proliferation in a MOI-dependent manner, while the osteogenesis was also strengthened as indicated by enhanced calcium nodule formation with the strongest effects achieved at MOI = 250. Blocking the activities of c-MET or its downstream signaling pathways, WNT, ERK1/2, and PI3K/AKT led to differential consequents. Specifically, blockage of the WNT pathway significantly promoted osteogenic differentiation, which also showed additive effects when combined application with rAd-HGF. Our data demonstrated the pro-osteogenic effects of optimized MOIs of rAd-HGF, while inhibition of WNT pathway or activation of PI3K/AKT pathway may act as candidate adjuvant modalities for promoting osteogenic differentiation in rAd-HGF-modified hBMSC treatment on ANFH.


Assuntos
Diferenciação Celular , Proliferação de Células , Fator de Crescimento de Hepatócito/genética , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Adenoviridae/genética , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Vetores Genéticos/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Via de Sinalização Wnt
11.
J Cell Mol Med ; 20(10): 1984-98, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27113787

RESUMO

Tuberculosis (TB) and human immunodeficiency virus type 1 (HIV-1) infection are closely intertwined, with one-quarter of TB/HIV coinfected deaths among people died of TB. Effector CD8(+) T cells play a crucial role in the control of Mycobacterium tuberculosis (MTB) and HIV-1 infection in coinfected patients. Adoptive transfer of a multitude of effector CD8(+) T cells is an appealing strategy to impose improved anti-MTB/HIV-1 activity onto coinfected individuals. Due to extensive existence of heterologous immunity, that is, T cells cross-reactive with peptides encoded by related or even very dissimilar pathogens, it is reasonable to find a single T cell receptor (TCR) recognizing both MTB and HIV-1 antigenic peptides. In this study, a single TCR specific for both MTB Ag85B199-207 peptide and HIV-1 Env120-128 peptide was screened out from peripheral blood mononuclear cells of a HLA-A*0201(+) healthy individual using complementarity determining region 3 spectratype analysis and transferred to primary CD8(+) T cells using a recombinant retroviral vector. The bispecificity of the TCR gene-modified CD8(+) T cells was demonstrated by elevated secretion of interferon-γ, tumour necrosis factor-α, granzyme B and specific cytolytic activity after antigen presentation of either Ag85B199-207 or Env120-128 by autologous dendritic cells. To the best of our knowledge, this study is the first report proposing to produce responses against two dissimilar antigenic peptides of MTB and HIV-1 simultaneously by transfecting CD8(+) T cells with a single TCR. Taken together, T cells transduced with the additional bispecific TCR might be a useful strategy in immunotherapy for MTB/HIV-1 coinfected individuals.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos/imunologia , HIV-1/imunologia , Mycobacterium tuberculosis/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução Genética , Sequência de Aminoácidos , Antígenos/imunologia , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Sequência de Bases , Citotoxicidade Imunológica , Vetores Genéticos/metabolismo , Humanos , Interferon gama/metabolismo , Lectinas Tipo C/metabolismo , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
J Transl Med ; 13: 141, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25943357

RESUMO

INTRODUCTION: The invariant natural killer T (iNKT) cell has been shown to play a central role in early stages immune responses against Mycobacterium tuberculosis (Mtb) infection, which become nonresponsive (anergic) and fails to control the growth of Mtb in patients with active tuberculosis. Enhancement of iNKT cell responses to Mtb antigens can help to resist infection. STUDY DESIGN AND METHODS: In the present study, an Mtb 38-kDa antigen-specific T cell receptor (TCR) was isolated from human CD8(+) T cells stimulated by 38-kDa antigen in vitro, and then transduced into primary iNKT cells by retrovirus vector. RESULTS: The TCR gene-modified iNKT cells are endowed with new features to behave as a conventional MHC class I restricted CD8(+) T lymphocyte by displaying specific antigen recognition and anti-Mtb antigen activity in vitro. At the same time, the engineered iNKT cells retaining its original capacity to be stimulated proliferation by non-protein antigens α-Gal-Cer. CONCLUSIONS: This work is the first attempt to engineer iNKT cells by exogenous TCR genes and demonstrated that iNKT cell, as well as CD4(+) and CD8(+) T cells, can be genetically engineered to confer them a defined and alternative specificity, which provides new insights into TCR gene therapy for tuberculosis patients, especially those infected with drug-resistant Mtb.


Assuntos
Antígenos de Bactérias/imunologia , Lipoproteínas/imunologia , Mycobacterium tuberculosis/imunologia , Células T Matadoras Naturais/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Tuberculose/terapia , Anticorpos Monoclonais/imunologia , Linfócitos T CD8-Positivos/citologia , Proliferação de Células , Citocinas/metabolismo , Engenharia Genética/métodos , Antígenos HLA-A/metabolismo , Voluntários Saudáveis , Humanos , Ativação Linfocitária/imunologia , Microscopia de Fluorescência , Distribuição Normal , Receptores de Antígenos de Linfócitos T/metabolismo , Retroviridae/genética
13.
J Transl Med ; 12: 114, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24885252

RESUMO

BACKGROUND: Autologous transplantation of modified mesenchymal stem cells (MSCs) is a promising candidate for the treatment of the refractory clinical disease, avascular necrosis of the femoral head (ANFH). Our previous attempts by compounding MSCs with medical fibrin glue to treat ANFH in animal model have achieved excellent effects. However, the underlying molecular mechanism is unclear, especially on the transgenic gene expression. METHODS: Rabbit MSCs were isolated and compounded with fibrin glue. Following degrading of fibrin glue, proliferation, viability, expression of transgenic hepatocyte growth factor gene as well as osteogenic differentiation of MSCs were evaluated together with that of uncompounded MSCs. Fibrin glue-compounded MSCs were transplanted into the lesion of ANFH model, and the therapeutic efficacy was compared with uncompounded MSCs. One-Way ANOVA was used to determine the statistical significance among treatment groups. RESULTS: Fibrin glue compounding will not affect molecular activities of MSCs, including hepatocyte growth factor (HGF) secretion, cell proliferation and viability, and osteogenic differentiation in vitro. When applying fibrin glue-compounded MSCs for the therapy of ANFH in vivo, fibrin glue functioned as a drug delivery system and provided a sustaining microenvironment for MSCs which helped the relatively long-term secretion of HGF in the femoral head lesion and resulted in improved therapeutic efficacy when compared with uncompounded MSCs as indicated by hematoxylin-eosin staining and immunohistochemistry of osteocalcin, CD105 and HGF. CONCLUSION: Transplantation of fibrin glue-compounding MSCs is a promising novel method for ANFH therapy.


Assuntos
Necrose da Cabeça do Fêmur/terapia , Adesivo Tecidual de Fibrina , Fator de Crescimento de Hepatócito/genética , Transplante de Células-Tronco Mesenquimais , Osteogênese , Análise de Variância , Animais , Animais Geneticamente Modificados , Ensaio de Imunoadsorção Enzimática , Microscopia Eletrônica de Varredura , Coelhos
14.
JCI Insight ; 9(1)2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38016036

RESUMO

Tuberculosis has the highest mortality rate worldwide for a chronic infectious disease caused by a single pathogen. RNA-binding proteins (RBPs) are involved in autophagy - a key defense mechanism against Mycobacterium tuberculosis (M. tuberculosis) infection - by modulating RNA stability and forming intricate regulatory networks. However, the functions of host RBPs during M. tuberculosis infection remain relatively unexplored. Zinc finger NFX1-type containing 1 (ZNFX1), a conserved RBP critically involved in immune deficiency diseases and mycobacterial infections, is significantly upregulated in M. tuberculosis-infected macrophages. Here, we aimed to explore the immunoregulatory functions of ZNFX1 during M. tuberculosis infection. We observed that Znfx1 knockout markedly compromised the multifaceted immune responses mediated by macrophages. This compromise resulted in reduced phagocytosis, suppressed macrophage activation, increased M. tuberculosis burden, progressive lung tissue injury, and chronic inflammation in M. tuberculosis-infected mice. Mechanistic investigations revealed that the absence of ZNFX1 inhibited autophagy, consequently mediating immune suppression. ZNFX1 critically maintained AMPK-regulated autophagic flux by stabilizing protein kinase AMP-activated catalytic subunit alpha 2 mRNA, which encodes a key catalytic α subunit of AMPK, through its zinc finger region. This process contributed to M. tuberculosis growth suppression. These findings reveal a function of ZNFX1 in establishing anti-M. tuberculosis immune responses, enhancing our understanding of the roles of RBPs in tuberculosis immunity and providing a promising approach to bolster antituberculosis immunotherapy.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Animais , Camundongos , Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/genética , Macrófagos/metabolismo
15.
AIDS ; 37(1): 33-42, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36281689

RESUMO

OBJECTIVE: Mycobacterium tuberculosis /human immunodeficiency virus (MTB/HIV) coinfection has become an urgent problem in the field of prevention and control of infectious diseases in recent years. Adoptive cellular immunotherapy using antigen-specific T-cell receptor (TCR) engineered T cells which recognize the specific antigen artificially may have tremendous potential in anti-MTB/HIV coinfection. We have previously successfully identified a MTB Ag85B 199-207 and HIV-1 Env 120-128 peptide-bispecific TCR screened out from peripheral blood mononuclear cells of a HLA-A∗0201 + healthy individual and have further studied that how residues on the predicted complementarity determining region (CDR) 3 of the ß chain contribute to the bispecific TCR contact with the peptide-MHC. However, it is not clear which amino acids in the predicted CDR3α of the bispecific TCR play a crucial role in ligand recognition. METHODS: The variants in the CDR3α of the bispecific TCR were generated using alanine substitution. We then evaluated the immune effects of the five variants on T-cell recognition upon encounter with the MTB or HIV-1 antigen. RESULTS: Mutation of two amino acids (E112A, Y115A) in CDR3α of the bispecific TCR caused a markedly diminished T-cell response to antigen, whereas mutation of the other three amino acids (S113A, P114A, S116A) resulted in completely eliminated response. CONCLUSION: This study demonstrates that Ser 113 , Pro 114 and Ser 116 in CDR3α of the bispecific TCR are especially important for antigen recognition. These results will pave the way for the future development of an improved high-affinity bispecific TCR for use in adoptive cellular immunotherapy for MTB/HIV coinfected patients.


Assuntos
Infecções por HIV , HIV-1 , Mycobacterium tuberculosis , Humanos , Regiões Determinantes de Complementaridade/genética , Leucócitos Mononucleares , Infecções por HIV/terapia , Aminoácidos , Sítios de Ligação
16.
Int Immunopharmacol ; 120: 110291, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37182451

RESUMO

OBJECTIVE: Tuberculosis is the leading killer among the chronic single-source infectious diseases. Mycobacterium tuberculosis can induce necrotic-dominant multiple modes of cell death in macrophages, which accelerates bacterium dissemination and expands tissue injury in host lungs. Mining drugs to counteract Mycobacterium tuberculosis-induced cell death would be beneficial to tuberculosis patients. METHODS: In this study, the protective drug was screened out from the FDA-approved drug library in Mycobacterium tuberculosis-infected macrophages with CCK-8 assay. The death mode regulated by the drug was identified using transcriptomic sequencing, cytomorphological observation, and in the experimental mouse Mycobacterium tuberculosis-infection model. The functional mechanism was explored using western blot, co-immunoprecipitation, and DARTS assay. The intracellular bacterial survival was detected using colony forming unit assays. RESULTS: Cisatracurium besylate was identified to be highly protective for the viability of macrophages during Mycobacterium tuberculosis infection via inhibiting necroptosis. Cisatracurium besylate prevented RIPK3 to be associated with the executive molecule MLKL for forming the necroptotic complex, resulting in the inhibition of MLKL phosphorylation and pore formation on cell membrane. However, Cisatracurium besylate did not interfere with the association between RIPK3 with its upstream kinase RIPK1 or ZBP1 but regulated RIPK3 autophosphorylation. Moreover, Cisatracurium besylate significantly inhibited the expansion of intracellular Mycobacterium tuberculosis both in vitro and in vivo, which also displayed a strong auxiliary bacteriostatic effect to support the therapeutic efficacy of isoniazid and rifampicin, the first-line anti-tubercular drugs. CONCLUSION: Cisatracurium besylate performs anti-Mycobacterium tuberculosis and anti-necroptotic roles, which potentiates its application to be an adjuvant drug for antituberculosis therapy to assist the battle against drug-resistant tuberculosis.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Camundongos , Animais , Apoptose , Mycobacterium tuberculosis/metabolismo , Isoniazida/farmacologia , Isoniazida/uso terapêutico , Necroptose , Proteínas Quinases/metabolismo , Tuberculose/tratamento farmacológico , Tuberculose/metabolismo , Antibacterianos/farmacologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Macrófagos/metabolismo
17.
Sci Signal ; 16(788): eabm1756, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37279284

RESUMO

Innate immune signaling in macrophages during viral infection is regulated by ISGylation, the covalent attachment of the ubiquitin-like protein interferon-stimulated gene 15 (ISG15) to protein targets. Here, we explored the role of ISGylation in the macrophage response to infection with Mycobacterium tuberculosis. In human and mouse macrophages, the E3 ubiquitin ligases HERC5 and mHERC6, respectively, mediated the ISGylation of the phosphatase PTEN, which promoted its degradation. The decreased abundance of PTEN led to an increase in the activity of the PI3K-AKT signaling pathway, which stimulated the synthesis of proinflammatory cytokines. Bacterial growth was increased in culture and in vivo when human or mouse macrophages were deficient in the major E3 ISG15 ligase. The findings expand the role of ISGylation in macrophages to antibacterial immunity and suggest that HERC5 signaling may be a candidate target for adjunct host-directed therapy in patients with tuberculosis.


Assuntos
Fosfatidilinositol 3-Quinases , Ubiquitina-Proteína Ligases , Animais , Humanos , Camundongos , Antibacterianos , Citocinas/metabolismo , Interferons , Peptídeos e Proteínas de Sinalização Intracelular/genética , PTEN Fosfo-Hidrolase/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinas/metabolismo
18.
J Cell Mol Med ; 16(6): 1260-73, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21831134

RESUMO

Mesenchymal stem cells (MSCs) play a crucial role in tissue repair by secretion of tissue nutrient factors such as hepatocyte growth factor (HGF). However, studies examining the effects of HGF on the proliferation and differentiation of MSCs used different concentrations of HGF and reported conflicting conclusions. This study aimed to determine the mechanisms by which different concentrations of HGF regulate MSC proliferation and osteogenic differentiation, and validate the mechanism in an animal model of early stage avascular necrosis of femoral head (ANFH). Our results demonstrate that a low concentration of HGF (20 ng/ml) preferentially promotes MSC osteogenic differentiation through increased c-Met expression and phosphorylation, Akt pathway activation, and increased expression of p27, Runx2 and Osterix. In contrast, a high concentration of HGF (100 ng/ml) strongly induced proliferation by inducing strong activation of the ERK1/2 signalling pathway. As validated by animal experiments, high localized expression of HGF achieved by transplantation of HGF transgenic MSCs into ANFH rabbits increased the number of MSCs. Subsequently, 2 weeks after transplantation, HGF levels decreased and MSCs differentiated into osteoblasts and resulted in efficient tissue repair. Our results demonstrate that sequential concentration changes in HGF control the proliferation and osteogenic differentiation of MSCs in vivo. This phenomenon can be exploited therapeutically to induce bone regeneration and, in turn, improve the efficacy of pharmacological intervention for ANFH treatment.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Necrose da Cabeça do Fêmur/tratamento farmacológico , Células-Tronco Mesenquimais/metabolismo , Modelos Animais , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Coelhos , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
J Inflamm Res ; 15: 735-746, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35153498

RESUMO

BACKGROUND: As deubiquitinases (DUBs), ubiquitin C-terminal hydrolase (UCH)-L1 has been shown to play a crucial role in regulating diverse biological processes. However, its function in macrophage polarization remains unclear. METHODS: We performed in vivo and in vitro experiments to investigate the role of ubiquitin carboxyl-terminal hydrolase L1 (UCHL1), a kind of DUBs, in macrophage differentiation by using UCHL1-deficiency mice. RESULTS: We demonstrated that LPS stimulation induced UCHL1 expression in macrophages. The deficiency of UCHL1 expression decreased the expression of CD80 and CD86 but increased the expression of CD206. The expression of TNF-α, IL-6, iNOS, and IL-10 was downregulated, while that of Arg1, Ym1, and Fizz1 was upregulated in UCHL1 deficient macrophages. Moreover, we observed that UCHL1 promoted the degradation of p110α through autophagy, but paradoxically increased the activity of AKT, thereby promoting polarization of macrophages into pro-inflammatory states. CONCLUSION: In this study, we identified UCHL1 as a positive regulator of M1 macrophage polarization. Our findings may help in developing therapeutic interventions for the treatment of inflammatory diseases and pathogenic infections.

20.
Sci Signal ; 15(754): eabe1621, 2022 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-36194648

RESUMO

Mycobacterium tuberculosis (Mtb) infection is a long-standing public health threat, and the development of host-directed therapy for eradicating Mtb infection requires better insights into Mtb-host interactions. Viperin [virus-inhibitory protein, endoplasmic reticulum-associated, interferon (IFN) inducible] is an IFN-inducible protein with broad antiviral activities. Here, we demonstrated that Viperin was increased in abundance in patients with lymphatic and pulmonary tuberculosis (TB). Viperin-deficient mice had decreased Mtb bacterial loads and enhanced macrophage responses compared with their wild-type counterparts. Viperin suppressed the formation of a complex containing interleukin-1 receptor-associated kinase 1, TNF receptor-associated factor 6, and transforming growth factor ß-activated kinase 1 (TAK1) and inhibited the TAK1-dependent activation of IκB kinase α/ß, thereby impairing the production of nitric oxide and proinflammatory cytokines. These results suggest that Viperin promotes Mtb infection by inhibiting host innate immune responses in macrophages, suggesting that Viperin may be a candidate target for adjunct host-directed therapy in patients with TB.


Assuntos
Quinases Associadas a Receptores de Interleucina-1 , Fator 6 Associado a Receptor de TNF , Animais , Antivirais/metabolismo , Citocinas/metabolismo , Quinase I-kappa B/metabolismo , Imunidade Inata , Interferons/metabolismo , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , MAP Quinase Quinase Quinases , Camundongos , Óxido Nítrico/metabolismo , Proteínas , Fator 6 Associado a Receptor de TNF/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA