Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
1.
Nat Commun ; 13(1): 105, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013224

RESUMO

Zika virus (ZIKV) infection can be associated with neurological pathologies, such as microcephaly in newborns and Guillain-Barre syndrome in adults. Effective therapeutics are currently not available. As such, a comprehensive understanding of virus-host interactions may guide the development of medications for ZIKV. Here we report a human genome-wide overexpression screen to identify host factors that regulate ZIKV infection and find TMEM120A as a ZIKV restriction factor. TMEM120A overexpression significantly inhibits ZIKV replication, while TMEM120A knockdown increases ZIKV infection in cell lines. Moreover, Tmem120a knockout in mice facilitates ZIKV infection in primary mouse embryonic fibroblasts (MEF) cells. Mechanistically, the antiviral activity of TMEM120A is dependent on STING, as TMEM120A interacts with STING, promotes the translocation of STING from the endoplasmic reticulum (ER) to ER-Golgi intermediate compartment (ERGIC) and enhances the phosphorylation of downstream TBK1 and IRF3, resulting in the expression of multiple antiviral cytokines and interferon-stimulated genes. In summary, our gain-of-function screening identifies TMEM120A as a key activator of the antiviral signaling of STING.


Assuntos
Interações Hospedeiro-Patógeno/genética , Canais Iônicos/genética , Proteínas de Membrana/genética , Infecção por Zika virus/genética , Zika virus/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Linhagem Celular Tumoral , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/virologia , Feminino , Regulação da Expressão Gênica , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Complexo de Golgi/virologia , Hepatócitos/imunologia , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Canais Iônicos/deficiência , Canais Iônicos/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Zika virus/crescimento & desenvolvimento , Zika virus/patogenicidade , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
2.
Immunol Cell Biol ; 99(10): 1067-1076, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34555867

RESUMO

The proinflammatory cytokine tumor necrosis factor (TNF) plays a central role in the host control of mycobacterial infections. Expression and release of TNF are tightly regulated, yet the molecular mechanisms that control the release of TNF by mycobacteria-infected host cells, in particular macrophages, are incompletely understood. Rab GTPases direct the transport of intracellular membrane-enclosed vesicles and are important regulators of macrophage cytokine secretion. Rab6b is known to be predominantly expressed in the brain where it functions in retrograde transport and anterograde vesicle transport for exocytosis. Whether it executes similar functions in the context of immune responses is unknown. Here we show that Rab6b is expressed by primary mouse macrophages, where it localized to the Golgi complex. Infection with Mycobacterium bovis bacille Calmette-Guérin (BCG) resulted in dynamic changes in Rab6b expression in primary mouse macrophages in vitro as well as in organs from infected mice in vivo. We further show that Rab6b facilitated TNF release by M. bovis BCG-infected macrophages, in the absence of discernible impact on Tnf messenger RNA and intracellular TNF protein expression. Our observations identify Rab6b as a positive regulator of M. bovis BCG-induced TNF trafficking and secretion by macrophages and positions Rab6b among the molecular machinery that orchestrates inflammatory cytokine responses by macrophages.


Assuntos
Complexo de Golgi/imunologia , Macrófagos/imunologia , Infecções por Mycobacterium , Fator de Necrose Tumoral alfa/imunologia , Proteínas rab de Ligação ao GTP/imunologia , Animais , Camundongos , Infecções por Mycobacterium/imunologia , Mycobacterium bovis
3.
J Immunol ; 207(2): 421-435, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34233909

RESUMO

Intracellular ion fluxes emerge as critical actors of immunoregulation but still remain poorly explored. In this study, we investigated the role of the redundant cation channels TMEM176A and TMEM176B (TMEM176A/B) in retinoic acid-related orphan receptor γt+ cells and conventional dendritic cells (DCs) using germline and conditional double knockout mice. Although Tmem176a/b appeared surprisingly dispensable for the protective function of Th17 and group 3 innate lymphoid cells in the intestinal mucosa, we found that they were required in conventional DCs for optimal Ag processing and presentation to CD4+ T cells. Using a real-time imaging method, we show that TMEM176A/B accumulate in dynamic post-Golgi vesicles preferentially linked to the late endolysosomal system and strongly colocalize with HLA-DM. Taken together, our results suggest that TMEM176A/B ion channels play a direct role in the MHC class II compartment of DCs for the fine regulation of Ag presentation and naive CD4+ T cell priming.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Proteínas de Membrana/imunologia , Animais , Endossomos/imunologia , Feminino , Genes MHC da Classe II/imunologia , Complexo de Golgi/imunologia , Imunidade Inata/imunologia , Mucosa Intestinal/imunologia , Canais Iônicos/imunologia , Linfócitos/imunologia , Lisossomos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th17/imunologia , Tretinoína/imunologia
4.
Nat Immunol ; 22(4): 497-509, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33790474

RESUMO

Classic major histocompatibility complex class I (MHC-I) presentation relies on shuttling cytosolic peptides into the endoplasmic reticulum (ER) by the transporter associated with antigen processing (TAP). Viruses disable TAP to block MHC-I presentation and evade cytotoxic CD8+ T cells. Priming CD8+ T cells against these viruses is thought to rely solely on cross-presentation by uninfected TAP-functional dendritic cells. We found that protective CD8+ T cells could be mobilized during viral infection even when TAP was absent in all hematopoietic cells. TAP blockade depleted the endosomal recycling compartment of MHC-I molecules and, as such, impaired Toll-like receptor-regulated cross-presentation. Instead, MHC-I molecules accumulated in the ER-Golgi intermediate compartment (ERGIC), sequestered away from Toll-like receptor control, and coopted ER-SNARE Sec22b-mediated vesicular traffic to intersect with internalized antigen and rescue cross-presentation. Thus, when classic MHC-I presentation and endosomal recycling compartment-dependent cross-presentation are impaired in dendritic cells, cell-autonomous noncanonical cross-presentation relying on ERGIC-derived MHC-I counters TAP dysfunction to nevertheless mediate CD8+ T cell priming.


Assuntos
Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Vírus da Influenza A/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Modelos Animais de Doenças , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Feminino , Complexo de Golgi/imunologia , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/patogenicidade , Ativação Linfocitária , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/genética
5.
Mol Cell ; 81(12): 2656-2668.e8, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33930332

RESUMO

A deficient interferon (IFN) response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been implicated as a determinant of severe coronavirus disease 2019 (COVID-19). To identify the molecular effectors that govern IFN control of SARS-CoV-2 infection, we conducted a large-scale gain-of-function analysis that evaluated the impact of human IFN-stimulated genes (ISGs) on viral replication. A limited subset of ISGs were found to control viral infection, including endosomal factors inhibiting viral entry, RNA binding proteins suppressing viral RNA synthesis, and a highly enriched cluster of endoplasmic reticulum (ER)/Golgi-resident ISGs inhibiting viral assembly/egress. These included broad-acting antiviral ISGs and eight ISGs that specifically inhibited SARS-CoV-2 and SARS-CoV-1 replication. Among the broad-acting ISGs was BST2/tetherin, which impeded viral release and is antagonized by SARS-CoV-2 Orf7a protein. Overall, these data illuminate a set of ISGs that underlie innate immune control of SARS-CoV-2/SARS-CoV-1 infection, which will facilitate the understanding of host determinants that impact disease severity and offer potential therapeutic strategies for COVID-19.


Assuntos
Antígenos CD/genética , Interações Hospedeiro-Patógeno/genética , Fatores Reguladores de Interferon/genética , Interferon Tipo I/genética , SARS-CoV-2/genética , Proteínas Virais/genética , Animais , Antígenos CD/química , Antígenos CD/imunologia , Sítios de Ligação , Linhagem Celular Tumoral , Chlorocebus aethiops , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/virologia , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Regulação da Expressão Gênica , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Complexo de Golgi/virologia , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Fatores Reguladores de Interferon/classificação , Fatores Reguladores de Interferon/imunologia , Interferon Tipo I/imunologia , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , SARS-CoV-2/imunologia , Transdução de Sinais , Células Vero , Proteínas Virais/química , Proteínas Virais/imunologia , Internalização do Vírus , Liberação de Vírus/genética , Liberação de Vírus/imunologia , Replicação Viral/genética , Replicação Viral/imunologia
6.
J Clin Invest ; 131(3)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33529166

RESUMO

The coat protein I (COPI) complex mediates retrograde trafficking from the Golgi to the endoplasmic reticulum (ER). Five siblings with persistent bacterial and viral infections and defective humoral and cellular immunity had a homozygous p.K652E mutation in the γ1 subunit of COPI (γ1-COP). The mutation disrupts COPI binding to the KDEL receptor and impairs the retrieval of KDEL-bearing chaperones from the Golgi to the ER. Homozygous Copg1K652E mice had increased ER stress in activated T and B cells, poor antibody responses, and normal numbers of T cells that proliferated normally, but underwent increased apoptosis upon activation. Exposure of the mutants to pet store mice caused weight loss, lymphopenia, and defective T cell proliferation that recapitulated the findings in the patients. The ER stress-relieving agent tauroursodeoxycholic acid corrected the immune defects of the mutants and reversed the phenotype they acquired following exposure to pet store mice. This study establishes the role of γ1-COP in the ER retrieval of KDEL-bearing chaperones and thereby the importance of ER homeostasis in adaptive immunity.


Assuntos
Apoptose/imunologia , Linfócitos B/imunologia , Estresse do Retículo Endoplasmático/imunologia , Ativação Linfocitária , Mutação de Sentido Incorreto , Imunodeficiência Combinada Severa/imunologia , Linfócitos T/imunologia , Substituição de Aminoácidos , Animais , Apoptose/genética , Proteína Coatomer/genética , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Estresse do Retículo Endoplasmático/genética , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Humanos , Camundongos , Camundongos Mutantes , Receptores de Peptídeos/genética , Receptores de Peptídeos/imunologia , Imunodeficiência Combinada Severa/genética
7.
J Clin Invest ; 131(2)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33463537

RESUMO

T cell-mediated responses are dependent on their secretion of key effector molecules. However, the critical molecular determinants of the secretion of these proteins are largely undefined. Here, we demonstrate that T cell activation increases trafficking via the ER-to-Golgi pathway. To study the functional role of this pathway, we generated mice with a T cell-specific deletion in SEC23B, a core subunit of coat protein complex II (COPII). We found that SEC23B critically regulated the T cell secretome following activation. SEC23B-deficient T cells exhibited a proliferative defect and reduced effector functions in vitro, as well as in experimental models of allogeneic and xenogeneic hematopoietic cell transplantation in vivo. However, T cells derived from 3 patients with congenital dyserythropoietic anemia II (CDAII), which results from Sec23b mutation, did not exhibit a similar phenotype. Mechanistic studies demonstrated that unlike murine KO T cells, T cells from patients with CDAII harbor increased levels of the closely related paralog, SEC23A. In vivo rescue of murine KO by expression of Sec23a from the Sec23b genomic locus restored T cell functions. Together, our data demonstrate a critical role for the COPII pathway, with evidence for functional overlap in vivo between SEC23 paralogs in the regulation of T cell immunity in both mice and humans.


Assuntos
Autoimunidade , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/imunologia , Retículo Endoplasmático/imunologia , Complexo de Golgi/imunologia , Linfócitos T/imunologia , Animais , Transporte Biológico Ativo/genética , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/genética , Retículo Endoplasmático/genética , Complexo de Golgi/genética , Humanos , Camundongos , Camundongos Knockout
8.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796072

RESUMO

Guanylate binding protein 5 (GBP5) belongs to the GTPase subfamily, which is mainly induced by interferon gamma (IFN-γ) and is involved in many important cellular processes, including inflammasome activation and innate immunity against a wide variety of microbial pathogens. However, it is unknown whether GBP5 inhibits respiratory syncytial virus (RSV) infection. In this study, we identified GBP5 as an effector of the anti-RSV activity of IFN-γ and found that in children, the weaker immune response, especially the weaker IFN-γ response and the decreased GBP5 expression, leads to RSV susceptibility. Furthermore, we revealed that GBP5 reduced the cell-associated levels of the RSV small hydrophobic (SH) protein, which was identified as a viroporin. In contrast, overexpression of the SH protein rescued RSV replication in the presence of GBP5. The GBP5-induced decrease in intracellular SH protein levels is because GBP5 promotes the release of the SH protein into the cell culture. Moreover, the GBP5 C583A mutants with changes at the C terminus or the GBP5 ΔC mutant lacking the C-terminal region, which impairs GBP5 localization in the Golgi, could not inhibit RSV infection, whereas the GTPase-defective GBP5 maintained RSV inhibition, suggesting that Golgi localization but not the GTPase activity of GBP5 is required for RSV inhibition. Interestingly, we found that RSV infection or RSV G protein downregulates GBP5 expression by upregulating DZIP3, an E3 ligase, which induces GBP5 degradation through the K48 ubiquitination and proteasomal pathways. Thus, this study reveals a complicated interplay between host restrictive factor GBP5 and RSV infection and provides important information for understanding the pathogenesis of RSV.IMPORTANCE RSV is a highly contagious virus that causes multiple infections in infants within their first year of life. It can also easily cause infection in elderly or immunocompromised individuals, suggesting that individual differences in immunity play an important role in RSV infection. Therefore, exploring the pathogenic mechanisms of RSV and identifying essential genes which inhibit RSV infection are necessary to develop an effective strategy to control RSV infection. Here, we report that the IFN-inducible gene GBP5 potently inhibits RSV replication by reducing the cell-associated levels of the RSV small hydrophobic (SH) protein, which is a viroporin. In contrast, the RSV G protein was shown to upregulate the expression of the DZIP3 protein, an E3 ligase that degrades GBP5 through the proteasomal pathway. Our study provides important information for the understanding of the pathogenic mechanisms of RSV and host immunity as well as the complicated interplay between the virus and host.


Assuntos
Proteínas de Ligação ao GTP/genética , Interações Hospedeiro-Patógeno/genética , Interferon gama/genética , Infecções por Vírus Respiratório Sincicial/genética , Vírus Sincicial Respiratório Humano/genética , Proteínas Oncogênicas de Retroviridae/genética , Adulto , Criança , Células Epiteliais/imunologia , Células Epiteliais/virologia , Feminino , Proteínas de Ligação ao GTP/imunologia , Regulação da Expressão Gênica , Complexo de Golgi/imunologia , Complexo de Golgi/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Inflamassomos/genética , Inflamassomos/imunologia , Interferon gama/imunologia , Masculino , Mutação , Complexo de Endopeptidases do Proteassoma/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/crescimento & desenvolvimento , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia , Ubiquitinação , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
9.
FEBS Lett ; 594(17): 2782-2799, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32484234

RESUMO

Intracellular pathogens affect diverse host cellular defence and metabolic pathways. Here, we used infection with Francisella tularensis to identify SON DNA-binding protein as a central determinant of macrophage activities. RNAi knockdown of SON increases survival of human macrophages following F. tularensis infection or inflammasome stimulation. SON is required for macrophage autophagy, interferon response factor 3 expression, type I interferon response and inflammasome-associated readouts. SON knockdown has gene- and stimulus-specific effects on inflammatory gene expression. SON is required for accurate splicing and expression of GBF1, a key mediator of cis-Golgi structure and function. Chemical GBF1 inhibition has similar effects to SON knockdown, suggesting that SON controls macrophage functions at least in part by controlling Golgi-associated processes.


Assuntos
Autofagia/genética , Proteínas de Ligação a DNA/genética , Francisella tularensis/patogenicidade , Complexo de Golgi/imunologia , Fatores de Troca do Nucleotídeo Guanina/genética , Interações Hospedeiro-Patógeno/genética , Macrófagos/imunologia , Antígenos de Histocompatibilidade Menor/genética , Autofagia/efeitos dos fármacos , Morte Celular , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/imunologia , Francisella tularensis/genética , Francisella tularensis/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/microbiologia , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Antígenos de Histocompatibilidade Menor/imunologia , Piridinas/farmacologia , Quinolinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Células THP-1 , Acetato de Tetradecanoilforbol/farmacologia
10.
J Allergy Clin Immunol ; 145(1): 324-334, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31604087

RESUMO

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is an X-linked primary immune deficiency disorder resulting from Wiskott-Aldrich syndrome protein (WASp) deficiency. Lymphocytes from patients with WAS manifest increased DNA damage and lymphopenia from cell death, yet how WASp influences DNA damage-linked cell survival is unknown. A recently described mechanism promoting cell survival after ionizing radiation (IR)-induced DNA damage involves fragmentation and dispersal of the Golgi apparatus, known as the Golgi-dispersal response (GDR), which uses the Golgi phosphoprotein 3 (GOLPH3)-DNA-dependent protein kinase (DNA-PK)-myosin XVIIIA-F-actin signaling pathway. OBJECTIVE: We sought to define WASp's role in the DNA damage-induced GDR and its disruption as a contributor to the development of radiosensitivity-linked immunodeficiency in patients with WAS. METHODS: In human TH and B-cell culture systems, DNA damage-induced GDR elicited by IR or radiomimetic chemotherapy was monitored in the presence or absence of WASp or GOLPH3 alone or both together. RESULTS: WASp deficiency completely prevents the development of IR-induced GDR in human TH and B cells, despite the high DNA damage load. Loss of WASp impedes nuclear translocation of GOLPH3 and its colocalization with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Surprisingly, however, depletion of GOLPH3 alone or depolymerization of F-actin in WASp-sufficient TH cells still allows development of robust GDR, suggesting that WASp, but not GOLPH3, is essential for GDR and cell survival after IR-induced DNA-damage in human lymphocytes. CONCLUSION: The study identifies WASp as a novel effector of the nucleus-to-Golgi cell-survival pathway triggered by IR-induced DNA damage in cells of the hematolymphoid lineage and proposes an impaired GDR as a new cause for development of a "radiosensitive" form of immune dysregulation in patients with WAS.


Assuntos
Linfócitos B/imunologia , Dano ao DNA/imunologia , Complexo de Golgi/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Família de Proteínas da Síndrome de Wiskott-Aldrich/imunologia , Dano ao DNA/genética , Proteína Quinase Ativada por DNA/genética , Proteína Quinase Ativada por DNA/imunologia , Complexo de Golgi/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/imunologia , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética
11.
J Immunol ; 203(6): 1560-1570, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31391232

RESUMO

STING plays central roles in the innate immune response to pathogens that contain DNA. Sensing cytoplasmic DNA by cyclic GMP-AMP synthase produces cyclic GMP-AMP, which binds to and activates STING and induces STING translocation from the endoplasmic reticulum to the perinuclear microsome. However, this trafficking process has not been fully elucidated yet. In this study, we identified YIPF5 as a positive regulator of STING trafficking. YIPF5 is essential for DNA virus- or intracellular DNA-triggered production of type I IFNs. Consistently, knockdown of YIPF5 impairs cellular antiviral responses to DNA virus. Mechanistically, YIPF5 interacts with both STING and components of COPII, facilitating STING recruitment to COPII in the presence of cytoplasmic dsDNA. Furthermore, knockdown of components of COPII inhibits DNA virus-triggered production of type I IFNs, suggesting that COPII is involved in innate immune responses to DNA viruses. Collectively, our findings demonstrate that YIPF5 positively regulates STING-mediated innate immune responses by recruiting STING to COPII-coated vesicles and facilitating STING trafficking from the endoplasmic reticulum to Golgi, providing important insights into the molecular mechanisms of intracellular DNA-stimulated STING trafficking and activation.


Assuntos
Vesículas Revestidas pelo Complexo de Proteína do Envoltório/imunologia , Vírus de DNA/imunologia , Imunidade Inata/imunologia , Proteínas de Membrana/imunologia , Transporte Proteico/imunologia , Transdução de Sinais/imunologia , Proteínas de Transporte Vesicular/imunologia , Animais , DNA Viral/imunologia , Retículo Endoplasmático/imunologia , Complexo de Golgi/imunologia , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL
12.
Clin Chim Acta ; 496: 121-124, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31278896

RESUMO

OBJECTIVE: Anti-golgi antibodies (AGAs) are rarely encountered and often considered in relation to autoimmune diseases in clinical practice. This research was performed for studying the prevalence and clinical significance of AGAs in Chinese population. METHODS: We retrospectively reviewed 22,619 laboratory reports of AGAs detected by indirect immunofluorescence (IIF) were consecutively collected from the First People's Hospital of Wenling between June 2012 and June 2017. Eight patients with AGAs were followed up for relevant clinical and laboratory characteristics. RESULT: A total of 22,619 laboratory reports were collected. Of 19 patients with AGAs, 7 cases (all females) had autoimmune diseases (AID) and 12 cases (6 females and 6 males) had non-AID. High titer AGAs ranging from 1:1000 to 1:3200 were persistently present in AID patients, while low-titer AGAs ranging from 1:100 to 1:320 were transient in non-AID patients. CONCLUSION: This is the first study to assess the AGA positive rate and relevant clinical manifestations in a hospitalized Chinese population. AGAs were rare and occurred in a variety of diseases. They were persistently strongly positive in AID, whereas low-titered and transient in non-AID.


Assuntos
Autoanticorpos/sangue , Complexo de Golgi/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Autoanticorpos/imunologia , Doenças Autoimunes/imunologia , Criança , Pré-Escolar , China , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Prevalência , Estudos Retrospectivos , Adulto Jovem
13.
FASEB J ; 33(5): 6442-6455, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30776316

RESUMO

Cellular stress or injury induces release of endogenous danger signals such as ATP, which plays a central role in activating immune cells. ATP is essential for the release of nonclassically secreted cytokines such as IL-1ß but, paradoxically, has been reported to inhibit the release of classically secreted cytokines such as TNF. Here, we reveal that ATP does switch off soluble TNF (17 kDa) release from LPS-treated macrophages, but rather than inhibiting the entire TNF secretion, ATP packages membrane TNF (26 kDa) within microvesicles (MVs). Secretion of membrane TNF within MVs bypasses the conventional endoplasmic reticulum- and Golgi transport-dependent pathway and is mediated by acid sphingomyelinase. These membrane TNF-carrying MVs are biologically more potent than soluble TNF in vivo, producing significant lung inflammation in mice. Thus, ATP critically alters TNF trafficking and secretion from macrophages, inducing novel unconventional membrane TNF signaling via MVs without direct cell-to-cell contact. These data have crucial implications for this key cytokine, particularly when therapeutically targeting TNF in acute inflammatory diseases.-Soni, S., O'Dea, K. P., Tan, Y. Y., Cho, K., Abe, E., Romano, R., Cui, J., Ma, D., Sarathchandra, P., Wilson, M. R., Takata, M. ATP redirects cytokine trafficking and promotes novel membrane TNF signaling via microvesicles.


Assuntos
Trifosfato de Adenosina/imunologia , Membrana Celular/imunologia , Vesículas Extracelulares/imunologia , Macrófagos/imunologia , Pneumonia/imunologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia , Doença Aguda , Trifosfato de Adenosina/genética , Animais , Comunicação Celular/genética , Comunicação Celular/imunologia , Membrana Celular/genética , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Vesículas Extracelulares/genética , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Knockout , Pneumonia/induzido quimicamente , Pneumonia/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/genética
14.
Biol Pharm Bull ; 41(12): 1757-1768, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30504678

RESUMO

The pentacyclic triterpenoid ursolic acid was previously shown to inhibit the intracellular trafficking of intercellular adhesion molecule-1 (ICAM-1) from the endoplasmic reticulum (ER) to the Golgi apparatus. In the present study, we further investigated the biological activities of three pentacyclic triterpenoids closely related to ursolic acid on the interleukin 1α-induced expression and intracellular trafficking of ICAM-1. In human lung adenocarcinoma A549 cells, asiatic acid, corosolic acid, and maslinic acid interfered with the intracellular transport of ICAM-1 to the cell surface. Endoglycosidase H-sensitive glycans were linked to ICAM-1 in asiatic acid-, corosolic acid-, and maslinic acid-treated cells. Unlike corosolic acid, asiatic acid and maslinic acid increased the amount of the ICAM-1 protein. Moreover, asiatic acid increased the co-localization of ICAM-1 with calnexin (an ER marker), but not GM130 (a cis-Golgi marker). Asiatic acid, corosolic acid, and maslinic acid inhibited yeast α-glucosidase activity, but not Jack bean α-mannosidase activity. These results indicate that asiatic acid, corosolic acid, and maslinic acid interfere with the intracellular transport of ICAM-1 to the cell surface and cause the accumulation of ICAM-1 linked to endoglycosidase H-sensitive glycans.


Assuntos
Retículo Endoplasmático/metabolismo , Glicosilação/efeitos dos fármacos , Complexo de Golgi/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Triterpenos Pentacíclicos/farmacologia , Triterpenos/farmacologia , Células A549 , Citocinas/imunologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/imunologia , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/imunologia , Humanos , Molécula 1 de Adesão Intercelular/genética , Microscopia Confocal , Polissacarídeos/metabolismo , Transporte Proteico
15.
Cell Rep ; 23(5): 1387-1398, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29719252

RESUMO

Selenof (15-kDa selenoprotein; Sep15) is an endoplasmic reticulum (ER)-resident thioredoxin-like oxidoreductase that occurs in a complex with UDP-glucose:glycoprotein glucosyltransferase. We found that Selenof deficiency in mice leads to elevated levels of non-functional circulating plasma immunoglobulins and increased secretion of IgM during in vitro splenic B cell differentiation. However, Selenof knockout animals show neither enhanced bacterial killing capacity nor antigen-induced systemic IgM activity, suggesting that excess immunoglobulins are not functional. In addition, ER-to-Golgi transport of a target glycoprotein was delayed in Selenof knockout embryonic fibroblasts, and proteomic analyses revealed that Selenof deficiency is primarily associated with antigen presentation and ER-to-Golgi transport. Together, the data suggest that Selenof functions as a gatekeeper of immunoglobulins and, likely, other client proteins that exit the ER, thereby supporting redox quality control of these proteins.


Assuntos
Apresentação de Antígeno , Linfócitos B/imunologia , Retículo Endoplasmático/imunologia , Complexo de Golgi/imunologia , Imunoglobulina M/imunologia , Selenoproteínas/imunologia , Animais , Linfócitos B/citologia , Linhagem Celular , Retículo Endoplasmático/genética , Fibroblastos/citologia , Fibroblastos/imunologia , Complexo de Golgi/genética , Imunoglobulina M/genética , Camundongos , Camundongos Knockout , Selenoproteínas/genética , Baço/citologia , Baço/imunologia
16.
Front Immunol ; 9: 853, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29755463

RESUMO

After a viral infection and the stimulation of some pattern-recognition receptors as the toll-like receptor 3 in the endosomes or the RIG-I-like receptors in the cytosol, activation of the IKK-related kinase TBK1 leads to the production of type I interferons (IFNs) after phosphorylation of the transcription factors IRF3 and IRF7. Recent findings indicate an involvement of K63-linked polyubiquitination and of the Golgi-localized protein optineurin (OPTN) in the activation of this crucial kinase involved in innate antiviral immunity. This review summarizes the sensing of viruses and the signaling leading to type I IFN production following TBK1 activation through its ubiquitination and the sensing of ubiquitin chains by OPTN at the Golgi apparatus.


Assuntos
Imunidade Inata , Interferon Tipo I/metabolismo , Transdução de Sinais , Fator de Transcrição TFIIIA/imunologia , Ubiquitinação , Viroses/imunologia , Animais , Proteínas de Ciclo Celular , Complexo de Golgi/imunologia , Humanos , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/imunologia , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/imunologia , Proteínas de Membrana Transportadoras , Camundongos , Fosforilação , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição TFIIIA/genética
17.
Exp Parasitol ; 187: 75-85, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29499180

RESUMO

Like most intracellular pathogens, the apicomplexan parasites Besnoitia besnoiti, Toxoplasma gondii and Neospora caninum scavenge metabolites from their host cells. Recruitment of the Golgi complex to the vicinity of the parasitophorous vacuole (PV) is likely to aid in this process. In this work, we comparatively assessed B. besnoiti, T. gondii and N. caninum infected human retinal pigmented epithelial (hTERT-RPE-1) cells at 24 h post-infection and used antibodies to confirm Golgi ribbon compaction in B. besnoiti, and Golgi ribbon dispersion in T. gondii, while no alteration in Golgi morphology was seen in N. caninum infected cells. In either case, the Golgi stacks of infected cells contained both cis- (GM130) and trans- (TGN46) Golgi proteins. The localization of Rab9A, an important regulator of endosomal trafficking, was also studied. GFP-tagged Rab9A was recruited to the vicinity of the PV of all three parasites. Toxoplasma-infected cells exhibited increased expression of Rab9A in comparison to non-infected cells. However, Rab9A expression levels remained unaltered upon infection with N. caninum and B. besnoiti tachyzoites. In contrast to Rab9A, a GFP-tagged dominant negative mutant form of Rab9A (Rab9A DN), was not recruited to the PV, and the expression of Rab9A DN did not affect host cell invasion nor replication by all three parasites. Thus, B. besnoiti, T. gondii and N. caninum show similarities but also differences in how they affect constituents of the endosomal/secretory pathways.


Assuntos
Coccidiose/metabolismo , Complexo de Golgi/metabolismo , Neospora , Toxoplasmose/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Autoantígenos/imunologia , Western Blotting , Linhagem Celular , Coccidiose/enzimologia , Endossomos/parasitologia , Imunofluorescência , Complexo de Golgi/imunologia , Complexo de Golgi/ultraestrutura , Humanos , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/imunologia , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia de Interferência , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/parasitologia , Toxoplasmose/enzimologia , Rede trans-Golgi/imunologia , Rede trans-Golgi/metabolismo , Rede trans-Golgi/ultraestrutura
18.
Immunology ; 152(2): 232-242, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28518215

RESUMO

Mucosal-associated invariant T (MAIT) cells are conserved T cells that express a semi-invariant T-cell receptor (Vα7.2 in humans and Vα19 in mice). The development of MAIT cells requires the antigen-presenting MHC-related protein 1 (MR1), as well as commensal bacteria. The mechanisms that regulate the functional expression of MR1 molecules and their loading with bacterial antigen in antigen-presenting cells are largely unknown. We have found that treating B cells with the Toll-like receptor 9 (TLR9) agonist CpG increases MR1 surface expression. Interestingly, activation of TLR9 by CpG-A (but not CpG-B) enhances MR1 surface expression. This is limited to B cells and not other types of cells such as monocytes, T or natural killer cells. Knocking-down TLR9 expression by short hairpin RNA reduces MR1 surface expression and MR1-mediated bacterial antigen presentation. CpG-A triggers early endosomal TLR9 activation, whereas CpG-B is responsible for late endosomal/lysosomal activation of TLR9. Consistently, blocking endoplasmic reticulum to Golgi protein transport, rather than lysosomal acidification, suppressed MR1 antigen presentation. Overall, our results indicate that early endosomal TLR9 activation is important for MR1-mediated bacterial antigen presentation.


Assuntos
Apresentação de Antígeno , Antígenos de Bactérias/imunologia , Linfócitos B/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Ativação Linfocitária , Antígenos de Histocompatibilidade Menor/imunologia , Receptor Toll-Like 9/imunologia , Antígenos de Bactérias/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/microbiologia , Linhagem Celular Tumoral , Ilhas de CpG , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Endossomos/imunologia , Endossomos/metabolismo , Complexo de Golgi/imunologia , Complexo de Golgi/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Lisossomos/imunologia , Lisossomos/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Oligonucleotídeos/farmacologia , Transporte Proteico , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Receptor Toll-Like 9/efeitos dos fármacos , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Transfecção
19.
J Steroid Biochem Mol Biol ; 172: 29-35, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28549691

RESUMO

Investigating differentially expressed proteins in a milieu rich in cholesterol oxidation products, we found via mass spectrometry-based proteomics that surface levels of heat shock protein 60 (HSP60) were upregulated on monocytic cells in the presence of 27-hydroxycholesterol (27OHChol). The elevated levels of cytoplasmic membrane HSP60 were verified via Western blot analysis and visualized by confocal microscopy. Treatment with 27OHChol also resulted in increased levels of cellular HSP60 without altering its transcription. Cholesterol, however, did not affect cell-surface levels and cellular amount of HSP60. GSK 2033, an LXR antagonist, inhibited expression of live X receptor α, but not of HSP60, induced by 27OHChol. Treatment with 27OHChol also resulted in increased release of HSP60 from monocytic cells, but the release was significantly reduced by inhibitors of endoplasmic reticulum-Golgi protein trafficking, brefeldin A and monensin. Results of the current study indicate that 27OHChol upregulates not only cell-surface and cellular levels of HSP60 but also its release from monocytic cells, thereby contributing to activation of the immune system.


Assuntos
Chaperonina 60/genética , Hidroxicolesteróis/farmacocinética , Proteínas Mitocondriais/genética , Monócitos/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Brefeldina A/farmacologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/imunologia , Membrana Celular/metabolismo , Chaperonina 60/agonistas , Chaperonina 60/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/imunologia , Complexo de Golgi/metabolismo , Humanos , Hidroxicolesteróis/metabolismo , Imunidade Celular , Receptores X do Fígado/antagonistas & inibidores , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Proteínas Mitocondriais/agonistas , Proteínas Mitocondriais/metabolismo , Monensin/farmacologia , Monócitos/citologia , Monócitos/imunologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Sulfonamidas/farmacologia , Transcrição Gênica
20.
PLoS Biol ; 15(4): e2000653, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28394935

RESUMO

The actin cytoskeleton coordinates the organization of signaling microclusters at the immune synapse (IS); however, the mechanisms involved remain poorly understood. We show here that nitric oxide (NO) generated by endothelial nitric oxide synthase (eNOS) controls the coalescence of protein kinase C-θ (PKC-θ) at the central supramolecular activation cluster (c-SMAC) of the IS. eNOS translocated with the Golgi to the IS and partially colocalized with F-actin around the c-SMAC. This resulted in reduced actin polymerization and centripetal retrograde flow of ß-actin and PKC-θ from the lamellipodium-like distal (d)-SMAC, promoting PKC-θ activation. Furthermore, eNOS-derived NO S-nitrosylated ß-actin on Cys374 and impaired actin binding to profilin-1 (PFN1), as confirmed with the transnitrosylating agent S-nitroso-L-cysteine (Cys-NO). The importance of NO and the formation of PFN1-actin complexes on the regulation of PKC-θ was corroborated by overexpression of PFN1- and actin-binding defective mutants of ß-actin (C374S) and PFN1 (H119E), respectively, which reduced the coalescence of PKC-θ at the c-SMAC. These findings unveil a novel NO-dependent mechanism by which the actin cytoskeleton controls the organization and activation of signaling microclusters at the IS.


Assuntos
Actinas/metabolismo , Sinapses Imunológicas/enzimologia , Isoenzimas/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Profilinas/metabolismo , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional , Linfócitos T/metabolismo , Substituição de Aminoácidos , Linhagem Celular , Células Cultivadas , Cisteína/metabolismo , Ativação Enzimática , Complexo de Golgi/enzimologia , Complexo de Golgi/imunologia , Complexo de Golgi/metabolismo , Humanos , Sinapses Imunológicas/imunologia , Sinapses Imunológicas/metabolismo , Isoenzimas/química , Isoenzimas/genética , Proteínas Luminescentes/antagonistas & inibidores , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mutação , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/genética , Profilinas/genética , Proteína Quinase C/química , Proteína Quinase C/genética , Proteína Quinase C-theta , Transporte Proteico , Pseudópodes , Interferência de RNA , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...