Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.430
Filtrar
Mais filtros

Coleção CLAP
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 33: 823-74, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25706096

RESUMO

Patients with autoinflammatory diseases present with noninfectious fever flares and systemic and/or disease-specific organ inflammation. Their excessive proinflammatory cytokine and chemokine responses can be life threatening and lead to organ damage over time. Studying such patients has revealed genetic defects that have helped unravel key innate immune pathways, including excessive IL-1 signaling, constitutive NF-κB activation, and, more recently, chronic type I IFN signaling. Discoveries of monogenic defects that lead to activation of proinflammatory cytokines have inspired the use of anticytokine-directed treatment approaches that have been life changing for many patients and have led to the approval of IL-1-blocking agents for a number of autoinflammatory conditions. In this review, we describe the genetically characterized autoinflammatory diseases, we summarize our understanding of the molecular pathways that drive clinical phenotypes and that continue to inspire the search for novel treatment targets, and we provide a conceptual framework for classification.


Assuntos
Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Predisposição Genética para Doença , Inflamação/genética , Inflamação/imunologia , Animais , Doenças Autoimunes/metabolismo , Autoimunidade , Modelos Animais de Doenças , Humanos , Imunidade Inata , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/metabolismo , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Interferons/metabolismo , Interleucina-1/metabolismo , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais
2.
Cell ; 185(21): 3980-3991.e18, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36182704

RESUMO

Simian arteriviruses are endemic in some African primates and can cause fatal hemorrhagic fevers when they cross into primate hosts of new species. We find that CD163 acts as an intracellular receptor for simian hemorrhagic fever virus (SHFV; a simian arterivirus), a rare mode of virus entry that is shared with other hemorrhagic fever-causing viruses (e.g., Ebola and Lassa viruses). Further, SHFV enters and replicates in human monocytes, indicating full functionality of all of the human cellular proteins required for viral replication. Thus, simian arteriviruses in nature may not require major adaptations to the human host. Given that at least three distinct simian arteriviruses have caused fatal infections in captive macaques after host-switching, and that humans are immunologically naive to this family of viruses, development of serology tests for human surveillance should be a priority.


Assuntos
Arterivirus , Febres Hemorrágicas Virais , Animais , Arterivirus/fisiologia , Febres Hemorrágicas Virais/veterinária , Febres Hemorrágicas Virais/virologia , Humanos , Macaca , Primatas , Zoonoses Virais , Internalização do Vírus , Replicação Viral
3.
Cell ; 184(20): 5163-5178.e24, 2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34559985

RESUMO

Rift Valley fever virus (RVFV) is a zoonotic pathogen with pandemic potential. RVFV entry is mediated by the viral glycoprotein (Gn), but host entry factors remain poorly defined. Our genome-wide CRISPR screen identified low-density lipoprotein receptor-related protein 1 (mouse Lrp1/human LRP1), heat shock protein (Grp94), and receptor-associated protein (RAP) as critical host factors for RVFV infection. RVFV Gn directly binds to specific Lrp1 clusters and is glycosylation independent. Exogenous addition of murine RAP domain 3 (mRAPD3) and anti-Lrp1 antibodies neutralizes RVFV infection in taxonomically diverse cell lines. Mice treated with mRAPD3 and infected with pathogenic RVFV are protected from disease and death. A mutant mRAPD3 that binds Lrp1 weakly failed to protect from RVFV infection. Together, these data support Lrp1 as a host entry factor for RVFV infection and define a new target to limit RVFV infections.


Assuntos
Interações Hospedeiro-Patógeno , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Vírus da Febre do Vale do Rift/fisiologia , Internalização do Vírus , Animais , Especificidade de Anticorpos/imunologia , Sequência de Bases , Encéfalo/patologia , Encéfalo/virologia , Sistemas CRISPR-Cas/genética , Membrana Celular/metabolismo , Células Cultivadas , Glicoproteínas/metabolismo , Glicosaminoglicanos/metabolismo , Glicosilação , Humanos , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/metabolismo , Ligantes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Glicoproteínas de Membrana/metabolismo , Camundongos , Ligação Proteica , Desnaturação Proteica , Febre do Vale de Rift/patologia , Febre do Vale de Rift/prevenção & controle , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/imunologia
4.
Cell ; 184(13): 3486-3501.e21, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34077751

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is a World Health Organization priority pathogen. CCHFV infections cause a highly lethal hemorrhagic fever for which specific treatments and vaccines are urgently needed. Here, we characterize the human immune response to natural CCHFV infection to identify potent neutralizing monoclonal antibodies (nAbs) targeting the viral glycoprotein. Competition experiments showed that these nAbs bind six distinct antigenic sites in the Gc subunit. These sites were further delineated through mutagenesis and mapped onto a prefusion model of Gc. Pairwise screening identified combinations of non-competing nAbs that afford synergistic neutralization. Further enhancements in neutralization breadth and potency were attained by physically linking variable domains of synergistic nAb pairs through bispecific antibody (bsAb) engineering. Although multiple nAbs protected mice from lethal CCHFV challenge in pre- or post-exposure prophylactic settings, only a single bsAb, DVD-121-801, afforded therapeutic protection. DVD-121-801 is a promising candidate suitable for clinical development as a CCHFV therapeutic.


Assuntos
Anticorpos Neutralizantes/imunologia , Febre Hemorrágica da Crimeia/imunologia , Sobreviventes , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Antígenos Virais/metabolismo , Fenômenos Biofísicos , Chlorocebus aethiops , Mapeamento de Epitopos , Epitopos/metabolismo , Feminino , Vírus da Febre Hemorrágica da Crimeia-Congo/imunologia , Febre Hemorrágica da Crimeia/prevenção & controle , Humanos , Imunoglobulina G/metabolismo , Masculino , Camundongos , Testes de Neutralização , Ligação Proteica , Engenharia de Proteínas , Proteínas Recombinantes/imunologia , Células Vero , Proteínas Virais/química
5.
Immunity ; 54(6): 1245-1256.e5, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34004140

RESUMO

We examined how baseline CD4+ T cell repertoire and precursor states impact responses to pathogen infection in humans using primary immunization with yellow fever virus (YFV) vaccine. YFV-specific T cells in unexposed individuals were identified by peptide-MHC tetramer staining and tracked pre- and post-vaccination by tetramers and TCR sequencing. A substantial number of YFV-reactive T cells expressed memory phenotype markers and contained expanded clones in the absence of exposure to YFV. After vaccination, pre-existing YFV-specific T cell populations with low clonal diversity underwent limited expansion, but rare populations with a reservoir of unexpanded TCRs generated robust responses. These altered dynamics reorganized the immunodominance hierarchy and resulted in an overall increase in higher avidity T cells. Thus, instead of further increasing the representation of dominant clones, YFV vaccination recruits rare and more responsive T cells. Our findings illustrate the impact of vaccines in prioritizing T cell responses and reveal repertoire reorganization as a key component of effective vaccination.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vacina contra Febre Amarela/imunologia , Febre Amarela/imunologia , Vírus da Febre Amarela/imunologia , Adulto , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Células Cultivadas , Chlorocebus aethiops , Humanos , Receptores de Antígenos de Linfócitos T/imunologia , Vacinação/métodos , Células Vero , Febre Amarela/virologia
6.
Immunity ; 52(2): 328-341.e5, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32049050

RESUMO

Fever, an evolutionarily conserved physiological response to infection, is also commonly associated with many autoimmune diseases, but its role in T cell differentiation and autoimmunity remains largely unclear. T helper 17 (Th17) cells are critical in host defense and autoinflammatory diseases, with distinct phenotypes and pathogenicity. Here, we show that febrile temperature selectively regulated Th17 cell differentiation in vitro in enhancing interleukin-17 (IL-17), IL-17F, and IL-22 expression. Th17 cells generated under febrile temperature (38.5°C-39.5°C), compared with those under 37°C, showed enhanced pathogenic gene expression with increased pro-inflammatory activities in vivo. Mechanistically, febrile temperature promoted SUMOylation of SMAD4 transcription factor to facilitate its nuclear localization; SMAD4 deficiency selectively abrogated the effects of febrile temperature on Th17 cell differentiation both in vitro and ameliorated an autoimmune disease model. Our results thus demonstrate a critical role of fever in shaping adaptive immune responses with implications in autoimmune diseases.


Assuntos
Temperatura Corporal/imunologia , Febre/imunologia , Células Th17/imunologia , Imunidade Adaptativa , Animais , Diferenciação Celular/imunologia , Núcleo Celular/metabolismo , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Febre/genética , Regulação da Expressão Gênica , Resposta ao Choque Térmico/imunologia , Camundongos , Proteína Smad4/deficiência , Proteína Smad4/metabolismo , Sumoilação , Células Th17/metabolismo
7.
Immunity ; 50(1): 137-151.e6, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30650373

RESUMO

Fever is an evolutionarily conserved response that confers survival benefits during infection. However, the underlying mechanism remains obscure. Here, we report that fever promoted T lymphocyte trafficking through heat shock protein 90 (Hsp90)-induced α4 integrin activation and signaling in T cells. By inducing selective binding of Hsp90 to α4 integrins, but not ß2 integrins, fever increased α4-integrin-mediated T cell adhesion and transmigration. Mechanistically, Hsp90 bound to the α4 tail and activated α4 integrins via inside-out signaling. Moreover, the N and C termini of one Hsp90 molecule simultaneously bound to two α4 tails, leading to dimerization and clustering of α4 integrins on the cell membrane and subsequent activation of the FAK-RhoA pathway. Abolishment of Hsp90-α4 interaction inhibited fever-induced T cell trafficking to draining lymph nodes and impaired the clearance of bacterial infection. Our findings identify the Hsp90-α4-integrin axis as a thermal sensory pathway that promotes T lymphocyte trafficking and enhances immune surveillance during infection.


Assuntos
Febre/imunologia , Proteínas de Choque Térmico HSP90/metabolismo , Integrina alfa4/metabolismo , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Linfócitos T/imunologia , Animais , Carga Bacteriana , Adesão Celular , Movimento Celular , Dimerização , Quinase 1 de Adesão Focal/metabolismo , Vigilância Imunológica , Integrina alfa4/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo
8.
Immunity ; 49(2): 363-374.e10, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30029854

RESUMO

Ebolaviruses cause severe disease in humans, and identification of monoclonal antibodies (mAbs) that are effective against multiple ebolaviruses are important for therapeutics development. Here we describe a distinct class of broadly neutralizing human mAbs with protective capacity against three ebolaviruses infectious for humans: Ebola (EBOV), Sudan (SUDV), and Bundibugyo (BDBV) viruses. We isolated mAbs from human survivors of ebolavirus disease and identified a potent mAb, EBOV-520, which bound to an epitope in the glycoprotein (GP) base region. EBOV-520 efficiently neutralized EBOV, BDBV, and SUDV and also showed protective capacity in relevant animal models of these infections. EBOV-520 mediated protection principally by direct virus neutralization and exhibited multifunctional properties. This study identified a potent naturally occurring mAb and defined key features of the human antibody response that may contribute to broad protection. This multifunctional mAb and related clones are promising candidates for development as broadly protective pan-ebolavirus therapeutic molecules.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/farmacologia , Ebolavirus/imunologia , Glicoproteínas/imunologia , Doença pelo Vírus Ebola/imunologia , Células 3T3 , Adulto , Animais , Células CHO , Linhagem Celular , Chlorocebus aethiops , Cricetulus , Modelos Animais de Doenças , Drosophila , Feminino , Furões , Cobaias , Doença pelo Vírus Ebola/prevenção & controle , Doença pelo Vírus Ebola/virologia , Humanos , Imunoglobulina G/imunologia , Células Jurkat , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células THP-1 , Células Vero
9.
Proc Natl Acad Sci U S A ; 121(39): e2404781121, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39284055

RESUMO

Systemic inflammation elicits sickness behaviors and fever by engaging a complex neuronal circuitry that begins in the preoptic area of the hypothalamus. Ectotherms such as teleost fish display sickness behaviors in response to infection or inflammation, seeking warmer temperatures to enhance survival via behavioral fever responses. To date, the hypothalamus is the only brain region implicated in sickness behaviors and behavioral fever in teleosts. Yet, the complexity of neurobehavioral manifestations underlying sickness responses in teleosts suggests engagement of higher processing areas of the brain. Using in vivo models of systemic inflammation in rainbow trout, we find canonical pyrogenic cytokine responses in the hypothalamus whereas in the telencephalon and the optic tectum il-1b and tnfa expression is decoupled from il-6 expression. Polyamine metabolism changes, characterized by accumulation of putrescine and decreases in spermine and spermidine, are recorded in the telencephalon but not hypothalamus upon systemic injection of bacteria. While systemic inflammation causes canonical behavioral fever in trout, blockade of bacterial polyamine metabolism prior to injection abrogates behavioral fever, polyamine responses, and telencephalic but not hypothalamic cytokine responses. Combined, our work identifies the telencephalon as a neuronal substrate for brain responses to systemic inflammation in teleosts and uncovers the role of polyamines as critical chemical mediators in sickness behaviors.


Assuntos
Inflamação , Oncorhynchus mykiss , Poliaminas , Telencéfalo , Animais , Telencéfalo/metabolismo , Poliaminas/metabolismo , Inflamação/metabolismo , Oncorhynchus mykiss/metabolismo , Oncorhynchus mykiss/imunologia , Neurônios/metabolismo , Hipotálamo/metabolismo , Espermina/metabolismo , Putrescina/metabolismo , Comportamento de Doença/fisiologia , Espermidina/metabolismo
10.
Proc Natl Acad Sci U S A ; 121(10): e2312150121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38412127

RESUMO

African swine fever, one of the major viral diseases of swine, poses an imminent threat to the global pig industry. The high-efficient replication of the causative agent African swine fever virus (ASFV) in various organs in pigs greatly contributes to the disease. However, how ASFV manipulates the cell population to drive high-efficient replication of the virus in vivo remains unclear. Here, we found that the spleen reveals the most severe pathological manifestation with the highest viral loads among various organs in pigs during ASFV infection. By using single-cell-RNA-sequencing technology and multiple methods, we determined that macrophages and monocytes are the major cell types infected by ASFV in the spleen, showing high viral-load heterogeneity. A rare subpopulation of immature monocytes represents the major population infected at late infection stage. ASFV causes massive death of macrophages, but shifts its infection into these monocytes which significantly arise after the infection. The apoptosis, interferon response, and antigen-presentation capacity are inhibited in these monocytes which benefits prolonged infection of ASFV in vivo. Until now, the role of immature monocytes as an important target by ASFV has been overlooked due to that they do not express classical monocyte marker CD14. The present study indicates that the shift of viral infection from macrophages to the immature monocytes is critical for maintaining prolonged ASFV infection in vivo. This study sheds light on ASFV tropism, replication, and infection dynamics, and elicited immune response, which may instruct future research on antiviral strategies.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Suínos , Animais , Vírus da Febre Suína Africana/fisiologia , Baço/patologia , Replicação Viral , Macrófagos/patologia
11.
Proc Natl Acad Sci U S A ; 121(32): e2404536121, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39088396

RESUMO

Alcelaphine gammaherpesvirus 1 (AlHV-1) asymptomatically persists in its natural host, the wildebeest. However, cross-species transmission to cattle results in the induction of an acute and lethal peripheral T cell lymphoma-like disease (PTCL), named malignant catarrhal fever (MCF). Our previous findings demonstrated an essential role for viral genome maintenance in infected CD8+ T lymphocytes but the exact mechanism(s) leading to lymphoproliferation and MCF remained unknown. To decipher how AlHV-1 dysregulates T lymphocytes, we first examined the global phenotypic changes in circulating CD8+ T cells after experimental infection of calves. T cell receptor repertoire together with transcriptomics and epigenomics analyses demonstrated an oligoclonal expansion of infected CD8+ T cells displaying effector and exhaustion gene signatures, including GZMA, GNLY, PD-1, and TOX2 expression. Then, among viral genes expressed in infected CD8+ T cells, we uncovered A10 that encodes a transmembrane signaling protein displaying multiple tyrosine residues, with predicted ITAM and SH3 motifs. Impaired A10 expression did not affect AlHV-1 replication in vitro but rendered AlHV-1 unable to induce MCF. Furthermore, A10 was phosphorylated in T lymphocytes in vitro and affected T cell signaling. Finally, while AlHV-1 mutants expressing mutated forms of A10 devoid of ITAM or SH3 motifs (or both) were able to induce MCF, a recombinant virus expressing a mutated form of A10 unable to phosphorylate its tyrosine residues resulted in the lack of MCF and protected against a wild-type virus challenge. Thus, we could characterize the nature of this γ-herpesvirus-induced PTCL-like disease and identify an essential mechanism explaining its development.


Assuntos
Linfócitos T CD8-Positivos , Gammaherpesvirinae , Animais , Linfócitos T CD8-Positivos/imunologia , Gammaherpesvirinae/genética , Gammaherpesvirinae/imunologia , Bovinos , Febre Catarral Maligna/virologia , Febre Catarral Maligna/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia
12.
Immunity ; 47(1): 183-198.e6, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28723550

RESUMO

Tissue macrophages arise during embryogenesis from yolk-sac (YS) progenitors that give rise to primitive YS macrophages. Until recently, it has been impossible to isolate or derive sufficient numbers of YS-derived macrophages for further study, but data now suggest that induced pluripotent stem cells (iPSCs) can be driven to undergo a process reminiscent of YS-hematopoiesis in vitro. We asked whether iPSC-derived primitive macrophages (iMacs) can terminally differentiate into specialized macrophages with the help of growth factors and organ-specific cues. Co-culturing human or murine iMacs with iPSC-derived neurons promoted differentiation into microglia-like cells in vitro. Furthermore, murine iMacs differentiated in vivo into microglia after injection into the brain and into functional alveolar macrophages after engraftment in the lung. Finally, iPSCs from a patient with familial Mediterranean fever differentiated into iMacs with pro-inflammatory characteristics, mimicking the disease phenotype. Altogether, iMacs constitute a source of tissue-resident macrophage precursors that can be used for biological, pathophysiological, and therapeutic studies.


Assuntos
Técnicas de Cultura de Células/métodos , Hematopoese , Macrófagos/fisiologia , Neurônios/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese
13.
Proc Natl Acad Sci U S A ; 120(37): e2304722120, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37669378

RESUMO

Crimean-Congo hemorrhagic fever (CCHF) caused by CCHF virus (CCHFV) is one of the epidemic-prone diseases prioritized by the World Health Organisation as public health emergency with an urgent need for accelerated research. The trajectory of host response against CCHFV is multifarious and remains unknown. Here, we reported the temporal spectrum of pathogenesis following the CCHFV infection using genome-wide blood transcriptomics analysis followed by advanced systems biology analysis, temporal immune-pathogenic alterations, and context-specific progressive and postinfection genome-scale metabolic models (GSMM) on samples collected during the acute (T0), early convalescent (T1), and convalescent-phase (T2). The interplay between the retinoic acid-inducible gene-I-like/nucleotide-binding oligomerization domain-like receptor and tumor necrosis factor signaling governed the trajectory of antiviral immune responses. The rearrangement of intracellular metabolic fluxes toward the amino acid metabolism and metabolic shift toward oxidative phosphorylation and fatty acid oxidation during acute CCHFV infection determine the pathogenicity. The upregulation of the tricarboxylic acid cycle during CCHFV infection, compared to the noninfected healthy control and between the severity groups, indicated an increased energy demand and cellular stress. The upregulation of glycolysis and pyruvate metabolism potentiated energy generation through alternative pathways associated with the severity of the infection. The downregulation of metabolic processes at the convalescent phase identified by blood cell transcriptomics and single-cell type proteomics of five immune cells (CD4+ and CD8+ T cells, CD14+ monocytes, B cells, and NK cells) potentially leads to metabolic rewiring through the recovery due to hyperactivity during the acute phase leading to post-viral fatigue syndrome.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Humanos , Linfócitos T CD8-Positivos , Regulação para Cima , Metaboloma
14.
Trends Biochem Sci ; 46(5): 378-390, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33423940

RESUMO

Virion assembly is an important step in the life cycle of all viruses. For viruses of the Flavivirus genus, a group of enveloped positive-sense RNA viruses, the assembly step represents one of the least understood processes in the viral life cycle. While assembly is primarily driven by the viral structural proteins, recent studies suggest that several nonstructural proteins also play key roles in coordinating the assembly and packaging of the viral genome. This review focuses on describing recent advances in our understanding of flavivirus virion assembly, including the intermolecular interactions between the viral structural (capsid) and nonstructural proteins (NS2A and NS2B-NS3), host factors, as well as features of the viral genomic RNA required for efficient flavivirus virion assembly.


Assuntos
Flavivirus , RNA Viral/genética , Proteínas não Estruturais Virais/genética , Vírion , Montagem de Vírus
15.
J Biol Chem ; 300(6): 107307, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38657868

RESUMO

African swine fever, caused by the African swine fever virus (ASFV), is a viral hemorrhagic disease that affects domestic pigs and wild boars. ASFV infection causes extensive tissue damage, and the associated mechanism is poorly understood. Pyroptosis is characterized by the activation of inflammatory caspases and pore formation in the cellular plasma membrane, resulting in the release of inflammatory cytokines and cell damage. How ASFV infection regulates pyroptosis remains unclear. Here, using siRNA assay and overexpression methods, we report that ASFV infection regulated pyroptosis by cleaving the pyroptosis execution protein gasdermin A (GSDMA). ASFV infection activated caspase-3 and caspase-4, which specifically cleaved GSDMA at D75-P76 and D241-V242 to produce GSDMA into five fragments, including GSDMA-N1-75, GSDMA-N1-241, and GSDMA-N76-241 fragments at the N-terminal end of GSDMA. Only GSDMA-N1-241, which was produced in the late stage of ASFV infection, triggered pyroptosis and inhibited ASFV replication. The fragments, GSDMA-N1-75 and GSDMA-N76-241, lose the ability to induce pyroptosis. Overall ASFV infection differentially regulates pyroptosis by GSDMA in the indicated phase, which may be conducive to its own replication. Our findings reveal a novel molecular mechanism for the regulation of pyroptosis.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Caspase 3 , Caspases Iniciadoras , Piroptose , Vírus da Febre Suína Africana/metabolismo , Animais , Febre Suína Africana/metabolismo , Febre Suína Africana/virologia , Febre Suína Africana/patologia , Suínos , Caspase 3/metabolismo , Caspase 3/genética , Caspases Iniciadoras/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Ligação a Fosfato/metabolismo , Células HEK293 , Replicação Viral
16.
J Biol Chem ; 300(7): 107453, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38852886

RESUMO

Identification of a conserved G-quadruplex in E165R of ASFVAfrican swine fever virus (ASFV) is a double-stranded DNA arbovirus with high transmissibility and mortality rates. It has caused immense economic losses to the global pig industry. Currently, no effective vaccines or medications are to combat ASFV infection. G-quadruplex (G4) structures have attracted increasing interest because of their regulatory role in vital biological processes. In this study, we identified a conserved G-rich sequence within the E165R gene of ASFV. Subsequently, using various methods, we verified that this sequence could fold into a parallel G4. In addition, the G4-stabilizers pyridostatin and 5,10,15,20-tetrakis-(N-methyl-4-pyridyl) porphin (TMPyP4) can bind and stabilize this G4 structure, thereby inhibiting E165R gene expression, and the inhibitory effect is associated with G4 formation. Moreover, the G4 ligand pyridostatin substantially impeded ASFV proliferation in Vero cells by reducing gene copy number and viral protein expression. These compelling findings suggest that G4 structures may represent a promising and novel antiviral target against ASFV.


Assuntos
Vírus da Febre Suína Africana , Antivirais , Quadruplex G , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/metabolismo , Animais , Chlorocebus aethiops , Células Vero , Antivirais/farmacologia , Antivirais/química , Suínos , Febre Suína Africana/virologia , Febre Suína Africana/metabolismo , Porfirinas/química , Porfirinas/farmacologia , Ácidos Picolínicos/química , Ácidos Picolínicos/farmacologia , Ácidos Picolínicos/metabolismo , Replicação Viral/efeitos dos fármacos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais/química , Aminoquinolinas
17.
J Biol Chem ; : 107758, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39260696

RESUMO

S. Typhi, the cause of typhoid fever, is a bacterial pathogen of substantial global importance. Typhoid toxin is a secreted AB-type toxin that is a key S. Typhi virulence factor encoded within a 5-gene genetic islet. Four genes in this islet have well-defined roles in typhoid toxin biology, however the function of the fifth gene is unknown. Here, we investigate the function of this gene, which we name ttaP. We show that ttaP is co-transcribed with the typhoid toxin subunit cdtB, and we perform genomic analyses that indicate that TtaP is very highly conserved in typhoid toxin islets found in diverse salmonellae. We show that TtaP is a distant homolog of group XIV secreted phospholipase A2 (PLA2) enzymes, and experimentally demonstrate that TtaP is a bona fide PLA2. Sequence and structural analyses indicate that TtaP differs substantially from characterized PLA2s, and thus represents a novel class of PLA2. Secretion assays revealed that TtaP is neither co-secreted with typhoid toxin, nor is it required for toxin secretion. Although TtaP is a phospholipase that remains associated with the S. Typhi cell, assays that probed for altered cell envelope integrity failed to identify any differences between wild-type S. Typhi and a ttaP deletion strain. Collectively, this study identifies a biochemical activity for the lone uncharacterized typhoid toxin islet gene and lays the groundwork for exploring how this gene factors into S. Typhi pathogenesis. This study further identifies a novel class of PLA2, enzymes that have a wide range of industrial applications.

18.
J Biol Chem ; 300(7): 107472, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38879005

RESUMO

African swine fever virus (ASFV) causes severe disease in domestic pigs and wild boars, seriously threatening the development of the global pig industry. Type I interferon (IFN-I) is an important component of innate immunity, inducing the transcription and expression of antiviral cytokines by activating Janus-activated kinase-signal transducer and activator of transcription (STAT). However, the underlying molecular mechanisms by which ASFV antagonizes IFN-I signaling have not been fully elucidated. Therefore, using coimmunoprecipitation, confocal microscopy, and dual luciferase reporter assay methods, we investigated these mechanisms and identified a novel ASFV immunosuppressive protein, pB475L, which interacts with the C-terminal domain of STAT2. Consequently, pB475L inhibited IFN-I signaling by inhibiting STAT1 and STAT2 heterodimerization and nuclear translocation. Furthermore, we constructed an ASFV-B475L7PM mutant strain by homologous recombination, finding that ASFV-B475L7PM attenuated the inhibitory effects on IFN-I signaling compared to ASFV-WT. In summary, this study reveals a new mechanism by which ASFV impairs host innate immunity.


Assuntos
Vírus da Febre Suína Africana , Imunidade Inata , Interferon Tipo I , Fator de Transcrição STAT2 , Transdução de Sinais , Proteínas Virais , Animais , Humanos , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Febre Suína Africana/metabolismo , Febre Suína Africana/genética , Vírus da Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/genética , Células HEK293 , Evasão da Resposta Imune , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT2/metabolismo , Fator de Transcrição STAT2/genética , Suínos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais/imunologia
19.
Mol Biol Evol ; 41(8)2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39191515

RESUMO

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne virus recognized by the World Health Organization as an emerging infectious disease of growing concern. Utilizing phylodynamic and phylogeographic methods, we have reconstructed the origin and transmission patterns of SFTSV lineages and the roles demographic, ecological, and climatic factors have played in shaping its emergence and spread throughout Asia. Environmental changes and fluctuations in tick populations, exacerbated by the widespread use of pesticides, have contributed significantly to its geographic expansion. The increased adaptability of Lineage L2 strains to the Haemaphysalis longicornis vector has facilitated the dispersal of SFTSV through Southeast Asia. Increased surveillance and proactive measures are needed to prevent further spread to Australia, Indonesia, and North America.


Assuntos
Phlebovirus , Filogeografia , Febre Grave com Síndrome de Trombocitopenia , Phlebovirus/genética , Animais , Sudeste Asiático , Febre Grave com Síndrome de Trombocitopenia/virologia , Febre Grave com Síndrome de Trombocitopenia/transmissão , Humanos , Filogenia , Vetores Aracnídeos/virologia , Carrapatos/virologia , Ixodidae/virologia , Espécies Introduzidas
20.
Mol Microbiol ; 121(3): 513-528, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38115201

RESUMO

Intracellular bacteria have evolved mechanisms to invade host cells, establish an intracellular niche that allows survival and replication, produce progeny, and exit the host cell after completion of the replication cycle to infect new target cells. Bacteria exit their host cell by (i) initiation of apoptosis, (ii) lytic cell death, and (iii) exocytosis. While bacterial egress is essential for bacterial spreading and, thus, pathogenesis, we currently lack information about egress mechanisms for the obligate intracellular pathogen C. burnetii, the causative agent of the zoonosis Q fever. Here, we demonstrate that C. burnetii inhibits host cell apoptosis early during infection, but induces and/or increases apoptosis at later stages of infection. Only at later stages of infection did we observe C. burnetii egress, which depends on previously established large bacteria-filled vacuoles and a functional intrinsic apoptotic cascade. The released bacteria are not enclosed by a host cell membrane and can infect and replicate in new target cells. In summary, our data argue that C. burnetii egress in a non-synchronous way at late stages of infection. Apoptosis-induction is important for C. burnetii egress, but other pathways most likely contribute.


Assuntos
Coxiella burnetii , Febre Q , Humanos , Coxiella burnetii/metabolismo , Febre Q/metabolismo , Febre Q/microbiologia , Febre Q/patologia , Apoptose/fisiologia , Transdução de Sinais , Vacúolos/metabolismo , Interações Hospedeiro-Patógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA