Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Biol Chem ; 300(9): 107676, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39151726

RESUMEN

Innate immunity is the body's first line of defense against disease, and regulated cell death is a central component of this response that balances pathogen clearance and inflammation. Cell death pathways are generally categorized as non-lytic and lytic. While non-lytic apoptosis has been extensively studied in health and disease, lytic cell death pathways are also increasingly implicated in infectious and inflammatory diseases and cancers. Staurosporine (STS) is a well-known inducer of non-lytic apoptosis. However, in this study, we observed that STS also induces lytic cell death at later timepoints. Using biochemical assessments with genetic knockouts, pharmacological inhibitors, and gene silencing, we identified that STS triggered PANoptosis via the caspase-8/RIPK3 axis, which was mediated by RIPK1. PANoptosis is a lytic, innate immune cell death pathway initiated by innate immune sensors and driven by caspases and RIPKs through PANoptosome complexes. Deletion of caspase-8 and RIPK3, core components of the PANoptosome complex, protected against STS-induced lytic cell death. Overall, our study identifies STS as a time-dependent inducer of lytic cell death, PANoptosis. These findings emphasize the importance of understanding trigger- and time-specific activation of distinct cell death pathways to advance our understanding of the molecular mechanisms of innate immunity and cell death for clinical translation.


Asunto(s)
Caspasa 8 , Inflamación , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Estaurosporina , Estaurosporina/farmacología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Caspasa 8/metabolismo , Caspasa 8/genética , Animales , Ratones , Humanos , Inflamación/metabolismo , Inflamación/patología , Apoptosis/efectos de los fármacos , Necroptosis/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos
2.
J Immunol ; 210(3): 283-296, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36548461

RESUMEN

Foot-and-mouth disease virus (FMDV) is the causative agent of foot-and-mouth disease, one of the most highly infectious animal viruses throughout the world. The JAK-STAT signaling pathway is a highly conserved pathway for IFN-ß-induced antiviral gene expression. Previous studies have shown that FMDV can strongly suppress the innate immune response. Moreover, although STAT1 and STAT2 (STAT1/2) have been well established in JAK-STAT signaling-induced antiviral gene expression, whether FMDV proteins inhibit IFN-ß-induced JAK-STAT signaling remains poorly understood. In this study, we described the Lb leader protease (Lbpro) of FMDV as a candidate for inhibiting IFN-ß-induced signaling transduction via directly interacting with STAT1/2. We further showed that Lbpro colocalized with STAT1/2 to inhibit their nuclear translocation. Importantly, Lbpro cleaved STAT1/2 to inhibit IFN-ß-induced signal transduction, whereas the catalytically inactive mutant of LC51A (Lbpro with cysteine substituted with alanine at amino acid residue 51) had no effect on the stability of STAT1/2 proteins. The cleavage of the STAT1/2 proteins was also determined during FMDV infection in vitro. Lbpro could cleave the residues between 252 and 502 aa for STAT1 and the site spanning residues 140 - 150 aa (QQHEIESRIL) for STAT2. The in vivo results showed that Lbpro can cleave STAT1/2 in pigs. Overall, our findings suggest that FMDV Lbpro-mediated targeting of STAT1/2 may reveal a novel mechanism for viral immune evasion.


Asunto(s)
Endopeptidasas , Virus de la Fiebre Aftosa , Interferón beta , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Animales , Virus de la Fiebre Aftosa/enzimología , Inmunidad Innata , Péptido Hidrolasas , Transducción de Señal , Porcinos , Interferón beta/inmunología
3.
PLoS One ; 19(2): e0299577, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38412164

RESUMEN

Regulated cell death is a key component of the innate immune response, which provides the first line of defense against infection and homeostatic perturbations. However, cell death can also drive pathogenesis. The most well-defined cell death pathways can be categorized as nonlytic (apoptosis) and lytic (pyroptosis, necroptosis, and PANoptosis). While specific triggers are known to induce each of these cell death pathways, it is unclear whether all cell types express the cell death proteins required to activate these pathways. Here, we assessed the protein expression and compared the responses of immune and non-immune cells of human and mouse origin to canonical pyroptotic (LPS plus ATP), apoptotic (staurosporine), necroptotic (TNF-α plus z-VAD), and PANoptotic (influenza A virus infection) stimuli. When compared to fibroblasts, both mouse and human innate immune cells, macrophages, expressed higher levels of cell death proteins and activated cell death effectors more robustly, including caspase-1, gasdermins, caspase-8, and RIPKs, in response to specific stimuli. Our findings highlight the importance of considering the cell type when examining the mechanisms regulating inflammation and cell death. Improved understanding of the cell types that contain the machinery to execute different forms of cell death and their link to innate immune responses is critical to identify new strategies to target these pathways in specific cellular populations for the treatment of infectious diseases, inflammatory disorders, and cancer.


Asunto(s)
Necroptosis , Piroptosis , Humanos , Animales , Ratones , Apoptosis , Muerte Celular , Caspasa 1
4.
Microbiol Spectr ; 10(2): e0209721, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35254168

RESUMEN

Senecavirus A (SVA) infection induces inflammation in animals, such as fever, diarrhea, vesicles and erosions, and even death. The inflammatory cytokine interleukin-1ß (IL-1ß) plays a pivotal role in inflammatory responses to combat microbes. Although SVA infection can produce inflammatory clinical symptoms, the modulation of IL-1ß production by SVA infection remains unknown at present. Here, both in vitro and in vivo, SVA robustly induced IL-1ß production in macrophages and pigs. Infection performed in NOD-, LRR-, and pyrin domain-containing three (NLRP3) knockdown cells indicated that NLRP3 is essential for SVA-induced IL-1ß secretion. Importantly, we identified that the 1 to 154 amino acid (aa) portion of SVA 3D binds to the NLRP3 NACHT domain to activate NLRP3 inflammasome assembly and IL-1ß secretion. In addition, the SVA 3D protein interacts with IKKα and IKKß to induce NF-κB activation, which facilitates pro-IL-1ß transcription. Meanwhile, 3D induces p65 nucleus entry. Moreover, SVA 3D induces calcium influx and potassium efflux, which triggers IL-1ß secretion. Ion channels might be related to 3D binding with NLRP3, resulting in NLRP3-ASC complex assembly. We found that 3D protein expression induced tissue hemorrhage and swelling in the mice model. Consistently, expression of 3D in mice caused IL-1ß maturation and secretion. In the natural host of pigs, we confirmed that 3D also induced IL-1ß production. Our data reveal a novel mechanism underlying the activation of the NLRP3 inflammasome after SVA 3D expression, which provides clues for controlling pig's inflammation during the SVA infection. IMPORTANCE Inflammation refers to the response of the immune system to viral, bacterial, and fungal infections or other foreign particles in the body, which can involve the production of a wide array of soluble inflammatory mediators. The NLRP3 inflammasome is one of the best-characterized inflammasome leading to IL-1ß production and maturation. Senecavirus A (SVA) is an oncolytic virus that can cause fever, vesicles and erosions, severe fatal diarrhea, and even the sudden death of piglets. In this study, we demonstrated that 1 to 154 aa of SVA polymerase protein 3D interacts with the NACHT domain of NLRP3 to induce IL-1ß production via the NF-κB signaling pathway and ion channel signal. Our study unveils the mechanism underlying the regulation of inflammasome assembly and production of IL-1ß in response to SVA infection that will help better understand the modulation of host inflammation in pathogens invasion and development of the vaccine.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Diarrea , Inflamasomas/metabolismo , Inflamación , Canales Iónicos , Ratones , Ratones Endogámicos NOD , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Picornaviridae , Porcinos
5.
Front Vet Sci ; 8: 697194, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34805327

RESUMEN

Pathogens of viral origin produce a large variety of infectious diseases in livestock. It is essential to establish the best practices in animal care and an efficient way to stop and prevent infectious diseases that impact animal husbandry. So far, the greatest way to combat the disease is to adopt a vaccine policy. In the fight against infectious diseases, vaccines are very popular. Vaccination's fundamental concept is to utilize particular antigens, either endogenous or exogenous to induce immunity against the antigens or cells. In light of how past emerging and reemerging infectious diseases and pandemics were handled, examining the vaccination methods and technological platforms utilized for the animals may provide some useful insights. New vaccine manufacturing methods have evolved because of developments in technology and medicine and our broad knowledge of immunology, molecular biology, microbiology, and biochemistry, among other basic science disciplines. Genetic engineering, proteomics, and other advanced technologies have aided in implementing novel vaccine theories, resulting in the discovery of new ruminant vaccines and the improvement of existing ones. Subunit vaccines, recombinant vaccines, DNA vaccines, and vectored vaccines are increasingly gaining scientific and public attention as the next generation of vaccines and are being seen as viable replacements to conventional vaccines. The current review looks at the effects and implications of recent ruminant vaccine advances in terms of evolving microbiology, immunology, and molecular biology.

6.
J Inflamm Res ; 14: 1145-1163, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33814921

RESUMEN

Inflammation refers to the response of the immune system to viral, bacterial, and fungal infections, or other foreign particles in the body, which can involve the production of a wide array of soluble inflammatory mediators. It is important for the development of many RNA virus-infected diseases. The primary factors through which the infection becomes inflammation involve inflammasome. Inflammasomes are proteins complex that the activation is responsive to specific pathogens, host cell damage, and other environmental stimuli. Inflammasomes bring about the maturation of various pro-inflammatory cytokines such as IL-18 and IL-1ß in order to mediate the innate immune defense mechanisms. Many RNA viruses and their components, such as encephalomyocarditis virus (EMCV) 2B viroporin, the viral RNA of hepatitis C virus, the influenza virus M2 viroporin, the respiratory syncytial virus (RSV) small hydrophobic (SH) viroporin, and the human rhinovirus (HRV) 2B viroporin can activate the Nod-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome to influence the inflammatory response. On the other hand, several viruses use virus-encoded proteins to suppress inflammation activation, such as the influenza virus NS1 protein and the measles virus (MV) V protein. In this review, we summarize how RNA virus infection leads to the activation or inhibition of the NLRP3 inflammasome.

7.
Viruses ; 14(1)2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-35062226

RESUMEN

Foot-and-mouth disease virus (FMDV) infection causes inflammatory clinical symptoms, such as high fever and vesicular lesions, even death of animals. Interleukin-1ß (IL-1ß) is an inflammatory cytokine that plays an essential role in inflammatory responses against viral infection. The viruses have developed multiple strategies to induce the inflammatory responses, including regulation of IL-1ß production. However, the molecular mechanism underlying the induction of IL-1ß by FMDV remains not fully understood. Here, we found that FMDV robustly induced IL-1ß production in macrophages and pigs. Infection of Casp-1 inhibitor-treated cells and NOD-, LRR- and pyrin domain-containing 3 (NLRP3)-knockdown cells indicated that NLRP3 is essential for FMDV-induced IL-1ß secretion. More importantly, we found that FMDV Lpro associates with the NACHT and LRR domains of NLRP3 to promote NLRP3 inflammasome assembly and IL-1ß secretion. Moreover, FMDV Lpro induces calcium influx and potassium efflux, which trigger NLRP3 activation. Our data revealed the mechanism underlying the activation of the NLRP3 inflammasome after FMDV Lpro expression, thus providing insights for the control of FMDV infection-induced inflammation.


Asunto(s)
Endopeptidasas/metabolismo , Virus de la Fiebre Aftosa/metabolismo , Fiebre Aftosa/inmunología , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Línea Celular , Fiebre Aftosa/virología , Humanos , Inflamasomas/metabolismo , Canales Iónicos/metabolismo , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/química , Dominios Proteicos , Porcinos
8.
Pathogens ; 9(9)2020 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-32899635

RESUMEN

Foot-and-mouth disease virus (FMDV) causes a highly contagious vesicular disease in cloven-hoofed livestock that results in severe consequences for international trade, posing a great economic threat to agriculture. The FMDV infection antagonizes the host immune responses via different signaling pathways to achieve immune escape. Strategies to escape the cell immune system are key to effective infection and pathogenesis. This review is focused on summarizing the recent advances to understand how the proteins encoded by FMDV antagonize the host innate and adaptive immune responses.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA