Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Immunity ; 49(5): 873-885.e7, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30366765

RESUMEN

Receptor interacting protein 2 (RIP2) plays a role in sensing intracellular pathogens, but its function in T cells is unclear. We show that RIP2 deficiency in CD4+ T cells resulted in chronic and severe interleukin-17A-mediated inflammation during Chlamydia pneumoniae lung infection, increased T helper 17 (Th17) cell formation in lungs of infected mice, accelerated atherosclerosis, and more severe experimental autoimmune encephalomyelitis. While RIP2 deficiency resulted in reduced conventional Th17 cell differentiation, it led to significantly enhanced differentiation of pathogenic (p)Th17 cells, which was dependent on RORα transcription factor and interleukin-1 but independent of nucleotide oligomerization domain (NOD) 1 and 2. Overexpression of RIP2 resulted in suppression of pTh17 cell differentiation, an effect mediated by its CARD domain, and phenocopied by a cell-permeable RIP2 CARD peptide. Our data suggest that RIP2 has a T cell-intrinsic role in determining the balance between homeostatic and pathogenic Th17 cell responses.


Asunto(s)
Diferenciación Celular/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Células Th17/citología , Células Th17/metabolismo , Animales , Aterosclerosis , Biomarcadores , Dominio de Reclutamiento y Activación de Caspasas , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/mortalidad , Expresión Génica , Inmunofenotipificación , Inflamación/genética , Inflamación/metabolismo , Interleucina-17/biosíntesis , Interleucina-1beta , Ratones , Ratones Noqueados , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
2.
BMC Cardiovasc Disord ; 24(1): 103, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38350853

RESUMEN

BACKGROUND: The Caspase activation and recruitment domain 8 (CARD8) protein is a component of innate immunity as a negative regulator of NF- ĸB, and has been associated with regulation of proteins involved in inflammation. Expression of CARD8 mRNA and protein has been identified in human atherosclerotic lesions, and the truncated T30A variant (rs2043211) of CARD8 has been associated with lower C-reactive (CRP) and MCP-1 levels in myocardial infarction patients. The present study examines the role of a genetic variation in the CARD8 gene in relation to a selection of markers of inflammation. METHODS: In a cross-sectional study of young healthy individuals (18.0-25.9 yrs, n = 744) the association between the rs2043211 variant in the CARD8 gene and protein markers of inflammation was assessed. Genotyping of the CARD8 C10X (rs2043211) polymorphism was performed with TaqMan real time PCR on DNA from blood samples. Protein levels were studied via Olink inflammation panel ( https://olink.com/ ). Using linear models, we analyzed men and two groups of women with and without estrogen containing contraceptives separately, due to previous findings indicating differences between estrogen users and non-estrogen using women. Genotypes were analyzed by additive, recessive and dominant models. RESULTS: The minor (A) allele of the rs2043211 polymorphism in the CARD8 gene was associated with lower levels of CCL20 and IL-6 in men (CCL20, Additive model: p = 0.023; Dominant model: p = 0.016. IL-6, Additive model: p = 0.042; Dominant model: p = 0.039). The associations remained significant also after adjustment for age and potential intermediate variables. CONCLUSIONS: Our data indicate that CARD8 may be involved in the regulation of CCL20 and IL-6 in men. No such association was observed in women. These findings strengthen and support previous in vitro data on IL-6 and CCL20 and highlight the importance of CARD8 as a factor in the regulation of inflammatory proteins. The reason to the difference between sexes is however not clear, and the influence of estrogen as a possible factor important for the inflammatory response needs to be further explored.


Asunto(s)
Dominio de Reclutamiento y Activación de Caspasas , Predisposición Genética a la Enfermedad , Masculino , Humanos , Femenino , Factores de Riesgo , Estudios Transversales , Interleucina-6/genética , Polimorfismo de Nucleótido Simple , Frecuencia de los Genes , Proteínas Adaptadoras de Señalización CARD/genética , Genotipo , Inflamación/diagnóstico , Inflamación/genética , Estrógenos , Proteínas de Neoplasias/genética
3.
Mol Cell ; 63(3): 498-513, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27494558

RESUMEN

The poly(ADP-ribose) polymerase (PARP) Tankyrase (TNKS and TNKS2) is paramount to Wnt-ß-catenin signaling and a promising therapeutic target in Wnt-dependent cancers. The pool of active ß-catenin is normally limited by destruction complexes, whose assembly depends on the polymeric master scaffolding protein AXIN. Tankyrase, which poly(ADP-ribosyl)ates and thereby destabilizes AXIN, also can polymerize, but the relevance of these polymers has remained unclear. We report crystal structures of the polymerizing TNKS and TNKS2 sterile alpha motif (SAM) domains, revealing versatile head-to-tail interactions. Biochemical studies informed by these structures demonstrate that polymerization is required for Tankyrase to drive ß-catenin-dependent transcription. We show that the polymeric state supports PARP activity and allows Tankyrase to effectively access destruction complexes through enabling avidity-dependent AXIN binding. This study provides an example for regulated signal transduction in non-membrane-enclosed compartments (signalosomes), and it points to novel potential strategies to inhibit Tankyrase function in oncogenic Wnt signaling.


Asunto(s)
Motivo alfa Estéril , Tanquirasas/metabolismo , Vía de Señalización Wnt , Proteína Axina/metabolismo , Sitios de Unión , Dominio de Reclutamiento y Activación de Caspasas , Catálisis , Cristalografía , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Mutación , Poli(ADP-Ribosa) Polimerasas/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Relación Estructura-Actividad , Tanquirasas/química , Tanquirasas/genética , Transfección
4.
J Biol Chem ; 298(3): 101566, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35007535

RESUMEN

ASC is an essential adaptor of the inflammasome, a micrometer-size multiprotein complex that processes proinflammatory cytokines. Inflammasome formation depends on ASC self-association into large assemblies via homotypic interactions of its two death domains, PYD and CARD. ASCb, an alternative splicing isoform, activates the inflammasome to a lesser extent compared with ASC. Thus, it has been postulated that adaptor isoforms differentially regulate inflammasome function. At the amino acid level, ASC and ASCb differ only in the length of the linker connecting the two death domains. To understand inflammasome regulation at the molecular level, we investigated the self-association properties of ASC and ASCb using real-time NMR, dynamic light scattering (DLS), size-exclusion chromatography, and transmission electron microscopy (TEM). The NMR data indicate that ASC self-association is faster than that of ASCb; a kinetic model for this oligomerization results in differing values for both the reaction order and the rate constants. Furthermore, DLS analysis indicates that ASC self-associates into more compact macrostructures compared with ASCb. Finally, TEM data show that ASCb has a reduced tendency to form densely packed filaments relative to ASC. Overall, these differences can only be explained by an effect of the linker length, as the NMR results show structural equivalence of the PYD and CARD in both proteins. The effect of linker length was corroborated by molecular docking with the procaspase-1 CARD domain. Altogether, our results indicate that ASC's faster and less polydisperse polymerization is more efficient, plausibly explaining inflammasome activation differences by ASC isoforms at the molecular level.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD , Inflamasomas , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Dominio de Reclutamiento y Activación de Caspasas , Inflamasomas/metabolismo , Simulación del Acoplamiento Molecular , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Isoformas de Proteínas , Dominio Pirina
5.
Stroke ; 54(5): 1377-1389, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37021569

RESUMEN

BACKGROUND: Neuroinflammation plays extremely crucial roles in the neurological damage mediated by ischemic stroke. TRIM29 (tripartite motif containing 29) has previously been proposed to contribute to the regulation of innate immunity, however, the effect of TRIM29 on ischemic stroke induced neurodegenerative processes and neuroinflammation still largely unexplored. In the current article, we aimed to investigate the function and the precise mechanisms of TRIM29 in ischemic stroke. METHODS: Middle cerebral artery occlusion mice model and oxygen-glucose deprivation cell model were established as in vivo and in vitro models of ischemic stroke. Quantitative real-time polymerase chain reaction (PCR), Western blot, and ELSIA were used to detect the expression levels of TRIM29, cytokines, and marker proteins. Immunofluorescence assay was performed to examine the extent of cell death. Different truncations were generated, and coimmunoprecipitation assays were used to confirm the protein interaction. Ubiquitination assay was performed to detect the ubiquitination levels. RESULTS: We found that the cerebral ischemia-reperfusion induced injury was aggravated in TRIM29 knockout mice after middle cerebral artery occlusion operation as well as the increased neurological deficits score. TRIM29 expression was also found to be up-regulated upon middle cerebral artery occlusion or OGD administration, and loss of TRIM29 promoted the apoptosis and pyroptosis of neurons and microglial cells induced by middle cerebral artery occlusion or OGD, consistent with the enhanced proinflammatory mediators production and activation of NLRC4 (NLR [NOD-like receptor] family CARD [caspase recruitment domain] domain containing protein 4) inflammasome. Furthermore, we observed that TRIM29 interacted with NLRC4 directly and promoted the K48-linked polyubiquitination of NLRC4, lead to the proteasomal degradation of NLRC4. CONCLUSIONS: In conclusion, for the first time, we revealed the role of TRIM29 in ischemic stroke and illustrated the direct relationship between TRIM29 and NLRC4.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , Animales , Ratones , Dominio de Reclutamiento y Activación de Caspasas , Infarto de la Arteria Cerebral Media , Inflamasomas/metabolismo , Microglía/metabolismo , Enfermedades Neuroinflamatorias , Daño por Reperfusión/metabolismo
6.
Cancer Sci ; 114(12): 4511-4520, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37991442

RESUMEN

Eribulin inhibits microtubule polymerization and improves the overall survival of patients with recurrent metastatic breast cancer. A subgroup analysis revealed a low neutrophil to lymphocyte ratio (NLR) (<3) to be a prognostic factor of eribulin treatment. We thus hypothesized that eribulin might be related to the immune response for breast cancer cells and we analyzed the effects of eribulin on the immune system. Immunohistochemical staining revealed that human leukocyte antigen (HLA) class I expression was increased in clinical samples after eribulin treatment. In vitro assays revealed that eribulin treatment increased HLA class I expression in breast cancer line cells. RNA-sequencing demonstrated that eribulin treatment increased the expression of the NOD-like family CARD domain-containing 5 (NLRC5), a master regulator of HLA class I expression. Eribulin treatment increased the NY-ESO-1-specific T-cell receptor (TCR) transduced T (TCR-T) cell response for New York oesophageal squamous cell carcinoma 1 (NY-ESO-1) overexpressed breast cancer cells. The eribulin and TCR-T combined therapy model revealed that eribulin and immunotherapy using TCR-T cells has a synergistic effect. In summary, eribulin increases the expression of HLA class 1 via HLA class 1 transactivatior NLRC5 and eribulin combination with immunotherapy can be effective for the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Proteínas NLR , Dominio de Reclutamiento y Activación de Caspasas , Recurrencia Local de Neoplasia , Receptores de Antígenos de Linfocitos T/metabolismo , Antígenos de Neoplasias , Antígenos HLA , Péptidos y Proteínas de Señalización Intracelular/metabolismo
7.
Apoptosis ; 28(3-4): 313-325, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36652128

RESUMEN

Apoptosis repressor with caspase recruitment domain (ARC) acts as a potent and multifunctional inhibitor of apoptosis, which is mainly expressed in postmitotic cells, including cardiomyocytes. ARC is special for its N-terminal caspase recruitment domain and caspase recruitment domain. Due to the powerful inhibition of apoptosis, ARC is mainly reported to act as a cardioprotective factor during ischaemia‒reperfusion (I/R) injury, preventing cardiomyocytes from being devastated by various catastrophes, including oxidative stress, calcium overload, and mitochondrial dysfunction in the circulatory system. However, recent studies have found that ARC also plays a potential regulatory role in tumorigenesis especially in colorectal cancer and renal cell carcinomas, through multiple apoptosis-associated pathways, which remains to be explored in further studies. Therefore, ARC regulates the body and maintains the balance of physiological activities with its interesting duplex. This review summarizes the current research progress of ARC in the field of tumorigenesis and ischaemia/reperfusion injury, to provide overall research status and new possibilities for researchers.


Asunto(s)
Apoptosis , Daño por Reperfusión , Humanos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Dominio de Reclutamiento y Activación de Caspasas , Daño por Reperfusión/genética , Carcinogénesis/genética , Transformación Celular Neoplásica , Reperfusión
8.
Int J Mol Sci ; 24(3)2023 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-36768499

RESUMEN

ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD)) is the only adaptor involved in the formation of multiple types of inflammasomes. Accumulating evidence demonstrates that ASC plays a critical role in the protection of the host against pathogen infection. In this study, we identified an ASC gene in the large yellow croaker (Larimichthys crocea), namely LcASC, and then investigated the expression characteristics and related signal pathways. On one hand, LcASC has several conserved protein modules, i.e., an N-terminal PYD region, a C-terminal CARD region, and twelve α-helix structures. On the other hand, it has a high variable linker between PYD and CARD domains. Moreover, LcASC has varying degrees of expression in different tissues, among which the highest expression is observed in the spleen followed by the gills and skin. It also shows induced expressions in the head kidney, liver, and spleen following immune stimulation, especially Vibrio Parahaemolyticus infection. Further subcellular localization analysis showed that LcASC formed a clear aggregated speck in the cytoplasm close to the nucleus. In addition, we found 46 DEGs in a comparative transcriptome analysis between the LcASC overexpression group and the control vector group. Notedly, the up-regulated gene Fos and down-regulated gene DOK3 in LcASC overexpressed cells play important roles in the immune system. How ASC contacts these two genes needs to be clarified in upcoming studies. These findings collectively provide new insights into finfish ASC and its potential regulatory signaling pathway as well.


Asunto(s)
Inflamasomas , Perciformes , Animales , Inflamasomas/metabolismo , Dominio de Reclutamiento y Activación de Caspasas , Apoptosis , Proteínas Adaptadoras de Señalización CARD/química , Perciformes/genética , Perciformes/metabolismo , Transducción de Señal
9.
Int J Mol Sci ; 24(21)2023 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-37958977

RESUMEN

CARD9, a scaffolding protein, has been implicated in the pathogenesis of metabolic diseases, including obesity and diabetes. We recently reported novel roles for CARD9 in islet ß-cell dysregulation under duress of gluco (HG)- and glucolipotoxic (GLT) stress. CARD9 expression was also increased in ß-cells following exposure to HG and GLT stress. The current study is aimed at understanding the putative roles of histone deacetylation in HG- and GLT-induced expression of CARD9. Using two structurally distinct inhibitors of histone deacetylases (HDACs), namely trichostatin (TSA) and suberoylanilide hydroxamic acid (SAHA), we provide the first evidence to suggest that the increased expression of CARD9 seen under duress of HG and GLT stress is under the regulatory control of histone deacetylation. Interestingly, the expression of protein kinase Cδ (PKCδ), a known upstream regulator of CARD9 activation, is also increased under conditions of metabolic stress. However, it is resistant to TSA and SAHA, suggesting that it is not regulated via histone deacetylation. Based on these data, we propose that targeting the appropriate HDACs, which mediate the expression (and function) of CARD9, might be the next step to further enhance our current understanding of the roles of CARD9 in islet dysfunction under metabolic stress and diabetes.


Asunto(s)
Diabetes Mellitus , Inhibidores de Histona Desacetilasas , Humanos , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Ácidos Hidroxámicos/farmacología , Dominio de Reclutamiento y Activación de Caspasas , Vorinostat , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Estrés Fisiológico , Proteínas Adaptadoras de Señalización CARD/metabolismo
10.
Zhonghua Yi Xue Za Zhi ; 103(10): 746-751, 2023 Mar 14.
Artículo en Zh | MEDLINE | ID: mdl-36889688

RESUMEN

Objective: To investigate the role of caspase recruitment domain protein 9 (CARD9) in airway injury and inflammation of steroid resistant asthma in C57BL/6 mice. Methods: C57BL/6 mice were divided into A group (control group), B group (model group) and C group (dexamethasone treatment group), with 6 mouse in each group using random number table. The mouse asthma model was established in B and C group by subcutaneous injection of ovalbumin (OVA)/complete Freund adjuvant (CFA) in the abdomen and OVA aerosol challenge, the pathological change and cell count in broncho alveolar lavage fluid (BALF) were detected in order to confirm the model as steroid resistant asthma, and the lung tissue inflammatory infiltration was scored. Western blot was used to detect the changes of CARD9 protein between the group A and B; then wild-type and CARD9 knockout mice were divided into D group (wild-type control group), E group (wild-type model group), F group (CARD9 knockout control group) and G group (CARD9 knockout model group), the following indicators were observed and compared after establishing steroid resistant asthma model separately: HE staining was used to observe the pathological changes of lung tissue, ELISA was used to detect the protein levels of interleukin-4 (IL-4), interleukin-5 (IL-5) and interleukin-17(IL-17) in BALF, and RT-PCR was used to detect the mRNA levels of CXC motif chemokine ligand-10 (CXCL-10) and IL-17 in lung. Results: The inflammatory score (3.33±0.82 vs 0.67±0.52) and BALF total cell count [(10.13±4.83) ×105/ml vs (3.76±0.84) ×105/ml] in B group were higher than those in the A group with statistical significance (P<0.05). There was no significant difference between group C and group B in inflammatory infiltration score (2.83±0.75 vs 3.33±0.82) and BALF total cell count [(9.80±3.19) ×105/ml vs (10.13±4.83) ×105/ml] (P>0.05). Moreover the protein level of CARD9 was increased in the B group than A group (0.245±0.090 vs 0.047±0.014, P=0.004). Compared to E group and F group, more obviously inflammatory cells, neutrophils, eosinophils infiltration and tissue injury were observed in G group (P<0.05), so did the expression of IL-4 (P<0.05), IL-5 and IL-17. Meanwhile the mRNA expression levels of IL-17 and CXCL-10 also increased in lung tissue (P<0.05) of G group. Conclusion: CARD9 gene deletion may aggravate the steroid resistant of asthma by increasing neutrophil chemokines, such as IL-17 and CXCL-10, therefore increasing infiltration of neutrophils in C57BL/6 mice asthma model.


Asunto(s)
Asma , Interleucina-4 , Ratones , Animales , Interleucina-5 , Interleucina-17 , Dominio de Reclutamiento y Activación de Caspasas , Técnicas de Inactivación de Genes , Ratones Endogámicos C57BL , Asma/terapia , Pulmón/patología , Líquido del Lavado Bronquioalveolar , Esteroides , Inflamación , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Ovalbúmina
11.
J Biol Chem ; 295(4): 1153-1164, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31843969

RESUMEN

A critical role of influenza A virus nonstructural protein 1 (NS1) is to antagonize the host cellular antiviral response. NS1 accomplishes this role through numerous interactions with host proteins, including the cytoplasmic pathogen recognition receptor, retinoic acid-inducible gene I (RIG-I). Although the consequences of this interaction have been studied, the complete mechanism by which NS1 antagonizes RIG-I signaling remains unclear. We demonstrated previously that the NS1 RNA-binding domain (NS1RBD) interacts directly with the second caspase activation and recruitment domain (CARD) of RIG-I. We also identified that a single strain-specific polymorphism in the NS1RBD (R21Q) completely abrogates this interaction. Here we investigate the functional consequences of an R21Q mutation on NS1's ability to antagonize RIG-I signaling. We observed that an influenza virus harboring the R21Q mutation in NS1 results in significant up-regulation of RIG-I signaling. In support of this, we determined that an R21Q mutation in NS1 results in a marked deficit in NS1's ability to antagonize TRIM25-mediated ubiquitination of the RIG-I CARDs, a critical step in RIG-I activation. We also observed that WT NS1 is capable of binding directly to the tandem RIG-I CARDs, whereas the R21Q mutation in NS1 significantly inhibits this interaction. Furthermore, we determined that the R21Q mutation does not impede the interaction between NS1 and TRIM25 or NS1RBD's ability to bind RNA. The data presented here offer significant insights into NS1 antagonism of RIG-I and illustrate the importance of understanding the role of strain-specific polymorphisms in the context of this specific NS1 function.


Asunto(s)
Dominio de Reclutamiento y Activación de Caspasas , Proteína 58 DEAD Box/química , Proteína 58 DEAD Box/metabolismo , Proteínas no Estructurales Virales/metabolismo , Células A549 , Secuencia de Aminoácidos , Animales , Perros , Regulación de la Expresión Génica , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Cinética , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Modelos Animales , Modelos Biológicos , Mutación/genética , Fosforilación , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Viral/metabolismo , Especificidad de la Especie , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Replicación Viral
12.
Apoptosis ; 26(1-2): 24-37, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33604728

RESUMEN

Apoptosis repressor with caspase recruitment domain (ARC) is a highly effective and multifunctional inhibitor of apoptosis that is mainly expressed in postmitotic cells such as cardiomyocytes and skeletal muscle cells. ARC contains a C-terminal region rich in proline and glutamic acid residues and an N-terminal caspase recruitment domain (CARD). The CARD is originally described as a protein-binding motif that interacts with caspase through a CARD-CARD interaction. Initially, the inhibitory effect of ARC was only found in apoptosis, however, it was later found that ARC also played a regulatory role in other types of cell death. As a powerful cardioprotective factor, ARC can protect the heart by inhibiting the death of cardiomyocytes in various ways. ARC can reduce the cardiomyocyte apoptotic response to various stresses and injuries, including extrinsic apoptosis induced by death receptor ligands, cellular Ca2+ homeostasis and the dysregulation of endoplasmic reticulum (ER) stress, oxidative stress and hypoxia. In addition, changes in ARC transcription and translation levels in the heart can cause a series of physiological and pathological changes, and ARC can also perform corresponding functions through interactions with other molecules. Although there has been much research on ARC, the functional redundancy among proteins shows that ARC still has much research value. This review summarizes the molecular characteristics of ARC, its roles in the various death modes in cardiomyocytes and the roles of ARC in cardiac pathophysiology. This article also describes the potential therapeutic effect and research prospects of ARC.


Asunto(s)
Apoptosis , Enfermedades Cardiovasculares/fisiopatología , Dominio de Reclutamiento y Activación de Caspasas , Animales , Calcio/metabolismo , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/genética , Caspasas/genética , Caspasas/metabolismo , Muerte Celular , Estrés del Retículo Endoplásmico , Humanos , Estrés Oxidativo
13.
J Biol Chem ; 294(2): 439-452, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30459235

RESUMEN

The inflammasome is a multiprotein complex necessary for the onset of inflammation. The adapter protein ASC assembles inflammasome components by acting as a molecular glue between danger-signal sensors and procaspase-1. The assembly is mediated by ASC self-association and protein interactions via its two Death Domains, PYD and CARD. Truncated versions of ASC have been shown to form filaments, but information on the filaments formed by full-length ASC is needed to construct a meaningful model of inflammasome assembly. To gain insights into this system, we used a combination of transmission EM, NMR, and computational analysis to investigate intact ASC structures. We show that ASC forms ∼6-7-nm-wide filaments that stack laterally to form bundles. The structural characteristics and dimensions of the bundles indicate that both PYD and CARD are integral parts of the filament. A truncated version of ASC with only the CARD domain (ASCCARD) forms different filaments (∼3-4-nm width), providing further evidence that both domains work in concert in filament assembly. Ring-shaped protein particles bound to pre-existing filaments match the size of ASC dimer structures generated by NMR-based protein docking, suggesting that the ASC dimer could be a basic building block for filament formation. Solution NMR binding studies identified the protein surfaces involved in the ASCCARD-ASCCARD interaction. These data provide new insights into the structural underpinnings of the inflammasome and should inform future efforts to interrogate this important biological system.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/química , Proteínas Adaptadoras de Señalización CARD/ultraestructura , Dominio de Reclutamiento y Activación de Caspasas , Dominio Pirina , Apoptosis , Proteínas Adaptadoras de Señalización CARD/inmunología , Humanos , Concentración de Iones de Hidrógeno , Inflamasomas/inmunología , Modelos Moleculares , Conformación Proteica , Conformación Proteica en Hélice alfa , Multimerización de Proteína
14.
J Virol ; 93(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30814289

RESUMEN

Infection by enteroviruses can cause severe neurological complications in humans. The interactions between the enteroviral and host proteins may facilitate the virus replication and be involved in the pathogenicity of infected individuals. It has been shown that human enteroviruses possess various mechanisms to suppress host innate immune responses in infected cells. Previous studies showed that infection by enterovirus 71 (EV71) causes the degradation of MDA5, which is a critical cytoplasmic pathogen sensor in the recognition of picornaviruses for initiating transcription of type I interferons. In the present study, we demonstrated that the RNA-dependent RNA polymerase (RdRP; also denoted 3Dpol) encoded by EV71 interacts with the caspase activation and recruitment domains (CARDs) of MDA5 and plays a role in the inhibition of MDA5-mediated beta interferon (IFN-ß) promoter activation and mRNA expression. In addition, we found that the 3Dpol protein encoded by coxsackievirus B3 also interacted with MDA5 and downregulated the antiviral signaling initiated by MDA5. These findings indicate that enteroviral RdRP may function as an antagonist against the host antiviral innate immune response.IMPORTANCE Infection by enteroviruses causes severe neurological complications in humans. Human enteroviruses possess various mechanisms to suppress the host type I interferon (IFN) response in infected cells to establish viral replication. In the present study, we found that the enteroviral 3Dpol protein (or RdRP), which is a viral RNA-dependent RNA polymerase for replicating viral RNA, plays a role in the inhibition of MDA5-mediated beta interferon (IFN-ß) promoter activation. We further demonstrated that enteroviral 3Dpol protein interacts with the caspase activation and recruitment domains (CARDs) of MDA5. These findings indicate that enteroviral RdRP functions as an antagonist against the host antiviral response.


Asunto(s)
Enterovirus Humano A/metabolismo , Helicasa Inducida por Interferón IFIH1/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Dominio de Reclutamiento y Activación de Caspasas/genética , Dominio de Reclutamiento y Activación de Caspasas/fisiología , Enterovirus/genética , Enterovirus/metabolismo , Enterovirus Humano A/genética , Enterovirus Humano B/metabolismo , Infecciones por Enterovirus/virología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Interferón beta/metabolismo , Interferones/metabolismo , Interferones/fisiología , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , Transducción de Señal , Replicación Viral
15.
Proc Natl Acad Sci U S A ; 114(7): 1542-1547, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28143931

RESUMEN

Mammalian intrinsic apoptosis requires activation of the initiator caspase-9, which then cleaves and activates the effector caspases to execute cell killing. The heptameric Apaf-1 apoptosome is indispensable for caspase-9 activation by together forming a holoenzyme. The molecular mechanism of caspase-9 activation remains largely enigmatic. Here, we report the cryoelectron microscopy (cryo-EM) structure of an apoptotic holoenzyme and structure-guided biochemical analyses. The caspase recruitment domains (CARDs) of Apaf-1 and caspase-9 assemble in two different ways: a 4:4 complex docks onto the central hub of the apoptosome, and a 2:1 complex binds the periphery of the central hub. The interface between the CARD complex and the central hub is required for caspase-9 activation within the holoenzyme. Unexpectedly, the CARD of free caspase-9 strongly inhibits its proteolytic activity. These structural and biochemical findings demonstrate that the apoptosome activates caspase-9 at least in part through sequestration of the inhibitory CARD domain.


Asunto(s)
Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Caspasa 9/metabolismo , Holoenzimas/metabolismo , Apoptosis , Apoptosomas/química , Apoptosomas/ultraestructura , Factor Apoptótico 1 Activador de Proteasas/química , Factor Apoptótico 1 Activador de Proteasas/genética , Caspasa 9/química , Caspasa 9/genética , Dominio de Reclutamiento y Activación de Caspasas/genética , Microscopía por Crioelectrón , Activación Enzimática , Holoenzimas/química , Holoenzimas/ultraestructura , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Dominios Proteicos , Multimerización de Proteína
16.
J Struct Biol ; 205(2): 189-195, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30625366

RESUMEN

Caspase recruitment domain (CARD)-only proteins (COPs), regulate apoptosis, inflammation, and innate immunity. They inhibit the assembly of NOD-like receptor complexes such as the inflammasome and NODosome, which are molecular complexes critical for caspase-1 activation. COPs are known to interact with either caspase-1 CARD or RIP2 CARD via a CARD-CARD interaction, and inhibit caspase-1 activation or further downstream signaling. In addition to the human COPs, Pseudo-ICE, INCA, and ICEBERG, several viruses also contain viral COPs that help them escape the host immune system. To elucidate the molecular mechanism of host immunity inhibition by viral COPs, we solved the structure of a viral COP for the first time. Our structure showed that viral COP forms a structural transformation-mediated dimer, which is unique and has not been reported in any structural study of a CARD domain. Based on the current structure, and the previously solved structures of other death domain superfamily members, we propose that structural transformation-mediated dimerization might be a new strategy for dimer assembly in the death domain superfamily.


Asunto(s)
Proteínas/química , Proteínas/metabolismo , Ranavirus/química , Ranavirus/metabolismo , Apoptosis , Dominio de Reclutamiento y Activación de Caspasas , Dimerización , Humanos
17.
J Cell Biochem ; 120(8): 12534-12543, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30912187

RESUMEN

The innate immune system offers the first line of defense against invading microbial pathogens through the recognition of conserved pathogen-associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs). The host innate immune system through PRRs, the sensors for PAMPs, induces the production of cytokines. Among different families of PRRs, the retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), and its mitochondrial adaptor ie, the mitochondrial antiviral-signaling (MAVS) protein, are crucial for RLR-triggered interferon (IFN) antiviral immunity. Recent studies have shown that the N-terminal caspase recruitment domain (CARD) and transmembrane domain play a pivotal role in oligomerization of black carp MAVS (BcMAVS), crucial for the host innate immune response against viral invasion. In this study, we have used molecular modeling, docking, and molecular dynamics (MD) simulation approaches to shed molecular insights into the oligomerization mechanism of BcMAVSCARD . MD simulation and interaction analysis portrayed that the type-I surface patches of BcMAVS CARD  make the major contribution to the interaction. Moreover, the evidence from surface patches and critical residues involved in the said interaction is found to be similar to that of the human counterpart and requires further investigation for legitimacy. Altogether, our study provided crucial information on oligomerization of BcMAVS CARDs and might be helpful for clarifying the innate immune response against pathogens and downstream signaling in fishes.


Asunto(s)
Dominio de Reclutamiento y Activación de Caspasas , Inmunidad Innata , Proteínas Mitocondriales/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Animales , Carpas/inmunología , Carpas/metabolismo , Biología Computacional , Proteínas de Peces/metabolismo , Conformación Proteica
18.
PLoS Pathog ; 13(1): e1006126, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28125719

RESUMEN

Tuberculosis is a global health problem and at least one-third of the world's population is infected with Mycobacterium tuberculosis (MTB). MTB is a successful pathogen that enhances its own intracellular survival by inhibiting inflammation and arresting phago-lysosomal fusion. We previously demonstrated that Toxoplasma gondii (T. gondii) dense granule antigen (GRA) 7 interacts with TNF receptor-associated factor 6 via Myeloid differentiation primary response gene 88, enabling innate immune responses in macrophages. To extend these studies, we found that GRA7 interacts with host proteins involved in antimicrobial host defense mechanisms as a therapeutic strategy for tuberculosis. Here, we show that protein kinase C (PKC)α-mediated phosphorylation of T. gondii GRA7-I (Ser52) regulates the interaction of GRA7 with PYD domain of apoptosis-associated speck-like protein containing a carboxy-terminal CARD, which is capable of oligomerization and inflammasome activation can lead to antimicrobial defense against MTB. Furthermore, GRA7-III interacted with the PX domain of phospholipase D1, facilitating its enzyme activity, phago-lysosomal maturation, and subsequent antimicrobial activity in a GRA7-III (Ser135) phosphorylation-dependent manner via PKCα. Taken together, these results underscore a previously unrecognized role of GRA7 in modulating antimicrobial host defense mechanism during mycobacterial infection.


Asunto(s)
Antígenos de Protozoos/metabolismo , Mycobacterium/inmunología , Proteína Quinasa C-alfa/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/fisiología , Tuberculosis/inmunología , Animales , Antígenos de Protozoos/genética , Dominio de Reclutamiento y Activación de Caspasas , Diferenciación Celular , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/inmunología , Fosforilación , Proteína Quinasa C-alfa/genética , Proteínas Protozoarias/genética , Dominio Pirina , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Toxoplasma/genética , Toxoplasma/inmunología , Tuberculosis/microbiología
19.
EMBO Rep ; 18(5): 733-744, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28336776

RESUMEN

Caspases are key enzymes responsible for mediating apoptotic cell death. Across species, caspase-2 is the most conserved caspase and stands out due to unique features. Apart from cell death, caspase-2 also regulates autophagy, genomic stability and ageing. Caspase-2 requires dimerization for its activation which is primarily accomplished by recruitment to high molecular weight protein complexes in cells. Here, we demonstrate that apoptosis inhibitor 5 (API5/AAC11) is an endogenous and direct inhibitor of caspase-2. API5 protein directly binds to the caspase recruitment domain (CARD) of caspase-2 and impedes dimerization and activation of caspase-2. Interestingly, recombinant API5 directly inhibits full length but not processed caspase-2. Depletion of endogenous API5 leads to an increase in caspase-2 dimerization and activation. Consistently, loss of API5 sensitizes cells to caspase-2-dependent apoptotic cell death. These results establish API5/AAC-11 as a direct inhibitor of caspase-2 and shed further light onto mechanisms driving the activation of this poorly understood caspase.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 2/metabolismo , Inhibidores de Caspasas/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas Nucleares/metabolismo , Apoptosis , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Autofagia , Caspasa 2/química , Dominio de Reclutamiento y Activación de Caspasas , Cisteína Endopeptidasas/química , Activación Enzimática , Células HeLa , Humanos , Espectrometría de Masas , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Unión Proteica , Multimerización de Proteína
20.
Biochem J ; 475(6): 1177-1196, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29500231

RESUMEN

Caspase-9 is a critical factor in the initiation of apoptosis and as a result is tightly regulated by many mechanisms. Caspase-9 contains a Caspase Activation and Recruitment Domain (CARD), which enables caspase-9 to form a tight interaction with the apoptosome, a heptameric activating platform. The caspase-9 CARD has been thought to be principally involved in recruitment to the apoptosome, but its roles outside this interaction have yet to be uncovered. In this work, we show that the CARD is involved in physical interactions with the catalytic core of caspase-9 in the absence of the apoptosome; this interaction requires a properly formed caspase-9 active site. The active sites of caspases are composed of four extremely mobile loops. When the active-site loops are not properly ordered, the CARD and core domains of caspase-9 do not interact and behave independently, like loosely tethered beads. When the active-site loop bundle is properly ordered, the CARD domain interacts with the catalytic core, forming a single folding unit. Taken together, these findings provide mechanistic insights into a new level of caspase-9 regulation, prompting speculation that the CARD may also play a role in the recruitment or recognition of substrate.


Asunto(s)
Caspasa 9/química , Caspasa 9/metabolismo , Dominio de Reclutamiento y Activación de Caspasas/fisiología , Pliegue de Proteína , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Caspasa 9/genética , Dominio Catalítico/genética , Activación Enzimática/genética , Humanos , Modelos Moleculares , Mutación Missense , Unión Proteica , Multimerización de Proteína/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA