Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.296
Filtrar
1.
Immunobiology ; 229(3): 152810, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38772101

RESUMEN

BACKGROUND AND AIMS: Activation of the cGAS-STING pathway induces the production of type I interferons, initiating the antiviral immune response, which contributes to the clearance of pathogens. Previous studies have shown that STING agonists promote hepatitis B virus (HBV) clearance; however, few studies have investigated the effect of activating the cGAS-STING pathway in macrophages on HBV. METHODS: The polarization status of HBV particle-stimulated RAW264.7 macrophages was analyzed. After stimulation with HBV particles, the analysis focused on determining whether the DNA sensors in RAW264.7 macrophages recognized the viral double-stranded DNA (dsDNA) and evaluating the activation of the cGAS-STING pathway. Coculture of mouse macrophages and hepatocytes harboring HBV was used to study the antiviral activity of HBV-stimulated RAW264.7 macrophages. RESULTS: After stimulation with HBV particles, HBV relaxed circular DNA (rcDNA) was detected in RAW264.7 macrophages, and the protein expression of phospho-STING, phospho-TBK1, and phospho-IRF3 in the STING pathway was increased, as shown by Western blot analysis, which revealed that M1 polarization of macrophages was caused by increased expression of CD86. RT-PCR analyses revealed elevated expression of M1 macrophage polarization-associated cytokines such as TNFα, IL-1ß, iNOS, and IFNα/ß. In the coculture experiment, both HBsAg and HBeAg expression levels were significantly decreased in AML12-HBV1.3 cells cocultured with the supernatants of HBV-stimulated RAW264.7 macrophages. CONCLUSION: The results suggest that macrophages can endocytose HBV particles. Additionally, viral dsDNA can be recognized by DNA pattern recognition receptors, which in turn activate the cGAS-STING pathway, promoting the M1 polarization of macrophages, while no significant M2 polarization is observed. Macrophages stimulated with HBV particles exhibit enhanced antiviral activity against HBV.


Asunto(s)
ADN Viral , Virus de la Hepatitis B , Macrófagos , Proteínas de la Membrana , Nucleotidiltransferasas , Transducción de Señal , Virus de la Hepatitis B/fisiología , Virus de la Hepatitis B/inmunología , Animales , Nucleotidiltransferasas/metabolismo , Ratones , Macrófagos/inmunología , Macrófagos/virología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Células RAW 264.7 , Hepatitis B/inmunología , Hepatitis B/virología , Humanos , Activación de Macrófagos/inmunología , Hepatocitos/virología , Hepatocitos/inmunología , Hepatocitos/metabolismo , Factor 3 Regulador del Interferón/metabolismo
2.
Chin J Nat Med ; 22(5): 402-415, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38796214

RESUMEN

In the realm of autoimmune and inflammatory diseases, the cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) signaling pathway has been thoroughly investigated and established. Despite this, the clinical approval of drugs targeting the cGAS-STING pathway has been limited. The Total glucosides of paeony (TGP) is highly anti-inflammatory and is commonly used in the treatment of rheumatoid arthritis (RA), emerged as a subject of our study. We found that the TGP markedly reduced the activation of the cGAS-STING signaling pathway, triggered by various cGAS-STING agonists, in mouse bone marrow-derived macrophages (BMDMs) and Tohoku Hospital Pediatrics-1 (THP-1) cells. This inhibition was noted alongside the suppression of interferon regulatory factor 3 (IRF3) phosphorylation and the expression of interferon-beta (IFN-ß), C-X-C motif chemokine ligand 10 (CXCL10), and inflammatory mediators such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). The mechanism of action appeared to involve the TGP's attenuation of the STING-IRF3 interaction, without affecting STING oligomerization, thereby inhibiting the activation of downstream signaling pathways. In vivo, the TGP hindered the initiation of the cGAS-STING pathway by the STING agonist dimethylxanthenone-4-acetic acid (DMXAA) and exhibited promising therapeutic effects in a model of acute liver injury induced by lipopolysaccharide (LPS) and D-galactosamine (D-GalN). Our findings underscore the potential of the TGP as an effective inhibitor of the cGAS-STING pathway, offering a new treatment avenue for inflammatory and autoimmune diseases mediated by this pathway.


Asunto(s)
Glucósidos , Factor 3 Regulador del Interferón , Proteínas de la Membrana , Nucleotidiltransferasas , Paeonia , Transducción de Señal , Factor 3 Regulador del Interferón/metabolismo , Animales , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Glucósidos/farmacología , Ratones , Humanos , Paeonia/química , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Transducción de Señal/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Células THP-1
3.
Nat Commun ; 15(1): 4099, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816352

RESUMEN

Chronic inflammation is a major cause of cancer worldwide. Interleukin 33 (IL-33) is a critical initiator of cancer-prone chronic inflammation; however, its induction mechanism by environmental causes of chronic inflammation is unknown. Herein, we demonstrate that Toll-like receptor (TLR)3/4-TBK1-IRF3 pathway activation links environmental insults to IL-33 induction in the skin and pancreas inflammation. An FDA-approved drug library screen identifies pitavastatin to effectively suppress IL-33 expression by blocking TBK1 membrane recruitment/activation through the mevalonate pathway inhibition. Accordingly, pitavastatin prevents chronic pancreatitis and its cancer sequela in an IL-33-dependent manner. The IRF3-IL-33 axis is highly active in chronic pancreatitis and its associated pancreatic cancer in humans. Interestingly, pitavastatin use correlates with a significantly reduced risk of chronic pancreatitis and pancreatic cancer in patients. Our findings demonstrate that blocking the TBK1-IRF3-IL-33 signaling axis suppresses cancer-prone chronic inflammation. Statins present a safe and effective prophylactic strategy to prevent chronic inflammation and its cancer sequela.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Factor 3 Regulador del Interferón , Interleucina-33 , Neoplasias Pancreáticas , Proteínas Serina-Treonina Quinasas , Quinolinas , Transducción de Señal , Interleucina-33/metabolismo , Animales , Factor 3 Regulador del Interferón/metabolismo , Humanos , Neoplasias Pancreáticas/prevención & control , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Quinolinas/farmacología , Quinolinas/uso terapéutico , Inflamación/prevención & control , Inflamación/metabolismo , Pancreatitis Crónica/prevención & control , Pancreatitis Crónica/metabolismo , Receptor Toll-Like 3/metabolismo , Ratones Endogámicos C57BL , Receptor Toll-Like 4/metabolismo , Ácido Mevalónico/metabolismo , Masculino , Femenino , Ratones Noqueados
4.
PLoS Pathog ; 20(5): e1012230, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38776321

RESUMEN

While macrophage is one of the major type I interferon (IFN-I) producers in multiple tissues during viral infections, it also serves as an important target cell for many RNA viruses. However, the regulatory mechanism for the IFN-I response of macrophages to respond to a viral challenge is not fully understood. Here we report ADAP, an immune adaptor protein, is indispensable for the induction of the IFN-I response of macrophages to RNA virus infections via an inhibition of the conjugation of ubiquitin-like ISG15 (ISGylation) to RIG-I. Loss of ADAP increases RNA virus replication in macrophages, accompanied with a decrease in LPS-induced IFN-ß and ISG15 mRNA expression and an impairment in the RNA virus-induced phosphorylation of IRF3 and TBK1. Moreover, using Adap-/- mice, we show ADAP deficiency strongly increases the susceptibility of macrophages to RNA-virus infection in vivo. Mechanically, ADAP selectively interacts and functionally cooperates with RIG-I but not MDA5 in the activation of IFN-ß transcription. Loss of ADAP results in an enhancement of ISGylation of RIG-I, whereas overexpression of ADAP exhibits the opposite effect in vitro, indicating ADAP is detrimental to the RNA virus-induced ISGylation of RIG-I. Together, our data demonstrate a novel antagonistic activity of ADAP in the cell-intrinsic control of RIG-I ISGylation, which is indispensable for initiating and sustaining the IFN-I response of macrophages to RNA virus infections and replication.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteína 58 DEAD Box , Interferón Tipo I , Macrófagos , Ratones Noqueados , Infecciones por Virus ARN , Ubiquitinas , Animales , Macrófagos/virología , Macrófagos/metabolismo , Macrófagos/inmunología , Ratones , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/metabolismo , Ubiquitinas/metabolismo , Ubiquitinas/genética , Proteína 58 DEAD Box/metabolismo , Interferón Tipo I/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Citocinas/metabolismo , Ratones Endogámicos C57BL , Humanos , Receptores Inmunológicos/metabolismo , Interferón beta/metabolismo , Virus ARN/inmunología , Factor 3 Regulador del Interferón/metabolismo
5.
Mol Immunol ; 170: 156-169, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692097

RESUMEN

Type-I and -III interferons play a central role in immune rejection of pathogens and tumors, thus promoting immunogenicity and suppressing tumor recurrence. Double strand RNA is an important ligand that stimulates tumor immunity via interferon responses. Differentiation of embryonic stem cells to pluripotent epithelial cells activates the interferon response during development, raising the question of whether epithelial vs. mesenchymal gene signatures in cancer potentially regulate the interferon pathway as well. Here, using genomics and signaling approaches, we show that Grainyhead-like-2 (GRHL2), a master programmer of epithelial cell identity, promotes type-I and -III interferon responses to double-strand RNA. GRHL2 enhanced the activation of IRF3 and relA/NF-kB and the expression of IRF1; a functional GRHL2 binding site in the IFNL1 promoter was also identified. Moreover, time to recurrence in breast cancer correlated positively with GRHL2 protein expression, indicating that GRHL2 is a tumor recurrence suppressor, consistent with its enhancement of interferon responses. These observations demonstrate that epithelial cell identity supports interferon responses in the context of cancer.


Asunto(s)
Neoplasias de la Mama , Proteínas de Unión al ADN , Factores de Transcripción , Humanos , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Femenino , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Recurrencia Local de Neoplasia/inmunología , Interferones/metabolismo , Interferones/inmunología , Interferones/genética , Línea Celular Tumoral , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Animales , ARN Bicatenario/inmunología , Factor de Transcripción ReIA/metabolismo , Ratones , Regulación Neoplásica de la Expresión Génica , Transducción de Señal/inmunología , Factor 1 Regulador del Interferón/metabolismo , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología
6.
Arterioscler Thromb Vasc Biol ; 44(6): 1365-1378, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38695170

RESUMEN

BACKGROUND: Macrophages play a crucial role in atherosclerotic plaque formation, and the death of macrophages is a vital factor in determining the fate of atherosclerosis. GSDMD (gasdermin D)-mediated pyroptosis is a programmed cell death, characterized by membrane pore formation and inflammatory factor release. METHODS: ApoE-/- and Gsdmd-/- ApoE-/- mice, bone marrow transplantation, and AAV (adeno-associated virus serotype 9)-F4/80-shGSDMD (shRNA-GSDMD) were used to examine the effect of macrophage-derived GSDMD on atherosclerosis. Single-cell RNA sequencing was used to investigate the changing profile of different cellular components and the cellular localization of GSDMD during atherosclerosis. RESULTS: First, we found that GSDMD is activated in human and mouse atherosclerotic plaques and Gsdmd-/- attenuates the atherosclerotic lesion area in high-fat diet-fed ApoE-/- mice. We performed single-cell RNA sequencing of ApoE-/- and Gsdmd-/- ApoE-/- mouse aortas and showed that GSDMD is principally expressed in atherosclerotic macrophages. Using bone marrow transplantation and AAV-F4/80-shGSDMD, we identified the potential role of macrophage-derived GSDMD in aortic pyroptosis and atherosclerotic injuries in vivo. Mechanistically, GSDMD contributes to mitochondrial perforation and mitochondrial DNA leakage and subsequently activates the STING (stimulator of interferon gene)-IRF3 (interferon regulatory factor 3)/NF-κB (nuclear factor kappa B) axis. Meanwhile, GSDMD regulates the STING pathway activation and macrophage migration via cytokine secretion. Inhibition of GSDMD with GSDMD-specific inhibitor GI-Y1 (GSDMD inhibitor Y1) can effectively alleviate the progression of atherosclerosis. CONCLUSIONS: Our study has provided a novel macrophage-derived GSDMD mechanism in the promotion of atherosclerosis and demonstrated that GSDMD can be a potential therapeutic target for atherosclerosis.


Asunto(s)
Aterosclerosis , Modelos Animales de Enfermedad , Factor 3 Regulador del Interferón , Péptidos y Proteínas de Señalización Intracelular , Macrófagos , Proteínas de la Membrana , Ratones Endogámicos C57BL , Mitocondrias , FN-kappa B , Proteínas de Unión a Fosfato , Piroptosis , Transducción de Señal , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/genética , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de Unión a Fosfato/metabolismo , Proteínas de Unión a Fosfato/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Ratones , FN-kappa B/metabolismo , Mitocondrias/metabolismo , Mitocondrias/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones Noqueados para ApoE , Placa Aterosclerótica , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/prevención & control , Gasderminas
7.
Inflamm Res ; 73(6): 897-913, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38625657

RESUMEN

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


Asunto(s)
Factor 3 Regulador del Interferón , Interferón gamma , Macrófagos , Ratones Endogámicos C57BL , Mycobacterium tuberculosis , Proteínas Serina-Treonina Quinasas , Proteínas , Transducción de Señal , Animales , Interferón gamma/metabolismo , Interferón gamma/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Mycobacterium tuberculosis/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Ratones , Proteínas/genética , Proteínas/metabolismo , Quinasa I-kappa B/metabolismo , Quinasas Janus/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Ratones Noqueados , Tuberculosis/inmunología , Pulmón/inmunología , Pulmón/microbiología , Proteína Viperina
8.
Biochem Biophys Res Commun ; 712-713: 149915, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38663038

RESUMEN

Viral infections pose a significant threat to public health, and the production of interferons represents one of the most critical antiviral innate immune responses of the host. Consequently, the screening and identification of compounds or reagents that induce interferon production are of paramount importance. This study commenced with the cultivation of host bacterium 15,597, followed by the infection of Escherichia coli with the MS2 bacteriophage. Utilizing the J2 capture technique, a class of dsRNA mixtures (MS2+15,597) was isolated from the E. coli infected with the MS2 bacteriophage. Subsequent investigations were conducted on the immunostimulatory activity of the MS2+15,597 mixture. The results indicated that the dsRNA mixtures (MS2+15,597) extracted from E. coli infected with the MS2 bacteriophage possess the capability to activate innate immunity, thereby inducing the production of interferon-ß. These dsRNA mixtures can activate the RIG-I and TLR3 pattern recognition receptors, stimulating the expression of interferon stimulatory factors 3/7, which in turn triggers the NF-κB signaling pathway, culminating in the cellular production of interferon-ß to achieve antiviral effects. This study offers novel insights and strategies for the development of broad-spectrum antiviral drugs, potentially providing new modalities for future antiviral therapies.


Asunto(s)
Escherichia coli , Levivirus , ARN Bicatenario , Escherichia coli/virología , Escherichia coli/genética , Escherichia coli/metabolismo , ARN Bicatenario/metabolismo , Humanos , Levivirus/genética , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 3/genética , Inmunidad Innata , Interferón beta/metabolismo , Interferón beta/genética , FN-kappa B/metabolismo , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/genética , Transducción de Señal , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Receptores Inmunológicos , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética
9.
Cell Mol Biol Lett ; 29(1): 61, 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38671352

RESUMEN

BACKGROUND: Macrophage proinflammatory activation contributes to the pathology of severe acute pancreatitis (SAP) and, simultaneously, macrophage functional changes, and increased pyroptosis/necrosis can further exacerbate the cellular immune suppression during the process of SAP, where cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) plays an important role. However, the function and mechanism of cGAS-STING in SAP-induced lung injury (LI) remains unknown. METHODS: Lipopolysaccharide (LPS) was combined with caerulein-induced SAP in wild type, cGAS -/- and sting -/- mice. Primary macrophages were extracted via bronchoalveolar lavage and peritoneal lavage. Ana-1 cells were pretreated with LPS and stimulated with nigericin sodium salt to induce pyroptosis in vitro. RESULTS: SAP triggered NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation-mediated pyroptosis of alveolar and peritoneal macrophages in mouse model. Knockout of cGAS/STING could ameliorate NLRP3 activation and macrophage pyroptosis. In addition, mitochondrial (mt)DNA released from damaged mitochondria further induced macrophage STING activation in a cGAS- and dose-dependent manner. Upregulated STING signal can promote NLRP3 inflammasome-mediated macrophage pyroptosis and increase serum interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α levels and, thus, exacerbate SAP-associated LI (SAP-ALI). Downstream molecules of STING, IRF7, and IRF3 connect the mtDNA-cGAS-STING axis and the NLRP3-pyroptosis axis. CONCLUSIONS: Negative regulation of any molecule in the mtDNA-cGAS-STING-IRF7/IRF3 pathway can affect the activation of NLRP3 inflammasomes, thereby reducing macrophage pyroptosis and improving SAP-ALI in mouse model.


Asunto(s)
ADN Mitocondrial , Factor 3 Regulador del Interferón , Lesión Pulmonar , Macrófagos , Proteínas de la Membrana , Nucleotidiltransferasas , Pancreatitis , Piroptosis , Transducción de Señal , Animales , Piroptosis/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Ratones , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Pancreatitis/metabolismo , Pancreatitis/genética , Pancreatitis/patología , Pancreatitis/inducido químicamente , Macrófagos/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Inflamasomas/metabolismo , Lipopolisacáridos , Masculino , Modelos Animales de Enfermedad
10.
J Cell Sci ; 137(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38682259

RESUMEN

SARS-CoV-2 interferes with antigen presentation by downregulating major histocompatibility complex (MHC) II on antigen-presenting cells, but the mechanism mediating this process is unelucidated. Herein, analysis of protein and gene expression in human antigen-presenting cells reveals that MHC II is downregulated by the SARS-CoV-2 main protease, NSP5. This suppression of MHC II expression occurs via decreased expression of the MHC II regulatory protein CIITA. CIITA downregulation is independent of the proteolytic activity of NSP5, and rather, NSP5 delivers HDAC2 to the transcription factor IRF3 at an IRF-binding site within the CIITA promoter. Here, HDAC2 deacetylates and inactivates the CIITA promoter. This loss of CIITA expression prevents further expression of MHC II, with this suppression alleviated by ectopic expression of CIITA or knockdown of HDAC2. These results identify a mechanism by which SARS-CoV-2 limits MHC II expression, thereby delaying or weakening the subsequent adaptive immune response.


Asunto(s)
Antígenos de Histocompatibilidad Clase II , Histona Desacetilasa 2 , Proteínas Nucleares , Regiones Promotoras Genéticas , SARS-CoV-2 , Transactivadores , Humanos , Histona Desacetilasa 2/metabolismo , Histona Desacetilasa 2/genética , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Transactivadores/metabolismo , Transactivadores/genética , Regiones Promotoras Genéticas/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , COVID-19/virología , COVID-19/inmunología , COVID-19/genética , COVID-19/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/inmunología , Células HEK293 , Regulación hacia Abajo/genética , Presentación de Antígeno/genética , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética
11.
Int J Biol Macromol ; 267(Pt 1): 131336, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38583840

RESUMEN

Fucoidan, a sulfated polysaccharide of marine origin found in brown algae and sea cucumbers, has been identified as a neuroprotective compound. In this study, a novel fucoidan MF4 was extracted from Fucus vesiculosus and isolated using Q-Sepharose fast-flow ion-exchange chromatography. The physicochemical properties of MF4 were characterized. MF4 is primarily composed of fucose, xylose, galactose, glucose, and mannose in a molar ratio of 12.3: 4.9: 1.1: 1.0: 1.1, with an average molecular weight of 67.7 kDa. Notably, MF4 demonstrated suppression of LLC tumor growth in vivo. RNA-sequencing analysis revealed that MF4 enhanced the expression of type I interferon-associated downstream genes in macrophages. Furthermore, MF4 increased the levels of phosphorylated TBK1 and IRF3 proteins in vitro. By activating the STING-TBK1-IRF3 signaling pathway, MF4 may enhance the antitumor activity of macrophages. Taken together, MF4 has promising potential as an antitumor and immunomodulatory agent.


Asunto(s)
Carcinoma Pulmonar de Lewis , Factor 3 Regulador del Interferón , Polisacáridos , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Polisacáridos/farmacología , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Animales , Ratones , Factor 3 Regulador del Interferón/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Células RAW 264.7
12.
Int Immunopharmacol ; 132: 111971, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38565040

RESUMEN

DNA damage resulting from UV irradiation on the skin has been extensively documented in numerous studies. In our prior investigations, we demonstrated that UVB-induced DNA breakage from keratinocytes can activate the cGAS-STING pathway in macrophages. The cGAS-STING signaling pathway serves as the principal effector for detecting and responding to abnormal double-stranded DNA in the cytoplasm. Expanding on our previous findings, we have further validated that STING knockout significantly diminishes UVB-induced skin damage, emphasizing the critical role of cGAS-STING activation in this context. Salvianolic acid A, a principal active constituent of Salvia miltiorrhiza Burge, has been extensively studied for its therapeutic effects in conditions such as coronary heart disease, angina pectoris, and diabetic peripheral neuropathy. However, its effect on cGAS-STING pathway and its ability to alleviate skin damage have not been previously reported. In a co-culture system, supernatant from UVB-treated keratinocytes induced IRF3 activation in macrophages, and this activation was inhibited by salvianolic acid A. Our investigation, employing photodamage and photoaging models, establishes that salvianolic acid A effectively mitigates UV-induced epidermal thickening and collagen degeneration. Treatment with salvianolic acid A significantly reduced skin damage, epidermal thickness increase, and keratinocyte hyperproliferation compared to the untreated photo-damage and photoaging model groups. In summary, salvianolic acid A emerges as a promising candidate for preventing UV-induced skin damage by inhibiting cGAS-STING activation. This research enhances our understanding of the intricate mechanisms underlying skin photodamage and provides a potential avenue for the development of therapeutic interventions.


Asunto(s)
Ácidos Cafeicos , Queratinocitos , Lactatos , Proteínas de la Membrana , Nucleotidiltransferasas , Transducción de Señal , Piel , Rayos Ultravioleta , Rayos Ultravioleta/efectos adversos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Animales , Transducción de Señal/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Queratinocitos/efectos de la radiación , Piel/efectos de los fármacos , Piel/patología , Piel/efectos de la radiación , Nucleotidiltransferasas/metabolismo , Ácidos Cafeicos/farmacología , Humanos , Ratones , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones Endogámicos C57BL , Envejecimiento de la Piel/efectos de los fármacos , Envejecimiento de la Piel/efectos de la radiación , Daño del ADN/efectos de los fármacos , Factor 3 Regulador del Interferón/metabolismo , Femenino , Células RAW 264.7
13.
Mol Immunol ; 170: 131-143, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38663254

RESUMEN

Mammalian reovirus (MRV) is a non-enveloped, gene segmented double-stranded RNA (dsRNA) virus. It is an important zoonotic pathogen that infects many mammals and vertebrates that act as natural hosts and causes respiratory and digestive tract diseases. Studies have reported that RIG-I and MDA5 in the innate immune cytoplasmic RNA-sensing RIG-like receptor (RLR) signaling pathway can recognize dsRNA from MRV and promote antiviral type I interferon (IFN) responses. However, the mechanism by which many MRV-encoded proteins evade the host innate immune response remains unclear. Here, we show that exogenous µ1 protein promoted the proliferation of MRV in vitro, while knockdown of MRV µ1 protein expression by shRNA could impair MRV proliferation. Specifically, µ1 protein inhibited MRV or poly(I:C)-induced IFN-ß expression, and attenuated RIG-I/MDA5-mediated signaling axis transduction during MRV infection. Importantly, we found that µ1 protein significantly decreased IFN-ß mRNA expression induced by MDA5, RIG-I, MAVS, TBK1, IRF3(5D), and degraded the protein expression of exogenous MDA5, RIG-I, MAVS, TBK1 and IRF3 via the proteasomal and lysosomal pathways. Additionally, we show that µ1 protein can physically interact with MDA5, RIG-I, MAVS, TBK1, and IRF3 and attenuate the RIG-I/MDA5-mediated signaling cascades by blocking the phosphorylation and nuclear translocation of IRF3. In conclusion, our findings reveal that MRV outer capsid protein µ1 is a key factor in antagonizing RLRs signaling cascades and provide new strategies for effective prevention and treatment of MRV infection.


Asunto(s)
Proteína 58 DEAD Box , Factor 3 Regulador del Interferón , Helicasa Inducida por Interferón IFIH1 , Orthoreovirus de los Mamíferos , Receptores Inmunológicos , Transducción de Señal , Helicasa Inducida por Interferón IFIH1/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Factor 3 Regulador del Interferón/metabolismo , Proteína 58 DEAD Box/metabolismo , Transducción de Señal/inmunología , Humanos , Fosforilación , Orthoreovirus de los Mamíferos/inmunología , Orthoreovirus de los Mamíferos/fisiología , Células HEK293 , Interferón beta/metabolismo , Interferón beta/inmunología , Animales , Núcleo Celular/metabolismo , Infecciones por Reoviridae/inmunología , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Inmunidad Innata/inmunología , Proteínas Serina-Treonina Quinasas
14.
Cancer Lett ; 589: 216836, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38556105

RESUMEN

Despite the approval of immune checkpoint blockade (ICB) therapy for various tumor types, its effectiveness is limited to only approximately 15% of patients with microsatellite instability-high (MSI-H) or mismatch repair deficiency (dMMR) colorectal cancer (CRC). Approximately 80%-85% of CRC patients have a microsatellite stability (MSS) phenotype, which features a rare T-cell infiltration. Thus, elucidating the mechanisms underlying resistance to ICB in patients with MSS CRC is imperative. In this study, we demonstrate that ubiquitin-specific peptidase 4 (USP4) is upregulated in MSS CRC tumors and negatively regulates the immune response against tumors in CRC. Additionally, USP4 represses the cellular interferon (IFN) response and antigen presentation and impairs PRR signaling-mediated cell death. Mechanistically, USP4 impedes the nuclear localization of interferon regulator Factor 3 (IRF3) by deubiquitinating the K63-polyubiquitin chain of TRAF6 and IRF3. Knockdown of USP4 enhances the infiltration of T cells in CRC tumors and overcomes ICB resistance in an MC38 syngeneic mouse model. Moreover, published datasets revealed that patients showing higher USP4 expression exhibited decreased responsiveness to anti-PD-L1 therapy. These findings highlight an essential role of USP4 in the suppression of antitumor immunity in CRC.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Colorrectales , Interferones , Síndromes Neoplásicos Hereditarios , Animales , Ratones , Humanos , Interferones/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Inestabilidad de Microsatélites , Enzimas Desubicuitinizantes/genética , Factor 3 Regulador del Interferón/genética , Proteasas Ubiquitina-Específicas/genética , Proteasas Ubiquitina-Específicas/metabolismo
15.
Biochemistry ; 63(6): 767-776, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38439718

RESUMEN

Interferon regulatory factor 4 (IRF4) is a crucial transcription factor that plays a vital role in lymphocyte development, including in the fate-determining steps in terminal differentiation. It is also implicated in the development of lymphoid tumors such as multiple myeloma and adult T-cell leukemia. IRF4 can form a homodimer and multiple heterocomplexes with other transcription factors such as purine-rich box1 and activator protein 1. Each protein complex binds to specific DNA sequences to regulate a distinct set of genes. However, the precise relationship among these complex formations remains unclear. Herein, we investigated the abilities of IRF4 proteins with functional mutations in the IRF-association domain and autoinhibitory region to form complexes using luciferase reporter assays. The assays allowed us to selectively assess the activity of each complex. Our results revealed that certain IRF-association domain mutants, previously known to have impaired heterocomplex formation, maintained or even enhanced homodimer activity. This discrepancy suggests that the mutated amino acid residues selectively influence homodimer activity. Conversely, a phosphomimetic serine mutation in the autoinhibitory region displayed strong activating effects in all complexes. Furthermore, we observed that partner proteins involved in heterocomplex formation could disrupt the activity of the homodimer, suggesting a potential competition between homocomplexes and heterocomplexes. Our findings provide new insights into the mechanistic function of IRF4.


Asunto(s)
Regulación de la Expresión Génica , Factores Reguladores del Interferón , Secuencia de Bases , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Mutación , Factor de Transcripción AP-1/metabolismo , Humanos , Células HEK293
16.
J Innate Immun ; 16(1): 226-247, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38527452

RESUMEN

INTRODUCTION: While TLR ligands derived from microbial flora and pathogens are important activators of the innate immune system, a variety of factors such as intracellular bacteria, viruses, and parasites can induce a state of hyperreactivity, causing a dysregulated and potentially life-threatening cytokine over-response upon TLR ligand exposure. Type I interferon (IFN-αß) is a central mediator in the induction of hypersensitivity and is strongly expressed in splenic conventional dendritic cells (cDC) and marginal zone macrophages (MZM) when mice are infected with adenovirus. This study investigates the ability of adenoviral infection to influence the activation state of the immune system and underlines the importance of considering this state when planning the treatment of patients. METHODS: Infection with adenovirus-based vectors (Ad) or pretreatment with recombinant IFN-ß was used as a model to study hypersensitivity to lipopolysaccharide (LPS) in mice, murine macrophages, and human blood samples. The TNF-α, IL-6, IFN-αß, and IL-10 responses induced by LPS after pretreatment were measured. Mouse knockout models for MARCO, IFN-αßR, CD14, IRF3, and IRF7 were used to probe the mechanisms of the hypersensitive reaction. RESULTS: We show that, similar to TNF-α and IL-6 but not IL-10, the induction of IFN-αß by LPS increases strongly after Ad infection. This is true both in mice and in human blood samples ex vivo, suggesting that the regulatory mechanisms seen in the mouse are also present in humans. In mice, the scavenger receptor MARCO on IFN-αß-producing cDC and splenic marginal zone macrophages is important for Ad uptake and subsequent cytokine overproduction by LPS. Interestingly, not all IFN-αß-pretreated macrophage types exposed to LPS exhibit an enhanced TNF-α and IL-6 response. Pretreated alveolar macrophages and alveolar macrophage-like murine cell lines (MPI cells) show enhanced responses, while bone marrow-derived and peritoneal macrophages show a weaker response. This correlates with the respective absence or presence of the anti-inflammatory IL-10 response in these different macrophage types. In contrast, Ad or IFN-ß pretreatment enhances the subsequent induction of IFN-αß in all macrophage types. IRF3 is dispensable for the LPS-induced IFN-αß overproduction in infected MPI cells and partly dispensable in infected mice, while IRF7 is required. The expression of the LPS co-receptor CD14 is important but not absolutely required for the elicitation of a TNF-α over-response to LPS in Ad-infected mice. CONCLUSION: Viral infections or application of virus-based vaccines induces type I interferon and can tip the balance of the innate immune system in the direction of hyperreactivity to a subsequent exposure to TLR ligands. The adenoviral model presented here is one example of how multiple factors, both environmental and genetic, affect the physiological responses to pathogens. Being able to measure the current reactivity state of the immune system would have important benefits for infection-specific therapies and for the prevention of vaccination-elicited adverse effects.


Asunto(s)
Adenoviridae , Citocinas , Factor 3 Regulador del Interferón , Lipopolisacáridos , Macrófagos , Ratones Noqueados , Animales , Ratones , Lipopolisacáridos/inmunología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Macrófagos/inmunología , Citocinas/metabolismo , Ratones Endogámicos C57BL , Factor 7 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Vectores Genéticos , Infecciones por Adenoviridae/inmunología , Interferón Tipo I/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Interferón beta/metabolismo
17.
Arch Virol ; 169(4): 74, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38480558

RESUMEN

Triple motif protein 21 (TRIM21) has an antiviral function that inhibits various viral infections. However, its role in the progress of influenza A virus (IAV) infection is unclear. In this study, we investigated the role and molecular mechanism of TRIM21 in IAV infection. RT-qPCR was used to determine the level of TRIM21 mRNA, and ELISA was used to measure the levels of IFN-α, IFN-ß, IL-6, and TNF-α. The levels of the TRIM21, NP, TBK1, IRF3, p-TBK1, and p-IRF3 proteins were estimated by Western blot. The results showed that, after IAV infection, TRIM21 was upregulated in clinical patient serum and A549 cells, and this was correlated with the IFN response. Overexpression of TRIM21 reduced IAV replication and transcription in in vitro cell experiments. TRIM21 also increased IFN-α and IFN-ß levels and decreased IL-6 and TNF-α levels in A549 cells. In addition, overexpression of TRIM21 inhibited IAV-induced apoptosis. Further experiments demonstrated that TBK1-IRF3 signaling was activated by TRIM21 and was involved in the inhibitory effect of TRIM21 on virus replication. In summary, our study suggests that TRIM21 inhibits viral replication by activating the TBK1-IRF3 signaling pathway, further inhibiting the infection process of IAV.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Humana , Humanos , Células A549 , Inflamación , Virus de la Influenza A/metabolismo , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Gripe Humana/genética , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón-alfa/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
18.
Immunology ; 172(2): 295-312, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38453210

RESUMEN

Hyperactivation of the cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signalling pathway has been shown to be associated with the development of a variety of inflammatory diseases, and the discovery of an inhibitor of the cGAS-STING signalling pathway holds great promise in the therapeutic interventions. Epimedium flavonoid (EF), a major active ingredient isolated from the medicinal plant Epimedium, has been reported to have good anti-inflammatory activity, but its exact mechanism of action remains unclear. In the present study, we found that EF in mouse bone marrow-derived macrophages (BMDMs), THP-1 (Tohoku Hospital Pediatrics-1) as well as in human peripheral blood mononuclear cells (hPBMC) inhibited the activation of the cGAS-STING signalling pathway, which subsequently led to a decrease in the expression of type I interferon (IFN-ß, CXCL10 and ISG15) and pro-inflammatory cytokines (IL-6 and TNF-α). Mechanistically, EF does not affect STING oligomerization, but inhibits the formation of functional STING signalosome by attenuating the interaction of interferon regulatory factor 3 (IRF3) with STING and TANK-binding kinase 1 (TBK1). Importantly, in vivo experiments, EF has shown promising therapeutic effects on inflammatory diseases mediated by the cGAS-STING pathway, which include the agonist model induced by DMXAA stimulation, the autoimmune inflammatory disease model induced by three prime repair exonuclease 1 (Trex1) deficiency, and the non-alcoholic steatohepatitis (NASH) model induced by a pathogenic amino acid and choline deficiency diet (MCD). To summarize, our study suggests that EF is a potent potential inhibitor component of the cGAS-STING signalling pathway for the treatment of inflammatory diseases mediated by the cGAS-STING signalling pathway.


Asunto(s)
Epimedium , Flavonoides , Proteínas de la Membrana , Nucleotidiltransferasas , Transducción de Señal , Nucleotidiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Transducción de Señal/efectos de los fármacos , Humanos , Ratones , Flavonoides/farmacología , Epimedium/química , Factor 3 Regulador del Interferón/metabolismo , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Citocinas/metabolismo , Células THP-1 , Proteínas Serina-Treonina Quinasas/metabolismo , Antiinflamatorios/farmacología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/efectos de los fármacos
19.
Phytomedicine ; 128: 155404, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38507852

RESUMEN

BACKGROUND: The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon (IFN) genes (STING) pathway is critical in the innate immune system and can be mobilized by cytosolic DNA. The various inflammatory and autoimmune diseases progression is highly correlated with aberrant cGAS-STING pathway activation. While some cGAS-STING pathway inhibitor were identified, there are no drugs that can be applied to the clinic. Compound Danshen Dripping Pill (CDDP) has been successfully used in clinic around the world, but the most common application is limited to cardiovascular disease. Therefore, the purpose of the present investigation was to examine whether CDDP inhibits the cGAS-STING pathway and could be used as a therapeutic agent for multiple cGAS-STING-triggered diseases. METHODS: BMDMs, THP1 cells or Trex1-/- BMDMs were stimulated with various cGAS-STING-agonists after pretreatment with CDDP to detect the function of CDDP on IFN-ß and ISGs productionn. Next, we detect the influence on IRF3 and P65 nuclear translocation, STING oligomerization and STING-TBK1-IRF3 complex formation of CDDP. Additionally, the DMXAA-mediated activation mice model of cGAS-STING pathway was used to study the effects of CDDP. Trex1-/- mice model and HFD-mediated obesity model were established to clarify the efficacy of CDDP on inflammatory and autoimmune diseases. RESULTS: CDDP efficacy suppressed the IRF3 phosphorylation or the generation of IFN-ß, ISGs, IL-6 and TNF-α. Mechanistically, CDDP did not influence the STING oligomerization and IRF3-TBK1 and STING-IRF3 interaction, but remarkably eliminated the STING-TBK1 interaction, ultimately blocking the downstream responses. In addition, we also clarified that CDDP could suppress cGAS-STING pathway activation triggered by DMXAA, in vivo. Consistently, CDDP could alleviate multi-organ inflammatory responses in Trex1-/- mice model and attenuate the inflammatory disorders, incleding obesity-induced insulin resistance. CONCLUSION: CDDP is a specifically cGAS-STING pathway inhibitor. Furthermore, we provide novel mechanism for CDDP and discovered a clinical agent for the therapy of cGAS-STING-triggered inflammatory and autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Canfanos , Medicamentos Herbarios Chinos , Inflamación , Panax notoginseng , Salvia miltiorrhiza , Ratones , Ratones Endogámicos C57BL , Salvia miltiorrhiza/química , Panax notoginseng/química , Inmunidad Innata , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/metabolismo , Transducción de Señal , Células THP-1 , Exodesoxirribonucleasas/genética , Fosfoproteínas/genética , Macrófagos , Interferón beta/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular , Enfermedades Autoinmunes/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Dieta Alta en Grasa , Proteínas Serina-Treonina Quinasas/metabolismo , Humanos
20.
EMBO J ; 43(5): 780-805, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38316991

RESUMEN

Inflammation is a common condition of prostate tissue, whose impact on carcinogenesis is highly debated. Microbial colonization is a well-documented cause of a small percentage of prostatitis cases, but it remains unclear what underlies the majority of sterile inflammation reported. Here, androgen- independent fluctuations of PSA expression in prostate cells have lead us to identify a prominent function of the Transient Receptor Potential Cation Channel Subfamily M Member 8 (TRPM8) gene in sterile inflammation. Prostate cells secret TRPM8 RNA into extracellular vesicles (EVs), which primes TLR3/NF-kB-mediated inflammatory signaling after EV endocytosis by epithelial cancer cells. Furthermore, prostate cancer xenografts expressing a translation-defective form of TRPM8 RNA contain less collagen type I in the extracellular matrix, significantly more infiltrating NK cells, and larger necrotic areas as compared to control xenografts. These findings imply sustained, androgen-independent expression of TRPM8 constitutes as a promoter of anticancer innate immunity, which may constitute a clinically relevant condition affecting prostate cancer prognosis.


Asunto(s)
Neoplasias de la Próstata , Canales Catiónicos TRPM , Humanos , Masculino , Andrógenos , Inflamación/genética , Factor 3 Regulador del Interferón , Proteínas de la Membrana , FN-kappa B/genética , Neoplasias de la Próstata/genética , Receptor Toll-Like 3/genética , Canales Catiónicos TRPM/genética , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA