Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Immunol ; 210(5): 640-652, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36651806

RESUMEN

IκBα is a critical protein that inhibits NF-κB nuclear translocation and impairs NF-κB-mediated signaling. The abundance of IκBα determines the activation and restoration of the inflammatory response. However, posttranslational regulation of IκBα remains to be fully understood. In this study, we identified ubiquitin-specific protease 39 (USP39) as a negative regulator in the NF-κB inflammatory response by stabilizing basal IκBα. The expression of USP39 in macrophages was reduced under LPS-induced inflammation. Knockdown or knockout of USP39 in macrophages significantly increased the expression and secretion of proinflammatory cytokines upon exposure to LPS or Escherichia coli, whereas reexpression of exogenous USP39 in USP39-deficient macrophages rescued the effect. Moreover, USP39-defective mice were more sensitive to LPS or E. coli-induced systemic sepsis. Mechanistically, USP39 interacted with and stabilized IκBα by reducing K48-linked polyubiquination of IκBα. Taken together, to our knowledge, our study for the first time revealed the inhibitory function of USP39 in the NF-κB inflammatory response, providing a previously unknown mechanism for control of inflammatory cytokine induction in the cellular anti-inflammatory response.


Asunto(s)
Lipopolisacáridos , FN-kappa B , Animales , Ratones , Citocinas/metabolismo , Escherichia coli/metabolismo , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Inhibidor NF-kappaB alfa
2.
Cell Commun Signal ; 22(1): 14, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38183076

RESUMEN

R-spondins (RSPOs) are secreted signaling molecules that potentiate the Wnt/ß-catenin pathway by cooperating with Wnt ligands. RSPO1 is crucial in tissue development and tissue homeostasis. However, the molecular mechanism by which RSPOs activate Wnt/ß-catenin signaling remains elusive. In this study, we found that RSPOs could mediate the degradation of Axin through the ubiquitin-proteasome pathway. The results of Co-IP showed that the recombinant RSPO1 protein promoted the interaction between Axin1 and CK1ε. Either knockout of the CK1ε gene or treatment with the CK1δ/CK1ε inhibitor SR3029 caused an increase in Axin1 protein levels and attenuated RSPO1-induced degradation of the Axin1 protein. Moreover, we observed an increase in the number of associations of LRP6 with CK1ε and Axin1 following RSPO1 stimulation. Overexpression of LRP6 further potentiated Axin1 degradation mediated by RSPO1 or CK1ε. In addition, recombinant RSPO1 and Wnt3A proteins synergistically downregulated the protein expression of Axin1 and enhanced the transcriptional activity of the SuperTOPFlash reporter. Taken together, these results uncover the novel mechanism by which RSPOs activate Wnt/ß-catenin signaling through LRP6/CK1ε-mediated degradation of Axin.


Asunto(s)
Proteína Axina , Trombospondinas , Vía de Señalización Wnt , beta Catenina , Transporte Biológico , Proteína Wnt3A , Humanos , Trombospondinas/metabolismo
3.
Nanomedicine ; 41: 102516, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35131469

RESUMEN

Two kinds of amphiphilic block copolymers of TfR-T12-PEG-PLGA and TATH7-PEG-PLGA were synthesized to self-assembly nano-composite micelles for encapsulating paclitaxel and imiquimod synchronously. TfR-T12 peptide modified nano-composite micelles can pass through BBB in a TfR-mediated way to achieve targeted delivery of chemotherapeutic drugs, and pH sensitive TATH7 peptide modified nano-composite micelles enhanced uptake efficiency more significantly under pH 5.5 medium than pH 7.4 medium. The results of pharmacodynamic evaluation in vivo showed that the nano-composite micelles had achieved good anti-tumor effect in subcutaneous and normotopia glioma models, and effectively prolonged the life cycle of tumor-bearing mice. The nano-composite micelles regulated the immunosuppression phenomenon of tumor microenvironment significantly, and promoted the M1 polarization of TAMs, then enhanced the proliferation and activation of CD8+ T cells in tumor microenvironment. It comes to conclusion that the nano-composite micelle achieves the purpose of effective treatment of glioma by chemotherapy combined with immunotherapy.


Asunto(s)
Glioma , Micelas , Animales , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Glioma/patología , Concentración de Iones de Hidrógeno , Ratones , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Péptidos/uso terapéutico , Polietilenglicoles/uso terapéutico , Microambiente Tumoral
4.
Pharmacol Res ; 164: 105386, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33352228

RESUMEN

Cellular inflammation is the underlying cause of several diseases and development of a safe and effective anti-inflammatory drug is need-of-the hour for treatment of diseases like lung inflammation. Callicarpa integerrima Champ. is a well-known herbal medicine with hemostatic and anti-inflammatory functions. However, the exact ingredient exhibiting anti-inflammatory activity in C. integerrima Champ. is largely unknown. Here, we first isolated, purified and characterized a novel clerodane-type diterpenoid Cintelactone A (CA) from C. integerrima Champ. We demonstrated that CA could significantly inhibit lipopolysaccharide (LPS)-induced pro-inflammatory cytokines and mediators production both in mouse peritoneal macrophages and THP1 cells. Consistently, CA also relieved inflammation and reduced LPS-induced lung injury in mice. We systematically elucidated the mechanism of action as well. CA interacted with Arg78 of tumor necrosis factor receptor-associated factor 6 (TRAF6) by hydrogen bonding. It further promoted the K48-linked ubiquitination and proteasomal degradation of TRAF6, and suppressed the activation of NF-κB and MAPKs signaling pathways. Collectively, our study reveals that new clerodane-type diterpenoid CA suppresses LPS-induced inflammation by promoting TRAF6 degradation, suggesting that CA as the potential therapeutic candidate for the treatment of inflammation associated diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Diterpenos de Tipo Clerodano/uso terapéutico , Factor 6 Asociado a Receptor de TNF/metabolismo , Animales , Antiinflamatorios/farmacología , Callicarpa , Células Cultivadas , Citocinas/sangre , Citocinas/genética , Citocinas/metabolismo , Diterpenos de Tipo Clerodano/farmacología , Femenino , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones Endogámicos C57BL , Fitoquímicos/farmacología , Fitoquímicos/uso terapéutico , Hojas de la Planta , Tallos de la Planta , Ubiquitinación/efectos de los fármacos
5.
Eur J Immunol ; 49(1): 42-53, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30466171

RESUMEN

Retinoic acid-inducible gene I (RIG-I) is a critical RNA virus sensor that initiates antiviral immune response through K63-linked ubiquitination. In this study, we demonstrated USP14, a deubiquitinating enzyme, as a negative regulator in antiviral responses by directly deubiquitinating K63-linked RIG-I. USP14 knockdown significantly enhanced RIG-I-triggered type I IFN signaling and inhibited vesicular stomatitis virus (VSV) replication both in mouse peritoneal macrophages and THP1 cells. USP14 overexpression in HeLa cells attenuated RIG-I-triggered IFN-ß expression and promoted VSV replication. Besides, USP14-specific inhibitor, IU1, increased RIG-I-mediated type I IFN production and antiviral responses in vitro and in vivo. In addition, USP14 could interact with RIG-I and remove RIG-I K63-linked polyubiquitination chains. This article is the first to report that USP14 acts as a negative regulator in antiviral response through deubiquitinating K63-linked RIG-I. These findings provide insights into a potential new therapy targeting USP14 for RNA virus-related diseases.


Asunto(s)
Macrófagos/inmunología , Infecciones por Rhabdoviridae/inmunología , Ubiquitina Tiolesterasa/metabolismo , Vesiculovirus/fisiología , Animales , Proteína 58 DEAD Box/metabolismo , Femenino , Células HeLa , Humanos , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , Receptores Inmunológicos , Transducción de Señal , Células THP-1 , Ubiquitina Tiolesterasa/genética , Ubiquitinación , Replicación Viral
6.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 44(5): 578-83, 2015 09.
Artículo en Zh | MEDLINE | ID: mdl-26713535

RESUMEN

Ubiquitin-specific protease(USP), which belongs to cysteine protease, is an important member of the deubiquitinating enzyme family(DUB). USP plays an important role in the immune response against viral infections, in which it can regulate the production of type I interferon through various ways to initiate or weaken the antiviral immune response. USP2b, USP3, USP18, USP25, UL36USP and HAUSP play a role of antivirus; while USP4, USP13, USP15 and USP17 negatively regulate antiviral immune response. In this article we review the recent progress on roles of USP family in antiviral immune response.


Asunto(s)
Proteasas Ubiquitina-Específicas/inmunología , Virosis/inmunología , Humanos , Interferón Tipo I/inmunología
7.
Transl Oncol ; 40: 101863, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38185060

RESUMEN

INTRODUCTION: Autophagy is an important mechanism of cell homeostasis maintenance. As essential serine/threonine-protein kinases, casein kinase I family members affect tumorigenesis by regulating a variety of cellular progression. However, the mechanism by which they regulate autophagy remains unclear. MATERIALS AND METHODS: We silenced CK1δ/ε in cancer cells and observed cell morphology, the expression of autophagy-related genes, and its impact on cancer cell growth and viability. By inhibiting CK1δ/ε-induced upregulation of autophagy genes, we profiled the regulatory mechanism of CK1δ/ε on autophagy and cancer cell growth. The impact of CK1δ/ε inhibition on tumor cell growth was also assessed in vivo. RESULTS: Here, we found that CK1δ/ε played an important role in ULK1-mediated autophagy regulation in both lung cancer and melanoma cells. Mechanically, silencing CK1δ/ε increased ULK1 expression with enhanced autophagic flux and suppressed cancer cell proliferation, while ULK1 knockdown blocked the activation of autophagy caused by CK1δ/ε inhibition. By silencing CK1δ/ε in syngeneic mouse model bearing LLC1 murine lung cancer cells in vivo, we observed tumor growth suppression mediated by CK1δ/ε inhibition. CONCLUSION: Our results provide evidence for the role of CK1δ/ε in the regulation of tumorigenesis via the ULK1-mediated autophagy, and also suggest the impact of CK1δ/ε inhibition on tumor growth and its significance as a potential therapeutic target.

8.
Mol Oncol ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38419282

RESUMEN

Casein kinase 1ε (CK1ε) and axis inhibitor 1 (AXIN1) are crucial components of the ß-catenin destruction complex in canonical Wnt signaling. CK1ε has been shown to interact with AXIN1, but its physiological function and role in tumorigenesis remain unknown. In this study, we found that CK1δ/ε inhibitors significantly enhanced AXIN1 protein level in colorectal cancer (CRC) cells through targeting CK1ε. Mechanistically, CK1ε promoted AXIN1 degradation by the ubiquitin-proteasome pathway by promoting the interaction of E3 ubiquitin-protein ligase SIAH1 with AXIN1. Genetic or pharmacological inhibition of CK1ε and knockdown of SIAH1 downregulated the expression of Wnt/ß-catenin-dependent genes, suppressed the viability of CRC cells, and restrained tumorigenesis and progression of CRC in vitro and in vivo. In summary, our results demonstrate that CK1ε exerted its oncogenic role in CRC occurrence and progression by regulating the stability of AXIN1. These findings reveal a novel mechanism by which CK1ε regulates the Wnt/ß-catenin signaling pathway and highlight the therapeutic potential of targeting the CK1ε/SIAH1 axis in CRC.

9.
Autophagy ; 19(3): 873-885, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35900990

RESUMEN

The NLRP3 inflammasome is involved in a diverse range of inflammatory diseases. The activation of inflammasomes must be tightly regulated to prevent excessive inflammation, and the protein ubiquitination system is reported to be one of the ways in which inflammasome activation is regulated. However, the deubiquitination regulatory mechanisms of inflammasome activation remain elusive. Here, we demonstrated that USP22 (ubiquitin specific peptidase 22) promotes NLRP3 degradation and inhibits NLRP3 inflammasome activation. USP22 deficiency or in vivo silencing significantly increases alum-induced peritonitis and lipopolysaccharide-induced systemic inflammation. Mechanistically, USP22 inhibits NLRP3 inflammasome activation via the promotion of ATG5-mediated macroautophagy/autophagy. USP22 stabilizes ATG5 via decreasing K27- and K48-linked ubiquitination of ATG5 at the Lys118 site. Taken together, these findings reveal the role USP22 plays in the regulation of NLRP3 inflammasome activation and suggest a potential therapeutic target to treat NLRP3 inflammasome-related diseases.Abbreviations: ATG5: autophagy related 5; ATP: adenosine triphosphate; CASP1: caspase 1; IL18: interleukin 18; IL1B/IL-1ß: interleukin 1 beta; LPS: lipopolysaccharide; NLRC4: NLR family, CARD domain containing 4; NLRP3: NLR family, pyrin domain containing 3; PYCARD/ASC: PYD and CARD domain containing; TNF/TNF-α: tumor necrosis factor; USP22: ubiquitin specific peptidase 22.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Animales , Ratones , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Autofagia , Lipopolisacáridos/farmacología , Inflamación/metabolismo , Caspasa 1/metabolismo , Proteasas Ubiquitina-Específicas , Interleucina-1beta/metabolismo , Ratones Endogámicos C57BL , Proteína 5 Relacionada con la Autofagia , Ubiquitina Tiolesterasa
10.
Front Immunol ; 14: 1174463, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37153555

RESUMEN

The NLRP3 inflammasome plays a critical role in the innate immune response, and its excessive activation will cause pyroptotic cell death and be associated with the onset of inflammatory diseases. However, NLRP3 inflammasome targeting therapies are still to be implemented in the clinic setting. Here, we first isolated, purified and characterized a novel Vitenegu acid from V. negundo L. herb that specifically inhibits NLRP3 inflammasome activation, without affecting NLRC4 or AIM2 inflammasomes. Vitenegu acid blocks the oligomerization of NLRP3, thus inhibiting NLRP3 inflammasome assembly and activation. In vivo data show that Vitenegu acid exerts therapeutic effects on NLRP3 inflammasome-dependent inflammation. Taken together, our results suggest that Vitenegu acid is a candidate therapeutic agent for treating NLRP3 inflammasome related diseases.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamación
11.
Transl Oncol ; 34: 101713, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37302347

RESUMEN

BACKGROUND: The elevated Cyclin B1 expression contributes to various tumorigenesis and poor prognosis. Cyclin B1 expression could be regulated by ubiquitination and deubiquitination. However, the mechanism of how Cyclin B1 is deubiquitinated and its roles in human glioma remain unclear. METHODS: Co-immunoprecipitation and other assays were performed to detect the interacting of Cyclin B1 and USP39. A series of in vitro and in vivo experiments were performed to investigate the effect of USP39 on the tumorigenicity of tumor cells. RESULTS: USP39 interacts with Cyclin B1 and stabilizes its expression by deubiquitinating Cyclin B1. Notably, USP39 cleaves the K29-linked polyubiquitin chain on Cyclin B1 at Lys242. Additionally, overexpression of Cyclin B1 rescues the arrested cell cycle at G2/M transition and the suppressed proliferation of glioma cells caused by USP39 knockdown in vitro. Furthermore, USP39 promotes the growth of glioma xenograft in subcutaneous and in situ of nude mice. Finally, in human tumor specimens, the expression levels of USP39 and Cyclin B1 are positively relevant. CONCLUSION: Our data support the evidence that USP39 acts a novel deubiquitinating enzyme of Cyclin B1 and promoted tumor cell proliferation at least in part through Cyclin B1 stabilization, represents a promising therapeutic strategy for tumor patients.

12.
Front Immunol ; 13: 816378, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309330

RESUMEN

Succinate is at the crossroads of multiple metabolic pathways and plays a role in several immune responses acting as an inflammation signal. However, whether succinate regulates antiviral immune response remains unclear. Here, we found that the production of succinate was reduced in RAW264.7 cells during vesicular stomatitis virus (VSV) infection. Using diethyl succinate to pretreat the mouse peritoneal macrophages and RAW264.7 cells before VSV infection, the production of interferon-ß (IFN-ß), chemokine (C-X-C motif) ligand 10 (CXCL-10), and IFN-stimulated genes 15 (ISG15) was significantly decreased, following which the VSV replication in diethyl succinate-pretreated cells was obviously increased. Moreover, succinate decreased the expression of IFN-ß in serum, lung, and spleen derived from the VSV-infected mice. The overall survival rate in the VSV-infected mice with diethyl succinate pretreatment was also remarkably downregulated. Furthermore, we identified that succinate inhibited the activation of MAVS-TBK1-IRF3 signaling by suppressing the formation of MAVS aggregates. Our findings provide previously unrecognized roles of succinate in antiviral immune response and establish a novel link between metabolism and innate immune response.


Asunto(s)
Ácido Succínico , Animales , Inmunidad Innata , Interferón beta/metabolismo , Ratones , Virus de la Estomatitis Vesicular Indiana
13.
Mol Oncol ; 16(2): 388-404, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33811456

RESUMEN

Glioma cells are characterized by high migration and invasion ability; however, the molecular mechanism behind both processes still remains to be investigated. Several studies have demonstrated that ubiquitin-specific protease 39 (USP39) plays an oncogenic role in various cancer types. Here, we investigated the expression and function of USP39 in patients with glioma. Oncomine database analysis revealed that high USP39 expression was significantly correlated with poor overall survival in patients with glioma. Knockdown of USP39 in U251 and U87 cell lines significantly inhibited their migration and invasion in vitro. Gene expression profiling of glioma cells transduced with short hairpin RNA (shRNA) against USP39 revealed that disintegrin and metalloproteinase domain-containing protein 9 (ADAM9), a molecule previously related to tumor cell migration and invasion, was significantly downregulated. Furthermore, USP39 induced ADAM9 messenger RNA (mRNA) maturation and decreased the expression of integrin ß1. Additionally, overexpression of ADAM9 inhibited the migration and invasion of glioma cells caused by USP39 depletion in vitro. USP39 promoted the invasion of glioma cells in vivo and reduced the overall survival of the mice. Altogether, our data show that USP39 induces mRNA maturation and elevates the expression of ADAM9 in glioma cells and may thus be considered potential target for treating patients with glioma.


Asunto(s)
Proteínas ADAM/genética , Neoplasias Encefálicas/patología , Movimiento Celular , Glioma/patología , Proteínas de la Membrana/genética , Invasividad Neoplásica , ARN Mensajero/genética , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Neoplasias Encefálicas/genética , Femenino , Técnicas de Silenciamiento del Gen , Glioma/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , ARN Interferente Pequeño/genética , Proteasas Ubiquitina-Específicas/genética
14.
Autophagy ; 18(4): 891-908, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34436957

RESUMEN

TBK1 (TANK-binding kinase 1) is an essential receptor protein required for the innate immune response, but the mechanisms underlying TBK1 stability, especially those regulated via autophagy, remain poorly understood. Here, we demonstrate that USP19 (ubiquitin specific peptidase 19) interacts with and promotes TBK1 lysosomal degradation via chaperone-mediated autophagy (CMA). We observed that TBK1 had a canonical CMA motif, knocking down key proteins involved in CMA (HSPA8/HSC70 or LAMP2A) or inhibiting CMA-prevented USP19-mediated TBK1 degradation. Furthermore, USP19 deficiency in macrophages caused an elevation of TBK1 and the activation of the type-I interferon signaling pathway after vesicular stomatitis virus (VSV) infection. Consistently, macrophage-specific usp19 knockout in mice resulted in attenuated VSV replication and resistance to VSV infection in vivo. Altogether, our results suggest that USP19 is a key regulator of TBK1 and uncovers a previously uncharacterized role for USP19 in CMA-mediated TBK1 degradation and infectious diseases.


Asunto(s)
Autofagia Mediada por Chaperones , Animales , Autofagia , Endopeptidasas/metabolismo , Lisosomas/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Proteasas Ubiquitina-Específicas/metabolismo
15.
Redox Biol ; 56: 102461, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36087429

RESUMEN

Fungal infections cause serious health problems, especially in patients with an immune-deficiency. Histone deacetylase 11 (HDAC11) mediates various immune functions, yet little is known about its role in regulating host immune responses to fungal infection. Here we report that HDAC11 negatively controls antifungal immunity in macrophages and dendritic cells. Deleting Hdac11 protects mice from morbidity and markedly improves their survival rate upon systemic infection with Candida albicans (C. albicans). Moreover, HDAC11 deficiency results in increased production of NO and reactive oxygen species, which enhances fungal killing. Mechanistically, loss of HDAC11 increases histone 3 and 4 acetylation at the Nos2 promoter and leads to enhanced Nos2 transcription and corresponding iNOS levels in macrophages. In addition, STAT3, a transcriptional repressor of Nos2, physically interacts with HDAC11, serving as a scaffold protein supporting the HDAC11 association with the Nos2 promoter. Notably, treatment with the HDAC11 inhibitor, FT895, exhibits antifungal therapeutic effects in both mouse and human cells challenged with C. albicans. These data support that HDAC11 may be a therapeutic target for fungal infection.


Asunto(s)
Antifúngicos , Histonas , Animales , Antifúngicos/farmacología , Candida albicans/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
16.
Front Immunol ; 13: 853194, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35865528

RESUMEN

Inflammatory bowel diseases (IBDs) are increasingly common diseases characterized by chronic and relapsing inflammation of the gastrointestinal tract. NLRP3 might be a crucial regulator of the homeostatic balance of the intestine, but its upregulation leads to pyroptosis. Munronoid I is extracted and purified from Munronia sinica, which has shown an anti-inflammatory effect, but the efficacy of Munronoid I in IBD remains unproven. In this study, we attempted to determine the effect of Munronoid I on NLRP3 to regulate the inflammasome activation and pyroptosis in IBD. Our data demonstrated that Munronoid I treatment attenuated DSS-induced body weight loss, pathological injury of the colon, the production of IL-1ß and IL-18, and the expression of pyroptosis-associated proteins in colon tissue in mice. Moreover, Munronoid I inhibited LPS/ATP-induced pyroptosis in mouse peritoneal macrophages, MODE-K cells, and DSS-induced pyroptosis in mouse colonic epithelial cells, and decreased the release of inflammatory cytokines IL-1ß and IL-18 in mouse peritoneal macrophages. Mechanically, Munronoid I could suppress the NLRP3 inflammasome activation and pyroptosis by promoting the K48-linked ubiquitination and NLRP3 degradation. It is suggested that Munronoid I might be a potential therapeutic candidate for IBD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Animales , Colitis/patología , Sulfato de Dextran/efectos adversos , Inflamasomas/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Interleucina-18/farmacología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis
17.
Cell Mol Immunol ; 19(4): 540-553, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35194174

RESUMEN

The polarization of macrophages to the M1 or M2 phenotype has a pivotal role in inflammation and host defense; however, the underlying molecular mechanism remains unclear. Here, we show that myocyte enhancer factor 2 C (MEF2C) is essential for regulating M1 macrophage polarization in response to infection and inflammation. Global gene expression analysis demonstrated that MEF2C deficiency in macrophages downregulated the expression of M1 phenotypic markers and upregulated the expression of M2 phenotypic markers. MEF2C significantly promoted the expression of interleukin-12 p35 subunit (Il12a) and interleukin-12 p40 subunit (Il12b). Myeloid-specific Mef2c-knockout mice showed reduced IL-12 production and impaired Th1 responses, which led to susceptibility to Listeria monocytogenes infection and protected against DSS-induced IBD in vivo. Mechanistically, we showed that MEF2C directly activated the transcription of Il12a and Il12b. These findings reveal a new function of MEF2C in macrophage polarization and Th1 responses and identify MEF2C as a potential target for therapeutic intervention in inflammatory and autoimmune diseases.


Asunto(s)
Factores de Transcripción MEF2 , Activación de Macrófagos , Macrófagos , Células TH1 , Animales , Biomarcadores/metabolismo , Inflamación/metabolismo , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Macrófagos/citología , Ratones , Ratones Noqueados , Células TH1/citología
18.
Mol Immunol ; 130: 69-76, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33360745

RESUMEN

Ubiquitin specific protease 14 (USP14) is a regulator of protein deubiquitination and proteasome activation, and has been implicated in negative regulation of type I IFN signaling pathway. However, the effect of USP14 on RNA virus-related inflammatory response has not been studied. Retinoic acid-inducible gene I (RIG-I) is the important pattern recognition receptor of the innate immunity to detect RNA viruses or intracellular Poly(I:C)-LMW. Here, we reported that USP14 knockdown increased pro-inflammatory cytokines production in macrophages upon VSV infection or intracellular Poly(I:C)-LMW stimulation. USP14-overexpressed HeLa cells exhibited a decrease in RIG-I-mediated IL-6 and TNF-α expression. IU1, USP14 inhibitor, significantly promotes pro-inflammatory cytokines production in VSV-infected mice in vivo. Furthermore, USP14 was also found to inhibit the RIG-I-triggered NF-κB activation by deubiquitinating K63-linked RIG-I. Thus, our results demonstrate that USP14 is a negative regulator of RIG-I-mediated inflammatory response.


Asunto(s)
Proteína 58 DEAD Box/genética , Interleucina-6/metabolismo , FN-kappa B/metabolismo , Receptores Inmunológicos/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina Tiolesterasa/fisiología , Animales , Células Cultivadas , Proteína 58 DEAD Box/fisiología , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Femenino , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Inflamación/genética , Inflamación/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Inmunológicos/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Células THP-1
19.
Biomed Pharmacother ; 138: 111543, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34311538

RESUMEN

Acute lung injury (ALI) is a severe lung disease with limited therapeutic strategies. Munronoid I, a limonoid, which is extracted and purified from Munronia sinica, exhibits effective anti-neoplastic activities. In this study, we attempted to determine the anti-inflammatory effects of Munronoid I using both the lipopolysaccharide (LPS)-induced in vivo murine ALI models and in vitro assays. Our results demonstrated that Munronoid I treatment ameliorated LPS-induced ALI and inflammation in mice. Moreover, it also significantly inhibited LPS-induced pathological injuries, infiltration of inflammatory cells, and production of IL-1ß and IL-6. Furthermore, the in vitro assay showed that Munronoid I could inhibit the LPS-induced expression of inflammatory mediators such as iNOS, COX2, and production of pro-inflammatory cytokines by suppressing the activation of NF-κB signaling pathway in mouse peritoneal macrophages. Munronoid I reduced the LPS-, tumor necrosis factor alpha (TNF-α)- or interleukin 1 beta (IL-1ß)-induced transforming growth factor beta-activated kinase 1 (TAK1) phosphorylation and protein expression. Furthermore, the Munronoid I also promoted K48-linked ubiquitination and proteasomal degradation of TAK1. Taken together, these results demonstrated that Munronoid I exhibited anti-inflammatory activities both in vitro and in vivo, which might be a potential therapeutic candidate for the treatment of ALI and pulmonary inflammation.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios/farmacología , Limoninas/farmacología , Pulmón/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/patología , Animales , Antiinflamatorios/aislamiento & purificación , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Limoninas/aislamiento & purificación , Lipopolisacáridos , Pulmón/enzimología , Pulmón/patología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Proteolisis , Ubiquitinación
20.
Int Immunopharmacol ; 86: 106783, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32652505

RESUMEN

Cirsitakaoside, isolated and purified from the stems and leaves of Premna szemaoensis and Macaranga denticulata, is a natural compound with potential anti-inflammatory effects. However, the role of Cirsitakaoside in antiviral activity and the underlying mechanism remains largely unknown. In this study, we aimed to identify whether Cirsitakaoside has antiviral activity and investigated the underlying mechanisms. Mouse peritoneal macrophages were pretreated with Cir or DMSO, and then infected by Vesicular Stomatitis Virus (VSV) for indicated hours, Q-PCR and ELISA were used to detect the expression of interferons and pro-inflammatory cytokines, immunoblot assay were employed to investigate the involved signaling pathway in the antiviral effects of Cirsitakaoside. Furthermore, mice infected with VSV were used to investigate the antiviral activities of Cirsitakaoside in vivo. Our study demonstrated that Cirsitakaoside could promote type I IFN expression and inhibit pro-inflammatory cytokines such as IL-6 and TNF-α production in mouse peritoneal macrophages infected by VSV. Suppressive viral replication effects of Cirsitakaoside were observed on VSV-infected mouse peritoneal macrophages as well. Furthermore, Cirsitakaoside significantly increased the VSV-triggered phosphorylation of TBK1, IRF3 and reduced the phosphorylation of IκBα and p65 in mouse peritoneal macrophages. in vivo, the results showed that Cirsitakaoside-treated mice were more resistant to VSV infection by producing more IFN-ß and less pro-inflammatory cytokines. Our study indicates that Cirsitakaoside is a good candidate for the treatment of viral infection and inflammation-related diseases.


Asunto(s)
Antivirales/uso terapéutico , Productos Biológicos/uso terapéutico , Flavonas/uso terapéutico , Glicósidos/uso terapéutico , Macrófagos Peritoneales/inmunología , Estomatitis Vesicular/tratamiento farmacológico , Vesiculovirus/fisiología , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA