Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Immunity ; 54(4): 632-647.e9, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33667382

RESUMEN

Aging is associated with DNA accumulation and increased homeostatic proliferation of circulating T cells. Although these attributes are associated with aging-related autoimmunity, their direct contributions remain unclear. Conventionally, KU complex, the regulatory subunit of DNA-dependent protein kinase (DNA-PK), together with the catalytic subunit of DNA-PK (DNA-PKcs), mediates DNA damage repair in the nucleus. Here, we found KU complex abundantly expressed in the cytoplasm, where it recognized accumulated cytoplasmic DNA in aged human and mouse CD4+ T cells. This process enhanced T cell activation and pathology of experimental autoimmune encephalomyelitis (EAE) in aged mice. Mechanistically, KU-mediated DNA sensing facilitated DNA-PKcs recruitment and phosphorylation of the kinase ZAK. This activated AKT and mTOR pathways, promoting CD4+ T cell proliferation and activation. We developed a specific ZAK inhibitor, which dampened EAE pathology in aged mice. Overall, these findings demonstrate a KU-mediated cytoplasmic DNA-sensing pathway in CD4+ T cells that potentiates aging-related autoimmunity.


Asunto(s)
Envejecimiento/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Citoplasma/inmunología , Proteína Quinasa Activada por ADN/inmunología , ADN/inmunología , Inflamación/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Núcleo Celular/inmunología , Proliferación Celular/fisiología , Reparación del ADN/inmunología , Células HEK293 , Humanos , Células Jurkat , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Células U937
2.
EMBO Rep ; 24(4): e56932, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36862324

RESUMEN

Obesity is associated with metabolic disorders and chronic inflammation. However, the obesity-associated metabolic contribution to inflammatory induction remains elusive. Here, we show that, compared with lean mice, CD4+ T cells from obese mice exhibit elevated basal levels of fatty acid ß-oxidation (FAO), which promote T cell glycolysis and thus hyperactivation, leading to enhanced induction of inflammation. Mechanistically, the FAO rate-limiting enzyme carnitine palmitoyltransferase 1a (Cpt1a) stabilizes the mitochondrial E3 ubiquitin ligase Goliath, which mediates deubiquitination of calcineurin and thus enhances activation of NF-AT signaling, thereby promoting glycolysis and hyperactivation of CD4+ T cells in obesity. We also report the specific GOLIATH inhibitor DC-Gonib32, which blocks this FAO-glycolysis metabolic axis in CD4+ T cells of obese mice and reduces the induction of inflammation. Overall, these findings establish a role of a Goliath-bridged FAO-glycolysis axis in mediating CD4+ T cell hyperactivation and thus inflammation in obese mice.


Asunto(s)
Ácidos Grasos , Inflamación , Animales , Ratones , Ratones Obesos , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Glucólisis , Ubiquitina-Proteína Ligasas/metabolismo , Oxidación-Reducción
3.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 29-35, 2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33201175

RESUMEN

The two-drug combined chemotherapy of platinum and fluorouracil has been reported to efficiently kill tumor cells as the first-line treatment for advanced gastric cancer. However, the effect of these drugs on T cells remains unclear. Here, we showed that T cells including CD4+ T cells and CD8+ T cells of the patients with advanced gastric cancer after platinum and fluorouracil chemotherapy exhibited enhanced ex vivo proliferation ability as compared to that before chemotherapy. In addition, platinum and fluorouracil also promoted the differentiation of human T cells into Th1 and Th9 subtypes and cytotoxic T lymphocytes (CTLs) in vitro and in vivo. Accordingly, the combination therapy greatly suppressed tumor growth with increased tumor infiltration of Th1, Th9, and CTL cells in a mouse tumor model. Moreover, in activated T cells, long-term treatment with these two drugs further facilitates T cell activation along with promoted nuclear factor-κB (NF-κB) activation. Our findings demonstrate a previously unidentified function of platinum and fluorouracil combination chemotherapy in promoting T cell-mediated antitumor immunity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Fluorouracilo/farmacología , Neoplasias/inmunología , Platino (Metal)/farmacología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Quimioterapia Combinada/métodos , Fluorouracilo/uso terapéutico , Humanos , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Melanoma/patología , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/metabolismo , Oxaliplatino/farmacología , Oxaliplatino/uso terapéutico , Platino (Metal)/uso terapéutico , Neoplasias Gástricas/sangre , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos , Linfocitos T Colaboradores-Inductores/inmunología
5.
Microbiology (Reading) ; 160(Pt 9): 1893-1902, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24996826

RESUMEN

Previously, we observed an acid-induced short-term wall extension in Flammulina velutipes apical stipes during a 15 min period after a change from a neutral to an acidic pH. This acid-induced stipe wall extension was eliminated by heating and reconstituted by a snail expansin-like protein, although we failed to isolate any endogenous expansin-like protein from F. velutipes because of its limited 1 mm fast elongation region. In this study, we report that Coprinopsis cinerea stipes possess a 9 mm fast elongation apical region, which is suitable as a model material for wall extension studies. The elongating apical stipe showed two phases of acid-induced wall extension, an initial quick short-term wall extension during the first 15 min and a slower, gradually decaying long-term wall extension over the subsequent 2 h. After heating or protein inactivation pretreatment, apical stipes lost the long-term wall extension, retaining a slower short-term wall extension, which was reconstituted by an expansin-like snail protein. In contrast, the non-elongating basal stipes showed only a weaker short-term wall extension. We propose that the long-term wall extension is a protein-mediated process involved in stipe elongation, whereas the short-term wall extension is a non-protein mediated process not involved in stipe elongation.


Asunto(s)
Ácidos/toxicidad , Adaptación Fisiológica , Agaricales/efectos de los fármacos , Agaricales/crecimiento & desarrollo , Pared Celular/efectos de los fármacos , Pared Celular/fisiología , Estrés Fisiológico , Agaricales/efectos de la radiación , Pared Celular/efectos de la radiación , Calor , Concentración de Iones de Hidrógeno
6.
Cell Rep Med ; 5(5): 101550, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38723624

RESUMEN

Tumor recurrence after chemoradiotherapy is challenging to overcome, and approaches to predict the recurrence remain elusive. Here, human cervical cancer tissues before and after concurrent chemoradiotherapy (CCRT) analyzed by single-cell RNA sequencing reveal that CCRT specifically promotes CD8+ T cell senescence, driven by atypical chemokine receptor 2 (ACKR2)+ CCRT-resistant tumor cells. Mechanistically, ACKR2 expression is increased in response to CCRT and is also upregulated through the ligation of CC chemokines that are produced by activated myeloid and T cells. Subsequently, ACKR2+ tumor cells are induced to produce transforming growth factor ß to drive CD8+ T cell senescence, thereby compromising antitumor immunity. Moreover, retrospective analysis reveals that ACKR2 expression and CD8+ T cell senescence are enhanced in patients with cervical cancer who experienced recurrence after CCRT, indicating poor prognosis. Overall, we identify a subpopulation of CCRT-resistant ACKR2+ tumor cells driving CD8+ T cell senescence and tumor recurrence and highlight the prognostic value of ACKR2 and CD8+ T cell senescence for chemoradiotherapy recurrence.


Asunto(s)
Linfocitos T CD8-positivos , Senescencia Celular , Quimioradioterapia , Recurrencia Local de Neoplasia , Neoplasias del Cuello Uterino , Humanos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Femenino , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/terapia , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Quimioradioterapia/métodos , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/genética , Animales , Ratones , Línea Celular Tumoral , Pronóstico , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento Transformador beta/metabolismo , Senescencia de Células T
7.
Cancer Res ; 84(5): 688-702, 2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38199791

RESUMEN

Detection of cytoplasmic DNA is an essential biological mechanism that elicits IFN-dependent and immune-related responses. A better understanding of the mechanisms regulating cytoplasmic DNA sensing in tumor cells could help identify immunotherapeutic strategies to improve cancer treatment. Here we identified abundant cytoplasmic DNA accumulated in lung squamous cell carcinoma (LUSC) cells. DNA-PK, but not cGAS, functioned as a specific cytoplasmic DNA sensor to activate downstream ZAK/AKT/mTOR signaling, thereby enhancing the viability, motility, and chemoresistance of LUSC cells. DNA-PK-mediated cytoplasmic DNA sensing boosted glycolysis in LUSC cells, and blocking glycolysis abolished the tumor-promoting activity of cytoplasmic DNA. Elevated DNA-PK-mediated cytoplasmic DNA sensing was positively correlated with poor prognosis of human patients with LUSC. Targeting signaling activated by cytoplasmic DNA sensing with the ZAK inhibitor iZAK2 alone or in combination with STING agonist or anti-PD-1 antibody suppressed the tumor growth and improved the survival of mouse lung cancer models and human LUSC patient-derived xenografts model. Overall, these findings established DNA-PK-mediated cytoplasmic DNA sensing as a mechanism that supports LUSC malignancy and highlight the potential of targeting this pathway for treating LUSC. SIGNIFICANCE: DNA-PK is a cytoplasmic DNA sensor that activates ZAK/AKT/mTOR signaling and boosts glycolysis to enhance malignancy and chemoresistance of lung squamous cell carcinoma.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Animales , Ratones , Humanos , Resistencia a Antineoplásicos , Proteínas Proto-Oncogénicas c-akt , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Proteína Quinasa Activada por ADN , Glucólisis , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Pulmón , Serina-Treonina Quinasas TOR , Pronóstico
8.
Exp Hematol Oncol ; 13(1): 52, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760861

RESUMEN

BACKGROUND: Non-small cell lung cancer (NSCLC) is one of the predominant malignancies globally. Percutaneous thermal ablation (PTA) has gained widespread use among NSCLC patients, with the potential to elicit immune responses but limited therapeutic efficacies for advanced-stage disease. T-helper type 9 (Th9) cells are a subset of CD4+ effector T cells with robust and persistent anti-tumor effects. This study proposes to develop PTA-Th9 cell integrated therapy as a potential strategy for NSCLC treatment. METHODS: The therapeutic efficacies were measured in mice models with subcutaneously transplanted, recurrence, or lung metastatic tumors. The tumor microenvironments (TMEs) were evaluated by flow cytometry. The cytokine levels were assessed by ELISA. The signaling molecules were determined by quantitative PCR and Western blotting. The translational potential was tested in the humanized NSCLC patient-derived xenograft (PDX) model. RESULTS: We find that PTA combined with adoptive Th9 cell transfer therapy substantially suppresses tumor growth, recurrence, and lung metastasis, ultimately extending the survival of mice with NSCLC grafts, outperforming both PTA and Th9 cell transfer monotherapy. Analysis of TMEs indicates that combinatorial therapy significantly augments tumor-infiltrating Th9 cells, boosts anti-tumor effects of CD8+ T cells, and remodels tumor immunosuppressive microenvironments. Moreover, combinatorial therapy significantly strengthens the regional and circulation immune response of CD8+ T cells in mice with tumor lung metastasis and induces peripheral CD8+ T effector memory cells in mice with tumor recurrence. Mechanically, PTA reinforces the anti-tumor ability of Th9 cells primarily through upregulating interleukin (IL)-1ß and subsequently activating the downstream STAT1/IRF1 pathway, which could be effectively blocked by intercepting IL-1ß signaling. Finally, the enhanced therapeutic effect of combinatorial therapy is validated in humanized NSCLC PDX models. CONCLUSIONS: Collectively, this study demonstrates that combinatorial therapy displays robust and durable anti-tumor efficacy and excellent translational potential, offering excellent prospects for translation and emerging as a promising approach for NSCLC treatment.

9.
iScience ; 26(2): 105954, 2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36718369

RESUMEN

Protein phosphatase 1 regulatory subunit 15A (PPP1R15A) is an important factor in the integrated stress response (ISR) in mammals and may play a crucial role in tumorigenesis. In our studies, we found an inhibitor of PPP1R15A, Sephin1, plays a protumorigenic role in mouse tumor models. By analyzing the single-cell transcriptome data of the mouse tumor models, we found that in C57BL/6 mice, Sephin1 treatment could lead to higher levels of ISR activity and lower levels of antitumor immune activities. Specifically, Sephin1 treatment caused reductions in antitumor immune cell types and lower expression levels of cytotoxicity-related genes. In addition, T cell receptor (TCR) repertoire analysis demonstrated that the clonal expansion of tumor-specific T cells was inhibited by Sephin1. A special TCR + macrophage subtype in tumor was identified to be significantly depleted upon Sephin1 treatment, implying its key antitumor role. These results suggest that PPP1R15A has the potential to be an effective target for tumor therapy.

10.
STAR Protoc ; 4(4): 102616, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37756156

RESUMEN

Here, we present a protocol for exploring the effects of PPP1R15A inhibitor, Sephin1, on antitumor immunity of B16F1 subcutaneous tumor in mice. We describe steps for constructing single-cell transcriptome and TCR libraries, sequencing, and using sequencing data for the integration of expression and TCR data. We then detail procedures for gene differentiation, regulon and cell-cell communication analysis, and validation of single-cell analysis results. For complete details on the use and execution of this protocol, please refer to Wang et al.1.


Asunto(s)
Comunicación Celular , Neoplasias , Animales , Ratones , Modelos Animales de Enfermedad , Análisis de la Célula Individual , Receptores de Antígenos de Linfocitos T
11.
Front Physiol ; 13: 898659, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35685283

RESUMEN

Background: The impairment of microvascular injury on prognosis has increasingly drawn extensive awareness along with the high morbidity and mortality of ST-segment elevation myocardial infarction (STEMI) over recent years. The prognostic significance of computational pressure-fluid dynamics applied to index of microcirculatory resistance, derived from coronary angiography (CPFD-caIMR) in microvascular injury evaluation of STEMI patients remained inconclusive. Methods: A total of 213 patients who met the inclusion criteria were selected retrospectively from 1003 STEMI patients from February 2018 to February 2020. Propensity score matching (PSM) was thereafter finished. CPFD-caIMR of all patients was obtained off-line using the software (FlashAngio, Rainmed Ltd., Suzhou, China) after PPCI. The primary endpoint was to compare the CPFD-caIMR and the incidence of major adverse cardiovascular events (MACEs) between drug-coated balloons (DCB) and drug-eluting stents (DES) groups. The correlation between CPFD-caIMR and MACEs was analyzed, and the prognosis of patients with STEMI was evaluated by CPFD-caIMR by multivariate regression analysis. Results: Totally 213 STEMI patients with successful primary percutaneous coronary intervention (PPCI) were included, of whom 84 patients accepted DCB and 129 patients accepted DES respectively. Baseline characteristics and CPFD-caIMR were comparable between DCB and DES groups after PSM (62 patients in each group). CPFD-caIMR was not significantly different between two groups (DES vs. DCB: mean difference: 2.26, 95% CI -4.05 to 8.57, p = 0.45), and so was it when re-grouped by whether CPFD-caIMR > 40U or not (DES vs. DCB: 34.17% vs. 27.16%, p = 0.29). After a follow-up of 1 year, more MACEs occurred in DES group than DCB group (relative risk: 2.50, 95% CI 1.04 to 6.02, p = 0.04). The predictors of MACEs by multi-variate analysis found that, only time from symptom to balloon (p = 0.03) and time from door to balloon (p < 0.01) were independent predictors of MACEs, independent of treatment with DCB or DES intervention. Furthermore, CPFD-caIMR > 40U became an independent predictor of the combined events including cardiovascular deaths or heart failure readmission irrespective of PSM (odds ratio: 4.07, 95% CI: 1.06 to 7.66, p = 0.04). Conclusion: CPFD-caIMR was a promising method for prognosis, which can predict CV death or heart failure readmission in STEMI patients. DCB was a possible strategy in PPCI of STEMI patients, not inferior to DES based on microvascular injury evaluated by CPFD-caIMR.

12.
Cell Death Differ ; 29(10): 2009-2023, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35568718

RESUMEN

Inflammasome contributes to the pathogenesis of various inflammatory diseases, but the epigenetic mechanism controlling its activation remains elusive. Here, we found that the histone methyltransferase Ezh2 mediates the activation of multiple types of inflammasomes in macrophages/microglia independent of its methyltransferase activity and thus promotes inflammasome-related pathologies. Mechanistically, Ezh2 functions through its SANT2 domain to maintain the enrichment of H3K27 acetylation in the promoter region of the long noncoding RNA (lncRNA) Neat1, thereby promoting chromatin accessibility and facilitating p65-mediated transcription of Neat1, which is a critical mediator of inflammasome assembly and activation. In addition, the tumour suppressor protein p53 competes with Ezh2 for the same binding region in the Neat1 promoter and thus antagonises Ezh2-induced Neat1 transcription and inflammasome activation. Therefore, loss of Ezh2 strongly promotes the binding of p53, which recruits the deacetylase SIRT1 for H3K27 deacetylation of the Neat1 promoter and thus suppresses Neat1 transcription and inflammasome activation. Overall, our study demonstrates an epigenetic mechanism involved in modulating inflammasome activation through an Ezh2/p53 competition model and highlights a novel function of Ezh2 in maintaining H3K27 acetylation to support lncRNA Neat1 transcription.


Asunto(s)
ARN Largo no Codificante , Cromatina , Histona Metiltransferasas/genética , Histona Metiltransferasas/metabolismo , Inflamasomas/metabolismo , ARN Largo no Codificante/metabolismo , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
J Clin Invest ; 132(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35230972

RESUMEN

IFN-γ-stimulated MHC class I (MHC-I) antigen presentation underlies the core of antitumor immunity. However, sustained IFN-γ signaling also enhances the programmed death ligand 1 (PD-L1) checkpoint pathway to dampen antitumor immunity. It remains unclear how these opposing effects of IFN-γ are regulated. Here, we report that loss of the histone dimethyltransferase WHSC1 impaired the antitumor effect of IFN-γ signaling by transcriptional downregulation of the MHC-I machinery without affecting PD-L1 expression in colorectal cancer (CRC) cells. Whsc1 loss promoted tumorigenesis via a non-cell-autonomous mechanism in an Apcmin/+ mouse model, CRC organoids, and xenografts. Mechanistically, we found that the IFN-γ/STAT1 signaling axis stimulated WHSC1 expression and, in turn, that WHSC1 directly interacted with NLRC5 to promote MHC-I gene expression, but not that of PD-L1. Concordantly, silencing Whsc1 diminished MHC-I levels, impaired antitumor immunity, and blunted the effect of immune checkpoint blockade. Patient cohort analysis revealed that WHSC1 expression positively correlated with enhanced MHC-I expression, tumor-infiltrating T cells, and favorable disease outcomes. Together, our findings establish a tumor-suppressive function of WHSC1 that relays IFN-γ signaling to promote antigen presentation on CRC cells and provide a rationale for boosting WHSC1 activity in immunotherapy.


Asunto(s)
Antígeno B7-H1 , N-Metiltransferasa de Histona-Lisina , Neoplasias , Proteínas Represoras , Animales , Presentación de Antígeno , Antígeno B7-H1/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas , Humanos , Interferón gamma , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones
14.
Nat Commun ; 13(1): 7281, 2022 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-36435834

RESUMEN

Chronic inflammation and an immunosuppressive microenvironment promote prostate cancer (PCa) progression and diminish the response to immune checkpoint blockade (ICB) therapies. However, it remains unclear how and to what extent these two events are coordinated. Here, we show that ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, functions downstream of inflammation-induced IKKß activation to shape the immunosuppressive tumor microenvironment (TME). Prostate-specific deletion of Arid1a cooperates with Pten loss to accelerate prostate tumorigenesis. We identify polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) as the major infiltrating immune cell type that causes immune evasion and reveal that neutralization of PMN-MDSCs restricts the progression of Arid1a-deficient tumors. Mechanistically, inflammatory cues activate IKKß to phosphorylate ARID1A, leading to its degradation via ß-TRCP. ARID1A downregulation in turn silences the enhancer of A20 deubiquitinase, a critical negative regulator of NF-κB signaling, and thereby unleashes CXCR2 ligand-mediated MDSC chemotaxis. Importantly, our results support the therapeutic strategy of anti-NF-κB antibody or targeting CXCR2 combined with ICB for advanced PCa. Together, our findings highlight that the IKKß/ARID1A/NF-κB feedback axis integrates inflammation and immunosuppression to promote PCa progression.


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias de la Próstata , Masculino , Humanos , Próstata/metabolismo , Quimiotaxis , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Neoplasias de la Próstata/metabolismo , FN-kappa B/metabolismo , Receptores de Interleucina-8B/metabolismo , Proteínas Serina-Treonina Quinasas , Inflamación/genética , Inflamación/metabolismo , Microambiente Tumoral/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Sci China Life Sci ; 65(12): 2354-2454, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36066811

RESUMEN

Aging is characterized by a progressive deterioration of physiological integrity, leading to impaired functional ability and ultimately increased susceptibility to death. It is a major risk factor for chronic human diseases, including cardiovascular disease, diabetes, neurological degeneration, and cancer. Therefore, the growing emphasis on "healthy aging" raises a series of important questions in life and social sciences. In recent years, there has been unprecedented progress in aging research, particularly the discovery that the rate of aging is at least partly controlled by evolutionarily conserved genetic pathways and biological processes. In an attempt to bring full-fledged understanding to both the aging process and age-associated diseases, we review the descriptive, conceptual, and interventive aspects of the landscape of aging composed of a number of layers at the cellular, tissue, organ, organ system, and organismal levels.


Asunto(s)
Enfermedades Cardiovasculares , Neoplasias , Humanos , Envejecimiento/genética , Envejecimiento/metabolismo , Neoplasias/genética
16.
Cell Mol Immunol ; 18(4): 969-978, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33707688

RESUMEN

T follicular helper (Tfh) cells are crucial for regulating autoimmune inflammation and protective immunity against viral infection. However, the molecular mechanism controlling Tfh cell differentiation is poorly understood. Here, through two mixed bone marrow chimeric experiments, we identified Peli1, a T cell-enriched E3 ubiquitin ligase, as an intrinsic regulator that inhibits Tfh cell differentiation. Peli1 deficiency significantly promoted c-Rel-mediated inducible T-cell costimulator (ICOS) expression, and PELI1 mRNA expression was negatively associated with ICOS expression on human CD4+ T cells. Mechanistically, increased ICOS expression on Peli1-KO CD4+ T cells enhanced the activation of PI3K-AKT signaling and thus suppressed the expression of Klf2, a transcription factor that inhibits Tfh differentiation. Therefore, reconstitution of Klf2 abolished the differences in Tfh differentiation and germinal center reaction between WT and Peli1-KO cells. As a consequence, Peli1-deficient CD4+ T cells promoted lupus-like autoimmunity but protected against H1N1 influenza virus infection in mouse models. Collectively, our findings established Peli1 as a critical negative regulator of Tfh differentiation and indicated that targeting Peli1 may have beneficial therapeutic effects in Tfh-related autoimmunity or infectious diseases.


Asunto(s)
Autoinmunidad , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Lupus Eritematoso Sistémico/patología , Activación de Linfocitos/inmunología , Proteínas Nucleares/fisiología , Infecciones por Orthomyxoviridae/prevención & control , Células T Auxiliares Foliculares/inmunología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Diferenciación Celular , Femenino , Regulación de la Expresión Génica , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Lupus Eritematoso Sistémico/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología
17.
J Exp Med ; 218(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33914044

RESUMEN

TGFß is essential for the generation of anti-tumor Th9 cells; on the other hand, it causes resistance against anti-tumor immunity. Despite recent progress, the underlying mechanism reconciling the double-edged effect of TGFß signaling in Th9-mediated cancer immunotherapy remains elusive. Here, we find that TGFß-induced down-regulation of bifunctional apoptosis regulator (BFAR) represents the key mechanism preventing the sustained activation of TGFß signaling and thus impairing Th9 inducibility. Mechanistically, BFAR mediates K63-linked ubiquitination of TGFßR1 at K268, which is critical to activate TGFß signaling. Thus, BFAR deficiency or K268R knock-in mutation suppresses TGFßR1 ubiquitination and Th9 differentiation, thereby inhibiting Th9-mediated cancer immunotherapy. More interestingly, BFAR-overexpressed Th9 cells exhibit promising therapeutic efficacy to curtail tumor growth and metastasis and promote the sensitivity of anti-PD-1-mediated checkpoint immunotherapy. Thus, our findings establish BFAR as a key TGFß-regulated gene to fine-tune TGFß signaling that causes Th9 induction insensitivity, and they highlight the translational potential of BFAR in promoting Th9-mediated cancer immunotherapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de la Membrana/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Diferenciación Celular/inmunología , Regulación hacia Abajo/inmunología , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Colaboradores-Inductores/inmunología
18.
Mitochondrial DNA B Resour ; 5(3): 2228-2230, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33366984

RESUMEN

Camellia nitidissima is an endangered species. This species contains two varieties. Here, we report on the chloroplast genomes of C. nitidissima var. nitidissima from Fangcheng (GenBank accession MT157617) and Nanning (MT157618), as well as one sample of C. nitidissima var. microcarpa (MT157619) from Nanning. The total chloroplast genomes of C. nitidissima var. nitidissima Fangcheng and Nanning samples are 156,596 bp and 157,567 bp in length, respectively. C. nitidissima var. microcarpa (MT157619) genome is 157,407 bp in length. The three samples possess GC contents of 37.3%, 128 genes, comprising 86 protein-coding genes, 34 tRNA genes, and 8 rRNA genes.

19.
Plant Divers ; 42(5): 343-350, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33134617

RESUMEN

Camellia huana is an endangered species with a narrow distribution in limestone hills of northern Guangxi and southern Guizhou provinces, China. We used one chloroplast DNA (cpDNA) fragment and 12 pairs of microsatellite (simple sequence repeat; SSR) markers to assess the genetic diversity and structure of 12 C. huana populations. A total of 99 alleles were detected for 12 polymorphic loci, and eight haplotypes and nine polymorphic sites were detected within 5200 bp of cpDNA. C. huana populations showed a low level of genetic diversity (n = 8, Hd = 0.759, Pi = 0.00042 for cpDNA, N A  = 3.931, H E  = 0.466 for SSRs), but high genetic differentiation between populations (F ST  = 0.2159 for SSRs, F ST  = 0.9318 for cpDNA). This can be attributed to the narrow distribution and limestone habitat of C. huana. STRUCTURE analysis divided natural C. huana populations into two groups, consistent with their geographical distribution. Thus, we suggest that five natural C. huana populations should be split into two units to be managed effectively.

20.
J Exp Med ; 217(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32324863

RESUMEN

Ubiquitination is an essential mechanism in the control of antiviral immunity upon virus infection. Here, we identify a series of ubiquitination-modulating enzymes that are modulated by vesicular stomatitis virus (VSV). Notably, TRIM24 is down-regulated through direct transcriptional suppression induced by VSV-activated IRF3. Reducing or ablating TRIM24 compromises type I IFN (IFN-I) induction upon RNA virus infection and thus renders mice more sensitive to VSV infection. Mechanistically, VSV infection induces abundant TRIM24 translocation to mitochondria, where TRIM24 binds with TRAF3 and directly mediates K63-linked TRAF3 ubiquitination at K429/K436. This modification of TRAF3 enables its association with MAVS and TBK1, which consequently activates downstream antiviral signaling. Together, these findings establish TRIM24 as a critical positive regulator in controlling the activation of antiviral signaling and describe a previously unknown mechanism of TRIM24 function.


Asunto(s)
Antivirales/metabolismo , Inmunidad , Lisina/metabolismo , Proteínas Nucleares/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinación , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Regulación hacia Abajo , Células HEK293 , Humanos , Inflamación/genética , Interferón Tipo I/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Modelos Biológicos , Proteínas Nucleares/química , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Dominios RING Finger , Transducción de Señal , Factor 3 Asociado a Receptor de TNF/química , Factor 3 Asociado a Receptor de TNF/genética , Factores de Transcripción/química , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transcripción Genética , Virus de la Estomatitis Vesicular Indiana/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA