Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 257
Filtrar
1.
Front Immunol ; 12: 769167, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956195

RESUMEN

Inflammatory bowel disease (IBD), which include Crohn's disease (CD) and ulcerative colitis (UC), exhibits a complex multifactorial pathogenesis involving genetic susceptibility, imbalance of gut microbiota, mucosal immune disorder and environmental factors. Recent studies reported associations between ubiquitination and deubiquitination and the occurrence and development of inflammatory bowel disease. Ubiquitination modification, one of the most important types of post-translational modifications, is a multi-step enzymatic process involved in the regulation of various physiological processes of cells, including cell cycle progression, cell differentiation, apoptosis, and innate and adaptive immune responses. Alterations in ubiquitination and deubiquitination can lead to various diseases, including IBD. Here, we review the role of E3 ubiquitin ligases and deubiquitinases (DUBs) and their mediated ubiquitination and deubiquitination modifications in the pathogenesis of IBD. We highlight the importance of this type of posttranslational modification in the development of inflammation, and provide guidance for the future development of targeted therapeutics in IBD.


Asunto(s)
Enzimas Desubicuitinizantes/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación/inmunología , Inmunidad Adaptativa/inmunología , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/enzimología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Humanos , Inmunidad Innata/inmunología , Inflamación/enzimología , Inflamación/inmunología , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
2.
Front Immunol ; 12: 698236, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34650549

RESUMEN

Recent publications have revealed that N6-methyladenosine (m6A) modification is critically involved in tumorigenesis and metastasis. However, the correlation of m6A modification and immune infiltration in early-stage lung adenocarcinoma (LUAD) is still uncertain. We performed NMF clustering based on 23 m6A regulators and identify three distinct m6A clusters and three m6A related genes clusters (m6A cluster-R) in early-stage LUAD. The immune infiltrating levels were calculated using CIBERSORT, MCPcounter and ssGSEA algorithms. And we established the m6A-predictive score to quantify m6A modified phenotypes and predict immunotherapeutic responses. Based on the TME characteristics, different immune profiles were also identified among three m6A gene-related clusters. And the m6A-R-C2 was related to a favorable overall survival (OS), whereas m6A-R-C3 had unfavorable overall survival. The m6A-predictive score was built according to the expression levels of m6A-related genes, and patients could be stratified into subgroups with low/high scores. Patients with high scores had poor overall survival, enhanced immune infiltration, high tumor mutation burden and increased level of somatic mutation. Besides, patients with high scores had unfavorable overall survival in the anti-PD-1 cohort, whereas the overall survival of high-score patients was better in the adoptive T cell therapy cohort. Our work highlights that m6A modification is closely related to immune infiltration in early-stage LUAD, which also contributes to the development of more effective immunotherapy strategies.


Asunto(s)
Adenocarcinoma del Pulmón , Adenosina/análogos & derivados , Neoplasias Pulmonares , Microambiente Tumoral/inmunología , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/metabolismo , Adenosina/genética , Adenosina/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/inmunología
3.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638916

RESUMEN

Rheumatoid arthritis (RA) is caused by prolonged periodic interactions between genetic, environmental, and immunologic factors. Posttranslational modifications (PTMs) such as citrullination, carbamylation, and acetylation are correlated with the pathogenesis of RA. PTM and cell death mechanisms such as apoptosis, autophagy, NETosis, leukotoxic hypercitrullination (LTH), and necrosis are related to each other and induce autoantigenicity. Certain microbial infections, such as those caused by Porphyromonasgingivalis, Aggregatibacter actinomycetemcomitans, and Prevotella copri, can induce autoantigens in RA. Anti-modified protein antibodies (AMPA) containing anti-citrullinated protein/peptide antibodies (ACPAs), anti-carbamylated protein (anti-CarP) antibodies, and anti-acetylated protein antibodies (AAPAs) play a role in pathogenesis as well as in prediction, diagnosis, and prognosis. Interestingly, smoking is correlated with both PTMs and AMPAs in the development of RA. However, there is lack of evidence that smoking induces the generation of AMPAs.


Asunto(s)
Artritis Reumatoide/inmunología , Citrulinación/inmunología , Carbamilación de Proteína/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Acetilación , Artritis Reumatoide/diagnóstico , Artritis Reumatoide/metabolismo , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Reacciones Cruzadas/inmunología , Humanos
4.
Commun Biol ; 4(1): 825, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34211107

RESUMEN

Extracellular vesicles can modulate diverse processes ranging from proliferation and tissue repair, to chemo-resistance and cellular differentiation. With the advent of tissue and immunological targeting, extracellular vesicles are also increasingly viewed as promising vectors to deliver peptide-based cancer antigens to the human immune system. Despite the clinical relevance and therapeutic potential of such 'cell-free' approaches, the natural antigen presentation landscape exported in extracellular vesicles is still largely uncharted, due to the challenging nature of such preparations and analyses. In the context of therapeutic vesicle production, a critical evaluation of the similarity in vesicular antigen presentation is also urgently needed. In this work, we compared the HLA-I peptide ligandomes of extracellular vesicles against that of whole-cells of the same cell line. We found that extracellular vesicles not only over-represent HLA-B complexes and peptide ligands, but also cysteinylated peptides that may modulate immune responses. Collectively, these findings describe the pre-existing provision of vesicular HLA complexes that may be utilized to carry peptide vaccines, as well as the propensity for different peptide and post-translationally modified ligands to be presented, and will outline critical considerations in devising novel EV vaccination strategies.


Asunto(s)
Presentación de Antígeno/inmunología , Cisteína/inmunología , Vesículas Extracelulares/inmunología , Antígenos HLA-B/inmunología , Línea Celular , Cromatografía Liquida/métodos , Cisteína/metabolismo , Vesículas Extracelulares/metabolismo , Antígenos HLA-B/metabolismo , Humanos , Ligandos , Péptidos/inmunología , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional/inmunología , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos
5.
Nat Commun ; 12(1): 2970, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34016972

RESUMEN

Activation of MAVS, an adaptor molecule in Rig-I-like receptor (RLR) signaling, is indispensable for antiviral immunity, yet the molecular mechanisms modulating MAVS activation are not completely understood. Ubiquitination has a central function in regulating the activity of MAVS. Here, we demonstrate that a mitochondria-localized deubiquitinase USP18 specifically interacts with MAVS, promotes K63-linked polyubiquitination and subsequent aggregation of MAVS. USP18 upregulates the expression and production of type I interferon following infection with Sendai virus (SeV) or Encephalomyocarditis virus (EMCV). Mice with a deficiency of USP18 are more susceptible to RNA virus infection. USP18 functions as a scaffold protein to facilitate the re-localization of TRIM31 and enhances the interaction between TRIM31 and MAVS in mitochondria. Our results indicate that USP18 functions as a post-translational modulator of MAVS-mediated antiviral signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Cardiovirus/inmunología , Infecciones por Respirovirus/inmunología , Ubiquitina Tiolesterasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/aislamiento & purificación , Animales , Infecciones por Cardiovirus/virología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/inmunología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Lisina/metabolismo , Masculino , Ratones , Ratones Noqueados , Procesamiento Proteico-Postraduccional/inmunología , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Infecciones por Respirovirus/virología , Virus Sendai/inmunología , Transducción de Señal/inmunología , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/aislamiento & purificación , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/inmunología
6.
Front Immunol ; 12: 652160, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859645

RESUMEN

Cancer cells are under the surveillance of the host immune system. Nevertheless, a number of immunosuppressive mechanisms allow tumors to escape protective responses and impose immune tolerance. Epigenetic alterations are central to cancer cell biology and cancer immune evasion. Accordingly, epigenetic modulating agents (EMAs) are being exploited as anti-neoplastic and immunomodulatory agents to restore immunological fitness. By simultaneously acting on cancer cells, e.g. by changing expression of tumor antigens, immune checkpoints, chemokines or innate defense pathways, and on immune cells, e.g. by remodeling the tumor stroma or enhancing effector cell functionality, EMAs can indeed overcome peripheral tolerance to transformed cells. Therefore, combinations of EMAs with chemo- or immunotherapy have become interesting strategies to fight cancer. Here we review several examples of epigenetic changes critical for immune cell functions and tumor-immune evasion and of the use of EMAs in promoting anti-tumor immunity. Finally, we provide our perspective on how EMAs could represent a game changer for combinatorial therapies and the clinical management of cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/terapia , Escape del Tumor/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Terapia Combinada/métodos , Metilación de ADN/efectos de los fármacos , Metilación de ADN/inmunología , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Epigénesis Genética/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia Adoptiva/métodos , Neoplasias/genética , Neoplasias/inmunología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/inmunología , Receptores Quiméricos de Antígenos/inmunología , Resultado del Tratamiento , Escape del Tumor/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
7.
Front Immunol ; 12: 646633, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679811

RESUMEN

Conjugation with the small ubiquitin-like modifier (SUMO) constitutes a key post-translational modification regulating the stability, activity, and subcellular localization of its target proteins. However, the vast numbers of identified SUMO substrates obscure a clear view on the function of SUMOylation in health and disease. This article presents a comprehensive review on the physiological relevance of SUMOylation by discussing how global SUMOylation levels-rather than specific protein SUMOylation-shapes the immune response. In particular, we highlight the growing body of work on SUMOylation in intestinal pathologies, because of the unique metabolic, infectious, and inflammatory challenges of this organ. Recent studies show that global SUMOylation can help restrain detrimental inflammation while maintaining immune defenses and tissue integrity. These results warrant further efforts to develop new therapeutic tools and strategies to control SUMOylation in infectious and inflammatory disorders.


Asunto(s)
Tracto Gastrointestinal/inmunología , Inflamación/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Estrés Fisiológico/inmunología , Animales , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/metabolismo , Humanos , Interferones/inmunología , Interferones/metabolismo , Macrófagos/inmunología , Neutrófilos/inmunología , Sumoilación/inmunología
8.
Int J Biol Macromol ; 171: 423-427, 2021 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-33428955

RESUMEN

Membrane-associated RING (really interesting new gene)-cysteine-histidine (CH) (MARCH) ubiquitin ligases belong to a RING finger domain E3 ligases family. So far, eleven members have been found in the MARCH family, which are MARCH 1 to 11. The members of the MARCH family are widely distributed and involve in a variety of cellular functions, including regulation of the immune system, transmembrane transport of proteins, protein stability, endoplasmic reticulum-related degradation, and endosome protein transport. Several seminal studies over the past decade have delineated that MARCH affects viral replication through various mechanisms by regulating the activity of signaling molecules and their expression in the antiviral innate immune responses. Here, we summarize the complex roles of MARCH ligases in the antiviral innate immune signaling pathway and its impact on viral replication in host immune defense systems. A better understanding of this interplay's molecular mechanisms is important concerning the development of new therapeutics targeting viral infections.


Asunto(s)
Inmunidad Innata/fisiología , Procesamiento Proteico-Postraduccional/inmunología , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación/inmunología , Virosis/enzimología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Antivirales/farmacología , ADN Viral/inmunología , Diseño de Fármacos , Interacciones Huésped-Patógeno , Humanos , Receptores Inmunológicos , Transducción de Señal , Receptores Toll-Like/fisiología , Virosis/inmunología , Replicación Viral/inmunología
9.
FASEB J ; 35(1): e21234, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33337557

RESUMEN

Emerging evidences highlight importance of epigenetic regulation and their integration with transcriptional and cell signaling machinery in determining tissue resident adult pluripotent mesenchymal stem/stromal cell (MSC) activity, lineage commitment, and multicellular development. Histone modifying enzymes and large multi-subunit chromatin remodeling complexes and their cell type-specific plasticity remain the central defining features of gene regulation and establishment of tissue identity. Modulation of transcription factor expression gradient ex vivo and concomitant flexibility of higher order chromatin architecture in response to signaling cues are exciting approaches to regulate MSC activity and tissue rejuvenation. Being an important constituent of the adult bone marrow microenvironment/niche, pathophysiological perturbation in MSC homeostasis also causes impaired hematopoietic stem/progenitor cell function in a non-cell autonomous mechanism. In addition, pluripotent MSCs can function as immune regulatory cells, and they reside at the crossroad of innate and adaptive immune response pathways. Research in the past few years suggest that MSCs/stromal fibroblasts significantly contribute to the establishment of immunosuppressive microenvironment in shaping antitumor immunity. Therefore, it is important to understand mesenchymal stromal epigenome and transcriptional regulation to leverage its applications in regenerative medicine, epigenetic memory-guided trained immunity, immune-metabolic rewiring, and precision immune reprogramming. In this review, we highlight the latest developments and prospects in chromatin biology in determining MSC function in the context of lineage commitment and immunomodulation.


Asunto(s)
Ensamble y Desensamble de Cromatina/inmunología , Células Madre Hematopoyéticas/inmunología , Histonas/inmunología , Células Madre Mesenquimatosas/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Nicho de Células Madre/inmunología , Animales , Células Madre Hematopoyéticas/citología , Humanos , Células Madre Mesenquimatosas/citología
10.
Front Immunol ; 12: 786857, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069561

RESUMEN

Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric Ser/Thr phosphatase that regulates many cellular processes. The role of PP2A as a tumor suppressor has been extensively studied and reviewed. However, emerging evidence suggests PP2A constrains inflammatory responses and is important in autoimmune and neuroinflammatory diseases. Here, we reviewed the existing literature on the role of PP2A in T-cell differentiation and autoimmunity. We have also discussed the modulation of PP2A activity by endogenous inhibitors and its small-molecule activators as potential therapeutic approaches against autoimmunity.


Asunto(s)
Autoinmunidad , Diferenciación Celular/inmunología , Proteína Fosfatasa 2/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Humanos , Procesamiento Proteico-Postraduccional/inmunología
11.
Front Immunol ; 11: 586613, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329564

RESUMEN

Dendritic cells (DCs) are specialized antigen-presenting cells that play a key role in immune homeostasis and the adaptive immune response. DC-induced immune tolerance or activation is strictly dependent on the distinct maturation stages and migration ability of DCs. Ubiquitination is a reversible protein post-translational modification process that has emerged as a crucial mechanism that regulates DC maturation and function. Recent studies have shown that ubiquitin enzymes, including E3 ubiquitin ligases and deubiquitinases (DUBs), are pivotal regulators of DC-mediated immune function and serve as potential targets for DC-based immunotherapy of immune-related disorders (e.g., autoimmune disease, infections, and tumors). In this review, we summarize the recent progress regarding the molecular mechanisms and function of ubiquitination in DC-mediated immune homeostasis and immune response.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ubiquitinación/fisiología , Animales , Diferenciación Celular/inmunología , Humanos , Procesamiento Proteico-Postraduccional/inmunología
12.
Int Rev Cell Mol Biol ; 357: 35-55, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33234244

RESUMEN

The Tyro3, Axl, and MerTK (TAM) receptors are three homologous Type I Receptor Tyrosine Kinases that have important homeostatic functions in multicellular organisms by regulating the clearance of apoptotic cells (efferocytosis). Pathologically, TAM receptors are overexpressed in a wide array of human cancers, and often associated with aggressive tumor grade and poor overall survival. In addition to their expression on tumor cells, TAMs are also expressed on infiltrating myeloid-derived cells in the tumor microenvironment, where they appear to act akin to negative immune checkpoints that impair host anti-tumor immunity. The ligands for TAMs are two endogenous proteins, Growth Arrest-Specific 6 (Gas6) and Protein S (Pros1), that function as bridging molecules between externalized phosphatidylserine (PtdSer) on apoptotic cells and the TAM ectodomains. One interesting feature of TAMs biology is that their ligand proteins require specific post-translational modifications to acquire activities. This chapter summarized these important modifications and explained the molecular mechanisms behind such phenomenon. Current evidences suggest that these modifications help Gas6/Pros1 to achieve optimal PtdSer-binding capacities. In addition, this chapter included recent discovery of regulating machineries of PtdSer dynamic across the plasma membrane, as well as their potential impacts in the tumor microenvironment. Taken together, this review highlights the importance of the upstream PtdSer and Gas6 in regulating TAMs' function and hope to provide researchers with new perspectives to inspire future studies of TAM receptors in human disease models.


Asunto(s)
Neoplasias/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Proteínas Proto-Oncogénicas/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Microambiente Tumoral/inmunología , Tirosina Quinasa c-Mer/inmunología , Animales , Apoptosis/inmunología , Activación Enzimática/inmunología , Humanos , Péptidos y Proteínas de Señalización Intercelular/inmunología , Neoplasias/patología , Proteína S/inmunología , Tirosina Quinasa del Receptor Axl
13.
Future Oncol ; 16(36): 3085-3094, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32976029

RESUMEN

Indoleamine 2,3 dioxygenase (IDO), first discovered in the 1960s, is an enzyme that has become a highly investigated metabolic target in cancer research. IDO is the rate-limiting step in tryptophan metabolism catabolism into its byproducts - kynurenines. Both IDO and kynurenines have been implicated in altering the tumor microenvironment, allowing for a tolerogenesis by affecting T-cell maturation and proliferation, and more specifically by inducing differentiation into T regulatory cells. Two mechanisms have been suspected in creating this environment: tryptophan starvation and metabolite toxicity. IDO has been shown to be expressed not only in cancer cells but also in antigen-presenting cells. The exact mechanisms underlying the two different sites of expression have not been fully elucidated. To date, most literature has focused on the role of IDO in solid tumors; we provide a review of IDO and its impact on hematological malignancies - more specifically, acute myeloid leukemia. The pathophysiology of IDO will be discussed, including a summarization of the literature to date on how IDO expression effects prognosis and disease progression in acute myeloid leukemia, along with current IDO-specific therapeutics with future considerations.


Asunto(s)
Antineoplásicos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Leucemia Mieloide Aguda/inmunología , Microambiente Tumoral/inmunología , Antineoplásicos/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Progresión de la Enfermedad , Regulación Leucémica de la Expresión Génica/inmunología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Quinurenina/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Pronóstico , Supervivencia sin Progresión , Procesamiento Proteico-Postraduccional/inmunología , Tasa de Supervivencia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Triptófano/metabolismo , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología , Microambiente Tumoral/efectos de los fármacos , Regulación hacia Arriba/inmunología
14.
Mol Pharm ; 17(10): 3649-3653, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32857512

RESUMEN

Adeno-associated virus (AAV)-based gene therapy is currently limited by (1) decline in therapeutic gene expression over time, (2) immune cell activation and (3) neutralization by pre-existing antibodies. Hence, studying the interaction of AAV vectors with various cellular pathways during the production and transduction process is necessary to overcome such barriers. Post-translational modifications (PTM) of AAV vectors during the production and transduction process is known to limit its transduction efficiency and further evoke the immune response. Further, AAV vectors are known to trigger cellular stress, resulting in an upregulation of distinct arms of the unfolded protein response (UPR) pathway. Recognition of the AAV genome by Toll-like receptor-9 triggers the myeloid differentiation primary response signaling cascade for innate (IL-6, IFN-α, IFN-ß) and adaptive (CD8+ T-cell, B-cell) immune response against the viral capsid and the transgene product. Herein, we highlight a potential intersection of the UPR, PTMs, and intracellular trafficking pathways, which could be fine-tuned to augment the outcome of AAV-based gene delivery.


Asunto(s)
Dependovirus/inmunología , Terapia Genética/métodos , Interacciones Microbiota-Huesped/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Transducción Genética/métodos , Inmunidad Adaptativa/genética , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Dependovirus/genética , Interacciones Microbiota-Huesped/genética , Humanos , Inmunidad Innata/genética , Procesamiento Proteico-Postraduccional/genética , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/inmunología
15.
Theranostics ; 10(16): 7178-7192, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32641986

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide and effective therapy remains a challenge. IFIT3 is an interferon-stimulated gene with antiviral and pro-inflammatory functions. Our previous work has shown that high expression of IFIT3 is correlated with poor survival in PDAC patients who receive chemotherapy suggesting a link between IFIT3 and chemotherapy resistance in PDAC. However, the exact role and molecular mechanism of IFIT3 in chemotherapy resistance in PDAC has been unclear. Methods: A group of transcriptome datasets were downloaded and analyzed for the characterization of IFIT3 in PDAC. Highly metastatic PDAC cell line L3.6pl and patient-derived primary cell TBO368 were used and IFIT3 knockdown and the corresponding knockin cells were established for in vitro studies. Chemotherapy-induced apoptosis, ROS production, confocal immunofluorescence, subcellular fractionation, chromatin-immunoprecipitation, co-immunoprecipitation and mass spectrometry analysis were determined to further explore the biological role of IFIT3 in chemotherapy resistance of PDAC. Results: Based on PDAC transcriptome data, we show that IFIT3 expression is associated with the squamous molecular subtype of PDAC and an increase in inflammatory response and apoptosis pathways. We further identify a crucial role for IFIT3 in the regulation of mitochondria-associated apoptosis during chemotherapy. Knockdown of IFIT3 attenuates the chemotherapy resistance of PDAC cells to gemcitabine, paclitaxel, and FOLFIRINOX regimen treatments, independent of individual chemotherapy regimens. While IFIT3 overexpression was found to promote drug resistance. Co-immunoprecipitation identified a direct interaction between IFIT3 and the mitochondrial channel protein VDAC2, an important regulator of mitochondria-associated apoptosis. It was subsequently found that IFIT3 regulates the post-translational modification-O-GlcNAcylation of VDAC2 by stabilizing the interaction of VDAC2 with O-GlcNAc transferase. Increased O-GlcNAcylation of VDAC2 protected PDAC cells from chemotherapy induced apoptosis. Conclusions: These results effectively demonstrate a central mechanism by which IFIT3 expression can affect chemotherapy resistance in PDAC. Targeting IFIT3/VDAC2 may represent a novel strategy to sensitize aggressive forms of pancreatic cancer to conventional chemotherapy regimens.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Canal Aniónico 2 Dependiente del Voltaje/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/genética , Apoptosis/inmunología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/mortalidad , Línea Celular Tumoral , Proliferación Celular/genética , Conjuntos de Datos como Asunto , Resistencia a Antineoplásicos/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Técnicas de Sustitución del Gen , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Estimación de Kaplan-Meier , Páncreas/inmunología , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/mortalidad , Cultivo Primario de Células , Pronóstico , Procesamiento Proteico-Postraduccional/inmunología , RNA-Seq
16.
Trends Cancer ; 6(7): 580-592, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32610068

RESUMEN

Epigenetic mechanisms, including DNA methylation, histone post-translational modifications, and chromatin structure regulation, are critical for the interactions between tumor and immune cells. Emerging evidence shows that tumors commonly hijack various epigenetic mechanisms to escape immune restriction. As a result, the pharmaceutical modulation of epigenetic regulators, including 'writers', 'readers', 'erasers', and 'remodelers', is able to normalize the impaired immunosurveillance and/or trigger antitumor immune responses. Thus, epigenetic targeting agents are attractive immunomodulatory drugs and will have major impacts on immuno-oncology. Here, we discuss epigenetic regulators of the cancer-immunity cycle and current advances in developing epigenetic therapies to boost anticancer immune responses, either alone or in combination with current immunotherapies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Epigénesis Genética/inmunología , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Microambiente Tumoral/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Metilación de ADN/inmunología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Epigénesis Genética/efectos de los fármacos , Histonas/genética , Histonas/metabolismo , Humanos , Inmunomodulación/efectos de los fármacos , Inmunomodulación/genética , Ratones , Neoplasias/genética , Neoplasias/inmunología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/inmunología , Escape del Tumor/efectos de los fármacos , Escape del Tumor/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
17.
Immunology ; 160(1): 24-37, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32022254

RESUMEN

The transcription factor Foxp3 controls the differentiation and function of regulatory T-cells (Treg). Studies in the past decades identified numerous Foxp3-interacting protein partners. However, it is still not clear how Foxp3 produces the Treg-type transcriptomic landscape through cooperating with its partners. Here I show the current understanding of how Foxp3 transcription factor complexes regulate the differentiation, maintenance and functional maturation of Treg. Importantly, T-cell receptor (TCR) signalling plays central roles in Treg differentiation and Foxp3-mediated gene regulation. Differentiating Treg will have recognized their cognate antigens and received TCR signals before initiating Foxp3 transcription, which is triggered by TCR-induced transcription factors including NFAT, AP-1 and NF-κB. Once expressed, Foxp3 seizes TCR signal-induced transcriptional and epigenetic mechanisms through interacting with AML1/Runx1 and NFAT. Thus, Foxp3 modifies gene expression dynamics of TCR-induced genes, which constitute cardinal mechanisms for Treg-mediated immune suppression. Next, I discuss the following key topics, proposing new mechanistic models for Foxp3-mediated gene regulation: (i) how Foxp3 transcription is induced and maintained by the Foxp3-inducing enhanceosome and the Foxp3 autoregulatory transcription factor complex; (ii) molecular mechanisms for effector Treg differentiation (i.e. Treg maturation); (iii) how Foxp3 activates or represses its target genes through recruiting coactivators and corepressors; (iv) the 'decision-making' Foxp3-containing transcription factor complex for Th17 and Treg differentiation; and (v) the roles of post-translational modification in Foxp3 regulation. Thus, this article provides cutting-edge understanding of molecular biology of Foxp3 and Treg, integrating findings by biochemical and genomic studies.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Modelos Biológicos , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Epigénesis Genética/inmunología , Humanos , Activación de Linfocitos , Ratones , Procesamiento Proteico-Postraduccional/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Transcripción Genética/inmunología
18.
Front Immunol ; 11: 618231, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33603747

RESUMEN

Inflammation is a host protection mechanism that eliminates invasive pathogens from the body. However, chronic inflammation, which occurs repeatedly and continuously over a long period, can directly damage tissues and cause various inflammatory and autoimmune diseases. Pattern recognition receptors (PRRs) respond to exogenous infectious agents called pathogen-associated molecular patterns and endogenous danger signals called danger-associated molecular patterns. Among PRRs, recent advancements in studies of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome have established its significant contribution to the pathology of various inflammatory diseases, including metabolic disorders, immune diseases, cardiovascular diseases, and cancer. The regulation of NLRP3 activation is now considered to be important for the development of potential therapeutic strategies. To this end, there is a need to elucidate the regulatory mechanism of NLRP3 inflammasome activation by multiple signaling pathways, post-translational modifications, and cellular organelles. In this review, we discuss the intracellular signaling events, post-translational modifications, small molecules, and phytochemicals participating in the regulation of NLRP3 inflammasome activation. Understanding how intracellular events and small molecule inhibitors regulate NLRP3 inflammasome activation will provide crucial information for elucidating the associated host defense mechanism and the development of efficient therapeutic strategies for chronic diseases.


Asunto(s)
Inflamasomas/inmunología , Inflamación/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Transducción de Señal/inmunología , Animales , Humanos , Inflamasomas/metabolismo , Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Procesamiento Proteico-Postraduccional/inmunología
19.
J Exp Med ; 217(1)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31653691

RESUMEN

The zinc finger transcription factor, Bcl11b, is expressed in T cells and group 2 innate lymphoid cells (ILC2s) among hematopoietic cells. In early T-lineage cells, Bcl11b directly binds and represses the gene encoding the E protein antagonist, Id2, preventing pro-T cells from adopting innate-like fates. In contrast, ILC2s co-express both Bcl11b and Id2. To address this contradiction, we have directly compared Bcl11b action mechanisms in pro-T cells and ILC2s. We found that Bcl11b binding to regions across the genome shows distinct cell type-specific motif preferences. Bcl11b occupies functionally different sites in lineage-specific patterns and controls totally different sets of target genes in these cell types. In addition, Bcl11b bears cell type-specific post-translational modifications and organizes different cell type-specific protein complexes. However, both cell types use the same distal enhancer region to control timing of Bcl11b activation. Therefore, although pro-T cells and ILC2s both need Bcl11b for optimal development and function, Bcl11b works substantially differently in these two cell types.


Asunto(s)
Linaje de la Célula/inmunología , Inmunidad Innata/inmunología , Linfocitos/inmunología , Proteínas Represoras/inmunología , Linfocitos T/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Línea Celular , Ratones , Ratones Endogámicos C57BL , Procesamiento Proteico-Postraduccional/inmunología
20.
Front Immunol ; 10: 2486, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31681337

RESUMEN

Regulatory T cells (Tregs) are engaged in maintaining immune homeostasis and preventing autoimmunity. Treg cells include thymic Treg cells and peripheral Treg cells, both of which can suppress the immune response via multiple distinct mechanisms. The differentiation, proliferation, suppressive function and survival of Treg cells are affected by distinct energy metabolic programs. Tissue-resident Treg cells hold unique features in comparison with the lymphoid organ Treg cells. Foxp3 transcription factor is a lineage master regulator for Treg cell development and suppressive activity. Accumulating evidence indicates that the activity of Foxp3 protein is modulated by various post-translational modifications (PTMs), including phosphorylation, O-GlcNAcylation, acetylation, ubiquitylation and methylation. These modifications affect multiple aspects of Foxp3 function. In this review, we define features of Treg cells and roles of Foxp3 in Treg biology, and summarize current research in PTMs of Foxp3 protein involved in modulating Treg function. This review also attempts to define Foxp3 dimer modifications relevant to mediating Foxp3 activity and Treg suppression. Understanding Foxp3 protein features and modulation mechanisms may help in the design of rational therapies for immune diseases and cancer.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Linfocitos T Reguladores/inmunología , Acetilación , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/genética , Homeostasis/genética , Homeostasis/inmunología , Humanos , Procesamiento Proteico-Postraduccional/genética , Linfocitos T Reguladores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA