Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.486
Filtrar
1.
Mol Biol Rep ; 51(1): 571, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662155

RESUMEN

Leukemia is a malignancy of the bone marrow and blood originating from self-renewing cancerous immature blast cells or transformed leukocytes. Despite improvements in treatments, leukemia remains still a serious disease with poor prognosis because of disease heterogeneity, drug resistance and relapse. There is emerging evidence that differentially expression of co-signaling molecules play a critical role in tumor immune evasion. Galectin-9 (Gal-9) is one of the key proteins that leukemic cells express, secrete, and use to proliferate, self-renew, and survive. It also suppresses host immune responses controlled by T and NK cells, enabling leukemic cells to evade immune surveillance. The present review provides the molecular mechanisms of Gal-9-induced immune evasion in leukemia. Understanding the complex immune evasion machinery driven by Gal-9 expressing leukemic cells will enable the identification of novel therapeutic strategies for efficient immunotherapy in leukemic patients. Combined treatment approaches targeting T-cell immunoglobulin and mucin domain-3 (Tim-3)/Gal-9 and other immune checkpoint pathways can be considered, which may enhance the efficacy of host effector cells to attack leukemic cells.


Asunto(s)
Transformación Celular Neoplásica , Galectinas , Receptor 2 Celular del Virus de la Hepatitis A , Leucemia , Humanos , Galectinas/metabolismo , Leucemia/inmunología , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/genética , Animales , Tolerancia Inmunológica , Transducción de Señal , Escape del Tumor , Proliferación Celular , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo
2.
Nucleic Acids Res ; 51(10): 5193-5209, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37070602

RESUMEN

The long non-coding RNA EPR is expressed in epithelial tissues, binds to chromatin and controls distinct biological activities in mouse mammary gland cells. Because of its high expression in the intestine, in this study we have generated a colon-specific conditional targeted deletion (EPR cKO) to evaluate EPR in vivo functions in mice. EPR cKO mice display epithelium hyperproliferation, impaired mucus production and secretion, as well as inflammatory infiltration in the proximal portion of the large intestine. RNA sequencing analysis reveals a rearrangement of the colon crypt transcriptome with strong reduction of goblet cell-specific factors including those involved in the synthesis, assembly, transport and control of mucus proteins. Further, colon mucosa integrity and permeability are impaired in EPR cKO mice, and this results in higher susceptibility to dextran sodium sulfate (DSS)-induced colitis and tumor formation. Human EPR is down-regulated in human cancer cell lines as well as in human cancers, and overexpression of EPR in a colon cancer cell line results in enhanced expression of pro-apoptotic genes. Mechanistically, we show that EPR directly interacts with select genes involved in mucus metabolism whose expression is reduced in EPR cKO mice and that EPR deletion causes tridimensional chromatin organization changes.


Asunto(s)
Transformación Celular Neoplásica , Inflamación , Moco , ARN Largo no Codificante , Animales , Humanos , Ratones , Transformación Celular Neoplásica/inmunología , Colon/metabolismo , Modelos Animales de Enfermedad , Inflamación/inmunología , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35121655

RESUMEN

The tumor microenvironment (TME) provides potential targets for cancer therapy. However, how signals originating in cancer cells affect tumor-directed immunity is largely unknown. Deletions in the CHUK locus, coding for IκB kinase α (IKKα), correlate with reduced lung adenocarcinoma (ADC) patient survival and promote KrasG12D-initiated ADC development in mice, but it is unknown how reduced IKKα expression affects the TME. Here, we report that low IKKα expression in human and mouse lung ADC cells correlates with increased monocyte-derived macrophage and regulatory T cell (Treg) scores and elevated transcription of genes coding for macrophage-recruiting and Treg-inducing cytokines (CSF1, CCL22, TNF, and IL-23A). By stimulating recruitment of monocyte-derived macrophages from the bone marrow and enforcing a TNF/TNFR2/c-Rel signaling cascade that stimulates Treg generation, these cytokines promote lung ADC progression. Depletion of TNFR2, c-Rel, or TNF in CD4+ T cells or monocyte-derived macrophages dampens Treg generation and lung tumorigenesis. Treg depletion also attenuates carcinogenesis. In conclusion, reduced cancer cell IKKα activity enhances formation of a protumorigenic TME through a pathway whose constituents may serve as therapeutic targets for KRAS-initiated lung ADC.


Asunto(s)
Adenocarcinoma del Pulmón/inmunología , Citocinas/inmunología , Quinasa I-kappa B/inmunología , Neoplasias Pulmonares/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Transformación Celular Neoplásica/inmunología , Humanos , Terapia de Inmunosupresión/métodos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología
4.
Mol Cancer ; 21(1): 27, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35062950

RESUMEN

Cellular metabolism constitutes a fundamental process in biology. During tumor initiation and progression, each cellular component in the cancerous niche undergoes dramatic metabolic reprogramming, adapting to a challenging microenvironment of hypoxia, nutrient deprivation, and other stresses. While the metabolic hallmarks of cancer have been extensively studied, the metabolic states of the immune cells are less well elucidated. Here we review the metabolic disturbance and fitness of the immune system in the tumor microenvironment (TME), focusing on the impact of oncometabolites to the function of immune cells and the clinical significance of targeting metabolism in anti-tumor immunotherapy. Metabolic alterations in the immune system of TME offer novel therapeutic insight into cancer treatment.


Asunto(s)
Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Metabolismo Energético , Neoplasias/etiología , Neoplasias/metabolismo , Microambiente Tumoral/inmunología , Adaptación Biológica , Animales , Transformación Celular Neoplásica/genética , Reprogramación Celular , Terapia Combinada , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunomodulación , Inmunoterapia , Neoplasias/diagnóstico , Neoplasias/terapia , Resultado del Tratamiento
5.
Eur J Surg Oncol ; 48(1): 245-252, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34620510

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is one of the most prevalent and life-threatening cancer among the world. Accumulated somatic mutations during malignant transformation process endow cancer cells with increased growth, invasiveness and immunogenicity. These highly immunogenic cancer cells develop multiple strategies to evade immune attack. Through post-transcriptional regulation, microRNAs (miRNAs) not only participate in cancer development and progression but also manipulate anti-cancer immune response. This study aims to identify miRNAs associated with the colorectal cell malignant transformation process and their association with immune cell population using synchronous adjacent normal, polyp and CRC specimens. METHODS: We conducted a Low Density Array to compare the miRNA expression profile of synchronous colorectal adenoma, adenocarcinoma and adjacent normal colon mucosa collected from 8 patients, in order to identify candidate miRNAs involved in CRC progression. These findings were further validated in 14 additional patients and GEO dataset GSE41655. The relative abundance of dendritic cells, natural killer cells, neutrophil and macrophage was determined and correlated with dysregulated miRNA levels. RESULTS: MicroRNA microarray identified 39 miRNAs aberrantly expressed during the colorectal cell transformation process. Seven novel miRNAs were shortlisted, and dysregulation of miR-149-3p, miR-192-3p, miR-335-5p and miR-425 were further validated by the qPCR validation experiment and data retrieved from the GEO dataset. Furthermore, these miRNAs demonstrated certain associations with level of dendritic cells, natural killer cells, neutrophil and macrophage within the polyp or CRC specimens. CONCLUSION: This study revealed miRNA dysregulated during stepwise malignant transformation of colorectal mucosal cells and their association with immune cell population.


Asunto(s)
Adenocarcinoma/genética , Adenoma/genética , Transformación Celular Neoplásica/genética , Pólipos del Colon/genética , Neoplasias Colorrectales/genética , MicroARNs/genética , Escape del Tumor/genética , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenoma/inmunología , Adenoma/metabolismo , Anciano , Anciano de 80 o más Años , Transformación Celular Neoplásica/inmunología , Colon/inmunología , Colon/metabolismo , Pólipos del Colon/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Células Dendríticas/inmunología , Femenino , Humanos , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/inmunología , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Neutrófilos/inmunología , Escape del Tumor/inmunología , Macrófagos Asociados a Tumores/inmunología
6.
Physiol Rev ; 102(1): 455-510, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34541899

RESUMEN

Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. Cancer is a multistep disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signaling in cancer in every step of disease progression. Rho GTPases contribute to tumor initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence, and cancer cell stemness. Rho GTPases play a major role in cell migration and in the metastatic process. They are also involved in interactions with the tumor microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias/tratamiento farmacológico , Microambiente Tumoral/fisiología , Proteínas de Unión al GTP rho/metabolismo , Animales , Movimiento Celular/fisiología , Transformación Celular Neoplásica/inmunología , Humanos , Transducción de Señal/genética
7.
Virology ; 566: 136-142, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34922257

RESUMEN

High mobility group box 1 (HMGB1) is an important chromatin protein and a pro-inflammatory molecule. Though shown to enhance target DNA binding by the Epstein-Barr virus (EBV) lytic switch protein ZEBRA, whether HMGB1 actually contributes to gammaherpesvirus biology is not known. In investigating the contribution of HMGB1 to the lytic phase of EBV, important for development of EBV-mediated diseases, we find that compared to latently-infected cells, lytic phase Burkitt lymphoma-derived cells and peripheral blood lytic cells during primary EBV infection express high levels of HMGB1. Our experiments place HMGB1 upstream of ZEBRA and reveal that HMGB1, through the NLRP3 inflammasome, sustains the expression of ZEBRA. These findings indicate that in addition to the NLRP3 inflammasome's recently discovered role in turning the EBV lytic switch on, NLRP3 cooperates with the danger molecule HMGB1 to also maintain ZEBRA expression, thereby sustaining the lytic signal.


Asunto(s)
Linfoma de Burkitt/genética , Infecciones por Virus de Epstein-Barr/genética , Proteína HMGB1/genética , Herpesvirus Humano 4/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Transactivadores/genética , Linfocitos B/inmunología , Linfocitos B/virología , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Linfoma de Burkitt/virología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Regulación Neoplásica de la Expresión Génica , Proteína HMGB1/inmunología , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/patogenicidad , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Cultivo Primario de Células , Transducción de Señal , Transactivadores/inmunología , Activación Viral/genética , Activación Viral/inmunología , Latencia del Virus/genética , Latencia del Virus/inmunología
8.
Front Endocrinol (Lausanne) ; 12: 785050, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925244

RESUMEN

Macrophages are one of the most common infiltrating immune cells and an essential component of tumor microenvironment. Macrophages and the soluble cytokines and chemokines produced play an important role in tumorigenesis, progression, invasion and metastasis in solid tumors. Despite the multiple studies in other solid tumors, there is little known about macrophages in pituitary adenomas. Recently, studies about pituitary adenoma-infiltrated macrophages have been emerging, including the immunohistochemical and immunophenotypic analysis of the pituitary adenomas and further studies into the mechanism of the crosstalk between macrophages and tumor cells in vivo and in vitro. These studies have offered us new insights into the polarization of macrophages and its role in tumorigenesis, progression and invasion of pituitary adenomas. This review describes the advances in the field of pituitary adenoma-infiltrated macrophages and the prospect of targeting macrophages as cancer therapy in pituitary adenoma.


Asunto(s)
Adenoma/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias Hipofisarias/metabolismo , Microambiente Tumoral/fisiología , Macrófagos Asociados a Tumores/metabolismo , Adenoma/inmunología , Adenoma/patología , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/patología , Humanos , Neoplasias Hipofisarias/inmunología , Neoplasias Hipofisarias/patología , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/patología
9.
PLoS One ; 16(12): e0261082, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34910746

RESUMEN

INTRODUCTION: Multiple previous studies have shown the monoclonal antibody Das-1 (formerly called 7E12H12) is specifically reactive towards metaplastic and carcinomatous lesions in multiple organs of the gastrointestinal system (e.g. Barrett's esophagus, intestinal-type metaplasia of the stomach, gastric adenocarcinoma, high-grade pancreatic intraepithelial neoplasm, and pancreatic ductal adenocarcinoma) as well as in other organs (bladder and lung carcinomas). Beyond being a useful biomarker in tissue, mAb Das-1 has recently proven to be more accurate than current paradigms for identifying cysts harboring advanced neoplasia. Though this antibody has been used extensively for clinical, basic science, and translational applications for decades, its epitope has remained elusive. METHODS: In this study, we chemically deglycosylated a standard source of antigen, which resulted in near complete loss of the signal as measured by western blot analysis. The epitope recognized by mAb Das-1 was determined by affinity to a comprehensive glycan array and validated by inhibition of a direct ELISA. RESULTS: The epitope recognized by mAb Das-1 is 3'-Sulfo-Lewis A/C (3'-Sulfo-LeA/C). 3'-Sulfo-LeA/C is broadly reexpressed across numerous GI epithelia and elsewhere during metaplastic and carcinomatous transformation. DISCUSSION: 3'-Sulfo-LeA/C is a clinically important antigen that can be detected both intracellularly in tissue using immunohistochemistry and extracellularly in cyst fluid and serum by ELISA. The results open new avenues for tumorigenic risk stratification of various gastrointestinal lesions.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Transformación Celular Neoplásica/inmunología , Epítopos de Linfocito B/inmunología , Neoplasias Gastrointestinales/inmunología , Mucosa Intestinal/inmunología , Antígenos del Grupo Sanguíneo de Lewis/inmunología , Oligosacáridos/inmunología , Especificidad de Anticuerpos , Biomarcadores de Tumor/inmunología , Línea Celular Tumoral , Humanos , Inmunohistoquímica
10.
Cell Rep ; 37(10): 110099, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34879282

RESUMEN

Pregnancy reprograms mammary epithelial cells (MECs) to control their responses to pregnancy hormone re-exposure and carcinoma progression. However, the influence of pregnancy on the mammary microenvironment is less clear. Here, we used single-cell RNA sequencing to profile the composition of epithelial and non-epithelial cells in mammary tissue from nulliparous and parous female mice. Our analysis indicates an expansion of γδ natural killer T-like immune cells (NKTs) following pregnancy and upregulation of immune signaling molecules in post-pregnancy MECs. We show that expansion of NKTs following pregnancy is due to elevated expression of the antigen-presenting molecule CD1d on MECs. Loss of CD1d expression on post-pregnancy MECs, or overall lack of activated NKTs, results in mammary oncogenesis. Collectively, our findings illustrate how pregnancy-induced changes modulate the communication between MECs and the immune microenvironment and establish a causal link between pregnancy, the immune microenvironment, and mammary oncogenesis.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/inmunología , Células Epiteliales/inmunología , Activación de Linfocitos , Glándulas Mamarias Animales/inmunología , Neoplasias Mamarias Experimentales/inmunología , Células T Asesinas Naturales/inmunología , Paridad , Animales , Antígenos CD1d/metabolismo , Comunicación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Genes BRCA1 , Genes myc , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células T Asesinas Naturales/metabolismo , Embarazo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Transducción de Señal , Microambiente Tumoral
11.
Front Immunol ; 12: 719954, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721383

RESUMEN

The constant exposure of the liver to gut derived foreign antigens has resulted in this organ attaining unique immunological characteristics, however it remains susceptible to immune mediated injury. Our understanding of this type of injury, in both the native and transplanted liver, has improved significantly in recent decades. This includes a greater awareness of the tolerance inducing CD4+ CD25+ CD127low T-cell lineage with the transcription factor FoxP3, known as regulatory T-Cells (Tregs). These cells comprise 5-10% of CD4+ T cells and are known to function as an immunological "braking" mechanism, thereby preventing immune mediated tissue damage. Therapies that aim to increase Treg frequency and function have proved beneficial in the setting of both autoimmune diseases and solid organ transplantations. The safety and efficacy of Treg therapy in liver disease is an area of intense research at present and has huge potential. Due to these cells possessing significant plasticity, and the potential for conversion towards a T-helper 1 (Th1) and 17 (Th17) subsets in the hepatic microenvironment, it is pre-requisite to modify the microenvironment to a Treg favourable atmosphere to maintain these cells' function. In addition, implementation of therapies that effectively increase Treg functional activity in the liver may result in the suppression of immune responses and will hinder those that destroy tumour cells. Thus, fine adjustment is crucial to achieve this immunological balance. This review will describe the hepatic microenvironment with relevance to Treg function, and the role these cells have in both native diseased and transplanted livers.


Asunto(s)
Inmunoterapia/métodos , Hepatopatías/terapia , Trasplante de Hígado , Linfocitos T Reguladores/inmunología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Microambiente Celular/inmunología , Enfermedad Crónica , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/etiología , Susceptibilidad a Enfermedades/metabolismo , Humanos , Inmunomodulación , Inmunoterapia/efectos adversos , Hepatopatías/diagnóstico , Hepatopatías/etiología , Hepatopatías/metabolismo , Trasplante de Hígado/efectos adversos , Trasplante de Hígado/métodos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Resultado del Tratamiento
12.
Front Immunol ; 12: 687874, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34675913

RESUMEN

Soluble tumor necrosis factor-α (sTNF-α) plays an important role in colitis-associated cancer (CAC); however, little is known about transmembrane TNF-α (tmTNF-α). Here, we observed an increase in sTNF-α mainly in colitis tissues from an azoxymethane/dextran sodium sulfate (DSS)-induced CAC mouse model whereas tmTNF-α levels were chiefly increased on epithelial cells at the tumor stage. The ratio of intracolonic tmTNF-α/sTNF-α was negatively correlated with the levels of pro-inflammatory mediators (IL-1ß, IL-6, and NO) and M1 macrophages but positively correlated with the infiltration of myeloid-derived suppressor cells, regulatory T cells, and the level of the anti-inflammatory cytokine IL-10, suggesting an anti-inflammatory effect of tmTNF-α. This effect of tmTNF-α was confirmed again by the induction of resistance to LPS in colonic epithelial cell lines NCM460 and HCoEpiC through the addition of exogenous tmTNF-α or transfection of the tmTNF-α leading sequence that lacks the extracellular segment but retains the intracellular domain of tmTNF-α. A tmTNF-α antibody was used to block tmTNF-α shedding after the first or second round of inflammation induction by DSS drinking to shift the time window of tmTNF-α expression ahead to the inflammation stage. Antibody treatment significantly alleviated inflammation and suppressed subsequent adenoma formation, accompanied by increased apoptosis. An antitumor effect was also observed when the antibody was administered at the malignant phase of CAC. Our results reveal tmTNF-α as a novel molecular marker for malignant transformation in CAC and provide a new insight into blocking the pathological process by targeting tmTNF-α processing.


Asunto(s)
Adenoma/prevención & control , Antiinflamatorios/farmacología , Anticuerpos/farmacología , Anticarcinógenos/farmacología , Membrana Celular/efectos de los fármacos , Neoplasias Asociadas a Colitis/prevención & control , Colon/efectos de los fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adenoma/inmunología , Adenoma/metabolismo , Adenoma/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Membrana Celular/inmunología , Membrana Celular/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/patología , Colon/inmunología , Colon/metabolismo , Colon/patología , Modelos Animales de Enfermedad , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Biomed Pharmacother ; 144: 112277, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34624674

RESUMEN

Increasing evidence suggested that cholesterol is an important integrant of cell membranes, that plays a key role in tumor progression, immune dysregulation, and pathological changes in epigenetic mechanisms. Based on these theories, there is a growing interest on targeting cholesterol in the treatment of cancer. Here, we comprehensively reviewed the major function of cholesterol on oncogenicity, the therapeutic targets of cholesterol and its metabolites in cancer, and provide detailed insight into the essential roles of cholesterol in mediating immune and epigenetic mechanisms of the tumor microenvironment. It is also worth mentioning that the gut microbiome is an indispensable component of cancer mediation because of its role in cholesterol metabolism. Finally, we summarized recent studies on the potential targets of cholesterol and their metabolism, to provide more therapeutic interventions in oncology.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Colesterol/metabolismo , Neoplasias/metabolismo , Animales , Bacterias/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Progresión de la Enfermedad , Epigénesis Genética , Microbioma Gastrointestinal , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/genética , Neoplasias/inmunología , Microambiente Tumoral/inmunología
14.
Cell Rep ; 37(3): 109858, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686350

RESUMEN

Cell death provides host defense and maintains homeostasis. Zα-containing molecules are essential for these processes. Z-DNA binding protein 1 (ZBP1) activates inflammatory cell death, PANoptosis, whereas adenosine deaminase acting on RNA 1 (ADAR1) serves as an RNA editor to maintain homeostasis. Here, we identify and characterize ADAR1's interaction with ZBP1, defining its role in cell death regulation and tumorigenesis. Combining interferons (IFNs) and nuclear export inhibitors (NEIs) activates ZBP1-dependent PANoptosis. ADAR1 suppresses this PANoptosis by interacting with the Zα2 domain of ZBP1 to limit ZBP1 and RIPK3 interactions. Adar1fl/flLysMcre mice are resistant to development of colorectal cancer and melanoma, but deletion of the ZBP1 Zα2 domain restores tumorigenesis in these mice. In addition, treating wild-type mice with IFN-γ and the NEI KPT-330 regresses melanoma in a ZBP1-dependent manner. Our findings suggest that ADAR1 suppresses ZBP1-mediated PANoptosis, promoting tumorigenesis. Defining the functions of ADAR1 and ZBP1 in cell death is fundamental to informing therapeutic strategies for cancer and other diseases.


Asunto(s)
Adenosina Desaminasa/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/enzimología , Melanoma Experimental/enzimología , Proteínas de Unión al ARN/metabolismo , Neoplasias Cutáneas/enzimología , Adenosina Desaminasa/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Muerte Celular , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Hidrazinas/farmacología , Interferón gamma/farmacología , Masculino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Necroptosis , Piroptosis , Proteínas de Unión al ARN/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Triazoles/farmacología
15.
Front Immunol ; 12: 680955, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566954

RESUMEN

Nasopharyngeal carcinoma (NPC) is a malignant tumor of the nasopharynx mainly characterized by geographic distribution and EBV infection. Metabolic reprogramming, one of the cancer hallmarks, has been frequently reported in NPCs to adapt to internal energy demands and external environmental pressures. Inevitably, the metabolic reprogramming within the tumor cell will lead to a decreased pH value and diverse nutritional supplements in the tumor-infiltrating micro-environment incorporating immune cells, fibroblasts, and endothelial cells. Accumulated evidence indicates that metabolic reprogramming derived from NPC cells may facilitate cancer progression and immunosuppression by cell-cell communications with their surrounding immune cells. This review presents the dysregulated metabolism processes, including glucose, fatty acid, amino acid, nucleotide metabolism, and their mutual interactions in NPC. Moreover, the potential connections between reprogrammed metabolism, tumor immunity, and associated therapy would be discussed in this review. Accordingly, the development of targets on the interactions between metabolic reprogramming and immune cells may provide assistances to overcome the current treatment resistance in NPC patients.


Asunto(s)
Susceptibilidad a Enfermedades , Metabolismo Energético , Evasión Inmune , Neoplasias Nasofaríngeas/etiología , Neoplasias Nasofaríngeas/metabolismo , Animales , Biomarcadores , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Redes y Vías Metabólicas , Mitocondrias/genética , Mitocondrias/inmunología , Mitocondrias/metabolismo , Neoplasias Nasofaríngeas/patología , Microambiente Tumoral
16.
Front Immunol ; 12: 720025, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484227

RESUMEN

Inborn Errors of Immunity (IEI) comprise more than 450 inherited diseases, from which selected patients manifest a frequent and early incidence of malignancies, mainly lymphoma and leukemia. Primary antibody deficiency (PAD) is the most common form of IEI with the highest proportion of malignant cases. In this review, we aimed to compare the oncologic hallmarks and the molecular defects underlying PAD with other IEI entities to dissect the impact of avoiding immune destruction, genome instability, and mutation, enabling replicative immortality, tumor-promoting inflammation, resisting cell death, sustaining proliferative signaling, evading growth suppressors, deregulating cellular energetics, inducing angiogenesis, and activating invasion and metastasis in these groups of patients. Moreover, some of the most promising approaches that could be clinically tested in both PAD and IEI patients were discussed.


Asunto(s)
Susceptibilidad a Enfermedades , Enfermedades Genéticas Congénitas/complicaciones , Enfermedades Genéticas Congénitas/genética , Enfermedades del Sistema Inmune/complicaciones , Neoplasias/etiología , Neoplasias/metabolismo , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Enfermedades de Inmunodeficiencia Primaria/inmunología , Animales , Biomarcadores , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Susceptibilidad a Enfermedades/inmunología , Metabolismo Energético , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Inestabilidad Genómica , Humanos , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/inmunología , Inmunidad/genética , Inflamación/complicaciones , Inflamación/etiología , Inflamación/metabolismo , Mutación , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Transducción de Señal
18.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209703

RESUMEN

The M1/M2 macrophage paradigm plays a key role in tumor progression. M1 macrophages are historically regarded as anti-tumor, while M2-polarized macrophages, commonly deemed tumor-associated macrophages (TAMs), are contributors to many pro-tumorigenic outcomes in cancer through angiogenic and lymphangiogenic regulation, immune suppression, hypoxia induction, tumor cell proliferation, and metastasis. The tumor microenvironment (TME) can influence macrophage recruitment and polarization, giving way to these pro-tumorigenic outcomes. Investigating TME-induced macrophage polarization is critical for further understanding of TAM-related pro-tumor outcomes and potential development of new therapeutic approaches. This review explores the current understanding of TME-induced macrophage polarization and the role of M2-polarized macrophages in promoting tumor progression.


Asunto(s)
Activación de Macrófagos/inmunología , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Animales , Biomarcadores , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Citocinas/metabolismo , Humanos , Hipoxia/genética , Hipoxia/inmunología , Hipoxia/metabolismo , Inmunofenotipificación , Inflamación/etiología , Inflamación/metabolismo , Activación de Macrófagos/genética , Metástasis de la Neoplasia , Estadificación de Neoplasias , Transducción de Señal , Macrófagos Asociados a Tumores/patología
19.
Biochem Soc Trans ; 49(3): 1467-1478, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34196360

RESUMEN

B-cells are antibody-producing cells of the adaptive immune system. Approximately 75% of all newly generated B-cells in the bone marrow are autoreactive and express potentially harmful autoantibodies. To prevent autoimmune disease, the immune system has evolved a powerful mechanism to eliminate autoreactive B-cells, termed negative B-cell selection. While designed to remove autoreactive clones during early B-cell development, our laboratory recently discovered that transformed B-cells in leukemia and lymphoma are also subject to negative selection. Indeed, besides the risk of developing autoimmune disease, B-cells are inherently prone to malignant transformation: to produce high-affinity antibodies, B-cells undergo multiple rounds of somatic immunoglobulin gene recombination and hypermutation. Reflecting high frequencies of DNA-breaks, adaptive immune protection by B-cells comes with a dramatically increased risk of development of leukemia and lymphoma. Of note, B-cells exist under conditions of chronic restriction of energy metabolism. Here we discuss how these metabolic gatekeeper functions during B-cell development provide a common mechanism for the removal of autoreactive and premalignant B-cells to safeguard against both autoimmune diseases and B-cell malignancies.


Asunto(s)
Inmunidad Adaptativa/inmunología , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Linfocitos B/inmunología , Animales , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/metabolismo , Linfocitos B/metabolismo , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Humanos , Leucemia de Células B/inmunología , Leucemia de Células B/metabolismo , Activación de Linfocitos/inmunología
20.
Front Immunol ; 12: 680441, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34234781

RESUMEN

The objective of this study was to investigate mechanisms of allergic inflammation both in vitro and in vivo in details. For this, RNA sequencing was performed. Early growth response 3 gene (Egr3) was one of the most highly upregulated genes in rat basophilic leukemia (RBL2H3) cells stimulated by antigen. The role of Egr3 in allergic inflammation has not been studied extensively. Egr3 was necessary for passive cutaneous anaphylaxis (PCA) and passive systemic anaphylaxis (PSA). Egr3 promoter sequences contained potential binding site for NF-κB p65. NF-κB p65 directly regulated Egr3 expression and mediated allergic inflammation in vitro. Histone deacetylases (HDACs) is known to be involved in allergic airway inflammation. HDAC6 promoter sequences contained potential binding site for EGR3. EGR3 showed binding to promoter sequences of HDAC6. EGR3 was necessary for increased expression of histone deacetylase 6 (HDAC6) in antigen-stimulated RBL2H3 cells. HDAC6 mediated allergic inflammation in vitro and PSA. TargetScan analysis predicted that miR-182-5p was a negative regulator of EGR3. Luciferase activity assay confirmed that miR-182-5p was a direct regulator of EGR3. MiR-182-5p mimic inhibited allergic inflammation both in vitro and in vivo. Cytokine array showed that HDAC6 was necessary for increased interleukin-27 (IL-27) expression in BALB/C mouse model of PSA. Antigen stimulation did not affect expression of EBI3, another subunit of IL-27 in RBL2H3 cells or BALB/C mouse model of PCA or PSA. IL-27 receptor alpha was shown to be able to bind to HDAC6. IL-27 p28 mediated allergic inflammation in vitro, PCA, and PSA. Mouse recombinant IL-27 protein promoted features of allergic inflammation in an antigen-independent manner. HDAC6 was necessary for tumorigenic and metastatic potential enhanced by PSA. PSA enhanced the metastatic potential of mouse melanoma B16F1 cells in an IL-27-dependent manner. Experiments employing culture medium and mouse recombinant IL-27 protein showed that IL-27 mediated and promoted cellular interactions involving B16F1 cells, lung macrophages, and mast cells during allergic inflammation. IL-27 was present in exosomes of antigen-stimulated RBL2H3 cells. Exosomes from antigen-stimulated RBL2H3 cells enhanced invasion of B16F1 melanoma cells in an IL-27-dependemt manner. These results present evidence that EGR3-HDAC6-IL-27 axis can regulate allergic inflammation by mediating cellular interactions.


Asunto(s)
Comunicación Celular , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Proteína 3 de la Respuesta de Crecimiento Precoz/metabolismo , Histona Desacetilasa 6/metabolismo , Interleucina-27/metabolismo , Transducción de Señal , Animales , Comunicación Celular/genética , Comunicación Celular/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Femenino , Hipersensibilidad/complicaciones , Hipersensibilidad/etiología , Hipersensibilidad/metabolismo , Melanoma Experimental , Ratones , MicroARNs/genética , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA