Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Biosci ; 14(1): 99, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39080804

RESUMEN

The PDZ-LIM domain-containing protein PDLIM2 is a common tumor suppressor and a key immune modulator. One main function of PDLIM2 is to promote the ubiquitination and proteasomal degradation of nuclear activated NF-κB RelA, a physiologically indispensable transcription factor whose persistent activation has been linked to almost all cancer types and inflammation-associated diseases. However, it remains unknown how PDLIM2 exerts this physiologically and pathogenically important function. Here, we show that PDLIM2 acts as a ubiquitin ligase enhancer, termed E5. It stabilizes ROC1, an essential component of SKP1/Cullin/F-box protein (SCF) ubiquitin ligases, and chaperones the ROC1-SCFß-TrCP ubiquitin ligase to ubiquitinate nuclear RelA for proteasomal degradation in the nucleus. Consistently, silencing of ROC1, Cullin 1 or the F-box protein ß-TrCP blocks RelA ubiquitination and degradation by PDLIM2. These data provide new mechanistic insights into how PDLIM2 promotes nuclear RelA ubiquitination and degradation, thereby serving as a critical tumor suppressor and a vital immune regulator. They also improve our understanding of the complex cascade of the ubiquitination and NF-κB pathways, particularly given the well-known role of the ROC1-SCFß-TrCP ubiquitin ligase in initiating NF-κB activation by directly binding to and ubiquitinating NF-κB inhibitors for the proteasomal degradation in the cytoplasm.

2.
JCI Insight ; 9(4)2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38385745

RESUMEN

Different from the well-studied canonical NF-κB member RelA, the role of the noncanonical NF-κB member NF-κB2 in solid tumors, and lung cancer in particular, is poorly understood. Here we report that in contrast to the tumor-promoting role of RelA, NF-κB2 intrinsic to lung epithelial and tumor cells had no marked effect on lung tumorigenesis and progression. On the other hand, NF-κB2 limited dendritic cell number and activation in the lung but protected lung macrophages and drove them to promote lung cancer through controlling activation of noncanonical and canonical NF-κB, respectively. NF-κB2 was also required for B cell maintenance and T cell activation. The antitumor activity of lymphocyte NF-κB2 was dominated by the protumor function of myeloid NF-κB2; thus, NF-κB2 has an overall tumor-promoting activity. These studies reveal a cell type-dependent role for NF-κB2 in lung cancer and help understand the complexity of NF-κB action and lung cancer pathogenesis for better design of NF-κB-targeted therapy against this deadliest cancer.


Asunto(s)
Neoplasias Pulmonares , FN-kappa B , Humanos , FN-kappa B/metabolismo , Subunidad p52 de NF-kappa B
3.
bioRxiv ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-37546791

RESUMEN

Background: Immune checkpoint inhibitors (ICIs) and their combination with other therapies such as chemotherapy, fail in most cancer patients. We previously identified the PDZ-LIM domain-containing protein 2 (PDLIM2) as a bona fide tumor suppressor that is repressed in lung cancer to drive cancer and its chemo and immunotherapy resistance, suggesting a new target for lung cancer therapy improvement. Methods: Human clinical samples and data were used to investigate PDLIM2 genetic and epigenetic changes in lung cancer. Using an endogenous mouse lung cancer model faithfully recapitulating refractory human lung cancer and a clinically feasible nano-delivery system, we investigated the therapeutic efficacy, action mechanism, and safety of systemically administrated PDLIM2 expression plasmids encapsulated in nanoparticles (nanoPDLIM2) and its combination with PD-1 antibody and chemotherapeutic drugs. Results: PDLIM2 repression in human lung cancer involves both genetic deletion and epigenetic alteration. NanoPDLIM2 showed low toxicity, high tumor specificity, antitumor activity, and greatly improved the efficacy of anti-PD-1 and chemotherapeutic drugs, with complete tumor remission in most mice and substantial tumor reduction in the remaining mice by their triple combination. Mechanistically, nanoPDLIM2 increased major histocompatibility complex class I (MHC-I) expression, suppressed multi-drug resistance 1 (MDR1) induction and survival genes and other tumor-related genes expression in tumor cells, and enhanced lymphocyte tumor infiltration, turning the cold tumors hot and sensitive to ICIs and rendering them vulnerable to chemotherapeutic drugs and activated tumor-infiltrating lymphocytes (TILs) including those unleashed by ICIs. Conclusions: These studies established a clinically applicable PDLIM2-based combination therapy with great efficacy for lung cancer and possibly other cold cancers.

4.
Methods Mol Biol ; 2366: 213-241, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34236641

RESUMEN

Macrophages are an abundant population in the tumor-infiltrating immune cells. The transcription factor NF-κB plays an important role in the response of tumor-associated macrophages (TAMs) to the tumor environmental cues. Detecting NF-κB activity in TAMs will help define the functional status of the TAMs. In this article, we describe several methods to detect NF-κB activity in TAM populations.


Asunto(s)
Macrófagos , Humanos , Macrófagos/metabolismo , FN-kappa B/metabolismo , Neoplasias/diagnóstico , Transducción de Señal , Macrófagos Asociados a Tumores
5.
J Immunol ; 207(1): 110-114, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34135059

RESUMEN

Macrophages play a central role in lung physiology and pathology. In this study, we show in mice that alveolar macrophages (AMs), unlike other macrophage types (interstitial, peritoneal, and splenic macrophages), constitutively express programmed death-1 ligand 1 (PD-L1), thereby possessing a superior phagocytic ability and the capacity to repress CTLs by cis- and trans-interacting with CD80 and programmed death-1 (PD-1), respectively. This extraordinary ability of AMs assures optimal protective immunity and tolerance within the lung. These findings uncover a unique characteristic of AMs and an innate immune function of PD-L1 and CD80 and therefore help in the understanding of lung physiology, diseases, and PD-L1/PD-1-based immunotherapy.


Asunto(s)
Antígeno B7-H1/inmunología , Macrófagos Alveolares/inmunología , Animales , Antígeno B7-1/inmunología , Ratones , Ratones Endogámicos , Ratones Noqueados
6.
J Neural Eng ; 18(5)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33784636

RESUMEN

Objective.Electrical vagus nerve stimulation (VNS) has the potential to treat a wide variety of diseases by modulating afferent and efferent communication to the heart, lungs, esophagus, stomach, and intestines. Although distal vagal nerve branches, close to end organs, could provide a selective therapeutic approach, these locations are often surgically inaccessible. In contrast, the cervical vagus nerve has been targeted for decades using surgically implantable helix electrodes to treat epileptic seizures and depression; however, to date, clinical implementation of VNS has relied on an electrode with contacts that fully wrap around the nerve, producing non-selective activation of the entire nerve. Here we demonstrate selective cervical VNS using cuff electrodes with multiple contacts around the nerve circumference to target different functional pathways.Approach.These flexible probes were adjusted to the diameter of the nerve using an adhesive hydrogel wrap to create a robust electrode interface. Our approach was verified in a rat model by demonstrating that cervical VNS produces neural activity in the abdominal vagus nerve while limiting effects on the cardiovascular system (i.e. changes in heart rate or blood pressure).Main results.This study demonstrates the potential for selective cervical VNS as a therapeutic approach for modulating distal nerve branches while reducing off target effects.Significance.This methodology could potentially be refined to treat gastrointestinal, metabolic, inflammatory, cardiovascular, and respiratory diseases amenable to vagal neuromodulatory control.


Asunto(s)
Estimulación del Nervio Vago , Animales , Electrodos Implantados , Frecuencia Cardíaca , Hidrogeles , Ratas , Nervio Vago
7.
JCI Insight ; 6(5)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33539325

RESUMEN

One of the most fundamental and challenging questions in the field of cancer is how immunity is transformed from tumor immunosurveillance to tumor-promoting inflammation. Here, we identified the tumor suppressor PDZ-LIM domain-containing protein 2 (PDLIM2) as a checkpoint of alveolar macrophages (AMs) important for lung tumor suppression. During lung tumorigenesis, PDLIM2 expression in AMs is downregulated by ROS-activated transcription repressor BTB and CNC homology 1 (BACH1). PDLIM2 downregulation leads to constitutive activation of the transcription factor STAT3, driving AM protumorigenic polarization/activation and differentiation from monocytes attracted from the circulation to suppress cytotoxic T lymphocytes and promote lung cancer. PDLIM2 downregulation also decreases AM phagocytosis. These findings establish ROS/BACH1/PDLIM2/STAT3 as a signaling pathway driving AMs for lung tumor promotion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas con Dominio LIM/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Macrófagos/citología , Ratones , Monocitos/citología , Monocitos/metabolismo , Factor de Transcripción STAT3/metabolismo
8.
J Immunother Cancer ; 8(1)2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32461344

RESUMEN

BACKGROUND: Programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) blockade therapy fails in the majority of patients with cancer. Oncolytic viruses represent a new class of therapeutic agents, yet the therapeutic efficacy is still disappointing. Moreover, intratumoral injection of viruses is the main approach and preclinical studies mainly employ syngeneic or xenograft models. METHODS: Use an endogenous mouse lung cancer model that faithfully recapitulates human lung cancer, and various in vivo, ex vivo and in vitro assays, to investigate the efficacy, mechanism of action and resistance of systemically administered oncolytic vaccinia virus (oVV), immunotherapy and their combination, to find an effective therapy for refractory lung cancer. RESULTS: Resembling human lung cancers, the majority of which are largely resistant to PD-1/PD-L1 blockade and with decreased PD-L1 expression and T-cell activation by our analysis, urethane-induced endogenous lung tumors in mice show reduced PD-L1 expression, low tumor-infiltrating lymphocytes and innate resistance to PD-1/PD-L1 blockade. Intravenous administration of oVV has efficacy and synergizes with simultaneous but not single blockade of PD-1 and T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) in this cancer model. Besides direct tumor cell killing, oVV induces T-cell lung recruitment, tumor infiltration, along with expression of PD-1 and TIM-3 on T cells and PD-1 and TIM-3 ligands on tumor cells and tumor-associated immune cells. Blockade of PD-1 or TIM-3 also causes their mutual induction on T cells. CONCLUSIONS: While systemic administration of oVV shows efficacy in lung cancer by killing tumor cells directly and recruiting and activating T cells for indirect tumor killing, its induction of PD-1 and TIM-3 on T cells and PD-1 and TIM-3 ligands on tumors and tumor-associated immune cells as well as mutual induction of PD-1 or TIM-3 on T cells by their blockade restricts the efficacy of oVV or its combination with single PD-1 or TIM-3 blockade. The triple combination therapy is more effective for refractory lung cancer, and possibly other cold cancers as well.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Inmunoterapia/métodos , Neoplasias Pulmonares/terapia , Neoplasias Experimentales/terapia , Viroterapia Oncolítica/métodos , Animales , Línea Celular Tumoral/trasplante , Terapia Combinada/métodos , Femenino , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Humanos , Inyecciones Intralesiones , Inyecciones Intravenosas , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/inmunología , Activación de Linfocitos , Ratones , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/inmunología , Virus Oncolíticos/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Uretano/administración & dosificación , Uretano/toxicidad
9.
Nat Commun ; 10(1): 5324, 2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31757943

RESUMEN

Most cancers are resistant to anti-PD-1/PD-L1 and chemotherapy. Herein we identify PDLIM2 as a tumor suppressor particularly important for lung cancer therapeutic responses. While PDLIM2 is epigenetically repressed in human lung cancer, associating with therapeutic resistance and poor prognosis, its global or lung epithelial-specific deletion in mice causes increased lung cancer development, chemoresistance, and complete resistance to anti-PD-1 and epigenetic drugs. PDLIM2 epigenetic restoration or ectopic expression shows antitumor activity, and synergizes with anti-PD-1, notably, with chemotherapy for complete remission of most lung cancers. Mechanistically, through repressing NF-κB/RelA and STAT3, PDLIM2 increases expression of genes involved in antigen presentation and T-cell activation while repressing multidrug resistance genes and cancer-related genes, thereby rendering cancer cells vulnerable to immune attacks and therapies. We identify PDLIM2-independent PD-L1 induction by chemotherapeutic and epigenetic drugs as another mechanism for their synergy with anti-PD-1. These findings establish a rationale to use combination therapies for cancer treatment.


Asunto(s)
Represión Epigenética/genética , Regulación Neoplásica de la Expresión Génica , Proteínas con Dominio LIM/genética , Neoplasias Pulmonares/genética , Proteínas de Microfilamentos/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular Tumoral , Metilación de ADN , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Técnicas de Silenciamiento del Gen , Genes Supresores de Tumor , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Noqueados , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas p21(ras)/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción ReIA/genética
10.
Oncoimmunology ; 7(6): e1435250, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29872577

RESUMEN

Activation of the inflammatory transcription factor NF-κB in tumor-associated macrophages (TAMs) is assumed to contribute to tumor promotion. However, whether and how NF-κB drives the antitumor macrophages to become pro-tumorigenic have not been determined in any cancer type yet. Similarly, how TAMs repress CD8+ cytotoxic T lymphocytes (CTLs) remains largely unknown, although their importance in regulatory T (Treg) cell regulation and tumor promotion has been well appreciated. Here, using an endogenous lung cancer model we uncover a direct crosstalk between TAMs and CTLs. TAMs suppress CTLs through the T-cell inhibitory molecule B7x (B7-H4/B7S1) in a cell-cell contact manner, whereas CTLs kill TAMs in a tumor antigen-specific manner. Remarkably, TAMs secrete the known T-cell suppressive cytokine interleukin-10 (IL-10) to activate, but not to repress, CTLs. Notably, one major role of cell-intrinsic NF-κB RelA is to drive TAMs to suppress CTLs for tumor promotion. It induces B7x expression in TAMs directly, and restricts IL-10 expression indirectly by repressing expression of the NF-κB cofactor Bcl3 and subsequent Bcl3/NF-κB1-mediated transcription of IL-10. It also renders TAMs resistant to CTLs by up-regulating anti-apoptotic genes. These studies help understand how immunity is shaped in lung tumorigenesis, and suggest a RelA-targeted immunotherapy for this deadliest cancer.

11.
Bio Protoc ; 7(10)2017 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-29082285

RESUMEN

A basic Bronchoalveolar lavage (BAL) procedure in mouse is described here. Cells and fluids obtained from BAL can be analyzed by Hema3-staining, immunostaining, Fluorescence-activated cell sorting (FACS), PCR, bicinchoninic acid protein assay, enzyme-linked immunosorbent assay (ELISA), luminex assays, etc., to examine the immune cells, pathogens, proteins such as cytokines/chemokines, and the expression levels of inflammation-related and other genes in the cells. This will help to understand the underlying mechanisms of these lung diseases and develop specific and effective drugs.

12.
Bio Protoc ; 7(10)2017 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-29082286

RESUMEN

We have optimized a protocol for isolation of alveolar type II epithelial cells from mouse lung. Lung cell suspensions are prepared by intratracheal instillation of dispase and agarose followed by mechanical disaggregation of the lungs. Alveolar type II epithelial cells are purified from these lung cell suspensions through magnetic-based negative selection using a Biotin-antibody, Streptavidin-MicroBeads system. The purified alveolar type II epithelial cells can be cultured and maintained on fibronectin-coated plates in DMEM with 10% FBS. This protocol enables specific investigation of alveolar type II epithelial cells at molecular and cellular levels and provides an important tool to investigate in vitro the mechanisms underlying lung pathogenesis.

13.
Cancer Immunol Res ; 5(3): 257-268, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28108629

RESUMEN

One of the most fundamental and challenging questions in the cancer field is how immunity in patients with cancer is transformed from tumor immunosurveillance to tumor-promoting inflammation. Here, we identify the transcription factor STAT3 as the culprit responsible for this pathogenic event in lung cancer development. We found that antitumor type 1 CD4+ T-helper (Th1) cells and CD8+ T cells were directly counter balanced in lung cancer development with tumor-promoting myeloid-derived suppressor cells (MDSCs) and suppressive macrophages, and that activation of STAT3 in MDSCs and macrophages promoted tumorigenesis through pulmonary recruitment and increased resistance of suppressive cells to CD8+ T cells, enhancement of cytotoxicity toward CD4+ and CD8+ T cells, induction of regulatory T cell (Treg), inhibition of dendritic cells (DC), and polarization of macrophages toward the M2 phenotype. The deletion of myeloid STAT3 boosted antitumor immunity and suppressed lung tumorigenesis. These findings increase our understanding of immune programming in lung tumorigenesis and provide a mechanistic basis for developing STAT3-based immunotherapy against this and other solid tumors. Cancer Immunol Res; 5(3); 257-68. ©2017 AACR.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Vigilancia Inmunológica , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Células Mieloides/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Antígenos CD8/deficiencia , Movimiento Celular/genética , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Vigilancia Inmunológica/genética , Vigilancia Inmunológica/inmunología , Inflamación/complicaciones , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/metabolismo , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Células Mieloides/inmunología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Pronóstico , Factor de Transcripción STAT3/genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
14.
Cancer Res ; 75(16): 3209-15, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26122841

RESUMEN

Recent studies suggest that high expression of the proinflammatory cytokine IL6 is associated with poor survival of lung cancer patients. Accordingly, IL6 has been a target of great interest for lung cancer therapy. However, the role of IL6 in lung cancer has not been determined yet. Here, we demonstrate that IL6 plays opposite roles in the initiation and growth of lung cancer in a mouse model of lung cancer induced by the K-Ras oncogene. We find that compared with wild-type mice, IL6-deficient mice developed much more lung tumors after an activating mutant of K-Ras was induced in the lungs. However, lung tumors developed in IL6-deficient mice were significantly smaller. Notably, both the lung tumor-suppressing and -promoting functions of IL6 involve its ability in activating the transcription factor STAT3. IL6/STAT3 signaling suppressed lung cancer initiation through maintaining lung homeostasis, regulating lung macrophages, and activating cytotoxic CD8 T cells under K-Ras oncogenic stress, whereas it promoted lung cancer cell growth through inducing the cell proliferation regulator cyclin D1. These studies reveal a previously unexplored role of IL6/STAT3 signaling in maintaining lung homeostasis and suppressing lung cancer induction. These studies also significantly improve our understanding of lung cancer and provide a molecular basis for designing IL6/STAT3-targeted therapies for this deadliest human cancer.


Asunto(s)
Interleucina-6/deficiencia , Neoplasias Pulmonares/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Quimiocinas/genética , Quimiocinas/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Progresión de la Enfermedad , Expresión Génica , Homeostasis , Inmunohistoquímica , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-6/genética , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Noqueados , Mutación , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Proteínas ras
15.
Methods Mol Biol ; 1280: 121-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25736747

RESUMEN

In unstimulated cells, NF-κB dimers usually exist as latent complexes in the cytoplasm with the IκB (inhibitor of NF-κB) proteins or IκB-like protein p100, the precursor of NF-κB2 mature form p52. Accordingly, there are two major mechanisms leading to NF-κB activation: inducible degradation of IκBs and processing of p100 to generate p52 (selective degradation of the C-terminal IκB-like sequence of p100), which are termed the canonical and noncanonical NF-κB pathways, respectively. While activation of the canonical NF-κB pathway plays critical roles in a wide range of biological processes, the noncanonical NF-κB pathway has important but more restricted roles in both normal and pathological processes. Systematic detection of the noncanonical NF-κB pathway activation is very important for understanding the physiological role of this pathway in biological processes, and for the diagnosis, prevention, and treatment of related diseases. We describe here the methods we employ to detect noncanonical NF-κB activation in cells and tissues. These methods are immunoblotting, co-immunoprecipitation, immunofluorescence, immunohistochemistry, chromatin immunoprecipitation (ChIP) analysis, and electrophoretic mobility shift assay (EMSA). Noncanonical NF-κB-induced gene expression changes can be determined by gene array analysis and quantitative real-time PCR.


Asunto(s)
FN-kappa B/metabolismo , Transducción de Señal , Western Blotting/métodos , Inmunoprecipitación de Cromatina/métodos , Ensayo de Cambio de Movilidad Electroforética/métodos , Activación Enzimática , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Inmunohistoquímica/métodos , Inmunoprecipitación/métodos , Técnicas In Vitro , Subunidad p52 de NF-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Oncogene ; 34(29): 3804-3814, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25284582

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is linked to multiple cancers, including pulmonary adenocarcinoma. However, the role of STAT3 in lung cancer pathogenesis has not been determined. Using lung epithelial-specific inducible knockout strategies, we demonstrate that STAT3 has contrasting roles in the initiation and growth of both chemically and genetically induced lung cancers. Selective deletion of lung epithelial STAT3 in mice before cancer induction by the smoke carcinogen, urethane, resulted in increased lung tissue damage and inflammation, K-Ras oncogenic mutations and tumorigenesis. Deletion of lung epithelial STAT3 after establishment of lung cancer inhibited cancer cell proliferation. Simultaneous deletion of STAT3 and expression of oncogenic K-Ras in mouse lung elevated pulmonary injury, inflammation and tumorigenesis, but reduced tumor growth. These studies indicate that STAT3 prevents lung cancer initiation by maintaining pulmonary homeostasis under oncogenic stress, whereas it facilitates lung cancer progression by promoting cancer cell growth. These studies also provide a mechanistic basis for targeting STAT3 to lung cancer therapy.


Asunto(s)
Proliferación Celular/genética , Neoplasias Pulmonares/genética , Neumonía/genética , Factor de Transcripción STAT3/genética , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Immunoblotting , Inmunohistoquímica , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/metabolismo , Ratones Noqueados , Ratones Transgénicos , Mutación , Neumonía/inducido químicamente , Neumonía/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Carga Tumoral/genética , Uretano
17.
Cell Biosci ; 4(1): 13, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24594309

RESUMEN

The canonical Wnt signaling pathway (or Wnt/ß-catenin pathway) plays a pivotal role in embryonic development and adult homeostasis; deregulation of the Wnt pathway contributes to the initiation and progression of human diseases including cancer. Despite its importance in human biology and disease, how regulation of the Wnt/ß-catenin pathway is achieved remains largely undefined. Increasing evidence suggests that post-translational modifications (PTMs) of Wnt pathway components are essential for the activation of the Wnt/ß-catenin pathway. PTMs create a highly dynamic relay system that responds to Wnt stimulation without requiring de novo protein synthesis and offer a platform for non-Wnt pathway components to be involved in the regulation of Wnt signaling, hence providing alternative opportunities for targeting the Wnt pathway. This review highlights the current status of PTM-mediated regulation of the Wnt/ß-catenin pathway with a focus on factors involved in Wnt-mediated stabilization of ß-catenin.

18.
J Biol Chem ; 288(50): 36094-105, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24189064

RESUMEN

The etiology of human T cell leukemia virus 1 (HTLV-1)-mediated adult T cell leukemia is associated with the ability of viral oncoprotein Tax to induce sustained NF-κB activation and the expression of many NF-κB target genes. Acetylation of the RelA subunit of NF-κB and the subsequent recruitment of bromodomain-containing factor Brd4 are important for the expression of NF-κB target genes in response to various stimuli. However, their contributions to Tax-mediated NF-κB target gene expression and tumorigenesis remain unclear. Here we report that Tax induced the acetylation of lysine 310 of RelA and the binding of Brd4 to acetylated RelA to facilitate Tax-mediated transcriptional activation of NF-κB. Depletion of Brd4 down-regulated Tax-mediated NF-κB target gene expression and cell proliferation. Inhibiting the interaction of Brd4 and acetylated RelA with the bromodomain extraterminal protein inhibitor JQ1 suppressed the proliferation of Tax-expressing rat fibroblasts and Tax-positive HTLV-1-infected cells and Tax-mediated cell transformation and tumorigenesis. Moreover, JQ1 attenuated the Tax-mediated transcriptional activation of NF-κB, triggering the polyubiquitination and proteasome-mediated degradation of constitutively active nuclear RelA. Our results identify Brd4 as a key regulator for Tax-mediated NF-κB gene expression and suggest that targeting epigenetic regulators such as Brd4 with the bromodomain extraterminal protein inhibitor might be a potential therapeutic strategy for cancers and other diseases associated with HTLV-1 infection.


Asunto(s)
Carcinogénesis , Productos del Gen tax/metabolismo , Virus Linfotrópico T Tipo 1 Humano/fisiología , Proteínas Nucleares/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Acetilación/efectos de los fármacos , Animales , Azepinas/farmacología , Carcinogénesis/efectos de los fármacos , Proteínas de Ciclo Celular , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Lisina/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/química , Triazoles/farmacología
19.
Neoplasia ; 15(9): 1110-24, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24027435

RESUMEN

Human T cell leukemia virus type I (HTLV-I) is the etiologic agent of adult T cell leukemia (ATL) and various inflammatory disorders including HTLV-I-associated myelopathy/tropical spastic paraparesis. HTLV-I oncoprotein Tax is known to cause permanent activation of many cellular transcription factors including nuclear factor-κB (NF-κB), cyclic adenosine 3',5'-monophosphate response element-binding protein, and activator protein 1 (AP-1). Here, we show that NF-κB-binding cofactor inhibitor of NF-κB-ζ (IκB-ζ) is constitutively expressed in HTLV-I-infected T cell lines and ATL cells, and Tax transactivates the IκB-ζ gene, mainly through NF-κB. Microarray analysis of IκB-ζ-expressing uninfected T cells demonstrated that IκB-ζ induced the expression of NF-κB. and interferon-regulatory genes such as B cell CLL/lymphoma 3 (Bcl3), guanylate-binding protein 1, and signal transducer and activator of transcription 1. The transcriptional activation domain, nuclear localization signal, and NF-κB-binding domain of IκB-ζ were required for Bcl3 induction, and IκB-ζ synergistically enhanced Tax-induced Bcl3 transactivation in an NF-κB-dependent manner. Interestingly, IκB-ζ inhibited Tax-induced NF-κB, AP-1 activation, and HTLV-I transcription. Furthermore, IκB-ζ interacted with Tax in vitro and this interaction was also observed in an HTLV-I-transformed T cell line. These results suggest that IκB-ζ modulates Tax-dependent and Tax-independent gene transcription in T cells. The function of IκB-ζ may be of significance in ATL genesis and pathogenesis of HTLV-I-associated diseases.


Asunto(s)
Productos del Gen tax/metabolismo , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Linfocitos T/metabolismo , Proteínas del Linfoma 3 de Células B , Línea Celular , Activación Enzimática , Proteínas de Unión al GTP/biosíntesis , Regulación Leucémica de la Expresión Génica , Células HEK293 , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Proteínas I-kappa B/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Factor de Transcripción STAT1/biosíntesis , Transducción de Señal , Factor de Transcripción AP-1/biosíntesis , Factores de Transcripción/biosíntesis , Transcripción Genética , Activación Transcripcional
20.
Retrovirology ; 9: 78, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-23009565

RESUMEN

Activation of the NF-κB transcription factors by the viral protein Tax plays a vital role in the pathogenesis of diseases associated with human T-cell lymphotropic virus type I (HTLV-I). The Tax oncoprotein undergoes constitutive K63-linked ubiquitination and sumoylation; however, the roles and molecular mechanisms of these post-translational modifications in Tax-mediated NF-κB activation are being debated. Here, we discuss our current understanding of Tax activation of NF-κB, with a focus on the controversies and the challenges that we are facing.


Asunto(s)
Productos del Gen tax/metabolismo , Interacciones Huésped-Patógeno , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , FN-kappa B/inmunología , FN-kappa B/metabolismo , Sumoilación , Ubiquitinación , Humanos , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA