Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 46(1): 51-64, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28099864

RESUMEN

Despite the importance of programmed cell death-1 (PD-1) in inhibiting T cell effector activity, the mechanisms regulating its expression remain poorly defined. We found that the chromatin organizer special AT-rich sequence-binding protein-1 (Satb1) restrains PD-1 expression induced upon T cell activation by recruiting a nucleosome remodeling deacetylase (NuRD) complex to Pdcd1 regulatory regions. Satb1 deficienct T cells exhibited a 40-fold increase in PD-1 expression. Tumor-derived transforming growth factor ß (Tgf-ß) decreased Satb1 expression through binding of Smad proteins to the Satb1 promoter. Smad proteins also competed with the Satb1-NuRD complex for binding to Pdcd1 enhancers, releasing Pdcd1 expression from Satb1-mediated repression, Satb1-deficient tumor-reactive T cells lost effector activity more rapidly than wild-type lymphocytes at tumor beds expressing PD-1 ligand (CD274), and these differences were abrogated by sustained CD274 blockade. Our findings suggest that Satb1 functions to prevent premature T cell exhaustion by regulating Pdcd1 expression upon T cell activation. Dysregulation of this pathway in tumor-infiltrating T cells results in diminished anti-tumor immunity.


Asunto(s)
Represión Epigenética/inmunología , Regulación de la Expresión Génica/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/biosíntesis , Receptor de Muerte Celular Programada 1/biosíntesis , Animales , Ensayo de Immunospot Ligado a Enzimas , Humanos , Inmunoprecipitación , Activación de Linfocitos/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/inmunología , Neoplasias/metabolismo
2.
PLoS Genet ; 16(2): e1008641, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32059012

RESUMEN

Men of predominantly African Ancestry (AA) have higher prostate cancer (CaP) incidence and worse survival than men of predominantly European Ancestry (EA). While socioeconomic factors drive this disparity, genomic factors may also contribute to differences in the incidence and mortality rates. To compare the prevalence of prostate tumor genomic alterations and transcriptomic profiles by patient genetic ancestry, we evaluated genomic profiles from The Cancer Genome Atlas (TCGA) CaP cohort (n = 498). Patient global and local genetic ancestry were estimated by computational algorithms using genotyping data; 414 (83.1%) were EA, 61 (12.2%) were AA, 11 (2.2%) were East Asian Ancestry (EAA), 10 (2.0%) were Native American (NA), and 2 (0.4%) were other ancestry. Genetic ancestry was highly concordant with self-identified race/ethnicity. Subsequent analyses were limited to 61 AA and 414 EA cases. Significant differences were observed by ancestry in the frequency of SPOP mutations (20.3% AA vs. 10.0% EA; p = 5.6×10-03), TMPRSS2-ERG fusions (29.3% AA vs. 39.6% EA; p = 4.4×10-02), and PTEN deletions/losses (11.5% AA vs. 30.2% EA; p = 3.5×10-03). Differentially expressed genes (DEGs) between AAs and EAs showed significant enrichment for prostate eQTL target genes (p = 8.09×10-48). Enrichment of highly expressed DEGs for immune-related pathways was observed in AAs, and for PTEN/PI3K signaling in EAs. Nearly one-third of DEGs (31.3%) were long non-coding RNAs (DE-lncRNAs). The proportion of DE-lncRNAs with higher expression in AAs greatly exceeded that with lower expression in AAs (p = 1.2×10-125). Both ChIP-seq and RNA-seq data suggested a stronger regulatory role for AR signaling pathways in DE-lncRNAs vs. non-DE-lncRNAs. CaP-related oncogenic lncRNAs, such as PVT1, PCAT1 and PCAT10/CTBP1-AS, were found to be more highly expressed in AAs. We report substantial heterogeneity in the prostate tumor genome and transcriptome between EA and AA. These differences may be biological contributors to racial disparities in CaP incidence and outcomes.


Asunto(s)
Biomarcadores de Tumor/genética , Negro o Afroamericano/genética , Disparidades en el Estado de Salud , Neoplasias de la Próstata/genética , Población Blanca/genética , Biomarcadores de Tumor/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina , Estudios de Cohortes , Regulación Neoplásica de la Expresión Génica , Genoma Humano/genética , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Mutación , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/epidemiología , ARN Largo no Codificante/metabolismo , RNA-Seq , Receptores Androgénicos/genética , Proteínas Represoras/genética , Transcriptoma/genética
3.
Nature ; 532(7598): 250-4, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27042933

RESUMEN

Cancer is a disease of ageing. Clinically, aged cancer patients tend to have a poorer prognosis than young. This may be due to accumulated cellular damage, decreases in adaptive immunity, and chronic inflammation. However, the effects of the aged microenvironment on tumour progression have been largely unexplored. Since dermal fibroblasts can have profound impacts on melanoma progression, we examined whether age-related changes in dermal fibroblasts could drive melanoma metastasis and response to targeted therapy. Here we find that aged fibroblasts secrete a Wnt antagonist, sFRP2, which activates a multi-step signalling cascade in melanoma cells that results in a decrease in ß-catenin and microphthalmia-associated transcription factor (MITF), and ultimately the loss of a key redox effector, APE1. Loss of APE1 attenuates the response of melanoma cells to DNA damage induced by reactive oxygen species, rendering the cells more resistant to targeted therapy (vemurafenib). Age-related increases in sFRP2 also augment both angiogenesis and metastasis of melanoma cells. These data provide an integrated view of how fibroblasts in the aged microenvironment contribute to tumour progression, offering new possibilities for the design of therapy for the elderly.


Asunto(s)
Envejecimiento/metabolismo , Resistencia a Antineoplásicos , Melanoma/tratamiento farmacológico , Melanoma/patología , Proteínas de la Membrana/metabolismo , Metástasis de la Neoplasia , Microambiente Tumoral , Adulto , Animales , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Daño del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Progresión de la Enfermedad , Fibroblastos/metabolismo , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Masculino , Melanoma/irrigación sanguínea , Melanoma/genética , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Persona de Mediana Edad , Terapia Molecular Dirigida , Neovascularización Patológica , Estrés Oxidativo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Vemurafenib , Vía de Señalización Wnt , Proteína Wnt1/antagonistas & inhibidores , beta Catenina/metabolismo
4.
Cancer Immunol Immunother ; 69(9): 1929-1936, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32435850

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are widely implicated in negative regulation of immune responses in cancer. Inhibition of class I histone deacetylases (HDAC) with entinostat has anti-MDSC activity. However, as single agent, it did not delay tumor growth in EL4 and LLC tumor models. Here, we found that entinostat reduced immune suppressive activity of only one type of MDSC-polymorphonuclear, PMN-MDSC, whereas it had no effect on monocytic M-MDSC or macrophages. M-MDSC had high amount of class II HDAC-HDAC6, which was further increased after the treatment of mice with entinostat. Inhibition of HDAC6 with ricolinostat reduced suppressive activity of M-MDSC, but did not affect PMN-MDSC or delayed tumor growth. However, combination of entinostat and ricolinostat abrogated suppressive activity of both populations of MDSC and substantially delayed tumor progression. Thus, inactivation of MDSC required targeting of both major subsets of these cells via inhibitors of class I and class II HDAC.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Células Mieloides/efectos de los fármacos , Células Supresoras de Origen Mieloide/efectos de los fármacos , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Femenino , Humanos , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Piridinas/farmacología , Pirimidinas/farmacología
5.
Cancer ; 125(12): 1963-1972, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30835824

RESUMEN

Substantial progress has been made in understanding ovarian cancer at the molecular and cellular level. Significant improvement in 5-year survival has been achieved through cytoreductive surgery, combination platinum-based chemotherapy, and more effective treatment of recurrent cancer, and there are now more than 280,000 ovarian cancer survivors in the United States. Despite these advances, long-term survival in late-stage disease has improved little over the last 4 decades. Poor outcomes relate, in part, to late stage at initial diagnosis, intrinsic drug resistance, and the persistence of dormant drug-resistant cancer cells after primary surgery and chemotherapy. Our ability to accelerate progress in the clinic will depend on the ability to answer several critical questions regarding this disease. To assess current answers, an American Association for Cancer Research Special Conference on "Critical Questions in Ovarian Cancer Research and Treatment" was held in Pittsburgh, Pennsylvania, on October 1-3, 2017. Although clinical, translational, and basic investigators conducted much of the discussion, advocates participated in the meeting, and many presentations were directly relevant to patient care, including treatment with poly adenosine diphosphate ribose polymerase (PARP) inhibitors, attempts to improve immunotherapy by overcoming the immune suppressive effects of the microenvironment, and a better understanding of the heterogeneity of the disease.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia/métodos , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/tratamiento farmacológico , Atención Dirigida al Paciente , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Congresos como Asunto , Resistencia a Antineoplásicos , Femenino , Humanos , Sociedades Científicas , Microambiente Tumoral
6.
Mol Cell ; 42(1): 36-49, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21474066

RESUMEN

Mutations in both RAS and the PTEN/PIK3CA/AKT signaling module are found in the same human tumors. PIK3CA and AKT are downstream effectors of RAS, and the selective advantage conferred by mutation of two genes in the same pathway is unclear. Based on a comparative molecular analysis, we show that activated PIK3CA/AKT is a weaker inducer of senescence than is activated RAS. Moreover, concurrent activation of RAS and PIK3CA/AKT impairs RAS-induced senescence. In vivo, bypass of RAS-induced senescence by activated PIK3CA/AKT correlates with accelerated tumorigenesis. Thus, not all oncogenes are equally potent inducers of senescence, and, paradoxically, a weak inducer of senescence (PIK3CA/AKT) can be dominant over a strong inducer of senescence (RAS). For tumor growth, one selective advantage of concurrent mutation of RAS and PTEN/PIK3CA/AKT is suppression of RAS-induced senescence. Evidence is presented that this new understanding can be exploited in rational development and targeted application of prosenescence cancer therapies.


Asunto(s)
Genes ras , Neoplasias/enzimología , Neoplasias/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Anciano , Animales , Línea Celular Transformada , Proliferación Celular , Senescencia Celular/genética , Senescencia Celular/fisiología , Fosfatidilinositol 3-Quinasa Clase I , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Mutación , Neoplasias/patología , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción de Señal
7.
Mol Cell Proteomics ; 15(11): 3348-3360, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27654507

RESUMEN

Inactivating mutations in ARID1A, which encodes a subunit of the SWI/SNF chromatin-remodeling complex, are found in over half of ovarian clear cell carcinoma cases and more broadly across most types of cancers. To identify ARID1A-dependent changes in intracellular signaling pathways, we performed proteome analyses of isogenic ovarian clear cell carcinoma cell lines with or without ARID1A expression. Knockout of ARID1A in an ovarian clear cell carcinoma cell line with wild-type ARID1A, OVCA429, primarily resulted in downregulation of the mevalonate pathway, an important metabolic pathway involved in isoprenoid synthesis, cholesterol synthesis, and other downstream pathways. In a complementary experiment, expression of wild-type ARID1A in an ovarian clear cell carcinoma cell line containing mutated ARID1A, OVISE, affected the mevalonate pathway in a reciprocal manner. A striking aspect of these analyses was that, although only 5% of the detected proteome showed significant abundance changes, most proteins in the mevalonate pathway were coordinately affected by ARID1A status. There were generally corresponding changes when comparing the proteomics data to our previously published microarray data for ectopic expression of ARID1A in the OVISE cell line. However, ARID1A-dependent changes were not detected for genes within the mevalonate pathway. This discrepancy suggests that the mevalonate pathway is not regulated directly by ARID1A-mediated transcription and may be regulated post-transcriptionally. We conclude that ARID1A status indirectly influences the mevalonate pathway and probably influences other processes including glycogen metabolism and 14-3-3-mediated signaling. Further, our findings demonstrate that changes in mRNA levels are sometimes poor indicators of signaling pathways affected by gene manipulations in cancer cells.


Asunto(s)
Adenocarcinoma de Células Claras/metabolismo , Regulación hacia Abajo , Ácido Mevalónico/metabolismo , Proteínas Nucleares/genética , Neoplasias Ováricas/metabolismo , Proteoma/metabolismo , Factores de Transcripción/genética , Adenocarcinoma de Células Claras/genética , Línea Celular Tumoral , Cromatografía Liquida , Proteínas de Unión al ADN , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Redes y Vías Metabólicas , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/genética , Proteoma/genética , Espectrometría de Masas en Tándem
8.
Proc Natl Acad Sci U S A ; 112(46): E6293-300, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578789

RESUMEN

Telomeric repeat-containing RNA (TERRA) has been identified as a telomere-associated regulator of chromosome end protection. Here, we report that TERRA can also be found in extracellular fractions that stimulate innate immune signaling. We identified extracellular forms of TERRA in mouse tumor and embryonic brain tissue, as well as in human tissue culture cell lines using RNA in situ hybridization. RNA-seq analyses revealed TERRA to be among the most highly represented transcripts in extracellular fractions derived from both normal and cancer patient blood plasma. Cell-free TERRA (cfTERRA) could be isolated from the exosome fractions derived from human lymphoblastoid cell line (LCL) culture media. cfTERRA is a shorter form (∼200 nt) of cellular TERRA and copurifies with CD63- and CD83-positive exosome vesicles that could be visualized by cyro-electron microscopy. These fractions were also enriched for histone proteins that physically associate with TERRA in extracellular ChIP assays. Incubation of cfTERRA-containing exosomes with peripheral blood mononuclear cells stimulated transcription of several inflammatory cytokine genes, including TNFα, IL6, and C-X-C chemokine 10 (CXCL10) Exosomes engineered with elevated TERRA or liposomes with synthetic TERRA further stimulated inflammatory cytokines, suggesting that exosome-associated TERRA augments innate immune signaling. These findings imply a previously unidentified extrinsic function for TERRA and a mechanism of communication between telomeres and innate immune signals in tissue and tumor microenvironments.


Asunto(s)
Exosomas/inmunología , Inmunidad Innata , Neoplasias/inmunología , ARN no Traducido/inmunología , Transducción de Señal/inmunología , Telómero , Animales , Antígenos CD/sangre , Antígenos CD/genética , Antígenos CD/inmunología , Línea Celular Tumoral , Citocinas/sangre , Citocinas/genética , Citocinas/inmunología , Exosomas/genética , Exosomas/metabolismo , Histonas/sangre , Histonas/genética , Histonas/inmunología , Humanos , Inmunoglobulinas/sangre , Inmunoglobulinas/genética , Inmunoglobulinas/inmunología , Inflamación/sangre , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Neoplasias/sangre , Neoplasias/genética , Neoplasias/patología , ARN no Traducido/sangre , ARN no Traducido/genética , Transducción de Señal/genética , Tetraspanina 30/sangre , Tetraspanina 30/genética , Tetraspanina 30/inmunología , Antígeno CD83
9.
Proc Natl Acad Sci U S A ; 112(28): 8638-43, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26124089

RESUMEN

Molecular therapies are hallmarks of "personalized" medicine, but how tumors adapt to these agents is not well-understood. Here we show that small-molecule inhibitors of phosphatidylinositol 3-kinase (PI3K) currently in the clinic induce global transcriptional reprogramming in tumors, with activation of growth factor receptors, (re)phosphorylation of Akt and mammalian target of rapamycin (mTOR), and increased tumor cell motility and invasion. This response involves redistribution of energetically active mitochondria to the cortical cytoskeleton, where they support membrane dynamics, turnover of focal adhesion complexes, and random cell motility. Blocking oxidative phosphorylation prevents adaptive mitochondrial trafficking, impairs membrane dynamics, and suppresses tumor cell invasion. Therefore, "spatiotemporal" mitochondrial respiration adaptively induced by PI3K therapy fuels tumor cell invasion, and may provide an important antimetastatic target.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Mitocondrias/efectos de los fármacos , Invasividad Neoplásica , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transporte Biológico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Citoesqueleto/metabolismo , Metabolismo Energético , Humanos , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
11.
Blood ; 120(18): 3764-73, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22976955

RESUMEN

Ribosomal protein (RP) mutations in diseases such as 5q- syndrome both disrupt hematopoiesis and increase the risk of developing hematologic malignancy. However, the mechanism by which RP mutations increase cancer risk has remained an important unanswered question. We show here that monoallelic, germline inactivation of the ribosomal protein L22 (Rpl22) predisposes T-lineage progenitors to transformation. Indeed, RPL22 was found to be inactivated in ∼ 10% of human T-acute lymphoblastic leukemias. Moreover, monoallelic loss of Rpl22 accelerates development of thymic lymphoma in both a mouse model of T-cell malignancy and in acute transformation assays in vitro. We show that Rpl22 inactivation enhances transformation potential through induction of the stemness factor, Lin28B. Our finding that Rpl22 inactivation promotes transformation by inducing expression of Lin28B provides the first insight into the mechanistic basis by which mutations in Rpl22, and perhaps some other RP genes, increases cancer risk.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/metabolismo , Neoplasias Hematológicas/genética , Proteínas de Unión al ARN/genética , Proteínas Ribosómicas/genética , Linfocitos T/metabolismo , Animales , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Silenciador del Gen , Neoplasias Hematológicas/metabolismo , Humanos , Immunoblotting , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Zhonghua Yi Xue Za Zhi ; 94(22): 1744-6, 2014 Jun 10.
Artículo en Zh | MEDLINE | ID: mdl-25151909

RESUMEN

OBJECTIVE: To evaluate the feasibility and safety of endoscopic closure of esophageal perforation with an over-the-scope clip (OTSC) system in a surviving canine model. METHODS: Esophageal perforations (20-mm diameter) were created by an endoscopic needle-knife in 10 dogs. Then the perforations were closed by the OTSC system. And gastroscopy was performed weekly to evaluate the postoperative perforation healing. RESULTS: Esophageal perforations could primarily be closed with one OTSC in each experimental dog. The mean handling time was 24 (18-33) min. All animals survived without postoperative complications. The retention of OTSC was observed in all dogs at the end of 3 weeks. CONCLUSION: The OTSC system can reliably close esophageal perforations without complications.


Asunto(s)
Perforación del Esófago/etiología , Instrumentos Quirúrgicos/efectos adversos , Animales , Perros , Estudios de Factibilidad , Gastroscopía
13.
bioRxiv ; 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38979222

RESUMEN

Patients with metastatic ovarian cancer (OvCa) have a 5-year survival rate of less than 30% due to persisting dissemination of chemoresistant cells in the peritoneal fluid and the immunosuppressive microenvironment in the peritoneal cavity. Here, we report that intraperitoneal administration of ß-glucan and IFNγ (BI) induced robust tumor regression in clinically relevant models of metastatic OvCa. BI induced tumor regression by controlling fluid tumor burden and activating localized antitumor immunity. ß-glucan alone cleared ascites and eliminated fluid tumor cells by inducing intraperitoneal clotting in the fluid and Dectin-1-Syk-dependent NETosis in the omentum. In omentum tumors, BI expanded a novel subset of immunostimulatory IL27+ macrophages and neutralizing IL27 impaired BI efficacy in vivo. Moreover, BI directly induced IL27 secretion in macrophages where single agent treatment did not. Finally, BI extended mouse survival in a chemoresistant model and significantly improved chemotherapy response in a chemo-sensitive model. In summary, we propose a new therapeutic strategy for the treatment of metastatic OvCa.

14.
Nat Aging ; 4(2): 185-197, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38267705

RESUMEN

Sterile inflammation, also known as 'inflammaging', is a hallmark of tissue aging. Cellular senescence contributes to tissue aging, in part, through the secretion of proinflammatory factors collectively known as the senescence-associated secretory phenotype (SASP). The genetic variability of thioredoxin reductase 1 (TXNRD1) is associated with aging and age-associated phenotypes such as late-life survival, activity of daily living and physical performance in old age. TXNRD1's role in regulating tissue aging has been attributed to its enzymatic role in cellular redox regulation. Here, we show that TXNRD1 drives the SASP and inflammaging through the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) innate immune response pathway independently of its enzymatic activity. TXNRD1 localizes to cytoplasmic chromatin fragments and interacts with cGAS in a senescence-status-dependent manner, which is necessary for the SASP. TXNRD1 enhances the enzymatic activity of cGAS. TXNRD1 is required for both the tumor-promoting and immune surveillance functions of senescent cells, which are mediated by the SASP in vivo in mouse models. Treatment of aged mice with a TXNRD1 inhibitor that disrupts its interaction with cGAS, but not with an inhibitor of its enzymatic activity alone, downregulated markers of inflammaging in several tissues. In summary, our results show that TXNRD1 promotes the SASP through the innate immune response, with implications for inflammaging. This suggests that the TXNRD1-cGAS interaction is a relevant target for selectively suppressing inflammaging.


Asunto(s)
Transducción de Señal , Tiorredoxina Reductasa 1 , Animales , Ratones , Senescencia Celular/genética , Inmunidad Innata/genética , Inflamación/genética , Nucleotidiltransferasas/genética , Tiorredoxina Reductasa 1/metabolismo
15.
Nat Commun ; 15(1): 2853, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38565883

RESUMEN

Aberrant glycosylation is a crucial strategy employed by cancer cells to evade cellular immunity. However, it's unclear whether homologous recombination (HR) status-dependent glycosylation can be therapeutically explored. Here, we show that the inhibition of branched N-glycans sensitizes HR-proficient, but not HR-deficient, epithelial ovarian cancers (EOCs) to immune checkpoint blockade (ICB). In contrast to fucosylation whose inhibition sensitizes EOCs to anti-PD-L1 immunotherapy regardless of HR-status, we observe an enrichment of branched N-glycans on HR-proficient compared to HR-deficient EOCs. Mechanistically, BRCA1/2 transcriptionally promotes the expression of MGAT5, the enzyme responsible for catalyzing branched N-glycans. The branched N-glycans on HR-proficient tumors augment their resistance to anti-PD-L1 by enhancing its binding with PD-1 on CD8+ T cells. In orthotopic, syngeneic EOC models in female mice, inhibiting branched N-glycans using 2-Deoxy-D-glucose sensitizes HR-proficient, but not HR-deficient EOCs, to anti-PD-L1. These findings indicate branched N-glycans as promising therapeutic targets whose inhibition sensitizes HR-proficient EOCs to ICB by overcoming immune evasion.


Asunto(s)
Proteína BRCA1 , Neoplasias Ováricas , Humanos , Femenino , Animales , Ratones , Proteína BRCA1/metabolismo , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Linfocitos T CD8-positivos/metabolismo , Glicosilación , Proteína BRCA2/metabolismo , Neoplasias Ováricas/patología , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Antígeno B7-H1/metabolismo
16.
Nat Commun ; 15(1): 5410, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926365

RESUMEN

METTL3 is the catalytic subunit of the methyltransferase complex, which mediates m6A modification to regulate gene expression. In addition, METTL3 regulates transcription in an enzymatic activity-independent manner by driving changes in high-order chromatin structure. However, how these functions of the methyltransferase complex are coordinated remains unknown. Here we show that the methyltransferase complex coordinates its enzymatic activity-dependent and independent functions to regulate cellular senescence, a state of stable cell growth arrest. Specifically, METTL3-mediated chromatin loops induce Hexokinase 2 expression through the three-dimensional chromatin organization during senescence. Elevated Hexokinase 2 expression subsequently promotes liquid-liquid phase separation, manifesting as stress granule phase separation, by driving metabolic reprogramming. This correlates with an impairment of translation of cell-cycle related mRNAs harboring polymethylated m6A sites. In summary, our results report a coordination of m6A-dependent and -independent function of the methyltransferase complex in regulating senescence through phase separation driven by metabolic reprogramming.


Asunto(s)
Senescencia Celular , Cromatina , Metiltransferasas , Gránulos de Estrés , Metiltransferasas/metabolismo , Metiltransferasas/genética , Cromatina/metabolismo , Humanos , Gránulos de Estrés/metabolismo , Gránulos de Estrés/genética , Hexoquinasa/metabolismo , Hexoquinasa/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Adenosina/metabolismo , Adenosina/análogos & derivados , Células HEK293 , Reprogramación Metabólica , Separación de Fases
17.
bioRxiv ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38798466

RESUMEN

Ovarian cancer remains a major health threat with limited treatment options available. It is characterized by immunosuppressive tumor microenvironment (TME) maintained by tumor- associated macrophages (TAMs) hindering anti-tumor responses and immunotherapy efficacy. Here we show that targeting retinoblastoma protein (Rb) by disruption of its LxCxE cleft pocket, causes cell death in TAMs by induction of ER stress, p53 and mitochondria-related cell death pathways. A reduction of pro-tumor Rb high M2-type macrophages from TME in vivo enhanced T cell infiltration and inhibited cancer progression. We demonstrate an increased Rb expression in TAMs in women with ovarian cancer is associated with poorer prognosis. Ex vivo, we show analogous cell death induction by therapeutic Rb targeting in TAMs in post-surgery ascites from ovarian cancer patients. Overall, our data elucidates therapeutic targeting of the Rb LxCxE cleft pocket as a novel promising approach for ovarian cancer treatment through depletion of TAMs and re-shaping TME immune landscape. Statement of significance: Currently, targeting immunosuppressive myeloid cells in ovarian cancer microenvironment is the first priority need to enable successful immunotherapy, but no effective solutions are clinically available. We show that targeting LxCxE cleft pocket of Retinoblastoma protein unexpectedly induces preferential cell death in M2 tumor-associated macrophages. Depletion of immunosuppressive M2 tumor-associated macrophages reshapes tumor microenvironment, enhances anti-tumor T cell responses, and inhibits ovarian cancer. Thus, we identify a novel paradoxical function of Retinoblastoma protein in regulating macrophage viability as well as a promising target to enhance immunotherapy efficacy in ovarian cancer.

18.
Gastrointest Endosc ; 77(5): 774-83, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23453129

RESUMEN

BACKGROUND: Reliable closure of the gastrotomy after transgastric natural orifice transluminal endoscopic surgery (NOTES) remains unresolved. OBJECTIVE: To compare the technical aspects and clinical and histologic outcomes of NOTES gastrotomy closure techniques. DESIGN: Experimental study. SETTING: Animal laboratory. PATIENTS: Thirty-four dogs, 14 for nonsurvival study and 20 for survival study. INTERVENTIONS: The animals randomly received different gastrotomy closures after NOTES: endoclip, omentoplasty, over-the-scope-clip (OTSC), and hand-suturing. MAIN OUTCOME MEASUREMENTS: Procedure time, closure strength, survival, postoperative adverse events, and histologic evaluation of wound healing. RESULTS: Omentoplasty and OTSC groups needed shorter procedure times and fewer clips than the endoclip group. The endoclip and omentoplasty groups generated similar leakage pressures (34.5 ± 2.6 vs 42.2 ± 4.1 mm Hg, P > .05), both lower than OTSC and hand-suturing groups (81.5 ± 2.1 and 87.0 ± 3.0 mm Hg, respectively, P < .001). Of the 20 animals in the survival study (all 4 groups), only 2 of 6 in the endoclip group were killed prematurely due to sepsis. Necropsy revealed the OTSC group reached a 100% clip retention rate, higher than the endoclip (47.9%) and omentoplasty groups (44.4%, P < .05) rates. Complete healing, defined as intact and continuous gastric layers microscopically, was seen in 83.3% of animals (5 of 6) in the omentoplasty group, comparable with OTSC (4 of 6, 66.7%, P = .500) but higher than the endoclip group (1 of 6, 16.7%, P = .04). LIMITATIONS: Animal study. CONCLUSIONS: Omentoplasty is easier and safer for NOTES gastrotomy closure than endoclips and offers safety profile and efficacy similar to OTSC and hand-suturing.


Asunto(s)
Cirugía Endoscópica por Orificios Naturales , Estómago/cirugía , Técnicas de Cierre de Heridas , Animales , Perros , Femenino , Masculino , Epiplón/cirugía , Tempo Operativo , Distribución Aleatoria , Estadísticas no Paramétricas , Estómago/fisiología , Instrumentos Quirúrgicos , Técnicas de Sutura , Técnicas de Cierre de Heridas/efectos adversos , Técnicas de Cierre de Heridas/instrumentación , Cicatrización de Heridas
19.
Zhonghua Yi Xue Za Zhi ; 93(17): 1321-3, 2013 May 07.
Artículo en Zh | MEDLINE | ID: mdl-24029481

RESUMEN

OBJECTIVE: To explore the surgical treatment and prognostic factors of bronchopulmonary carcinoid tumors (BPC). METHODS: The clinical data of 65 patients undergoing surgery for BPC from May 1999 to December 2007 were reviewed retrospectively. The predictors of univariate and multivariate analyses included gender, age, smoking history, pathological type and tumor stage. RESULTS: The procedures included segmentectomy (n = 1), lobectomy (n = 36), bilobectomy (n = 6), sleeve resection (n = 4), pneumonectomy (n = 11), carinal resection (n = 2), bronchoplastic resection (n = 4) and exploratory thoracotomy (n = 1). The 1- , 3- and 5-year overall survival rates were 86.2%, 73.8% and 64.6% respectively. Univariate analysis showed that gender (P = 0.029), age (P = 0.003), smoking history (P = 0.039), pathological type (P < 0.01), tumor stage (P < 0.01), postoperative radiochemotherapy (P < 0.01), lymph node metastasis (P < 0.01) and surgical type (P = 0.042) were prognostic factors. And multivariate analysis revealed that pathological type (P = 0.019) and lymph node metastasis (P < 0.01) were independent prognostic factors. CONCLUSION: Surgery remains a first-choice for BPC. The major resection procedure is anatomical lobectomy or pneumonectomy. Both pathological type and lymph node metastasis are independent prognostic factors.


Asunto(s)
Tumor Carcinoide/diagnóstico , Tumor Carcinoide/cirugía , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/cirugía , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neumonectomía , Pronóstico , Estudios Retrospectivos
20.
Cancer Res Commun ; 3(6): 1067-1077, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37377614

RESUMEN

The arginine methyltransferase CARM1 exhibits high expression levels in several human cancers, with the trend also observed in ovarian cancer. However, therapeutic approaches targeting tumors that overexpress CARM1 have not been explored. Cancer cells exploit metabolic reprogramming such as fatty acids for their survival. Here we report that CARM1 promotes monounsaturated fatty acid synthesis and fatty acid reprogramming represents a metabolic vulnerability for CARM1-expressing ovarian cancer. CARM1 promotes the expression of genes encoding rate-limiting enzymes of de novo fatty acid metabolism such as acetyl-CoA carboxylase 1 (ACC1) and fatty acid synthase (FASN). In addition, CARM1 upregulates stearoyl-CoA desaturase 1 (SCD1) that produces monounsaturated fatty acid by desaturation. Thus, CARM1 enhances de novo fatty acids synthesis which was subsequently utilized for synthesis of monounsaturated fatty acids. Consequently, inhibition of SCD1 suppresses the growth of ovarian cancer cells in a CARM1 status-dependent manner, which was rescued by the addition of monounsaturated fatty acids. Consistently, CARM1-expressing cells were more tolerant to the addition of saturated fatty acids. Indeed, SCD1 inhibition demonstrated efficacy against ovarian cancer in both orthotopic xenograft and syngeneic mouse models in a CARM1-dependent manner. In summary, our data show that CARM1 reprograms fatty acid metabolism and targeting SCD1 through pharmacological inhibition can serve as a potent therapeutic approach for CARM1-expressing ovarian cancers. Significance: CARM1 reprograms fatty acid metabolism transcriptionally to support ovarian cancer growth by producing monounsaturated fatty acids, supporting SCD1 inhibition as a rational strategy for treating CARM1-expressing ovarian cancer.


Asunto(s)
Ácidos Grasos , Neoplasias Ováricas , Animales , Ratones , Humanos , Femenino , Ácidos Grasos/metabolismo , Estearoil-CoA Desaturasa/genética , Neoplasias Ováricas/genética , Ácidos Grasos Monoinsaturados/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA