Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Commun ; 14(1): 887, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36797248

RESUMEN

Epigenetic mechanisms oversee epidermal homeostasis and oncogenesis. The identification of kinases controlling these processes has direct therapeutic implications. We show that ULK3 is a nuclear kinase with elevated expression levels in squamous cell carcinomas (SCCs) arising in multiple body sites, including skin and Head/Neck. ULK3 loss by gene silencing or deletion reduces proliferation and clonogenicity of human keratinocytes and SCC-derived cells and affects transcription impinging on stem cell-related and metabolism programs. Mechanistically, ULK3 directly binds and regulates the activity of two histone arginine methyltransferases, PRMT1 and PRMT5 (PRMT1/5), with ULK3 loss compromising PRMT1/5 chromatin association to specific genes and overall methylation of histone H4, a shared target of these enzymes. These findings are of translational significance, as downmodulating ULK3 by RNA interference or locked antisense nucleic acids (LNAs) blunts the proliferation and tumorigenic potential of SCC cells and promotes differentiation in two orthotopic models of skin cancer.


Asunto(s)
Arginina , Epigenoma , Humanos , Arginina/metabolismo , Queratinocitos/metabolismo , Histonas/metabolismo , Diferenciación Celular/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
2.
Nat Commun ; 14(1): 6498, 2023 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-37838724

RESUMEN

Melanoma provides a primary benchmark for targeted drug therapy. Most melanomas with BRAFV600 mutations regress in response to BRAF/MEK inhibitors (BRAFi/MEKi). However, nearly all relapse within the first two years, and there is a connection between BRAFi/MEKi-resistance and poor response to immune checkpoint therapy. We reported that androgen receptor (AR) activity is required for melanoma cell proliferation and tumorigenesis. We show here that AR expression is markedly increased in BRAFi-resistant melanoma cells, and in sensitive cells soon after BRAFi exposure. Increased AR expression is sufficient to render melanoma cells BRAFi-resistant, eliciting transcriptional changes of BRAFi-resistant subpopulations, including elevated EGFR and SERPINE1 expression, of likely clinical significance. Inhibition of AR expression or activity blunts changes in gene expression and suppresses proliferation and tumorigenesis of BRAFi-resistant melanoma cells, promoting clusters of CD8+ T cells infiltration and cancer cells killing. Our findings point to targeting AR as possible co-therapeutical approach in melanoma treatment.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Humanos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Linfocitos T CD8-positivos/metabolismo , Receptores Androgénicos/genética , Resistencia a Antineoplásicos/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos , Carcinogénesis , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral
3.
Am J Physiol Cell Physiol ; 301(5): C1046-56, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21775709

RESUMEN

Left ventricular remodeling, including the deposition of excess extracellular matrix, is key to the pathogenesis of heart failure. The stress-inducible transcriptional regulator p8 is increased in failing human hearts and is required both for agonist-stimulated cardiomyocyte hypertrophy and for cardiac fibroblasts matrix metalloprotease-9 (MMP9) induction. In the heart, upregulation of autophagy is an adaptive response to stress and plays a causative role in cardiomyopathies. We have recently shown that p8 ablation in cardiac cells upregulates autophagy and that, in vivo, loss of p8 results in a decrease of cardiac function. Here we investigated the effects of p8 genetic deletion in mediating adverse myocardial remodeling. Unstressed p8-/- mouse hearts manifested complex alterations in the expression of fibrosis markers. In addition, these mice displayed elevated autophagy and apoptosis compared with p8+/+ mice. Transverse aortic constriction (TAC) induced left ventricular p8 expression in p8+/+ mice. Pressure overload caused left ventricular remodeling in both genotypes, however, p8-/- mice showed less cardiac fibrosis induction. Consistent with this, although MMP9 induction was attenuated in the p8-/- mice, induction of MMP2 and MMP3 were strikingly upregulated while TIMP2 was downregulated. Left ventricular autophagy increased after TAC and was significantly higher in the p8-/- mice. Thus p8-deletion results in reduced collagen fibrosis after TAC, but in turn, is associated with a detrimental higher increase in autophagy. These findings suggest a role for p8 in regulating in vivo key signaling pathways involved in the pathogenesis of heart failure.


Asunto(s)
Autofagia , Proteínas de Unión al ADN/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Miocardio/patología , Proteínas de Neoplasias/metabolismo , Remodelación Ventricular , Animales , Proteínas de Unión al ADN/genética , Femenino , Fibrosis , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 3 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Proteínas de Neoplasias/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
4.
J Biol Chem ; 285(3): 1577-81, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19926786

RESUMEN

p8 (NUPR1 (nuclear protein-1), Com1 (candidate of metastasis-1)) is a protein related to the high mobility group of transcriptional regulators. It is a key player in the cellular stress response and is involved in metastasis. p8 was first identified as a gene induced in pancreatitis but has been since found overexpressed in several cancers and pathological conditions. Despite its small size and apparently simple structure, p8 functions in several biochemical and genetic pathways, and its expression is crucial for in vivo metastasis in mice, for cytokine induction of metalloproteases, and for stress-induced cardiomyocyte hypertrophy. Understanding p8 functions will provide new opportunities for developing more effective therapeutic approaches to cancer and cardiovascular diseases.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Enfermedad , Estrés Fisiológico , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Neoplasias/metabolismo , Transcripción Genética
5.
Mol Cell Biol ; 27(3): 993-1006, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17116693

RESUMEN

Cardiomyocyte hypertrophy and extracellular matrix remodeling, primarily mediated by inflammatory cytokine-stimulated cardiac fibroblasts, are critical cellular events in cardiac pathology. The molecular components governing these processes remain nebulous, and few genes have been linked to both hypertrophy and matrix remodeling. Here we show that p8, a small stress-inducible basic helix-loop-helix protein, is required for endothelin- and alpha-adrenergic agonist-induced cardiomyocyte hypertrophy and for tumor necrosis factor-stimulated induction, in cardiac fibroblasts, of matrix metalloproteases (MMPs) 9 and 13-MMPs linked to general inflammation and to adverse ventricular remodeling in heart failure. In a stimulus-dependent manner, p8 associates with chromatin containing c-Jun and with the cardiomyocyte atrial natriuretic factor (anf) promoter and the cardiac fibroblast mmp9 and mmp13 promoters, established activator protein 1 effectors. p8 is also induced strongly in the failing human heart by a process reversed upon therapeutic intervention. Our results identify an unexpectedly broad involvement for p8 in key cellular events linked to cardiomyocyte hypertrophy and cardiac fibroblast MMP production, both of which occur in heart failure.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Fibroblastos/enzimología , Metaloproteinasa 13 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Miocardio/citología , Miocitos Cardíacos/patología , Proteínas de Neoplasias/metabolismo , Animales , Factor Natriurético Atrial/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cromatina/efectos de los fármacos , Endotelina-1/farmacología , Inducción Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HeLa , Insuficiencia Cardíaca/terapia , Humanos , Hipertrofia , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Miocitos Cardíacos/efectos de los fármacos , Proteínas de Neoplasias/genética , Fenilefrina/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
6.
Nat Commun ; 11(1): 5126, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046701

RESUMEN

Cancer associated fibroblasts (CAFs) are a key component of the tumor microenvironment. Genomic alterations in these cells remain a point of contention. We report that CAFs from skin squamous cell carcinomas (SCCs) display chromosomal alterations, with heterogeneous NOTCH1 gene amplification and overexpression that also occur, to a lesser extent, in dermal fibroblasts of apparently unaffected skin. The fraction of the latter cells harboring NOTCH1 amplification is expanded by chronic UVA exposure, to which CAFs are resistant. The advantage conferred by NOTCH1 amplification and overexpression can be explained by NOTCH1 ability to block the DNA damage response (DDR) and ensuing growth arrest through suppression of ATM-FOXO3a association and downstream signaling cascade. In an orthotopic model of skin SCC, genetic or pharmacological inhibition of NOTCH1 activity suppresses cancer/stromal cells expansion. Here we show that NOTCH1 gene amplification and increased expression in CAFs are an attractive target for stroma-focused anti-cancer intervention.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Amplificación de Genes , Receptor Notch1/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Daño del ADN , Femenino , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Humanos , Ratones , Ratones SCID , Receptor Notch1/genética , Piel/metabolismo , Neoplasias Cutáneas/genética
7.
Autophagy ; 15(4): 738-739, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30653445

RESUMEN

In the tumor stroma, cancer-associated fibroblasts (CAFs) affect all aspects of tumor evolution. Whereas several programs leading to CAF activation have been elucidated, little is known about the impact of the microenvironment on the turnover of key CAF regulators. RBPJ/CSL is a transcriptional repressor that mediates NOTCH signaling and its down-modulation activates the gene expression program(s) leading to stromal senescence and CAF activation. We overview our evidence that conditions increasing macroautophagy/autophagy, as often found in the stroma of tumors, cause the down-modulation of the RBPJ protein. This event requires the autophagic machinery and is functionally relevant because it is associated with an increase of CAF effector gene expression. The mechanism involves the direct association with the autophagy receptor SQSTM1/p62, which is required for RBPJ down-modulation. As a reflection of increased autophagy in the stroma, both the RBPJ and SQSTM1 proteins are down-modulated in Squamous Cell Carcinoma (SCC) patient-derived CAFs. Increasing RBPJ cellular levels stabilizes SQSTM1 and down-modulates the autophagic process. Our findings identify an autophagy-initiated mechanism for RBPJ down-modulation leading to increased CAF gene expression.


Asunto(s)
Autofagia , Fibroblastos Asociados al Cáncer , Carcinoma de Células Escamosas , Fibroblastos , Humanos , Transducción de Señal , Microambiente Tumoral
8.
Nat Commun ; 10(1): 3884, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467287

RESUMEN

Genomic instability is a hallmark of cancer. Whether it also occurs in Cancer Associated Fibroblasts (CAFs) remains to be carefully investigated. Loss of CSL/RBP-Jκ, the effector of canonical NOTCH signaling with intrinsic transcription repressive function, causes conversion of dermal fibroblasts into CAFs. Here, we find that CSL down-modulation triggers DNA damage, telomere loss and chromosome end fusions that also occur in skin Squamous Cell Carcinoma (SCC)-associated CAFs, in which CSL is decreased. Separately from its role in transcription, we show that CSL is part of a multiprotein telomere protective complex, binding directly and with high affinity to telomeric DNA as well as to UPF1 and Ku70/Ku80 proteins and being required for their telomere association. Taken together, the findings point to a central role of CSL in telomere homeostasis with important implications for genomic instability of cancer stromal cells and beyond.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma de Células Escamosas/metabolismo , Fibroblastos/metabolismo , Inestabilidad Genómica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Telómero/metabolismo , Carcinoma de Células Escamosas/genética , Daño del ADN , Proteínas de Unión al ADN , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Homeostasis , Humanos , Autoantígeno Ku/metabolismo , Proteínas de la Membrana , Simulación del Acoplamiento Molecular , Mutagénesis , ARN Helicasas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Piel/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Transactivadores/metabolismo
9.
Cell Rep ; 24(12): 3108-3114.e4, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231994

RESUMEN

Cancer-associated fibroblasts (CAFs) are important at all tumor stages. CSL/RBPJκ suppresses the gene expression program leading to CAF activation and associated metabolic reprogramming, as well as autophagy. Little is known about CSL protein turnover, especially in the tumor microenvironment. We report that, in human dermal fibroblasts (HDFs), conditions inducing autophagy-often found in tumor stroma-down-regulate CSL protein levels but do not affect its mRNA levels. Genetic or pharmacologic targeting of the autophagic machinery blocks CSL down-modulation. Mechanistically, endogenous CSL associates with the autophagy and signaling adaptor p62/SQSTM1, which is required for CSL down-modulation by autophagy. This is functionally significant, because both CSL and p62 levels are lower in skin cancer-derived CAFs, in which autophagy is increased. Increasing cellular CSL levels stabilizes p62 and down-modulates the autophagic process. We reveal here an autophagy-initiated mechanism for CSL down-modulation, which could be targeted for stroma-focused cancer prevention and treatment.


Asunto(s)
Autofagia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Proteína Sequestosoma-1/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Línea Celular , Células Cultivadas , Regulación hacia Abajo , Fibroblastos/metabolismo , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Ratones , Unión Proteica , Estabilidad Proteica , Proteína Sequestosoma-1/genética , Transducción de Señal
10.
J Clin Invest ; 128(12): 5531-5548, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30395538

RESUMEN

The aging-associated increase of cancer risk is linked with stromal fibroblast senescence and concomitant cancer-associated fibroblast (CAF) activation. Surprisingly little is known about the role of androgen receptor (AR) signaling in this context. We have found downmodulated AR expression in dermal fibroblasts underlying premalignant skin cancer lesions (actinic keratoses and dysplastic nevi) as well as in CAFs from the 3 major skin cancer types, squamous cell carcinomas (SCCs), basal cell carcinomas, and melanomas. Functionally, decreased AR expression in primary human dermal fibroblasts (HDFs) from multiple individuals induced early steps of CAF activation, and in an orthotopic skin cancer model, AR loss in HDFs enhanced tumorigenicity of SCC and melanoma cells. Forming a complex, AR converged with CSL/RBP-Jκ in transcriptional repression of key CAF effector genes. AR and CSL were positive determinants of each other's expression, with BET inhibitors, which counteract the effects of decreased CSL, restoring AR expression and activity in CAFs. Increased AR expression in these cells overcame the consequences of CSL loss and was by itself sufficient to block the growth and tumor-enhancing effects of CAFs on neighboring cancer cells. As such, the findings establish AR as a target for stroma-focused cancer chemoprevention and treatment.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Neoplasias Cutáneas/metabolismo , Activación Transcripcional , Animales , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Proteínas de Neoplasias/genética , Receptores Androgénicos/genética , Proteínas Represoras/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
11.
Cell Rep ; 20(10): 2468-2479, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28877478

RESUMEN

The connection between signaling pathways activating cancer-associated fibroblasts (CAFs) remains to be determined. Metabolic alterations linked to autophagy have also been implicated in CAF activation. CSL/RBPJ, a transcriptional repressor that mediates Notch signaling, suppresses the gene expression program(s), leading to stromal senescence and CAF activation. Deregulated GLI signaling can also contribute to CAF conversion. Here, we report that compromised CSL function depends on GLI activation for conversion of human dermal fibroblasts into CAFs, separately from cellular senescence. Decreased CSL upregulates the expression of the ULK3 kinase, which binds and activates GLI2. Increased ULK3 also induces autophagy, which is unlinked from GLI and CAF activation. ULK3 upregulation occurs in the CAFs of several tumor types, and ULK3 silencing suppresses the tumor-enhancing properties of these cells. Thus, ULK3 links two key signaling pathways involved in CAF conversion and is an attractive target for stroma-focused anti-cancer intervention.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Autofagia/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/genética , Proteína con Dedos de Zinc GLI1/genética
12.
J Exp Med ; 214(8): 2349-2368, 2017 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-28684431

RESUMEN

Cancer-associated fibroblasts (CAFs) are important for tumor initiation and promotion. CSL, a transcriptional repressor and Notch mediator, suppresses CAF activation. Like CSL, ATF3, a stress-responsive transcriptional repressor, is down-modulated in skin cancer stromal cells, and Atf3 knockout mice develop aggressive chemically induced skin tumors with enhanced CAF activation. Even at low basal levels, ATF3 converges with CSL in global chromatin control, binding to few genomic sites at a large distance from target genes. Consistent with this mode of regulation, deletion of one such site 2 Mb upstream of IL6 induces expression of the gene. Observed changes are of translational significance, as bromodomain and extra-terminal (BET) inhibitors, unlinking activated chromatin from basic transcription, counteract the effects of ATF3 or CSL loss on global gene expression and suppress CAF tumor-promoting properties in an in vivo model of squamous cancer-stromal cell expansion. Thus, ATF3 converges with CSL in negative control of CAF activation with epigenetic changes amenable to cancer- and stroma-focused intervention.


Asunto(s)
Factor de Transcripción Activador 3/fisiología , Fibroblastos Asociados al Cáncer/fisiología , Cromatina/fisiología , Proteínas Musculares/fisiología , Animales , Carcinoma de Células Escamosas/fisiopatología , Queratinocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Cutáneas/fisiopatología
13.
Nat Cell Biol ; 17(9): 1193-204, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26302407

RESUMEN

Stromal fibroblast senescence has been linked to ageing-associated cancer risk. However, density and proliferation of cancer-associated fibroblasts (CAFs) are frequently increased. Loss or downmodulation of the Notch effector CSL (also known as RBP-Jκ) in dermal fibroblasts is sufficient for CAF activation and ensuing keratinocyte-derived tumours. We report that CSL silencing induces senescence of primary fibroblasts from dermis, oral mucosa, breast and lung. CSL functions in these cells as a direct repressor of multiple senescence- and CAF-effector genes. It also physically interacts with p53, repressing its activity. CSL is downmodulated in stromal fibroblasts of premalignant skin actinic keratosis lesions and squamous cell carcinomas, whereas p53 expression and function are downmodulated only in the latter, with paracrine FGF signalling as the probable culprit. Concomitant loss of CSL and p53 overcomes fibroblast senescence, enhances expression of CAF effectors and promotes stromal and cancer cell expansion. The findings support a CAF activation-stromal co-evolution model under convergent CSL-p53 control.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Fibroblastos/fisiología , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Neoplasias Cutáneas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Senescencia Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Ratones , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Neoplasias Cutáneas/patología , Proteína p53 Supresora de Tumor/genética
14.
Trends Cell Biol ; 23(12): 593-602, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24074947

RESUMEN

Multifocal and recurrent epithelial tumors, originating from either dormant or de novo cancer cells, are major causes of morbidity and mortality. The age-dependent increase of cancer incidence has long been assumed to result from the sequential accumulation of cancer-driving or -facilitating mutations with induction of cellular senescence as a protective mechanism. However, recent evidence suggests that the initiation and development of epithelial cancer results from a close interplay with its altered tissue microenvironment, with chronic inflammation, stromal senescence, autophagy, and the activation of cancer-associated fibroblasts (CAFs) playing possible primary roles. We will discuss recent progress in these areas, and highlight how this understanding may be used for devising novel preventive and therapeutic approaches to the epithelial cancer problem.


Asunto(s)
Mesodermo/patología , Terapia Molecular Dirigida , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Glandulares y Epiteliales/terapia , Animales , Autofagia , Senescencia Celular , Humanos , Células del Estroma/patología
15.
Autophagy ; 9(1): 95-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23047430

RESUMEN

The incidence of pancreatic adenocarcinoma is increasing with more than 43,000 predicted new cases in the US and 65,000 in Europe this year. Pancreatic cancer patients have a short life expectancy with less than 3-4% 5-y survival, which results in an equivalent incidence and mortality rate. One of the major challenges in pancreatic cancer is the identification of pharmacological approaches that overcome the resistance of this cancer to therapy. Intensive research in the past decades has led to the classification of pancreatic cancers and the identification of the driver key genetic events. Despite the advances in understanding the molecular mechanisms responsible for pancreatic cancer pathogenesis, this knowledge had little impact on significantly improving the treatment for this dismal disease. In particular, we know today that the lack of therapeutic response in pancreatic cancer is due to the intrinsic high resistance of these tumors to chemotherapy and radiation, rather than to the inappropriate design of these therapeutic approaches. Thus, in order to ensure a better outcome for pancreatic cancer patients, there is a strong need for research focused on the mechanism that determines this resistant phenotype and the means that might drive enhanced response to therapy.


Asunto(s)
Autofagia/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Proteínas de Neoplasias/metabolismo , Aurora Quinasas , Autofagia/efectos de los fármacos , Carcinoma Ductal Pancreático/terapia , Muerte Celular , Supervivencia Celular , Resistencia a Antineoplásicos , Humanos , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/metabolismo , Estrés Fisiológico/fisiología
16.
Clin Cancer Res ; 18(19): 5234-46, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22899799

RESUMEN

PURPOSE: The limited supply of oxygen and nutrients is thought to result in rigorous selection of cells that will eventually form the tumor. EXPERIMENTAL DESIGN: Nupr1 expression pattern was analyzed in human tissue microarray (TMA) and correlated with survival time of the patient. Microarray analysis was conducted on MiaPaCa2 cells subjected to metabolic stress in Nupr1-silenced conditions. DNA repair and cell cycle-associated gene expression was confirmed by real-time quantitative PCR (qRT-PCR). Nupr1 and AURKA protective role were analyzed using RNA interference (RNAi) silencing or overexpression. DNA damage and autophagy were analyzed by Western blot analysis and immunofluorescence. RESULTS: We showed that both Nupr1 and HIF1α are coexpressed in human pancreatic ductal adenocarcinoma (PDAC) samples and negatively correlate with survival time. PDAC-derived cells submitted to hypoxia and/or glucose starvation induce DNA damage-dependent cell death concomitantly to the overexpression of stress protein Nupr1. Affymetrix-based transcriptoma analysis reveals that Nupr1 knockdown enhances DNA damage and alters the expression of several genes involved in DNA repair and cell-cycle progression. Expression of some of these genes is common to hypoxia and glucose starvation, such as Aurka gene, suggesting that Nupr1 overexpression counteracts the transcriptional changes occurring under metabolic stress. The molecular mechanism by which hypoxia and glucose starvation induce cell death involves autophagy-associated, but not caspase-dependent, cell death. Finally, we have found that AURKA expression is partially regulated by Nupr1 and plays a major role in this response. CONCLUSIONS: Our data reveal that Nupr1 is involved in a defense mechanism that promotes pancreatic cancer cell survival when exposed to metabolic stress.


Asunto(s)
Adenocarcinoma , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Carcinoma Ductal Pancreático , Proteínas de Neoplasias , Proteínas Serina-Treonina Quinasas , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Aurora Quinasa A , Aurora Quinasas , Autofagia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Estrés Fisiológico
17.
Autophagy ; 6(5): 652-654, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28157434

RESUMEN

Through autophagy cells adapt to nutrient availability, recycle cellular material and eliminate toxic proteins and damaged cellular organelles. Dysregulation of autophagy is implicated in the pathogenesis of various diseases, including cancer, neurodegeneration and cardiomyopathies. The transcription factor FoxO3 activates autophagy by enhancing the expression of several genes. We find a role for the transcriptional regulator p8 in controling autophagy by repressing FoxO3 transcriptional activity. p8 silencing increases the association of FoxO3 with the bnip3 promoter, a known pro-autophagic FoxO3 target, and results in increasead basal autophagy and decreased cellular viability. Likewise, p8 overexpression inhibits Bnip3 upregulation after autophagy activation. Thus, p8 appears to antagonize the promotion of autophagy mediated by the FoxO3-Bnip3 axis. Consistent with this, bnip3 knockdown restores viability in p8-deficient cells. In vivo, hearts from p8-/- mice have higher basal autophagy and bnip3 levels. These mice develop left ventricular wall thinning and chamber dilation, with consequent impaired cardiac function.

18.
Mol Biol Cell ; 21(8): 1335-49, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20181828

RESUMEN

Autophagy is a cytoprotective pathway used to degrade and recycle cytoplasmic content. Dysfunctional autophagy has been linked to both cancer and cardiomyopathies. Here, we show a role for the transcriptional regulator p8 in autophagy. p8 RNA interference (RNAi) increases basal autophagy markers in primary cardiomyocytes, in H9C2 and U2OS cells, and decreases cellular viability after autophagy induction. This autophagy is associated with caspase activation and is blocked by atg5 silencing and by pharmacological inhibitors. FoxO3 transcription factor was reported to activate autophagy by enhancing the expression of autophagy-related genes. P8 expression represses FoxO3 transcriptional activity, and p8 knockdown affects FoxO3 nuclear localization. Thus, p8 RNAi increases FoxO3 association with bnip3 promoter, a known proautophagic FoxO3 target, resulting in higher bnip3 RNA and protein levels. Accordingly, bnip3 knockdown restores cell viability and blocks apoptosis of p8-deficient cells. In vivo, p8 -/- mice have higher autophagy and express higher cardiac bnip3 levels. These mice develop left ventricular wall thinning and chamber dilation, with consequent impaired cardiac function. Our studies provide evidence of a p8-dependent mechanism regulating autophagy by acting as FoxO3 corepressor, which may be relevant for diseases associated with dysregulated autophagy, as cardiovascular pathologies and cancer.


Asunto(s)
Apoptosis , Autofagia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Proteínas de Unión al ADN/deficiencia , Pruebas de Función Cardíaca , Corazón/fisiopatología , Proteínas de Neoplasias/deficiencia , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Portadoras , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Metabolismo Energético/genética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Silenciador del Gen , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fenotipo , Regiones Promotoras Genéticas/genética , Unión Proteica , Estabilidad Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Represoras/metabolismo , Estrés Fisiológico/genética , Activación Transcripcional/genética , Ubiquitina-Proteína Ligasas
20.
J Biol Chem ; 279(20): 20950-8, 2004 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-15016802

RESUMEN

Renal disease is a common complication of diabetes. The initiating events in diabetic nephropathy are triggered by hyperglycemia and, possibly, advanced glycation end products. Subsequently, excess levels of vasoactive peptides (especially endothelin-1 (ET-1)) accumulate in the diabetic kidney, and there is evidence that these peptides mediate many of the pathophysiological changes associated with diabetic renal disease. These changes include an excess deposition of extracellular matrix proteins into the glomerular basement membrane and renal mesangial cell hypertrophy. Our transcriptional profiling studies have revealed that the p8 gene, which encodes a putative basic helix-loop-helix protein, is strongly induced in ET-1-treated renal mesangial cells and in an animal model of diabetic nephropathy. RNA interference experiments indicated that the p8 gene is required for ET-1-induced mesangial cell hypertrophy. Here, we show that the p8 polypeptide is a phosphoprotein subject to constitutive degradation by the ubiquitin/proteasome system. This degradation is mediated by phosphatidylinositol 3-kinase and protein kinase B/Akt. By contrast, stabilization of the p8 protein requires glycogen synthase kinase-3. Finally, short interfering RNA-mediated RNA interference experiments indicated that ET-1-stimulated mesangial cell hypertrophy and p8 mRNA induction require the NFAT4 transcription factor. Thus, p8 levels in the cell are likely maintained by a balance between signal-dependent transcriptional induction and proteolysis.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al ADN/genética , Endotelinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Mesangio Glomerular/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Complejos Multienzimáticos/metabolismo , Proteínas de Neoplasias , Proteínas del Tejido Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Técnicas de Cultivo de Célula , Proteínas de Unión al ADN/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Mesangio Glomerular/efectos de los fármacos , Mesangio Glomerular/patología , Secuencias Hélice-Asa-Hélice/fisiología , Factores de Transcripción NFATC , Complejo de la Endopetidasa Proteasomal , Proteínas Proto-Oncogénicas c-akt , Ratas , Proteínas Recombinantes/metabolismo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA