Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
bioRxiv ; 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38260413

RESUMEN

Background: Hepatocellular carcinoma (HCC) incidence is increasing worldwide due to the obesity epidemic, which drives metabolic dysfunction-associated steatohepatitis (MASH) that can lead to HCC. However, the molecular pathways that lead to MASH-HCC are poorly understood. We have previously reported that male mice with global haploinsufficiency of hypoxia-associated factor, HAF ( SART1 +/ - ) spontaneously develop MASH/HCC. However, the cell type(s) responsible for HCC associated with HAF loss are unclear. Results: SART1 -floxed mice were crossed with mice expressing Cre-recombinase within hepatocytes (Alb-Cre; hepS -/- ) or macrophages (LysM-Cre, macS -/- ). Only hepS -/- mice (both male and female) developed HCC suggesting that HAF protects against HCC primarily within hepatocytes. HAF-deficient macrophages showed decreased P-p65 and P-p50 and in many major components of the NF-κB pathway, which was recapitulated using HAF siRNA in vitro . HAF depletion increased apoptosis both in vitro and in vivo , suggesting that HAF mediates a tumor suppressor role by suppressing hepatocyte apoptosis. We show that HAF regulates NF-κB activity by controlling transcription of TRADD and RIPK1 . Mice fed a high-fat diet (HFD) showed marked suppression of HAF, P-p65 and TRADD within their livers after 26 weeks, but manifest profound upregulation of HAF, P-65 and TRADD within their livers after 40 weeks of HFD, implicating deregulation of the HAF-NF-κB axis in the progression to MASH. In humans, HAF was significantly decreased in livers with simple steatosis but significantly increased in HCC compared to normal liver. Conclusions: HAF is novel transcriptional regulator of the NF-κB pathway that protects against hepatocyte apoptosis and is a key determinant of cell fate during progression to MASH and MASH-HCC.

2.
Cancer Res ; 83(24): 4161-4178, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-38098449

RESUMEN

Current treatment approaches for renal cell carcinoma (RCC) face challenges in achieving durable tumor responses due to tumor heterogeneity and drug resistance. Combination therapies that leverage tumor molecular profiles could offer an avenue for enhancing treatment efficacy and addressing the limitations of current therapies. To identify effective strategies for treating RCC, we selected ten drugs guided by tumor biology to test in six RCC patient-derived xenograft (PDX) models. The multitargeted tyrosine kinase inhibitor (TKI) cabozantinib and mTORC1/2 inhibitor sapanisertib emerged as the most effective drugs, particularly when combined. The combination demonstrated favorable tolerability and inhibited tumor growth or induced tumor regression in all models, including two from patients who experienced treatment failure with FDA-approved TKI and immunotherapy combinations. In cabozantinib-treated samples, imaging analysis revealed a significant reduction in vascular density, and single-nucleus RNA sequencing (snRNA-seq) analysis indicated a decreased proportion of endothelial cells in the tumors. SnRNA-seq data further identified a tumor subpopulation enriched with cell-cycle activity that exhibited heightened sensitivity to the cabozantinib and sapanisertib combination. Conversely, activation of the epithelial-mesenchymal transition pathway, detected at the protein level, was associated with drug resistance in residual tumors following combination treatment. The combination effectively restrained ERK phosphorylation and reduced expression of ERK downstream transcription factors and their target genes implicated in cell-cycle control and apoptosis. This study highlights the potential of the cabozantinib plus sapanisertib combination as a promising treatment approach for patients with RCC, particularly those whose tumors progressed on immune checkpoint inhibitors and other TKIs. SIGNIFICANCE: The molecular-guided therapeutic strategy of combining cabozantinib and sapanisertib restrains ERK activity to effectively suppress growth of renal cell carcinomas, including those unresponsive to immune checkpoint inhibitors.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Sistema de Señalización de MAP Quinasas , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Diana Mecanicista del Complejo 1 de la Rapamicina , Células Endoteliales/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Anilidas/farmacología , Anilidas/uso terapéutico , ARN Nuclear Pequeño/uso terapéutico
3.
J Pathol Inform ; 14: 100196, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36814440

RESUMEN

Immunohistochemistry (IHC) highlights specific cell types in tissues and traditionally involves antibody staining together with a hematoxylin counterstain. The intensity and pattern of hematoxylin staining differs between cell types and reveals morphological characteristics of cells. Here, we propose that features in the hematoxylin stain can be used to predict IHC labels, such as Neurofibromin (encoded by the gene NF1). The dataset consists of 7.2 million cells from benign and kidney cancer cores in a tissue microarray. Morphology and hematoxylin (H&M) features defined within QuPath are subjected to a clustering analysis in CytoMap. H&M features are also used to train 4 different XGBoost models to predict high, low, and negative NF1 stain classes in benign renal tubules, clear cell (ccRCC), papillary (PRCC), and chromophobe (ChRCC) renal carcinoma. The prediction accuracies of NF1 staining classes in benign, ccRCC, ChRCC, and PRCC range between 70% and 90% with areas under the precision recall curve PRAUCNF1-high = 0.82+0.12, PRAUCNF1-low = 0.62+0.25, and PRAUCNF1-negative = 0.83+0.16. The most important feature for predicting the NF1 class involves the minimum cellular hematoxylin staining intensity. Together, these results demonstrate the feasibility to predict NF1 expression solely from features in hematoxylin staining using open source software. Since the hematoxylin features can be obtained from regular H&E and IHC slides, the proposed workflow has broad applicability.

4.
Oncogene ; 41(42): 4709-4723, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36097192

RESUMEN

Clear cell renal cell carcinoma (ccRCC), the most common form of kidney cancer, is typically initiated by inactivation of the von Hippel Lindau (VHL) gene, which results in the constitutive activation of the hypoxia inducible factors, HIF-1α and HIF-2α. Using a high throughput screen, we identify novel compounds that decrease HIF-1/2α levels and induce ferroptosis by targeting Iron Sulfur Cluster Assembly 2 (ISCA2), a component of the late mitochondrial Iron Sulfur Cluster (L-ISC) assembly complex. ISCA2 inhibition either pharmacologically or using siRNA decreases HIF-2α protein levels by blocking iron-responsive element (IRE)-dependent translation, and at higher concentrations, also decreases HIF-1α translation through unknown mechanisms. Additionally, ISCA2 inhibition triggers the iron starvation response, resulting in iron/metals overload and death via ferroptosis. ISCA2 levels are decreased in ccRCC compared to normal kidney, and decreased ISCA2 levels are associated with pVHL loss and with sensitivity to ferroptosis induced by ISCA2 inhibition. Strikingly, pharmacological inhibition of ISCA2 using an orally available ISCA2 inhibitor significantly reduced ccRCC xenograft growth in vivo, decreased HIF-α levels and increased lipid peroxidation, suggesting increased ferroptosis in vivo. Thus, the targeting of ISCA2 may be a promising therapeutic strategy to inhibit HIF-1/2α and to induce ferroptosis in pVHL deficient cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Carcinoma de Células Renales , Ferroptosis , Proteínas Hierro-Azufre , Neoplasias Renales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , ARN Interferente Pequeño , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
5.
Trends Cancer ; 8(1): 28-42, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34743924

RESUMEN

Hypoxia is a hallmark of all solid tumors and their metastases. This leads to activation of the hypoxia-inducible factor (HIF) family of transcription factors, which modulate gene expression within both tumor cells and immune cells within the tumor microenvironment, influencing tumor progression and treatment response. The best characterized HIF isoforms, HIF-1α and HIF-2α, show nonoverlapping and often antagonistic roles. With the recent availability of inhibitors that target one or both HIFs, including the first-in-class selective HIF-2α inhibitor belzutifan, the prospect of HIF-α isoform-selective targeting is now a reality. Here, we summarize current knowledge on the unique contributions of the two HIF-α isoforms to tumor progression in the context of the complex tumor immune microenvironment, highlighting important considerations for therapy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias/genética , Neoplasias/inmunología , Microambiente Tumoral/genética
7.
Cancer Cell ; 39(5): 594-596, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33861995

RESUMEN

Intra- and interpatient heterogeneity of clear cell renal cell carcinoma (ccRCC) confounds the search for therapies associated with durable tumor responses and predictive biomarkers. Here, Braun et. al., Krishna et.al, and Bi et.al. use single-cell mRNA sequencing to characterize progressive immune dysfunction associated with ccRCC progression and resistance to immunotherapies.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Linfocitos T CD8-positivos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/terapia , Progresión de la Enfermedad , Humanos , Neoplasias Renales/genética , Neoplasias Renales/terapia , Macrófagos
8.
Clin Cancer Res ; 26(18): 4970-4982, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32586940

RESUMEN

PURPOSE: Clear cell renal cell carcinoma (ccRCC) is frequently associated with inactivation of the von Hippel-Lindau tumor suppressor, resulting in activation of HIF-1α and HIF-2α. The current paradigm, established using mechanistic cell-based studies, supports a tumor promoting role for HIF-2α, and a tumor suppressor role for HIF-1α. However, few studies have comprehensively examined the clinical relevance of this paradigm. Furthermore, the hypoxia-associated factor (HAF), which regulates the HIFs, has not been comprehensively evaluated in ccRCC. EXPERIMENTAL DESIGN: To assess the involvement of HAF/HIFs in ccRCC, we analyzed their relationship to tumor grade/stage/outcome using tissue from 380 patients, and validated these associations using tissue from 72 additional patients and a further 57 patients treated with antiangiogenic therapy for associations with response. Further characterization was performed using single-cell mRNA sequencing (scRNA-seq), RNA-in situ hybridization (RNA-ISH), and IHC. RESULTS: HIF-1α was primarily expressed in tumor-associated macrophages (TAMs), whereas HIF-2α and HAF were expressed primarily in tumor cells. TAM-associated HIF-1α was significantly associated with high tumor grade and increased metastasis and was independently associated with decreased overall survival. Furthermore, elevated TAM HIF-1α was significantly associated with resistance to antiangiogenic therapy. In contrast, high HAF or HIF-2α were associated with low grade, decreased metastasis, and increased overall survival. scRNA-seq, RNA-ISH, and Western blotting confirmed the expression of HIF-1α in M2-polarized CD163-expressing TAMs. CONCLUSIONS: These findings highlight a potential role of TAM HIF-1α in ccRCC progression and support the reevaluation of HIF-1α as a therapeutic target and marker of disease progression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/mortalidad , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/mortalidad , Macrófagos Asociados a Tumores/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores de Tumor/análisis , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/terapia , Línea Celular Tumoral , Quimioterapia Adyuvante , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Neoplasias Renales/diagnóstico , Neoplasias Renales/genética , Neoplasias Renales/terapia , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Nefrectomía , Pronóstico , RNA-Seq , Estudios Retrospectivos , Análisis de la Célula Individual , Análisis de Supervivencia , Macrófagos Asociados a Tumores/inmunología
9.
Cancer Res ; 79(22): 5839-5848, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31585939

RESUMEN

Cancer cells respond to hypoxia by upregulating the hypoxia-inducible factor 1α (HIF1A) transcription factor, which drives survival mechanisms that include metabolic adaptation and induction of angiogenesis by VEGF. Pancreatic tumors are poorly vascularized and severely hypoxic. To study the angiogenic role of HIF1A, and specifically probe whether tumors are able to use alternative pathways in its absence, we created a xenograft mouse tumor model of pancreatic cancer lacking HIF1A. After an initial delay of about 30 days, the HIF1A-deficient tumors grew as rapidly as the wild-type tumors and had similar vascularization. These changes were maintained in subsequent passages of tumor xenografts in vivo and in cell lines ex vivo. There were many cancer cells with a "clear-cell" phenotype in the HIF1A-deficient tumors; this was the result of accumulation of glycogen. Single-cell RNA sequencing (scRNA-seq) of the tumors identified hypoxic cancer cells with inhibited glycogen breakdown, which promoted glycogen accumulation and the secretion of inflammatory cytokines, including interleukins 1ß (IL1B) and 8 (IL8). scRNA-seq of the mouse tumor stroma showed enrichment of two subsets of myeloid dendritic cells (cDC), cDC1 and cDC2, that secreted proangiogenic cytokines. These results suggest that glycogen accumulation associated with a clear-cell phenotype in hypoxic cancer cells lacking HIF1A can initiate an alternate pathway of cytokine and DC-driven angiogenesis. Inhibiting glycogen accumulation may provide a treatment for cancers with the clear-cell phenotype. SIGNIFICANCE: These findings establish a novel mechanism by which tumors support angiogenesis in an HIF1α-independent manner.


Asunto(s)
Proliferación Celular/fisiología , Glucógeno/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Animales , Línea Celular Tumoral , Hipoxia/metabolismo , Hipoxia/patología , Inflamación/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Patológica/patología , Páncreas/metabolismo , Páncreas/patología , Transducción de Señal/fisiología
10.
Mol Cancer Res ; 17(5): 1220-1232, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30705246

RESUMEN

Low oxygen or hypoxia is a feature of all solid tumors and has been associated with aggressive disease. Here, we describe a novel mechanism for the hypoxia-dependent degradation of the Ras-GTPase-activating protein neurofibromin, by hypoxia-associated factor (HAF). We have previously characterized HAF as an oxygen-independent ubiquitin ligase for HIF-1α. Here, we show that HAF promotes neurofibromin ubiquitination and degradation independently of oxygen and pVHL, resulting in Ras-ERK pathway activation. Hypoxia enhanced HAF:neurofibromin binding independently of HAF-SUMOylation, whereas HAF knockdown increased neurofibromin levels primarily in hypoxia, supporting the role of HAF as a hypoxia-specific neurofibromin regulator. HAF overexpression increased p-ERK levels and promoted resistance of clear cell kidney cancer (ccRCC) cells to sorafenib and sunitinib in both normoxia and hypoxia. However, a greater-fold increase in sorafenib/sunitinib resistance was observed during hypoxia, particularly in pVHL-deficient cells. Intriguingly, HAF-mediated resistance was HIF-2α-dependent in normoxia, but HIF-2α-independent in hypoxia indicating two potential mechanisms of HAF-mediated resistance: a HIF-2α-dependent pathway dominant in normoxia, and the direct activation of the Ras-ERK pathway through neurofibromin degradation dominant in hypoxia. Patients with ccRCC with high HAF transcript or protein levels showed significantly decreased overall survival compared with those with low HAF. Thus, we establish a novel, nonmutational pathway of neurofibromin inactivation through hypoxia-induced HAF-mediated degradation, leading to Ras-ERK activation and poor prognosis in ccRCC. IMPLICATIONS: We describe a novel mechanism of neurofibromin degradation induced by hypoxia that leads to activation of the prooncogenic Ras-ERK pathway and resistance to therapy.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Resistencia a Antineoplásicos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Renales/metabolismo , Neurofibromina 1/química , Neurofibromina 1/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Sistema de Señalización de MAP Quinasas , Proteolisis , Ribonucleoproteínas Nucleares Pequeñas , Sorafenib , Sunitinib , Hipoxia Tumoral , Ubiquitinación , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Proteínas ras/metabolismo
12.
Cancer Res ; 76(14): 4259-4269, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27261507

RESUMEN

The hypoxia-inducible transcription factor HIF1α drives expression of many glycolytic enzymes. Here, we show that hypoxic glycolysis, in turn, increases HIF1α transcriptional activity and stimulates tumor growth, revealing a novel feed-forward mechanism of glycolysis-HIF1α signaling. Negative regulation of HIF1α by AMPK1 is bypassed in hypoxic cells, due to ATP elevation by increased glycolysis, thereby preventing phosphorylation and inactivation of the HIF1α transcriptional coactivator p300. Notably, of the HIF1α-activated glycolytic enzymes we evaluated by gene silencing, aldolase A (ALDOA) blockade produced the most robust decrease in glycolysis, HIF-1 activity, and cancer cell proliferation. Furthermore, either RNAi-mediated silencing of ALDOA or systemic treatment with a specific small-molecule inhibitor of aldolase A was sufficient to increase overall survival in a xenograft model of metastatic breast cancer. In establishing a novel glycolysis-HIF-1α feed-forward mechanism in hypoxic tumor cells, our results also provide a preclinical rationale to develop aldolase A inhibitors as a generalized strategy to treat intractable hypoxic cancer cells found widely in most solid tumors. Cancer Res; 76(14); 4259-69. ©2016 AACR.


Asunto(s)
Fructosa-Bifosfato Aldolasa/antagonistas & inhibidores , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias/tratamiento farmacológico , Transducción de Señal/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Proteína p300 Asociada a E1A/fisiología , Humanos , Ratones , Neoplasias/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Hepatology ; 63(5): 1576-91, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26799785

RESUMEN

UNLABELLED: The hypoxia-inducible factor (HIF), HIF-1, is a central regulator of the response to low oxygen or inflammatory stress and plays an essential role in survival and function of immune cells. However, the mechanisms regulating nonhypoxic induction of HIF-1 remain unclear. Here, we assess the impact of germline heterozygosity of a novel, oxygen-independent ubiquitin ligase for HIF-1α: hypoxia-associated factor (HAF; encoded by SART1). SART1(-/-) mice were embryonic lethal, whereas male SART1(+/-) mice spontaneously recapitulated key features of nonalcoholic steatohepatitis (NASH)-driven hepatocellular carcinoma (HCC), including steatosis, fibrosis, and inflammatory cytokine production. Male, but not female, SART1(+/-) mice showed significant up-regulation of HIF-1α in circulating and liver-infiltrating immune cells, but not in hepatocytes, before development of malignancy. Additionally, Kupffer cells derived from male, but not female, SART1(+/-) mice produced increased levels of the HIF-1-dependent chemokine, regulated on activation, normal T-cell expressed and secreted (RANTES), compared to wild type. This was associated with increased liver-neutrophilic infiltration, whereas infiltration of lymphocytes and macrophages were not significantly different. Neutralization of circulating RANTES decreased liver neutrophilic infiltration and attenuated HCC tumor initiation/growth in SART1(+/-) mice. CONCLUSION: This work establishes a new tumor-suppressor role for HAF in immune cell function by preventing inappropriate HIF-1 activation in male mice and identifies RANTES as a novel therapeutic target for NASH and NASH-driven HCC.


Asunto(s)
Carcinoma Hepatocelular/etiología , Quimiocina CCL5/fisiología , Haploinsuficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias Hepáticas/etiología , Transactivadores/genética , Animales , Ácidos Grasos/metabolismo , Hígado Graso/etiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Ribonucleoproteínas Nucleares Pequeñas
14.
Cancer Res ; 75(2): 316-29, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25421578

RESUMEN

Clear-cell renal cell cancer (CRCC) is initiated typically by loss of the tumor-suppressor VHL, driving constitutive activation of hypoxia-inducible factor-1 (HIF1) and HIF2. However, whereas HIF1 has a tumor-suppressor role, HIF2 plays a distinct role in driving CRCC. In this study, we show that the HIF1α E3 ligase hypoxia-associated factor (HAF) complexes with HIF2α at DNA to promote HIF2-dependent transcription through a mechanism relying upon HAF SUMOylation. HAF SUMOylation was induced by hypoxia, whereas HAF-mediated HIF1α degradation was SUMOylation independent. HAF overexpression in mice increased CRCC growth and metastasis. Clinically, HAF overexpression was associated with poor prognosis. Taken together, our results show that HAF is a specific mediator of HIF2 activation that is critical for CRCC development and morbidity.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias Renales/metabolismo , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/genética , Proteínas Portadoras/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Humanos , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Renales/enzimología , Neoplasias Renales/genética , Ratones , Ratones Desnudos , Ribonucleoproteínas Nucleares Pequeñas , Sumoilación , Transactivadores/genética , Activación Transcripcional , Ubiquitina-Proteína Ligasas/genética
15.
Mol Cell ; 48(5): 771-84, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23123196

RESUMEN

Many types of human tumor cells have overexpressed pyruvate kinase M2 (PKM2). However, the mechanism underlying this increased PKM2 expression remains to be defined. We demonstrate here that EGFR activation induces PLCγ1-dependent PKCε monoubiquitylation at Lys321 mediated by RINCK1 ubiquitin ligase. Monoubiquitylated PKCε interacts with a ubiquitin-binding domain in NEMO zinc finger and recruits the cytosolic IKK complex to the plasma membrane, where PKCε phosphorylates IKKß at Ser177 and activates IKKß. Activated RelA interacts with HIF1α, which is required for RelA to bind the PKM promoter. PKCε- and NF-κB-dependent PKM2 upregulation is required for EGFR-promoted glycolysis and tumorigenesis. In addition, PKM2 expression correlates with EGFR and IKKß activity in human glioblastoma specimens and with grade of glioma malignancy. These findings highlight the distinct regulation of NF-κB by EGF, in contrast to TNF-α, and the importance of the metabolic cooperation between the EGFR and NF-κB pathways in PKM2 upregulation and tumorigenesis.


Asunto(s)
Neoplasias Encefálicas/enzimología , Proteínas Portadoras/metabolismo , Transformación Celular Neoplásica/metabolismo , Receptores ErbB/metabolismo , Glioblastoma/enzimología , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteínas Portadoras/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Activación Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Glioblastoma/genética , Glioblastoma/patología , Glucosa/metabolismo , Glucólisis , Células HEK293 , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quinasa I-kappa B/metabolismo , Ácido Láctico/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Mutagénesis Sitio-Dirigida , Mutación , FN-kappa B/genética , Clasificación del Tumor , Trasplante de Neoplasias , Fosfolipasa C gamma/metabolismo , Fosforilación , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Pronóstico , Regiones Promotoras Genéticas , Proteína Quinasa C-epsilon/genética , Interferencia de ARN , Serina , Transducción de Señal , Hormonas Tiroideas/genética , Factor de Transcripción ReIA/metabolismo , Transfección , Ubiquitinación , Regulación hacia Arriba , Proteínas de Unión a Hormona Tiroide
16.
Trends Biochem Sci ; 37(9): 364-72, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22818162

RESUMEN

Hypoxia is an inadequate oxygen supply to tissues and cells, which can restrict their function. The hypoxic response is primarily mediated by the hypoxia-inducible transcription factors, HIF-1 and HIF-2, which have both overlapping and unique target genes. HIF target gene activation is highly context specific and is not a reliable indicator of which HIF-α isoform is active. For example, in some cell lines, the individual HIFs have specific temporal and functional roles: HIF-1 drives the initial response to hypoxia (<24h) and HIF-2 drives the chronic response (>24h). Here, we review the significance of the HIF switch and the relation between HIF-1 and HIF-2 under both physiological and pathophysiological conditions.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia/metabolismo , Animales , Hipoxia de la Célula , Humanos , Oxígeno/metabolismo , Células Madre/metabolismo , Activación Transcripcional
17.
Cancer Res ; 71(11): 4015-27, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21512133

RESUMEN

Most solid tumors and their metastases experience periods of low oxygen or hypoxia, which is of major clinical significance as it promotes both tumor progression and resistance to therapy. Critical mediators of the hypoxic response are the hypoxia-inducible factors HIF-1α and HIF-2α. The HIFs are nonredundant and regulate both overlapping and unique downstream target genes. Here, we describe a novel mechanism for the switch between HIF-1α- and HIF-2α-dependent transcription during tumor hypoxia caused by the hypoxia associated factor (HAF). HAF is overexpressed in a variety of tumors and its levels are decreased during acute hypoxia, but increased following prolonged hypoxia. We have previously identified HAF as an E3 ubiquitin ligase that binds and ubiquitinates HIF-1α by an oxygen and pVHL-independent mechanism, thus targeting HIF-1α for proteasomal degradation. Here, we show that HAF also binds to HIF-2α, but at a different site than HIF-1α, and increases HIF-2α transactivation without causing its degradation. HAF, thus, switches the hypoxic response of the cancer cell from HIF-1α-dependent to HIF-2α-dependent transcription and activates genes involved in invasion such as MMP9, PAI-1, and the stem cell factor OCT-3/4. The switch to HIF-2α-dependent gene expression caused by HAF also promotes an enriched tumor stem cell population, resulting in highly aggressive tumors in vivo. Thus, HAF, by causing a switch from a HIF-1α- to HIF-2α-dependent response to hypoxia, provides a mechanism for more aggressive growth of tumors under prolonged hypoxia.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Proteínas Portadoras/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Neoplasias/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Desnudos , Análisis por Micromatrices , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/patología , Ribonucleoproteínas Nucleares Pequeñas , Transducción de Señal , Transfección
18.
Recent Results Cancer Res ; 180: 15-34, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20033376

RESUMEN

Most solid tumors develop regions of hypoxia as they grow and outstrip their blood supply. In order to survive in the stressful hypoxic environment, tumor cells have developed a coordinated set of responses orchestrating their adaptation to hypoxia. The outcomes of the cellular responses to hypoxia are aggressive disease, resistance to therapy, and decreased patient survival. A critical mediator of the hypoxic response is the transcription factor hypoxia-inducible factor 1 (HIF-1) that upregulates expression of proteins that promote angiogenesis, anaerobic metabolism, and many other survival pathways. Regulation of HIF-1alpha, a component of the HIF-1 heterodimer, occurs at multiple levels including translation, degradation, and transcriptional activation, and serves as a testimony to the central role of HIF-1. Studies demonstrating the importance of HIF-1alpha expression for tumor survival have made HIF-1alpha an attractive target for cancer therapy. The growing l.ist of pharmacological inhibitors of HIF-1 and their varied targets mirrors the complex molecular mechanisms controlling HIF-1. In this chapter, we summarize recent findings regarding the regulation of HIF-1alpha and the progress made in identifying new therapeutic agents that inhibit HIF-1alpha.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias/tratamiento farmacológico , Animales , Hipoxia de la Célula , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Indazoles/uso terapéutico , Compuestos de Mostaza/uso terapéutico , Neoplasias/etiología , Fenilpropionatos/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteína p53 Supresora de Tumor/fisiología
19.
Clin Cancer Res ; 15(19): 5945-6, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19789327

RESUMEN

The hypoxia-inducible transcription factor (HIF)-1alpha inhibitor KC7F2 described in this issue of Clinical Cancer Research is the newest addition to an emerging class of antitumor agents targeting the hypoxia response. Here, we discuss the proposed mechanism of action of KC7F2 and its potential strengths and limitations in comparison with other promising HIF-1alpha inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Hipoxia de la Célula/fisiología , Disulfuros/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales/tendencias , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/fisiopatología , Sulfonamidas/uso terapéutico
20.
Cell Cycle ; 8(9): 1359-66, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19377289

RESUMEN

Adaptation to hypoxia is primarily mediated by the hypoxia-inducible transcription factor, HIF. The regulation of HIF activity by the oxygen-dependent degradation of the HIF-1alpha and HIF-2alpha subunits by the pVHL E3 ligase complex has been well characterized. We have recently described the hypoxia-associated factor, HAF, as an E3 ligase for HIF-1alpha that does not degrade HIF-2alpha. Here we summarize the mechanism of HAF-mediated HIF-1alpha degradation and the importance of oxygen-independent HIF-1alpha regulation in cancer. We also discuss the implications of the new HAF: HIF-1alpha degradation pathway with respect to other novel mediators of oxygen-independent HIF-alpha degradation. Finally, we review the significance of HAF as an isoform-specific E3 ligase in light of new information on the non-overlapping functions of HIF-1alpha and HIF-2alpha in cancer.


Asunto(s)
Proteínas Portadoras/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Portadoras/química , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA