Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(40): e2410356121, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39320914

RESUMEN

Loss-of-function germline von Hippel-Lindau (VHL) tumor suppressor mutations cause VHL disease, which predisposes individuals to kidney cancer, hemangioblastomas, and paragangliomas. The risk that a given VHL disease family will manifest some or all these tumor types is profoundly influenced by the VHL allele it carries. For example, almost all VHL disease families that develop paraganglioma have missense VHL mutations. VHL families with null VHL alleles develop kidney cancer and hemangioblastomas without a high risk of paraganglioma. The latter is surprising because the VHL gene product, pVHL, suppresses the HIF2 transcription factor and gain-of-function HIF2 mutations are also linked to paraganglioma. Paragangliomas arise from the sympathetic or parasympathetic nervous system. Given the lack of human paraganglioma cell lines, we studied the effects of inactivating VHL in neuroblastoma cell lines, which also arise from the sympathetic nervous system. We found that total loss of pVHL function profoundly impairs the fitness of neuroblastoma cell lines in a HIF2-dependent manner both ex vivo and in vivo. This fitness defect can be rescued by pVHL variants linked to paraganglioma, but not by pVHL variants associated with a low risk of paraganglioma. These findings suggest that HIF2 activity above a critical threshold prevents the development of paraganglioma.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau , Humanos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Línea Celular Tumoral , Animales , Ratones , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/patología , Paraganglioma/genética , Paraganglioma/metabolismo , Paraganglioma/patología
2.
Cancer ; 130(20): 3473-3479, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38824656

RESUMEN

BACKGROUND: Central nervous system hemangioblastomas are the most prevalent manifestation of von Hippel-Lindau (VHL) disease and remain the main cause of mortality. Surgical resection is the primary treatment strategy, but is not always possible, and should be used as restrictively as possible. There is an unmet need for less invasive treatment strategies, such as targeted therapy. Expression of somatostatin receptor 2A (SSTR2A) in VHL-related hemangioblastomas has been described earlier, but the extent of expression in a larger population has yet to be determined. The authors hypothesize that a substantial subset of VHL-related hemangioblastomas show SSTR2A expression, which may serve as a potential new treatment target. METHODS: Patients who were surgically treated for a VHL-related hemangioblastoma from 1990 until 2021 at the UMC Utrecht were included. Clinical data was derived from a clinical database. Tissue samples were histopathologically examined with use of hematoxylin and eosin staining, and immunohistochemical analysis of SSTR2A expression was performed. RESULTS: Forty-three tissue samples were obtained from 26 patients. Nine showed strong positivity for SSTR2A expression, whereas 13 showed moderate and 15 sparse expression. Three samples showed no expression of SSTR2A. The distribution showed right-skewedness favoring a strong expression. SSTR2A expression colocalized with endothelial markers and not with stromal cells. Additionally, within-patient variability for SSTR2A expression was described in 14 patients. CONCLUSION: SSTR2A is expressed in varying degrees in the majority of VHL-related hemangioblastomas. Future treatment with somatostatin analogues or even peptide receptor radionuclide treatment may be considered for SSTR2A-positive cases.


Asunto(s)
Hemangioblastoma , Receptores de Somatostatina , Enfermedad de von Hippel-Lindau , Humanos , Receptores de Somatostatina/metabolismo , Hemangioblastoma/metabolismo , Hemangioblastoma/patología , Enfermedad de von Hippel-Lindau/complicaciones , Enfermedad de von Hippel-Lindau/patología , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo , Femenino , Adulto , Masculino , Persona de Mediana Edad , Anciano , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/genética , Adulto Joven , Inmunohistoquímica
3.
J Biol Chem ; 297(2): 100910, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34174286

RESUMEN

Von Hippel-Lindau (VHL) disease is characterized by frequent mutation of VHL protein, a tumor suppressor that functions as the substrate recognition subunit of a Cullin2 RING E3 ligase complex (CRL2VHL). CRL2VHL plays important roles in oxygen sensing by targeting hypoxia-inducible factor-alpha (HIF-α) subunits for ubiquitination and degradation. VHL is also commonly hijacked by bifunctional molecules such as proteolysis-targeting chimeras to induce degradation of target molecules. We previously reported the design and characterization of VHL inhibitors VH032 and VH298 that block the VHL:HIF-α interaction, activate the HIF transcription factor, and induce a hypoxic response, which can be beneficial to treat anemia and mitochondrial diseases. How these compounds affect the global cellular proteome remains unknown. Here, we use unbiased quantitative MS to identify the proteomic changes elicited by the VHL inhibitor compared with hypoxia or the broad-spectrum prolyl-hydroxylase domain enzyme inhibitor IOX2. Our results demonstrate that VHL inhibitors selectively activate the HIF response similar to the changes induced in hypoxia and IOX2 treatment. Interestingly, VHL inhibitors were found to specifically upregulate VHL itself. Our analysis revealed that this occurs via protein stabilization of VHL isoforms and not via changes in transcript levels. Increased VHL levels upon VH298 treatment resulted in turn in reduced levels of HIF-1α protein. This work demonstrates the specificity of VHL inhibitors and reveals different antagonistic effects upon their acute versus prolonged treatment in cells. These findings suggest that therapeutic use of VHL inhibitors may not produce overt side effects from HIF stabilization as previously thought.


Asunto(s)
Ciclopropanos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Hipoxia/metabolismo , Proteómica/métodos , Pirrolidinas/farmacología , Tiazoles/farmacología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/antagonistas & inhibidores , Enfermedad de von Hippel-Lindau/patología , Línea Celular , Inhibidores Enzimáticos/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Unión Proteica , Estabilidad Proteica , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
4.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163250

RESUMEN

Von Hippel-Lindau (VHL) syndrome is a rare inherited cancer disease where the lack of VHL protein triggers the development of multisystemic tumors such us retinal hemangioblastomas (HBs), CNS-HBs, and clear cell renal cell carcinoma (ccRCC). Since standard therapies in VHL have shown limited response, leaving surgery as the only possible treatment, targeting of the ß2-adrenergic receptor (ADRB2) has shown therapeutic antitumor benefits on VHL-retinal HBs (clinical trial), VHL-CNS HBs, and VHL-ccRCC (in vitro and in vivo). In the present study, we wanted to look deep into the effects of the ADRB2 blockers propranolol and ICI-118,551 on two main aspects of cancer progression: (i) the changes on the inflammatory response of ccRCC cells; and (ii) the modulation on the Warburg effect (glycolytic metabolism), concretely, on the expression of genes involved in the cell reactive oxygen species (ROS) balance and levels. Accordingly, in vitro studies with primary VHL-ccRCC and 786-O cells measuring ROS levels, ROS-expression of detoxifying enzymes, and the expression of p65/NF-κB targets by RT-PCR were carried out. Furthermore, histological analyses of ccRCC samples from heterotopic mouse xenografts were performed. The obtained results show that ADRB2 blockade in ccRCC cells reduces the level of oxidative stress and stabilizes the inflammatory response. Thus, these data further support the idea of targeting ADRB2 as a promising strategy for the treatment of VHL and other non-VHL tumors.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Inflamación/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Estrés Oxidativo/efectos de los fármacos , Receptores Adrenérgicos beta 2/metabolismo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hemangioblastoma/tratamiento farmacológico , Hemangioblastoma/metabolismo , Humanos , Inflamación/metabolismo , Masculino , Ratones , Propanolaminas/farmacología , Propranolol/farmacología , Transducción de Señal/efectos de los fármacos , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/tratamiento farmacológico , Enfermedad de von Hippel-Lindau/metabolismo
5.
Am J Med Genet A ; 185(8): 2576-2581, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33973706

RESUMEN

Hypoxia-inducible factors (HIFs) activate gene transcription in response to reduced O2 availability and play critical roles in development, physiology, and disease pathogenesis. Mutations that dysregulate HIF activity are the genetic basis for tumor predisposition in the von Hippel-Lindau syndrome and excess red blood cell production in hereditary erythrocytosis.


Asunto(s)
Estudios de Asociación Genética , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Predisposición Genética a la Enfermedad , Oxígeno/metabolismo , Fenotipo , Biomarcadores , Diagnóstico Diferencial , Enfermedades Genéticas Congénitas/diagnóstico , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Policitemia/congénito , Policitemia/diagnóstico , Policitemia/metabolismo , Transducción de Señal , Enfermedad de von Hippel-Lindau/diagnóstico , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
6.
Am J Med Genet A ; 185(11): 3334-3339, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34655169

RESUMEN

Hypoxia-inducible factors (HIFs) activate gene transcription in response to reduced O2 availability and play critical roles in development, physiology, and disease pathogenesis. Mutations that dysregulate HIF activity are the genetic basis for tumor predisposition in the von Hippel-Lindau syndrome and excess red blood cell production in hereditary erythrocytosis.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Oxígeno/metabolismo , Policitemia/congénito , Enfermedad de von Hippel-Lindau/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Enfermedades Genéticas Congénitas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Mutación/genética , Policitemia/genética , Policitemia/metabolismo , Policitemia/patología , Enfermedad de von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/patología
7.
J Neurooncol ; 148(1): 29-38, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32356150

RESUMEN

INTRODUCTION: Central nervous system hemangioblastoma is a benign tumor associated with or without von Hippel-Lindau (VHL) disease which is an autosomal dominant hereditary disease that results from a germline mutation in the VHL gene. A main axis of signaling pathways in central nervous system hemangioblastoma is VHL-HIF signaling pathway. Here, we propose an alternative VHL-JAK-STAT signaling pathway in hemangioblastoma and discuss the role. METHODS: Using MACS method, Scl+ hemangioblast-like cells were isolated from multipotent nestin-expressing stem cells. Then, ubiquitination of JAK2 in those cells and immunoprecipitation between JAK2 and VHL were examined. Then, expressions of JAK2 and STAT3 in those cells and expressions of VHL-associated hemangioblastoma tissues were examined. In addition, the VHL genes of patients bearing hemangioblastoma were analyzed. RESULTS: JAK2 and STAT3 in Scl+ hemangioblast-like cells were ubiquitinated after VHL- expression vector was transferred to those cells. Expressions of JAK2 and STAT3 in those cells were well recognized before the transfer, but those disappeared after the transfer. Expressions of both JAK2 and STAT3 in hemangioblastoma tissues were well shown. The VHL gene analysis revealed that patients bearing hemangioblastoma carried missense mutations in 5, small deletions in 2, large deletions in 4, and nonsense mutation in 1 CONCLUSIONS: VHL-JAK-STAT signaling pathway might play an important role in proliferation, angiogenesis, and maintenance of stem-cell-nature in hemangioblastoma as an alternative signaling pathway to supplement VHL-HIF signaling pathway.


Asunto(s)
Neoplasias Cerebelosas/metabolismo , Hemangioblastoma/metabolismo , Transducción de Señal , Enfermedad de von Hippel-Lindau/metabolismo , Adulto , Neoplasias Cerebelosas/complicaciones , Neoplasias Cerebelosas/patología , Femenino , Hemangioblastoma/complicaciones , Hemangioblastoma/patología , Humanos , Janus Quinasa 2/metabolismo , Mutación , Factor de Transcripción STAT3/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Adulto Joven , Enfermedad de von Hippel-Lindau/complicaciones , Enfermedad de von Hippel-Lindau/patología
8.
PLoS Comput Biol ; 15(4): e1006478, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30943211

RESUMEN

Familiar cancers represent a privileged point of view for studying the complex cellular events inducing tumor transformation. Von Hippel-Lindau syndrome, a familiar predisposition to develop cancer is a clear example. Here, we present our efforts to decipher the role of von Hippel-Lindau tumor suppressor protein (pVHL) in cancer insurgence. We collected high quality information about both pVHL mutations and interactors to investigate the association between patient phenotypes, mutated protein surface and impaired interactions. Our data suggest that different phenotypes correlate with localized perturbations of the pVHL structure, with specific cell functions associated to different protein surfaces. We propose five different pVHL interfaces to be selectively involved in modulating proteins regulating gene expression, protein homeostasis as well as to address extracellular matrix (ECM) and ciliogenesis associated functions. These data were used to drive molecular docking of pVHL with its interactors and guide Petri net simulations of the most promising alterations. We predict that disruption of pVHL association with certain interactors can trigger tumor transformation, inducing metabolism imbalance and ECM remodeling. Collectively taken, our findings provide novel insights into VHL-associated tumorigenesis. This highly integrated in silico approach may help elucidate novel treatment paradigms for VHL disease.


Asunto(s)
Mutación , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Carcinoma de Células Renales/genética , Biología Computacional , Genes Supresores de Tumor , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Renales/genética , Simulación del Acoplamiento Molecular , Policitemia/genética , Dominios y Motivos de Interacción de Proteínas , Mapas de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/química , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
9.
Proc Natl Acad Sci U S A ; 114(37): 9948-9953, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28851829

RESUMEN

Glucocorticoid (GC) and hypoxic transcriptional responses play a central role in tissue homeostasis and regulate the cellular response to stress and inflammation, highlighting the potential for cross-talk between these two signaling pathways. We present results from an unbiased in vivo chemical screen in zebrafish that identifies GCs as activators of hypoxia-inducible factors (HIFs) in the liver. GCs activated consensus hypoxia response element (HRE) reporters in a glucocorticoid receptor (GR)-dependent manner. Importantly, GCs activated HIF transcriptional responses in a zebrafish mutant line harboring a point mutation in the GR DNA-binding domain, suggesting a nontranscriptional route for GR to activate HIF signaling. We noted that GCs increase the transcription of several key regulators of glucose metabolism that contain HREs, suggesting a role for GC/HIF cross-talk in regulating glucose homeostasis. Importantly, we show that GCs stabilize HIF protein in intact human liver tissue and isolated hepatocytes. We find that GCs limit the expression of Von Hippel Lindau protein (pVHL), a negative regulator of HIF, and that treatment with the c-src inhibitor PP2 rescued this effect, suggesting a role for GCs in promoting c-src-mediated proteosomal degradation of pVHL. Our data support a model for GCs to stabilize HIF through activation of c-src and subsequent destabilization of pVHL.


Asunto(s)
Glucocorticoides/farmacología , Glucocorticoides/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Animales , Hipoxia de la Célula/fisiología , Humanos , Hipoxia , Ligasas/metabolismo , Hígado/metabolismo , Unión Proteica , Transducción de Señal/fisiología , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Pez Cebra , Enfermedad de von Hippel-Lindau/metabolismo
10.
Am J Physiol Cell Physiol ; 316(5): C605-C620, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30758995

RESUMEN

Autophagy is a highly conserved, homeostatic process by which cytosolic components reach lysosomes for degradation. The roles played by different autophagic processes in cancer are complex and remain cancer type and stage dependent. Renal cell carcinoma (RCC) is the most common subtype of kidney cancer and is characterized by the inactivation of the von Hippel-Lindau (VHL) tumor suppressor. Our previous study identified a small compound, STF-62247, as an autophagy-modulating molecule causing selective cytotoxicity for VHL-inactivated cells. This present study investigates the effects of STF-62247 specifically on the macroautophagic flux to better characterize its mechanism of action in RCC. Our results clearly demonstrate that this compound is a potent blocker of late stages of autophagy. We show that inhibiting autophagy by CRISPR knockouts of autophagy-related genes rendered VHL-deficient cells insensitive to STF-62247, uncovering the importance of the autophagic pathway in STF-selective cell death. By exploiting the autofluorescence of STF-62247, we pinpointed its cellular localization to lysosomes. Finally, in response to prolonged STF treatments, we show that VHL-proficient cells are able to surmount the block in late stages of autophagy by restoring their lysosome numbers. Conversely, an increase in autophagic vesicles accompanied by a time-dependent decrease in lysosomes was observed in VHL-deficient cells. This is the first mechanistic study investigating STF-62447's effects on the autophagic flux in RCC. Importantly, our study reclassifies STF-62247 as a blocker of later stages of autophagy and highlights the possibility of blocking this process through lysosome disruption in VHL-mutated RCCs.


Asunto(s)
Autofagia/fisiología , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Lisosomas/metabolismo , Piridinas/metabolismo , Tiazoles/metabolismo , Enfermedad de von Hippel-Lindau/metabolismo , Autofagia/efectos de los fármacos , Carcinoma de Células Renales/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos/métodos , Células HeLa , Humanos , Neoplasias Renales/tratamiento farmacológico , Lisosomas/efectos de los fármacos , Piridinas/administración & dosificación , Tiazoles/administración & dosificación , Enfermedad de von Hippel-Lindau/tratamiento farmacológico
11.
Genes Chromosomes Cancer ; 56(12): 823-831, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28779490

RESUMEN

Inhibitors of molecular chaperones and the ubiquitin-proteasome system have already been clinically implemented to counter certain cancers, including multiple myeloma and mantle cell lymphoma. The efficacy of this treatment relies on genomic alterations in cancer cells causing a proteostatic imbalance, which makes them more dependent on protein quality control (PQC) mechanisms than normal cells. Accordingly, blocking PQC, e.g. by proteasome inhibitors, may cause a lethal proteotoxic crisis in cancer cells, while leaving normal cells unaffected. Evidence, however, suggests that the PQC system operates by following a better-safe-than-sorry principle and is thus prone to target proteins that are only slightly structurally perturbed, but still functional. Accordingly, implementing PQC inhibitors may also, through an entirely different mechanism, hold potential for other cancers. Several inherited cancer susceptibility syndromes, such as Lynch syndrome and von Hippel-Lindau disease, are caused by missense mutations in tumor suppressor genes, and in some cases, the resulting amino acid substitutions in the encoded proteins cause the cellular PQC system to target them for degradation, although they may still retain function. As a consequence of this over-meticulous PQC mechanism, the cell may end up with an insufficient amount of the abnormal, but functional, protein, which in turn leads to a loss-of-function phenotype and manifestation of the disease. Increasing the amounts of such proteins by stabilizing with chemical chaperones, or by targeting molecular chaperones or the ubiquitin-proteasome system, may thus avert or delay the disease onset. Here, we review the potential of targeting the PQC system in hereditary cancer susceptibility syndromes.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales Hereditarias sin Poliposis/tratamiento farmacológico , Inhibidores de Proteasoma/farmacología , Proteostasis , Enfermedad de von Hippel-Lindau/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/metabolismo , Humanos , Inhibidores de Proteasoma/uso terapéutico , Proteolisis , Ubiquitinación , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
12.
Pol Merkur Lekarski ; 44(263): 248-252, 2018 May 25.
Artículo en Polaco | MEDLINE | ID: mdl-29813043

RESUMEN

Von Hippel-Lindau disease (vHL, familial cerebello-retinal angiomatosis) is a rare genetic autosomal dominant disorder associated with predisposition to vascular tumors. Mutations of VHL tumor suppressor gene, located on chromosome 3p25-26, are responsible for clinical manifestation of the disease. The VHL gene product encodes VHL protein, which is responsible for HIF-1 (hypoxia-inducible factor-1) dependent cell cycle regulation and cellular pathways mediated by VEGF, PDGF, TGF-α, EPO. The mechanism substantiates the hypoxia dependent vascular tumor growth caused by loss of wild-type VHL protein. The clinical spectrum of vHL syndrome includes multiple tumors of various localization and low histologic grade, often bilateral. The most typical for the syndrome are: hemangioblastoma of central nervous system (typically posterior fossa or medulla), retinal hemangioblastoma, renal cell carcinoma and pheochromocytoma. The aim of the case report is to remind the typical clinical manifestation of von Hippel- Lindau syndrome, update the diagnostic criteria, recommended diagnostic and follow up methods.


Asunto(s)
Cuidados Paliativos , Eliminación de Secuencia , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/diagnóstico , Adulto , Exones , Femenino , Humanos , Neoplasias Renales/etiología , Neoplasias Renales/cirugía , Enfermedad de von Hippel-Lindau/complicaciones , Enfermedad de von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/terapia
13.
J Biol Chem ; 291(14): 7357-72, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26846855

RESUMEN

von Hippel-Lindau (VHL) disease is a rare familial cancer predisposition syndrome caused by a loss or mutation in a single gene,VHL, but it exhibits a wide phenotypic variability that can be categorized into distinct subtypes. The phenotypic variability has been largely argued to be attributable to the extent of deregulation of the α subunit of hypoxia-inducible factor α, a well established target of VHL E3 ubiquitin ligase, ECV (Elongins/Cul2/VHL). Here, we show that erythropoietin receptor (EPOR) is hydroxylated on proline 419 and 426 via prolyl hydroxylase 3. EPOR hydroxylation is required for binding to the ß domain of VHL and polyubiquitylation via ECV, leading to increased EPOR turnover. In addition, several type-specific VHL disease-causing mutants, including those that have retained proper binding and regulation of hypoxia-inducible factor α, showed a severe defect in binding prolyl hydroxylated EPOR peptides. These results identify EPOR as the secondbona fidehydroxylation-dependent substrate of VHL that potentially influences oxygen homeostasis and contributes to the complex genotype-phenotype correlation in VHL disease.


Asunto(s)
Oxígeno/metabolismo , Proteolisis , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Ubiquitinación , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Células HEK293 , Humanos , Receptores de Eritropoyetina/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/patología
14.
Kidney Int ; 87(4): 761-70, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25469848

RESUMEN

Renal endothelin-converting enzyme (ECE)-1 is induced in experimental diabetes and following radiocontrast administration, conditions characterized by renal hypoxia, hypoxia-inducible factor (HIF) stabilization, and enhanced endothelin synthesis. Here we tested whether ECE-1 might be a HIF-target gene in vitro and in vivo. ECE-1 transcription and expression increased in cultured vascular endothelial and proximal tubular cell lines, subject to hypoxia, to mimosine or cobalt chloride. These interventions are known to stabilize HIF signaling by inhibition of HIF-prolyl hydroxylases. In rats, HIF-prolyl-hydroxylase inhibition by mimosine or FG-4497 increased HIF-1α immunostaining in renal tubules, principally in distal nephron segments. This was associated with markedly enhanced ECE-1 protein expression, predominantly in the renal medulla. A progressive and dramatic increase in ECE-1 immunostaining over time, in parallel with enhanced HIF expression, was also noted in conditional von Hippel-Lindau knockout mice. Since HIF and STAT3 are cross-stimulated, we triggered HIF expression by STAT3 activation in mice, transfected by or injected with a chimeric IL-6/IL-6-receptor protein, and found a similar pattern of enhanced ECE-1 expression. Chromatin immunoprecipitation sequence (ChIP-seq) and PCR analysis in hypoxic endothelial cells identified HIF binding at the ECE-1 promoter and intron regions. Thus, our findings suggest that ECE-1 may be a novel HIF-target gene.


Asunto(s)
Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Animales , Hipoxia de la Célula/fisiología , Células Cultivadas , Cobalto/farmacología , Dioxigenasas/antagonistas & inhibidores , Enzimas Convertidoras de Endotelina , Células Endoteliales de la Vena Umbilical Humana , Humanos , Intrones , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mimosina/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidores de Prolil-Hidroxilasa/farmacología , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transcripción Genética , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
15.
Hum Mol Genet ; 21(26): 5456-71, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23001567

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) and von Hippel-Lindau (VHL) disease lead to large kidney cysts that share pathogenetic features. The polycystin-1 (PC1) and pVHL proteins may therefore participate in the same key signaling pathways. Jade-1 is a pro-apoptotic and growth suppressive ubiquitin ligase for beta-catenin and transcriptional coactivator associated with histone acetyltransferase activity that is stabilized by pVHL in a manner that correlates with risk of VHL renal disease. Thus, a relationship between Jade-1 and PC1 was sought. Full-length PC1 bound, stabilized and colocalized with Jade-1 and inhibited Jade-1 ubiquitination. In contrast, the cytoplasmic tail or the naturally occurring C-terminal fragment of PC1 (PC1-CTF) promoted Jade-1 ubiquitination and degradation, suggesting a dominant-negative mechanism. ADPKD-associated PC1 mutants failed to regulate Jade-1, indicating a potential disease link. Jade-1 ubiquitination was mediated by Siah-1, an E3 ligase that binds PC1. By controlling Jade-1 abundance, PC1 and the PC1-CTF differentially regulate Jade-1-mediated transcriptional activity. A key target of PC1, the cyclin-dependent kinase inhibitor p21, is also up-regulated by Jade-1. Through Jade-1, PC1 and PC1 cleaved forms may exert fine control of beta-catenin and canonical Wnt signaling, a critical pathway in cystic renal disease. Thus, Jade-1 is a transcription factor and ubiquitin ligase whose activity is regulated by PC1 in a manner that is physiologic and may correlate with disease. Jade-1 may be an important therapeutic target in renal cystogenesis.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Canales Catiónicos TRPP/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación , Secuencia de Aminoácidos , Apoptosis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células HEK293 , Semivida , Proteínas de Homeodominio/genética , Humanos , Riñón/citología , Riñón/patología , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Canales Catiónicos TRPP/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
16.
Nat Cell Biol ; 9(5): 588-95, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17450132

RESUMEN

Defects in the structure or function of the primary cilium, an antennae-like structure whose functional integrity has been linked to the suppression of uncontrolled kidney epithelial cell proliferation, are a common feature of genetic disorders characterized by kidney cysts. However, the mechanisms by which primary cilia are maintained remain poorly defined. von Hippel-Lindau (VHL) disease is characterized by the development of premalignant renal cysts and arises because of functional inactivation of the VHL tumour suppressor gene product, pVHL. Here, we show that pVHL and glycogen synthase kinase (GSK)3beta are key components of an interlinked signalling pathway that maintains the primary cilium. Although inactivation of either pVHL or GSK3beta alone did not affect cilia maintenance, their combined inactivation leads to loss of cilia. In VHL patients, GSK3beta is subjected to inhibitory phosphorylation in renal cysts, but not in early VHL mutant lesions, and these cysts exhibit reduced frequencies of primary cilia. We propose that pVHL and GSK3beta function together in a ciliary-maintenance signalling network, disruption of which enhances the vulnerability of cells to lose their cilia, thereby promoting cyst formation.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Enfermedades Renales Quísticas/metabolismo , Túbulos Renales/metabolismo , Transducción de Señal , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/metabolismo , Animales , Línea Celular , Cilios/metabolismo , Cilios/patología , Embrión de Mamíferos/citología , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Glucógeno Sintasa Quinasa 3/deficiencia , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Enfermedades Renales Quísticas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fosforilación , Interferencia de ARN , Transfección , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/patología
17.
Acta Neuropathol ; 125(3): 333-50, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23400300

RESUMEN

Patients with von Hippel-Lindau disease carry a germline mutation of the Von Hippel-Lindau (VHL) tumor-suppressor gene. We discuss the molecular consequences of loss of VHL gene function and their impact on the nervous system. Dysfunction of the VHL protein causes accumulation and activation of hypoxia inducible factor (HIF) which can be demonstrated in earliest stages of tumorigenesis and is followed by expression of VEGF, erythropoietin, nitric oxide synthase and glucose transporter 1 in VHL-deficient tumor cells. HIF-independent functions of VHL, epigenetic inactivation of VHL, pVHL proteostasis, and links between loss of VHL function and developmental arrest are also described. A most intriguing feature in VHL disease is the occurrence of primary hemangioblastic tumors in the nervous system, the origin of which has not yet been entirely clarified, and current hypotheses are discussed. Endolymphatic sac tumors may extend into the brain, but originally arise from proliferation of endolymphatic duct/sac epithelium; the exact nature of the proliferating epithelial cell, however, also has remained unclear, as well as the question why tumors almost consistently develop in the intraosseous portion of the endolymphatic sac/duct only. The epitheloid clear cell morphology of both advanced hemangioblastoma and renal clear cell carcinoma can make the differential diagnosis challenging, recent developments in immunohistochemical differentiation are discussed. Finally, metastasis to brain may not only be caused by renal carcinoma, but may derive from VHL disease-associated pheochromocytoma/paraganglioma, or pancreatic neuroendocrine tumor.


Asunto(s)
Neoplasias del Sistema Nervioso/patología , Sistema Nervioso/patología , Enfermedad de von Hippel-Lindau/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Saco Endolinfático/patología , Hemangioblastoma/complicaciones , Hemangioblastoma/etiología , Hemangioblastoma/patología , Humanos , Sistema Nervioso/fisiopatología , Neoplasias del Sistema Nervioso/etiología , Neuroimagen , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/complicaciones , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
18.
J Neurooncol ; 111(3): 313-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23203444

RESUMEN

Von Hippel-Lindau (VHL) is a tumor suppressor that negatively regulates the production of angiogenic factors. Mutations in the VHL gene cause VHL syndrome, which is characterized by highly vascularized tumors. Here we report a c.464T>A mutation of the VHL gene in three patients with hemangioblastoma from a Chinese family. This mutation was not reported previously and was absent in the unaffected family members. The mutation is predicted to cause Val to Glu substitution at VHL protein residue 155 in a conserved region. Previous biochemical studies demonstrated that residue Val-155 was critical for VHL protein binding to chaperonin TRiC/CCT, an essential step for proper VHL protein folding. Our finding of naturally occurring VHL V155E mutation in patients with VHL syndrome supports the functional importance of Val-155 residue in VHL protein and illustrates the diversity of VHL gene defects underlying VHL syndrome.


Asunto(s)
Salud de la Familia , Mutación/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/genética , Adulto , Anciano , Pueblo Asiatico/genética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Análisis Mutacional de ADN , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Mucina-1/metabolismo , Tomografía Computarizada por Rayos X , Enfermedad de von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/patología
19.
Nat Rev Cancer ; 2(9): 673-82, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12209156

RESUMEN

The von Hippel-Lindau hereditary cancer syndrome was first described about 100 years ago. The unusual clinical features of this disorder predicted a role for the von Hippel-Lindau gene (VHL) in the oxygen-sensing pathway. Indeed, recent studies of this gene have helped to decipher how cells sense changes in oxygen availability, and have revealed a previously unappreciated role of prolyl hydroxylation in intracellular signalling. These studies, in turn, are laying the foundation for the treatment of a diverse set of disorders, including cancer, myocardial infarction and stroke.


Asunto(s)
Predisposición Genética a la Enfermedad , Ligasas/genética , Neoplasias/complicaciones , Neoplasias/genética , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas , Enfermedad de von Hippel-Lindau/complicaciones , Enfermedad de von Hippel-Lindau/genética , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau , Enfermedad de von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/patología
20.
Proteins ; 80(3): 733-46, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22105711

RESUMEN

The von Hippel-Lindau tumor suppressor protein (pVHL) has an essential role in the regulation of the hypoxia response pathway in animal cells. Under normoxic conditions, the hypoxia-inducible factor (HIF) undergoes trans-4-prolyl hydroxylation and is subsequently recognised by the ß-domain of pVHL, leading to the ubiquitination and degradation of HIF. Mutations of pVHL alter the binding of HIF. A subset of relevant clinically observed mutations to pVHL are thought to cause weaker binding of HIF-1α and are associated with cancer and cardiovascular diseases. Here, we present computational studies analyzing the interaction of HIF with mutant forms of pVHL, describing at atomic detail the local structural reorganization caused by substitution of certain residues of pVHL. The results reveal that the canonical configuration in the wild-type system is vital for the efficient functioning of the complex and that mutation of any of the residues implicated in the h-bond network in the binding site disrupts HIF binding. Although the experimentally observed ordering of binding energies for mutants of Tyr98 is reproduced, our examination of a broader range of mutations does not support the hypothesis of a correlation between the degree of disruption of the pVHL/HIF-1α interaction caused by a mutation and the phenotype with which the mutation is associated. We suggest that disruption of the binding interaction is one of many factors behind the manifestation of VHL disease.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mutación Puntual , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Estabilidad Proteica , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/química , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA