Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 116(34): 16997-17006, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31375625

ABSTRACT

Despite the discovery of the oxygen-sensitive regulation of HIFα by the von Hippel-Lindau (VHL) protein, the mechanisms underlying the complex genotype/phenotype correlations in VHL disease remain unknown. Some germline VHL mutations cause familial pheochromocytoma and encode proteins that preserve their ability to down-regulate HIFα. While type 1, 2A, and 2B VHL mutants are defective in regulating HIFα, type 2C mutants encode proteins that preserve their ability to down-regulate HIFα. Here, we identified an oxygen-sensitive function of VHL that is abolished by VHL type 2C mutations. We found that BIM-EL, a proapoptotic BH3-only protein, is hydroxylated by EglN3 and subsequently bound by VHL. VHL mutants fail to bind hydroxylated BIM-EL, regardless of whether they have the ability to bind hydroxylated HIFα or not. VHL binding inhibits BIM-EL phosphorylation by extracellular signal-related kinase (ERK) on serine 69. This causes BIM-EL to escape from proteasomal degradation, allowing it to enhance EglN3-induced apoptosis. BIM-EL was rapidly degraded in cells lacking wild-type VHL or in which EglN3 was inactivated genetically or by lack of oxygen, leading to enhanced cell survival and chemotherapy resistance. Combination therapy using ERK inhibitors, however, resensitizes VHL- and EglN3-deficient cells that are otherwise cisplatin-resistant.


Subject(s)
Adrenal Gland Neoplasms , Bcl-2-Like Protein 11/metabolism , Drug Resistance, Neoplasm/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mutation , Pheochromocytoma , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Bcl-2-Like Protein 11/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Hydroxylation/drug effects , Hydroxylation/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , PC12 Cells , Pheochromocytoma/drug therapy , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Proteolysis/drug effects , Rats , Von Hippel-Lindau Tumor Suppressor Protein/genetics
3.
Proc Natl Acad Sci U S A ; 114(30): E6137-E6146, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696319

ABSTRACT

Neuroblastoma is a pediatric cancer characterized by variable outcomes ranging from spontaneous regression to life-threatening progression. High-risk neuroblastoma patients receive myeloablative chemotherapy with hematopoietic stem-cell transplant followed by adjuvant retinoid differentiation treatment. However, the overall survival remains low; hence, there is an urgent need for alternative therapeutic approaches. One feature of high-risk neuroblastoma is the high level of DNA methylation of putative tumor suppressors. Combining the reversibility of DNA methylation with the differentiation-promoting activity of retinoic acid (RA) could provide an alternative strategy to treat high-risk neuroblastoma. Here we show that treatment with the DNA-demethylating drug 5-Aza-deoxycytidine (AZA) restores high-risk neuroblastoma sensitivity to RA. Combined systemic distribution of AZA and RA impedes tumor growth and prolongs survival. Genome-wide analysis of treated tumors reveals that this combined treatment rapidly induces a HIF2α-associated hypoxia-like transcriptional response followed by an increase in neuronal gene expression and a decrease in cell-cycle gene expression. A small-molecule inhibitor of HIF2α activity diminishes the tumor response to AZA+RA treatment, indicating that the increase in HIF2α levels is a key component in tumor response to AZA+RA. The link between increased HIF2α levels and inhibited tumor growth is reflected in large neuroblastoma patient datasets. Therein, high levels of HIF2α, but not HIF1α, significantly correlate with expression of neuronal differentiation genes and better prognosis but negatively correlate with key features of high-risk tumors, such as MYCN amplification. Thus, contrary to previous studies, our findings indicate an unanticipated tumor-suppressive role for HIF2α in neuroblastoma.


Subject(s)
Azacitidine/analogs & derivatives , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Proliferation/genetics , Genetic Therapy/methods , Neuroblastoma/pathology , Tretinoin/therapeutic use , Animals , Azacitidine/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chemotherapy, Adjuvant , Decitabine , Female , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Mice, Nude
4.
Genes Dev ; 31(10): 1036-1053, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28637693

ABSTRACT

We recently identified pathogenic KIF1Bß mutations in sympathetic nervous system malignancies that are defective in developmental apoptosis. Here we deleted KIF1Bß in the mouse sympathetic nervous system and observed impaired sympathetic nervous function and misexpression of genes required for sympathoadrenal lineage differentiation. We discovered that KIF1Bß is required for nerve growth factor (NGF)-dependent neuronal differentiation through anterograde transport of the NGF receptor TRKA. Moreover, pathogenic KIF1Bß mutations identified in neuroblastoma impair TRKA transport. Expression of neuronal differentiation markers is ablated in both KIF1Bß-deficient mouse neuroblasts and human neuroblastomas that lack KIF1Bß. Transcriptomic analyses show that unfavorable neuroblastomas resemble mouse sympathetic neuroblasts lacking KIF1Bß independent of MYCN amplification and the loss of genes neighboring KIF1B on chromosome 1p36. Thus, defective precursor cell differentiation, a common trait of aggressive childhood malignancies, is a pathogenic effect of KIF1Bß loss in neuroblastomas. Furthermore, neuropathy-associated KIF1Bß mutations impede cargo transport, providing a direct link between neuroblastomas and neurodegeneration.


Subject(s)
Cell Differentiation/genetics , Kinesins/genetics , Kinesins/metabolism , Neuroblastoma/genetics , Neurons/cytology , Receptor, trkA/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation, Developmental , Gene Silencing , Mutation , Neuroblastoma/physiopathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , PC12 Cells , Rats , Signal Transduction/genetics , Sympathetic Nervous System/cytology , ras Proteins/genetics
5.
Hypoxia (Auckl) ; 4: 15-27, 2016.
Article in English | MEDLINE | ID: mdl-27525289

ABSTRACT

The cellular response to hypoxia is primarily regulated by the hypoxia-inducible factors (HIFs). HIF-1α is also a major mediator of tumor physiology, and its abundance is correlated with therapeutic resistance in a broad range of cancers. Accumulation of HIF-1α under hypoxia is mainly controlled by the oxygen-sensing HIF prolyl 4-hydroxylases (EGLNs, also known as PHDs). Here, we identified a high level of normoxic HIF-1α protein in various cancer cell lines. EGLNs require oxygen and 2-oxoglutarate for enzymatic activity. We tested the ability of several cell-permeable 2-oxoglutarate analogs to regulate the abundance of HIF-1α protein. We identified 3-oxoglutarate as a potent regulator of HIF-1α in normoxic conditions. In contrast to 2-oxoglutarate, 3-oxoglutarate decreased the abundance of HIF-1α protein in several cancer cell lines in normoxia and diminished HIF-1α levels independent of EGLN enzymatic activity. Furthermore, we observed that 3-oxoglutarate was detrimental to cancer cell survival. We show that esterified 3-oxoglutarate, in combination with the cancer chemotherapeutic drug vincristine, induces apoptosis and inhibits tumor growth in vitro and in vivo. Our data imply that a novel treatment strategy targeting HIF-1α in combination with the use of existing cytotoxic agents could serve as potent, future antitumor chemotherapies.

6.
Dev Cell ; 36(2): 164-78, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26812016

ABSTRACT

KIF1Bß is a candidate 1p36 tumor suppressor that regulates apoptosis in the developing sympathetic nervous system. We found that KIF1Bß activates the Ca(2+)-dependent phosphatase calcineurin (CN) by stabilizing the CN-calmodulin complex, relieving enzymatic autoinhibition and enabling CN substrate recognition. CN is the key mediator of cellular responses to Ca(2+) signals and its deregulation is implicated in cancer, cardiac, neurodegenerative, and immune disease. We show that KIF1Bß affects mitochondrial dynamics through CN-dependent dephosphorylation of Dynamin-related protein 1 (DRP1), causing mitochondrial fission and apoptosis. Furthermore, KIF1Bß actuates recognition of all known CN substrates, implying a general mechanism for KIF1Bß in Ca(2+) signaling and how Ca(2+)-dependent signaling is executed by CN. Pathogenic KIF1Bß mutations previously identified in neuroblastomas and pheochromocytomas all fail to activate CN or stimulate DRP1 dephosphorylation. Importantly, KIF1Bß and DRP1 are silenced in 1p36 hemizygous-deleted neuroblastomas, indicating that deregulation of calcineurin and mitochondrial dynamics contributes to high-risk and poor-prognosis neuroblastoma.


Subject(s)
Apoptosis/genetics , Calcineurin/genetics , GTP Phosphohydrolases/genetics , Kinesins/genetics , Microtubule-Associated Proteins/genetics , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Mutation/genetics , Dynamins , Genes, Tumor Suppressor/physiology , Humans , Kinesins/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Phosphorylation , Signal Transduction/genetics
7.
Cancer Discov ; 4(4): 434-51, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469107

ABSTRACT

UNLABELLED: Inherited KIF1B loss-of-function mutations in neuroblastomas and pheochromocytomas implicate the kinesin KIF1B as a 1p36.2 tumor suppressor. However, the mechanism of tumor suppression is unknown. We found that KIF1B isoform ß (KIF1Bß) interacts with RNA helicase A (DHX9), causing nuclear accumulation of DHX9, followed by subsequent induction of the proapoptotic XIAP-associated factor 1 (XAF1) and, consequently, apoptosis. Pheochromocytoma and neuroblastoma arise from neural crest progenitors that compete for growth factors such as nerve growth factor (NGF) during development. KIF1Bß is required for developmental apoptosis induced by competition for NGF. We show that DHX9 is induced by and required for apoptosis stimulated by NGF deprivation. Moreover, neuroblastomas with chromosomal deletion of 1p36 exhibit loss of KIF1Bß expression and impaired DHX9 nuclear localization, implicating the loss of DHX9 nuclear activity in neuroblastoma pathogenesis. SIGNIFICANCE: KIF1Bß has neuroblastoma tumor-suppressor properties and promotes and requires nuclear-localized DHX9 for its apoptotic function by activating XAF1 expression. Loss of KIF1Bß alters subcellular localization of DHX9 and diminishes NGF dependence of sympathetic neurons, leading to reduced culling of neural progenitors, and, therefore, might predispose to tumor formation.


Subject(s)
DEAD-box RNA Helicases/metabolism , Kinesins/metabolism , Neoplasm Proteins/metabolism , Nerve Growth Factor/genetics , Neuroblastoma/genetics , Animals , Apoptosis , Cell Nucleus/metabolism , Chromosomes, Human, Pair 1 , DEAD-box RNA Helicases/genetics , Humans , Karyopherins/metabolism , Kinesins/genetics , Mice , Mice, Inbred C57BL , Neoplasm Proteins/genetics , PC12 Cells , Rats , Sequence Deletion , Sympathetic Nervous System/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL