Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Physiol ; 15: 1351682, 2024.
Article in English | MEDLINE | ID: mdl-38444761

ABSTRACT

Molecular and physiological determinants of the timing of reproductive events, including the pre-ovulatory LH surge and seasonal fluctuations in fertility, are incompletely understood. We used the Cryptochrome 1-deficient duper mutant to examine the role of this core circadian clock gene in Syrian hamsters. We find that the phase of the LH surge and its stability upon shifts of the light: dark cycle are altered in duper mutants. The intensity of immunoreactive PER1 in GnRH cells of the preoptic area peaks earlier in the day in duper than wild type hamsters. We note that GnRH fibers coursing through the suprachiasmatic nucleus (SCN) contact vasopressin- and VIP-immunoreactive cells, suggesting a possible locus of circadian control of the LH surge. Unlike wild types, duper hamsters do not regress their gonads within 8 weeks of constant darkness, despite evidence of melatonin secretion during the subjective night. In light of the finding that the duper allele is a stop codon in Cryptochrome 1, our results suggest important neuroendocrine functions of this core circadian clock gene.

2.
Cancer Cell ; 39(9): 1262-1278.e7, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34329586

ABSTRACT

Fusion-transcription factors (fusion-TFs) represent a class of driver oncoproteins that are difficult to therapeutically target. Recently, protein degradation has emerged as a strategy to target these challenging oncoproteins. The mechanisms that regulate fusion-TF stability, however, are generally unknown. Using CRISPR-Cas9 screening, we discovered tripartite motif-containing 8 (TRIM8) as an E3 ubiquitin ligase that ubiquitinates and degrades EWS/FLI, a driver fusion-TF in Ewing sarcoma. Moreover, we identified TRIM8 as a selective dependency in Ewing sarcoma compared with >700 other cancer cell lines. Mechanistically, TRIM8 knockout led to an increase in EWS/FLI protein levels that was not tolerated. EWS/FLI acts as a neomorphic substrate for TRIM8, defining the selective nature of the dependency. Our results demonstrate that fusion-TF protein stability is tightly regulated and highlight fusion oncoprotein-specific regulators as selective therapeutic targets. This study provides a tractable strategy to therapeutically exploit oncogene overdose in Ewing sarcoma and potentially other fusion-TF-driven cancers.


Subject(s)
Bone Neoplasms/mortality , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Proteins, Fusion/chemistry , Proto-Oncogene Protein c-fli-1/chemistry , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/chemistry , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/mortality , Bone Neoplasms/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Knockout Techniques , HEK293 Cells , Humans , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Oncogene Proteins, Fusion/metabolism , Protein Stability , Proteolysis , Sarcoma, Ewing/metabolism , Trans-Activators/metabolism
3.
Cancer Cell ; 34(6): 922-938.e7, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30537514

ABSTRACT

Drug resistance represents a major challenge to achieving durable responses to cancer therapeutics. Resistance mechanisms to epigenetically targeted drugs remain largely unexplored. We used bromodomain and extra-terminal domain (BET) inhibition in neuroblastoma as a prototype to model resistance to chromatin modulatory therapeutics. Genome-scale, pooled lentiviral open reading frame (ORF) and CRISPR knockout rescue screens nominated the phosphatidylinositol 3-kinase (PI3K) pathway as promoting resistance to BET inhibition. Transcriptomic and chromatin profiling of resistant cells revealed that global enhancer remodeling is associated with upregulation of receptor tyrosine kinases (RTKs), activation of PI3K signaling, and vulnerability to RTK/PI3K inhibition. Large-scale combinatorial screening with BET inhibitors identified PI3K inhibitors among the most synergistic upfront combinations. These studies provide a roadmap to elucidate resistance to epigenetic-targeted therapeutics and inform efficacious combination therapies.


Subject(s)
Azepines/pharmacology , Drug Resistance, Neoplasm/drug effects , Indazoles/pharmacology , Molecular Targeted Therapy/methods , Neuroblastoma/drug therapy , Sulfonamides/pharmacology , Triazoles/pharmacology , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Disease-Free Survival , Epigenesis, Genetic/drug effects , Female , Humans , Mice, Nude , Neuroblastoma/genetics , Neuroblastoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteins/antagonists & inhibitors , Proteins/metabolism , Signal Transduction/drug effects
4.
Sci Rep ; 8(1): 10879, 2018 Jul 18.
Article in English | MEDLINE | ID: mdl-30022112

ABSTRACT

The self-assembling, mechanoenzymatic dynamin superfamily GTPase, dynamin-related protein 1 (Drp1), catalyzes mitochondrial and peroxisomal fission. Distinct intrinsically disordered regions (IDRs) in Drp1 substitute for the canonical pleckstrin homology (PH) domain and proline-rich domain (PRD) of prototypical dynamin, which cooperatively regulate endocytic vesicle scission. Whether the Drp1 IDRs function analogously to the corresponding dynamin domains however remains unknown. We show that an IDR unique to the Drp1 GTPase (G) domain, the 'extended 80-loop', albeit dissimilar in location, structure, and mechanism, functions akin to the dynamin PRD by enabling stable Drp1 mitochondrial recruitment and by suppressing Drp1 cooperative GTPase activity in the absence of specific partner-protein interactions. Correspondingly, we find that another IDR, the Drp1 variable domain (VD), in conjunction with the conserved stalk L1N loop, functions akin to the dynamin PH domain; first, in an 'auto-inhibitory' capacity that restricts Drp1 activity through a long-range steric inhibition of helical inter-rung G-domain dimerization, and second, as a 'fulcrum' for Drp1 self-assembly in the proper helical register. We show that the Drp1 VD is necessary and sufficient for specific Drp1-phospholipid interactions. We further demonstrate that the membrane-dependent VD conformational rearrangement essential for the alleviation of Drp1 auto-inhibition is contingent upon the basal GTP hydrolysis-dependent generation of Drp1 dimers from oligomers in solution. IDRs thus conformationally couple the enzymatic and membrane activities of Drp1 toward membrane fission.


Subject(s)
Dynamins/chemistry , GTP Phosphohydrolases/metabolism , Intrinsically Disordered Proteins/chemistry , Microtubule-Associated Proteins/chemistry , Mitochondrial Dynamics , Mitochondrial Proteins/chemistry , Amino Acid Sequence , Dynamins/metabolism , GTP Phosphohydrolases/chemistry , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Intrinsically Disordered Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/metabolism , Protein Multimerization , Protein Structure, Tertiary , Sequence Homology
5.
Cancer Cell ; 33(2): 202-216.e6, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29358035

ABSTRACT

Many cancer types are driven by oncogenic transcription factors that have been difficult to drug. Transcriptional inhibitors, however, may offer inroads into targeting these cancers. Through chemical genomics screening, we identified that Ewing sarcoma is a disease with preferential sensitivity to THZ1, a covalent small-molecule CDK7/12/13 inhibitor. The selective CDK12/13 inhibitor, THZ531, impairs DNA damage repair in an EWS/FLI-dependent manner, supporting a synthetic lethal relationship between response to THZ1/THZ531 and EWS/FLI expression. The combination of these molecules with PARP inhibitors showed striking synergy in cell viability and DNA damage assays in vitro and in multiple models of Ewing sarcoma, including a PDX, in vivo without hematopoietic toxicity.


Subject(s)
Cyclin-Dependent Kinases/drug effects , Phenylenediamines/pharmacology , Proto-Oncogene Protein c-fli-1/genetics , Pyrimidines/pharmacology , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oncogene Proteins, Fusion/drug effects , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/drug effects , RNA-Binding Protein EWS/drug effects , Synthetic Lethal Mutations/drug effects , Synthetic Lethal Mutations/genetics
6.
J Clin Invest ; 128(1): 446-462, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202477

ABSTRACT

Pharmacologically difficult targets, such as MYC transcription factors, represent a major challenge in cancer therapy. For the childhood cancer neuroblastoma, amplification of the oncogene MYCN is associated with high-risk disease and poor prognosis. Here, we deployed genome-scale CRISPR-Cas9 screening of MYCN-amplified neuroblastoma and found a preferential dependency on genes encoding the polycomb repressive complex 2 (PRC2) components EZH2, EED, and SUZ12. Genetic and pharmacological suppression of EZH2 inhibited neuroblastoma growth in vitro and in vivo. Moreover, compared with neuroblastomas without MYCN amplification, MYCN-amplified neuroblastomas expressed higher levels of EZH2. ChIP analysis showed that MYCN binds at the EZH2 promoter, thereby directly driving expression. Transcriptomic and epigenetic analysis, as well as genetic rescue experiments, revealed that EZH2 represses neuronal differentiation in neuroblastoma in a PRC2-dependent manner. Moreover, MYCN-amplified and high-risk primary tumors from patients with neuroblastoma exhibited strong repression of EZH2-regulated genes. Additionally, overexpression of IGFBP3, a direct EZH2 target, suppressed neuroblastoma growth in vitro and in vivo. We further observed strong synergy between histone deacetylase inhibitors and EZH2 inhibitors. Together, these observations demonstrate that MYCN upregulates EZH2, leading to inactivation of a tumor suppressor program in neuroblastoma, and support testing EZH2 inhibitors in patients with MYCN-amplified neuroblastoma.


Subject(s)
CRISPR-Cas Systems , Cell Differentiation , Enhancer of Zeste Homolog 2 Protein , Gene Amplification , Gene Expression Regulation, Neoplastic , N-Myc Proto-Oncogene Protein , Neuroblastoma , Up-Regulation , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , N-Myc Proto-Oncogene Protein/biosynthesis , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...