Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 52
1.
Acta Pharmacol Sin ; 44(5): 940-953, 2023 May.
Article En | MEDLINE | ID: mdl-36357669

Dopaminergic neuron degeneration is a hallmark of Parkinson's disease (PD). We previously reported that the inactivation of von Hippel‒Lindau (VHL) alleviated dopaminergic neuron degeneration in a C. elegans model. In this study, we investigated the specific effects of VHL loss and the underlying mechanisms in mammalian PD models. For in vivo genetic inhibition of VHL, AAV-Vhl-shRNA was injected into mouse lateral ventricles. Thirty days later, the mice received MPTP for 5 days to induce PD. Behavioral experiments were conducted on D1, D3, D7, D14 and D21 after the last injection, and the mice were sacrificed on D22. We showed that knockdown of VHL in mice significantly alleviated PD-like syndromes detected in behavioral and biochemical assays. Inhibiting VHL exerted similar protective effects in MPP+-treated differentiated SH-SY5Y cells and the MPP+-induced C. elegans PD model. We further demonstrated that VHL loss-induced protection against experimental parkinsonism was independent of hypoxia-inducible factor and identified the Dishevelled-2 (DVL-2)/ß-catenin axis as the target of VHL, which was evolutionarily conserved in both C. elegans and mammals. Inhibiting the function of VHL promoted the stability of ß-catenin by reducing the ubiquitination and degradation of DVL-2. Thus, in vivo overexpression of DVL-2, mimicking VHL inactivation, protected against PD. We designed a competing peptide, Tat-DDF-2, to inhibit the interaction between VHL and DVL-2, which exhibited pharmacological potential for protection against PD in vitro and in vivo. We propose the therapeutic potential of targeting the interaction between VHL and DVL-2, which may represent a strategy to alleviate neurodegeneration associated with PD.


Dishevelled Proteins , Parkinson Disease , Von Hippel-Lindau Tumor Suppressor Protein , Animals , Humans , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , beta Catenin/metabolism , Caenorhabditis elegans/metabolism , Disease Models, Animal , Dishevelled Proteins/drug effects , Dishevelled Proteins/metabolism , Dopamine/pharmacology , Dopaminergic Neurons/metabolism , Mammals , Mice, Inbred C57BL , Neuroblastoma/metabolism , Parkinson Disease/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Ubiquitination/drug effects , Ubiquitination/genetics , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
2.
Eur J Med Chem ; 227: 113906, 2022 Jan 05.
Article En | MEDLINE | ID: mdl-34656901

Proteolysis targeting chimeras (PROTACs), which hijack proteins of interest (POIs) and recruit E3 ligases for target degradation via the ubiquitin-proteasome pathway, are a novel drug discovery paradigm that has been widely used as biological tools and medicinal molecules with the potential of clinical application value. To date, a wide variety of small molecule PROTACs have been developed. Importantly, VHL-based PROTACs have emerged to be a promising approach for proteins, including those non-druggable ones, such as transcriptional factors and scaffold proteins. VHL-based PRTOACs have been developed for the treatment of diseases that are difficult to be dealt with by conventional methods, such as radiotherapy, chemotherapy, and small molecule inhibitors. In this review, the recent advances of VHL-based PRTOACs were summarized, and the chances and challenges associated with this area were also highlighted.


Small Molecule Libraries/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Humans , Ligands , Molecular Structure , Proteolysis/drug effects , Small Molecule Libraries/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
3.
Eur J Med Chem ; 227: 113871, 2022 Jan 05.
Article En | MEDLINE | ID: mdl-34638033

The ubiquitination of the hypoxia-inducible factor-1α (HIF-1α) is mediated by interacting with the von Hippel-Lindau protein (VHL), and is associated with cancer, chronic anemia, and ischemia. VHL, an E3 ligase, has been reported to degrade HIF-1 for decades, however, there are few successful inhibitors currently. Poor understanding of the binding pocket and a lack of in-depth exploration of the interactions between two proteins are the main reasons. Hence, we developed an effective strategy to identify and design new inhibitors for protein-protein interaction targets. The hydroxyproline (Hyp564) of HIF-1α contributed the key interaction between HIF-1α and VHL. In this study, detailed information of the binding pocket were explored by alanine scanning, site-directed mutagenesis and molecular dynamics simulations. Interestingly, we found the interaction(s) between Y565 and H110 played a key role in the binding of VHL/HIF-1α. Based on the interactions, 8 derivates of VH032, 16a-h, were synthesized by introducing various groups bounded to H110. Further assay on protein and cellular level exhibited that 16a-h accessed higher binding affinity to VHL and markable or modest improvement in stabilization of HIF-1α or HIF-1α-OH in HeLa cells. Our work provides a new orientation for the modification or design of VHL/HIF-1α protein-protein interaction inhibitors.


Drug Design , Hydroxyproline/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Cell Survival/drug effects , Dose-Response Relationship, Drug , HeLa Cells , Humans , Hydroxyproline/chemical synthesis , Hydroxyproline/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Dynamics Simulation , Molecular Structure , Protein Binding/drug effects , Structure-Activity Relationship , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
4.
J Med Chem ; 64(14): 10167-10184, 2021 07 22.
Article En | MEDLINE | ID: mdl-34196564

Traditional EZH2 inhibitors are developed to suppress the enzymatic methylation activity, and they may have therapeutic limitations due to the nonenzymatic functions of EZH2 in cancer development. Here, we report proteolysis-target chimera (PROTAC)-based EZH2 degraders to target the whole EZH2 in lymphoma. Two series of EZH2 degraders were designed and synthesized to hijack E3 ligase systems containing either von Hippel-Lindau (VHL) or cereblon (CRBN), and some VHL-based compounds were able to mediate EZH2 degradation. Two best degraders, YM181 and YM281, induced robust cell viability inhibition in diffuse large B-cell lymphoma (DLBCL) and other subtypes of lymphomas, outperforming a clinically used EZH2 inhibitor EPZ6438 (tazemetostat) that was only effective against DLBCL. The EZH2 degraders displayed promising antitumor activities in lymphoma xenografts and patient-derived primary lymphoma cells. Our study demonstrates that EZH2 degraders have better therapeutic activity than EZH2 inhibitors, which may provide a potential anticancer strategy to treat lymphoma.


Antineoplastic Agents/pharmacology , Drug Design , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Lymphoma/drug therapy , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Lymphoma/metabolism , Lymphoma/pathology , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
5.
J Biol Chem ; 297(2): 100910, 2021 08.
Article En | MEDLINE | ID: mdl-34174286

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.


Cyclopropanes/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia/metabolism , Proteomics/methods , Pyrrolidines/pharmacology , Thiazoles/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , von Hippel-Lindau Disease/pathology , Cell Line , Enzyme Inhibitors/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Binding , Protein Stability , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/metabolism
6.
Front Endocrinol (Lausanne) ; 11: 586857, 2020.
Article En | MEDLINE | ID: mdl-33329393

Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors originating from chromaffin cells in the adrenal medulla (PCCs) or extra-adrenal sympathetic or parasympathetic paraganglia (PGLs). About 40% of PPGLs result from germline mutations and therefore they are highly inheritable. Although dysfunction of any one of a panel of more than 20 genes can lead to PPGLs, mutations in genes involved in the VHL/HIF axis including PHD, VHL, HIF-2A (EPAS1), and SDHx are more frequently found in PPGLs. Multiple lines of evidence indicate that pseudohypoxia plays a crucial role in the tumorigenesis of PPGLs, and therefore PPGLs are also known as metabolic diseases. However, the interplay between VHL/HIF-mediated pseudohypoxia and metabolic disorder in PPGLs cells is not well-defined. In this review, we will first discuss the VHL/HIF axis and genetic alterations in this axis. Then, we will dissect the underlying mechanisms in VHL/HIF axis-driven PPGL pathogenesis, with special attention paid to the interplay between the VHL/HIF axis and cancer cell metabolism. Finally, we will summarize the currently available compounds/drugs targeting this axis which could be potentially used as PPGLs treatment, as well as their underlying pharmacological mechanisms. The overall goal of this review is to better understand the role of VHL/HIF axis in PPGLs development, to establish more accurate tools in PPGLs diagnosis, and to pave the road toward efficacious therapeutics against metastatic PPGLs.


Adrenal Gland Neoplasms/genetics , Apoptosis Regulatory Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Germ-Line Mutation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Pheochromocytoma/genetics , Repressor Proteins/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromaffin Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Pheochromocytoma/drug therapy , Pheochromocytoma/metabolism , Protein Kinase Inhibitors/therapeutic use , Repressor Proteins/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
7.
Life Sci ; 262: 118495, 2020 Dec 01.
Article En | MEDLINE | ID: mdl-32987061

BACKGROUND: The Von Hippel-Lindau (VHL) E3 ubiquitin ligase, which mediates its substrate hypoxia-inducible factor 1α (HIF-1α) for ubiquitination and subsequent degradation, is an attractive drug target in various diseases, such as anemia, inflammation, neurodegeneration and cancer. Proteolysis targeting chimeras (PROTACs) containing a VHL ligand that can hijack the E3 ligase activity to degrade the target protein has also been studied in academic and in industry areas recently. METHODS: Herein, by developing and optimizing the Bayesian Model, we report ensemble-based virtual screening as an effective strategy to discover potential VHL inhibitors from Specs database. RESULTS: The virtual screening protocol was developed, ten representative molecules were obtained and five compounds were selected for subsequent binding mode analysis to be potent VHL inhibitors.


Drug Discovery , Enzyme Inhibitors/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Bayes Theorem , Databases, Chemical , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
8.
Sci Rep ; 10(1): 1175, 2020 01 24.
Article En | MEDLINE | ID: mdl-31980715

Thrombospondin-1 (TSP-1) is a multifunctional matrix protein with antitumor activities due in part to its ability to inhibit angiogenesis, which in turn contributes to determine the fate of many tumours. Previous studies have shown that TSP-1 expression supports normal kidney angiostasis, and decreased TSP-1 levels contribute to the angiogenic phenotype of renal cell carcinomas (RCC). The loss of the von Hippel-Lindau tumour suppressor gene (VHL) in these tumours favours stabilization of the Hypoxia Inducible Factors (HIF), which in turn contribute to adapt tumour cells to hostile environments promoting tumour progression. However, HIF-independent regulation of certain genes might also be involved. We have previously shown that TSP-1 is regulated in hypoxia in clear cell RCC (ccRCC) in a HIF-independent manner; however, the effect of VHL protein (pVHL) on TSP-1 expression has not been evaluated. Our results proved that pVHL loss or mutation in its alpha or beta domain significantly decreased TSP-1 levels in ccRCC in a HIF-independent manner. Furthermore, this regulation proved to be important for ccRCC cells behaviour showing that decreased TSP-1 levels rendered ccRCC cells more migratory. This data substantiates a unique regulation pattern for TSP-1 in a pVHL-dependent manner, which may be relevant in the aggressiveness of ccRCC.


Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Neoplasm Proteins/physiology , Thrombospondin 1/biosynthesis , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Cell Line, Tumor , Cell Movement , Culture Media, Serum-Free , Down-Regulation , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Intercellular Junctions/metabolism , Mutation, Missense , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Protein Domains/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , Thrombospondin 1/genetics , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/genetics
9.
Carcinogenesis ; 41(6): 828-840, 2020 07 10.
Article En | MEDLINE | ID: mdl-31556451

The inactivation of the tumor suppressor gene, von Hippel-Lindau (VHL), has been identified as the earliest event in renal cell carcinoma (RCC) development. The loss of heterogeneity by chromosome 3p deletion followed by inactivating mutations on the second VHL copy are events present in close to 90% of patients. Our study illustrates a lysosomal vulnerability in VHL-inactivated RCC in vitro. By investigating the mechanism of action of the previously identified STF-62247, a small bioactive compound known for its selective cytotoxic properties towards VHL-defective models, we present the promising approach of targeting truncal-driven VHL inactivation through lysosome disruption. Furthermore, by analyzing the open platform for exploring cancer genomic data (cbioportal), we uncover the high alteration frequency of essential lysosomal and autophagic genes in sequenced biopsies from clear cell RCC patient primary tumors. By investigating lysosome physiology, we also identify VHL-inactivated cells' inability to maintain their lysosomes at the perinuclear localization in response to STF-62247-induced stress and accumulate cytoplasmic inclusion bodies in response to an inefficient lysosomal degradative capacity. Finally, by testing other known lysosomal-disrupting agents (LDAs), we show that these are selectively cytotoxic to cells lacking VHL functions. Our study builds a strong platform that could specifically link genetic clonal ccRCC evolution to lysosomal and trafficking vulnerabilities.


Autophagy , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Lysosomes/pathology , Mutation , Pyridines/pharmacology , Thiazoles/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Proliferation , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Tumor Cells, Cultured , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
10.
Prog Biophys Mol Biol ; 145: 65-77, 2019 08.
Article En | MEDLINE | ID: mdl-30528740

Renal cancer is the major cause of mortality due to abnormal functioning of von Hippel-Lindau (pVHL) and Jade Family PHD Finger 1 (JADE1) complex. E3 ubiquitin ligase JADE1 is stabilized by pVHL interaction through its plant homeodomains (PHDs). JADE1 acts as a renal tumor suppressor that promotes the ubiquitination and degradation of ß-catenin by inhibiting canonical WNT signalling. Current study focuses on the structural characterization of reported missense mutations in pVHL through in silico approaches. The predicted 3-dimensional structures of pVHLWT, pVHLY98H, pVHLY112H, pVHLL118P and pVHLR167W were subjected to binding analysis against JADE1 through molecular docking and simulation assays. In all cases, JADE1 binding was observed at the ß-domain, except pVHLL118P that exhibited binding with JADE1 through its α-domain. Our results signify that JADE1 stabilization is induced by pVHL α-domain, while ß-domain is required for JADE1 binding. pVHL binding was mediated through ß1 and ß2-strands against the concave surface of the JADE1-PHD domain. The pVHL-JADE1 complex was evaluated to scrutinize the ß-catenin-binding interface, which suggested the contribution of both α and ß-domains of pVHL in ß-catenin binding. The eleven-residue (Tyr30-Thr40) ß-catenin segment exhibited association in a bipartite manner with pVHL-JADE1 complex. The presented model depicts a pVHL-JADE1 interface for the cooperative regulation of ß-catenin binding. We propose that reduced JADE1 stabilization in case of pVHLL118P and pVHLR167W may correlate with the increased activity of ß-catenin that may lead to renal cancer through ß-catenin de-repression. Overall, ß-catenin targeting mechanism coupled with the structural knowledge of JADE1-pVHL complex will provide better understanding of renal cancer pathogenesis.


Homeodomain Proteins/chemistry , Kidney Neoplasms/metabolism , Tumor Suppressor Proteins/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/chemistry , beta Catenin/chemistry , Binding Sites , Homeodomain Proteins/metabolism , Humans , Kidney Neoplasms/etiology , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Wnt Signaling Pathway , beta Catenin/metabolism
12.
J Med Chem ; 61(2): 599-618, 2018 01 25.
Article En | MEDLINE | ID: mdl-28853884

The von Hippel-Lindau tumor suppressor protein is the substrate binding subunit of the VHL E3 ubiquitin ligase, which targets hydroxylated α subunit of hypoxia inducible factors (HIFs) for ubiquitination and subsequent proteasomal degradation. VHL is a potential target for treating anemia and ischemic diseases, motivating the development of inhibitors of the VHL:HIF-α protein-protein interaction. Additionally, bifunctional proteolysis targeting chimeras (PROTACs) containing a VHL ligand can hijack the E3 ligase activity to induce degradation of target proteins. We report the structure-guided design and group-based optimization of a series of VHL inhibitors with low nanomolar potencies and improved cellular permeability. Structure-activity relationships led to the discovery of potent inhibitors 10 and chemical probe VH298, with dissociation constants <100 nM, which induced marked HIF-1α intracellular stabilization. Our study provides new chemical tools to probe the VHL-HIF pathways and new VHL ligands for next-generation PROTACs.


Cyclopropanes/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Pyrrolidines/pharmacology , Structure-Activity Relationship , Thiazoles/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Animals , Cell Membrane Permeability/drug effects , Chemistry Techniques, Synthetic , Cyclopropanes/chemistry , Cyclopropanes/metabolism , Drug Evaluation, Preclinical/methods , Drug Stability , Enzyme Inhibitors/chemical synthesis , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Pyrrolidines/chemistry , Pyrrolidines/metabolism , Thiazoles/chemistry , Thiazoles/metabolism
13.
Sci Rep ; 7(1): 5463, 2017 07 14.
Article En | MEDLINE | ID: mdl-28710479

Inactivation of the VHL tumour suppressor gene is a highly frequent genetic event in the carcinogenesis of central nervous system-(CNS) hemangioblastomas (HBs). The patterning of the similar embryonic vasculogenesis is an increasing concern in HB-neovascularization, and the classic vascular endothelial growth factor (VEGF)-mediated angiogenesis driven by VHL loss-of-function from human endothelium have been questioned. With this regard, we identify a distinct, VHL silencing-driven mechanism in which human vascular endothelial cells by means of increasing cell proliferation and decreasing cell apoptosis, is concomitant with facilitating accumulation of Twist1 protein in vascular endothelial cells in vitro. Importantly, this molecular mechanism is also pinpointed in CNS-HBs, and associated with the process of HB-neovascularization. In contrast with recent studies of HB-neovascularization, these modified cells did not endow with the typical features of vasculogenesis, indicating that this is a common angiogenesis implementing the formation of the vascular network. Taken together, these findings suggest that vasculogenesis and angiogenesis may constitute complementary mechanisms for HB-neovascularization, and could provide a rational recognition of single anti-angiogenic intervention including targeting to the Twist1 signalling for HBs.


Cerebellar Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hemangioblastoma/genetics , Neovascularization, Pathologic/genetics , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Adolescent , Adult , Aged , Apoptosis , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/blood supply , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Female , Gene Expression Profiling , Gene Ontology , HEK293 Cells , Hemangioblastoma/blood supply , Hemangioblastoma/metabolism , Hemangioblastoma/pathology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Middle Aged , Molecular Sequence Annotation , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nuclear Proteins/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Twist-Related Protein 1/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
14.
PLoS One ; 12(6): e0178168, 2017.
Article En | MEDLINE | ID: mdl-28582447

The dioxonapthoimidazolium YM155 is a survivin suppressant which has been investigated as an anticancer agent in clinical trials. Here, we investigated its growth inhibitory properties on a panel of immortalized and patient derived renal cell carcinoma (RCC) cell lines which were either deficient in the tumour suppressor von Hippel-Lindau (VHL) protein or possessed a functional copy. Neither the VHL status nor the survivin expression levels of these cell lines influenced their susceptibility to growth inhibition by YM155. Of the various RCC lines, the papillary subtype was more resistant to YM155, suggesting that the therapeutic efficacy of YM155 may be restricted to clear cell subtypes. YM155 was equally potent in cells (RCC786.0) in which survivin expression had been stably silenced or overexpressed, implicating a limited reliance on survivin in the mode of action of YM155. A follow-up in-vitro high throughput RNA microarray identified possible targets of YM155 apart from survivin. Selected genes (ID1, FOXO1, CYLD) that were differentially expressed in YM155-sensitive RCC cells and relevant to RCC pathology were validated with real-time PCR and western immunoblotting analyses. Thus, there is corroboratory evidence that the growth inhibitory activity of YM155 in RCC cell lines is not exclusively mediated by its suppression of survivin. In view of the growing importance of combination therapy in oncology, we showed that a combination of YM155 and sorafenib at ½ x IC50 concentrations was synergistic on RCC786.0 cells. However, when tested intraperitoneally on a murine xenograft model derived from a nephrectomised patient with clear cell RCC, a combination of suboptimal doses of both drugs failed to arrest tumour progression. The absence of synergy in vivo highlighted the need to further optimize the dosing schedules of YM155 and sorafenib, as well as their routes of administration. It also implied that the expression of other oncogenic proteins which YM155 may target is either low or absent in this clear cell RCC.


Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Kidney Neoplasms/drug therapy , Naphthoquinones/pharmacology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Deubiquitinating Enzyme CYLD , Dose-Response Relationship, Drug , Drug Combinations , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mice , Mice, SCID , Niacinamide/pharmacology , Primary Cell Culture , Signal Transduction , Sorafenib , Survivin , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Xenograft Model Antitumor Assays
15.
Biochem J ; 474(7): 1127-1147, 2017 03 15.
Article En | MEDLINE | ID: mdl-28298557

Manipulation of the ubiquitin-proteasome system to achieve targeted degradation of proteins within cells using chemical tools and drugs has the potential to transform pharmacological and therapeutic approaches in cancer and other diseases. An increased understanding of the molecular mechanism of thalidomide and its analogues following their clinical use has unlocked small-molecule modulation of the substrate specificity of the E3 ligase cereblon (CRBN), which in turn has resulted in the advancement of new immunomodulatory drugs (IMiDs) into the clinic. The degradation of multiple context-specific proteins by these pleiotropic small molecules provides a means to uncover new cell biology and to generate future drug molecules against currently undruggable targets. In parallel, the development of larger bifunctional molecules that bring together highly specific protein targets in complexes with CRBN, von Hippel-Lindau, or other E3 ligases to promote ubiquitin-dependent degradation has progressed to generate selective chemical compounds with potent effects in cells and in vivo models, providing valuable tools for biological target validation and with future potential for therapeutic use. In this review, we survey recent breakthroughs achieved in these two complementary methods and the discovery of new modes of direct and indirect engagement of target proteins with the proteasome. We discuss the experimental characterisation that validates the use of molecules that promote protein degradation as chemical tools, the preclinical and clinical examples disclosed to date, and the future prospects for this exciting area of chemical biology.


Peptide Hydrolases/chemistry , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/pharmacology , Ubiquitin/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Drug Discovery , Drugs, Investigational/chemical synthesis , Drugs, Investigational/pharmacology , Gene Expression , Humans , Immunologic Factors/chemical synthesis , Immunologic Factors/pharmacology , Molecular Targeted Therapy , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemical synthesis , Proteolysis/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Substrate Specificity , Thalidomide/chemical synthesis , Thalidomide/pharmacology , Ubiquitin/genetics , Ubiquitin-Protein Ligases , Ubiquitination/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
16.
Oncotarget ; 8(9): 15085-15100, 2017 Feb 28.
Article En | MEDLINE | ID: mdl-28118605

We have recently shown that targeting Vascular Endothelial Growth Factor (VEGF) specifically in scar-infiltrating myeloid cells prevented remodeling of the sinusoidal vasculature and abrogated the resolution of murine liver fibrosis, thereby unmasking an unanticipated link between angiogenesis and resolution of fibrosis. In a gain of function approach, we wanted to test the impact of VEGF overexpression in myeloid cells on fibrolysis. We observe that genetic inactivation of the von Hippel Lindau protein (VHL), a negative regulator of Hypoxia-inducible factors (HIF) in myeloid cells, leads to increased VEGF expression and most importantly, accelerated matrix degradation and reduced myofibroblast numbers after CCl4 challenge. This is associated with enhanced expression of MMP-2 and -14 as well as lower expression of TIMP-2 in liver endothelial cells. In addition, we report increased expression of MMP-13 in scar-associated macrophages as well as improved liver regeneration upon ablation of VHL in myeloid cells. Finally, therapeutic infusion of macrophages nulli-zygous for VHL or treated with the pharmacologic hydroxylase inhibitor and HIF-inducer Dimethyloxalylglycine (DMOG) accelerates resolution of fibrosis. Hence, boosting the HIF-VEGF signaling axis in macrophages represents a promising therapeutic avenue for the treatment of liver fibrosis.


Cell Hypoxia/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Cirrhosis/prevention & control , Liver Regeneration/physiology , Myeloid Cells/physiology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Female , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Macrophages/cytology , Macrophages/metabolism , Male , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
17.
Biochim Biophys Acta Mol Cell Res ; 1864(4): 645-654, 2017 Apr.
Article En | MEDLINE | ID: mdl-28089774

Centrosome cohesion, mostly regarded as a proteinaceous linker between parental centrioles, ensures the interphase centrosome(s) to function as a single microtubule-organizing center. Maintenance of centrosome cohesion counts on a number of centrosomal linker proteins because depletion of any of those leads to premature centrosome separation in interphase, termed centrosome splitting. However, the underlying mechanisms of the dependence are unknown. Here, we show that absence of Rootletin triggers the von Hippel-Lindau tumour suppressor protein (VHL)-mediated proteasomal degradation of Cep68 and, in turn, results in centrosome splitting. The VHL E3 ligase complex ubiquitinates Cep68 in vitro and in vivo. Co-silencing of Rootletin and VHL reverts Cep68 loss and centrosome splitting. Expression of a stable mutant of Cep68, either diminishing its polyubiquitylation or eliminating binding to ß-domain of VHL, also suppresses centrosome splitting provoked by Rootletin depletion. We propose that the archetypal linker protein Rootletin maintains centrosome cohesion in part through inhibition of VHL-mediated Cep68 degradation.


Centrosome/metabolism , Cytoskeletal Proteins/genetics , Epithelial Cells/metabolism , Microtubule-Associated Proteins/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Binding Sites , Cell Line , Centrosome/ultrastructure , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/metabolism , Epithelial Cells/cytology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
18.
Nat Commun ; 7: 13312, 2016 11 04.
Article En | MEDLINE | ID: mdl-27811928

Chemical strategies to using small molecules to stimulate hypoxia inducible factors (HIFs) activity and trigger a hypoxic response under normoxic conditions, such as iron chelators and inhibitors of prolyl hydroxylase domain (PHD) enzymes, have broad-spectrum activities and off-target effects. Here we disclose VH298, a potent VHL inhibitor that stabilizes HIF-α and elicits a hypoxic response via a different mechanism, that is the blockade of the VHL:HIF-α protein-protein interaction downstream of HIF-α hydroxylation by PHD enzymes. We show that VH298 engages with high affinity and specificity with VHL as its only major cellular target, leading to selective on-target accumulation of hydroxylated HIF-α in a concentration- and time-dependent fashion in different cell lines, with subsequent upregulation of HIF-target genes at both mRNA and protein levels. VH298 represents a high-quality chemical probe of the HIF signalling cascade and an attractive starting point to the development of potential new therapeutics targeting hypoxia signalling.


Cell Hypoxia/physiology , Cyclopropanes/pharmacology , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pyrrolidines/pharmacology , Signal Transduction/drug effects , Thiazoles/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Animals , Cell Line, Tumor , Cyclopropanes/therapeutic use , Enzyme Inhibitors/therapeutic use , Humans , Hydroxylation , Mice , Primary Cell Culture , Procollagen-Proline Dioxygenase/metabolism , Pyrrolidines/therapeutic use , RNA, Messenger/metabolism , Signal Transduction/physiology , Thiazoles/therapeutic use , Up-Regulation
19.
Chem Commun (Camb) ; 52(87): 12837-12840, 2016 Oct 25.
Article En | MEDLINE | ID: mdl-27709157

We describe herein compound 1, which is similar to many known natural products, as an inhibitor of the VHL-HIF1α interaction via structure-based virtual screening. Compound 1 disrupts VHL-mediated HIF1α degradation, leading to significantly increased VEGF expression. To our knowledge, compound 1 is a member of only the second class of small molecule inhibitors of the VHL-HIF1α interaction.


High-Throughput Screening Assays/methods , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Protein Conformation/drug effects , Protein Interaction Maps/drug effects , Small Molecule Libraries/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Databases, Protein , HEK293 Cells , Humans , Molecular Docking Simulation
20.
Tumour Biol ; 37(10): 13649-13657, 2016 Oct.
Article En | MEDLINE | ID: mdl-27473082

Von Hippel-Lindau (VHL) is the most frequently mutated gene in clear cell renal carcinoma. Here, we identified a novel translational variant of VHL, termed VHLα, initiated from an alternative translational start site upstream and in frame with the ATG start codon. We showed that VHLα interacts with and regulates heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1), which consequently modulates pyruvate kinase transcript splicing and reprograms cellular glucose metabolism. Our study demonstrated that a novel VHL isoform may function as a tumor suppressor through inhibiting the Warburg effect.


Glycolysis , Neoplasms/pathology , Pyruvate Kinase/genetics , RNA Splicing/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , CRISPR-Cas Systems , Glucose/metabolism , Humans , Immunoprecipitation , Lactic Acid/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/genetics , Neoplasms/metabolism , Protein Isoforms , Sequence Homology, Nucleic Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/genetics
...