Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Commun ; 15(1): 275, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38177131

ABSTRACT

Targeted protein degradation (TPD) mediates protein level through small molecule induced redirection of E3 ligases to ubiquitinate neo-substrates and mark them for proteasomal degradation. TPD has recently emerged as a key modality in drug discovery. So far only a few ligases have been utilized for TPD. Interestingly, the workhorse ligase CRBN has been observed to be downregulated in settings of resistance to immunomodulatory inhibitory drugs (IMiDs). Here we show that the essential E3 ligase receptor DCAF1 can be harnessed for TPD utilizing a selective, non-covalent DCAF1 binder. We confirm that this binder can be functionalized into an efficient DCAF1-BRD9 PROTAC. Chemical and genetic rescue experiments validate specific degradation via the CRL4DCAF1 E3 ligase. Additionally, a dasatinib-based DCAF1 PROTAC successfully degrades cytosolic and membrane-bound tyrosine kinases. A potent and selective DCAF1-BTK-PROTAC (DBt-10) degrades BTK in cells with acquired resistance to CRBN-BTK-PROTACs while the DCAF1-BRD9 PROTAC (DBr-1) provides an alternative strategy to tackle intrinsic resistance to VHL-degrader, highlighting DCAF1-PROTACS as a promising strategy to overcome ligase mediated resistance in clinical settings.


Subject(s)
Carrier Proteins , Proteolysis Targeting Chimera , Ubiquitin-Protein Ligases , Carrier Proteins/metabolism , Proteolysis , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
ACS Med Chem Lett ; 14(12): 1631-1639, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38116426

ABSTRACT

Redirecting E3 ligases to neo-substrates, leading to their proteasomal disassembly, known as targeted protein degradation (TPD), has emerged as a promising alternative to traditional, occupancy-driven pharmacology. Although the field has expanded tremendously over the past years, the choice of E3 ligases remains limited, with an almost exclusive focus on CRBN and VHL. Here, we report the discovery of novel ligands to the PRY-SPRY domain of TRIM58, a RING ligase that is specifically expressed in erythroid precursor cells. A DSF screen, followed by validation using additional biophysical methods, led to the identification of TRIM58 ligand TRIM-473. A basic SAR around the chemotype was established by utilizing a competitive binding assay employing a short FP peptide probe derived from an endogenous TRIM58 substrate. The X-ray co-crystal structure of TRIM58 in complex with TRIM-473 gave insights into the binding mode and potential exit vectors for bifunctional degrader design.

3.
Blood Cancer J ; 12(7): 110, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35853853

ABSTRACT

Bromodomain-containing protein 9 (BRD9), an essential component of the SWI/SNF chromatin remodeling complex termed ncBAF, has been established as a therapeutic target in a subset of sarcomas and leukemias. Here, we used novel small molecule inhibitors and degraders along with RNA interference to assess the dependency on BRD9 in the context of diverse hematological malignancies, including acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), and multiple myeloma (MM) model systems. Following depletion of BRD9 protein, AML cells undergo terminal differentiation, whereas apoptosis was more prominent in ALL and MM. RNA-seq analysis of acute leukemia and MM cells revealed both unique and common signaling pathways affected by BRD9 degradation, with common pathways including those associated with regulation of inflammation, cell adhesion, DNA repair and cell cycle progression. Degradation of BRD9 potentiated the effects of several chemotherapeutic agents and targeted therapies against AML, ALL, and MM. Our findings support further development of therapeutic targeting of BRD9, alone or combined with other agents, as a novel strategy for acute leukemias and MM.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Multiple Myeloma , Transcription Factors , Antineoplastic Agents/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , RNA Interference , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Cell Chem Biol ; 29(1): 57-66.e6, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34499862

ABSTRACT

While there are hundreds of predicted E3 ligases, characterizing their applications for targeted protein degradation has proved challenging. Here, we report a chemical biology approach to evaluate the ability of modified recombinant E3 ligase components to support neo-substrate degradation. Bypassing the need for specific E3 ligase binders, we use maleimide-thiol chemistry for covalent functionalization followed by E3 electroporation (COFFEE) in live cells. We demonstrate that electroporated recombinant von Hippel-Lindau (VHL) protein, covalently functionalized at its ligandable cysteine with JQ1 or dasatinib, induces degradation of BRD4 or tyrosine kinases, respectively. Furthermore, by applying COFFEE to SPSB2, a Cullin-RING ligase 5 receptor, as well as to SKP1, the adaptor protein for Cullin-RING ligase 1 F box (SCF) complexes, we validate this method as a powerful approach to define the activity of previously uncharacterized ubiquitin ligase components, and provide further evidence that not only E3 ligase receptors but also adaptors can be directly hijacked for neo-substrate degradation.


Subject(s)
Ubiquitin-Protein Ligases/metabolism , Cell Line , Female , Humans , Male , Recombinant Proteins/metabolism
5.
Cell Chem Biol ; 28(10): 1407-1419.e6, 2021 10 21.
Article in English | MEDLINE | ID: mdl-33794192

ABSTRACT

Three limonoid natural products with selective anti-proliferative activity against BRAF(V600E) and NRAS(Q61K)-mutation-dependent melanoma cell lines were identified. Differential transcriptome analysis revealed dependency of compound activity on expression of the mitochondrial cytochrome P450 oxidase CYP27A1, a transcriptional target of melanogenesis-associated transcription factor (MITF). We determined that CYP27A1 activity is necessary for the generation of a reactive metabolite that proceeds to inhibit cellular proliferation. A genome-wide small interfering RNA screen in combination with chemical proteomics experiments revealed gene-drug functional epistasis, suggesting that these compounds target mitochondrial biogenesis and inhibit tumor bioenergetics through a covalent mechanism. Our work suggests a strategy for melanoma-specific targeting by exploiting the expression of MITF target gene CYP27A1 and inhibiting mitochondrial oxidative phosphorylation in BRAF mutant melanomas.


Subject(s)
Cholestanetriol 26-Monooxygenase/metabolism , Limonins/pharmacology , Mitochondria/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Biological Products/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cholestanetriol 26-Monooxygenase/antagonists & inhibitors , Cholestanetriol 26-Monooxygenase/genetics , Humans , Limonins/chemistry , Limonins/metabolism , Limonins/therapeutic use , Melanoma/drug therapy , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA Interference , RNA, Small Interfering/metabolism
6.
Sci Rep ; 10(1): 20044, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33208877

ABSTRACT

MYC oncoprotein is a multifunctional transcription factor that regulates the expression of a large number of genes involved in cellular growth, proliferation and metabolism. Altered MYC protein level lead to cellular transformation and tumorigenesis. MYC is deregulated in > 50% of human cancers, rendering it an attractive drug target. However, direct inhibition of this class of proteins using conventional small molecules is challenging due to their intrinsically disordered state. To discover novel posttranslational regulators of MYC protein stability and turnover, we established a genetic screen in mammalian cells by combining a fluorescent protein-based MYC abundance sensor, CRISPR/Cas9-based gene knockouts and next-generation sequencing. Our screen identifies UBR5, an E3 ligase of the HECT-type family, as a novel regulator of MYC degradation. Even in the presence of the well-described and functional MYC ligase, FBXW7, UBR5 depletion leads to accumulation of MYC in cells. We demonstrate interaction of UBR5 with MYC and reduced K48-linked ubiquitination of MYC upon loss of UBR5 in cells. Interestingly, in cancer cell lines with amplified MYC expression, depletion of UBR5 resulted in reduced cell survival, as a consequence of MYC stabilization. Finally, we show that MYC and UBR5 are co-amplified in more than 40% of cancer cells and that MYC copy number amplification correlates with enhanced transcriptional output of UBR5. This suggests that UBR5 acts as a buffer in MYC amplified settings and protects these cells from apoptosis.


Subject(s)
CRISPR-Cas Systems , Neoplasms/pathology , Proteolysis , Proto-Oncogene Proteins c-myc/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Apoptosis , Humans , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/genetics
8.
Nat Chem Biol ; 16(1): 50-59, 2020 01.
Article in English | MEDLINE | ID: mdl-31819276

ABSTRACT

The post-genomic era has seen many advances in our understanding of cancer pathways, yet resistance and tumor heterogeneity necessitate multiple approaches to target even monogenic tumors. Here, we combine phenotypic screening with chemical genetics to identify pre-messenger RNA endonuclease cleavage and polyadenylation specificity factor 3 (CPSF3) as the target of JTE-607, a small molecule with previously unknown target. We show that CPSF3 represents a synthetic lethal node in a subset of acute myeloid leukemia (AML) and Ewing's sarcoma cancer cell lines. Inhibition of CPSF3 by JTE-607 alters expression of known downstream effectors in AML and Ewing's sarcoma lines, upregulates apoptosis and causes tumor-selective stasis in mouse xenografts. Mechanistically, it prevents the release of newly synthesized pre-mRNAs, resulting in read-through transcription and the formation of DNA-RNA hybrid R-loop structures. This study implicates pre-mRNA processing, and specifically CPSF3, as a druggable target providing an avenue to therapeutic intervention in cancer.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/metabolism , Leukemia, Myeloid, Acute/metabolism , RNA Precursors/metabolism , Sarcoma, Ewing/metabolism , Animals , Apoptosis/drug effects , Binding Sites , Carboxylic Ester Hydrolases/metabolism , Cell Line, Tumor , Cell Survival , Cleavage And Polyadenylation Specificity Factor/genetics , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/drug therapy , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Phenotype , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Piperazines/pharmacology , Protein Binding , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sarcoma, Ewing/drug therapy
9.
PLoS Comput Biol ; 15(12): e1007403, 2019 12.
Article in English | MEDLINE | ID: mdl-31860671

ABSTRACT

Computational approaches have shown promise in contextualizing genes of interest with known molecular interactions. In this work, we evaluate seventeen previously published algorithms based on characteristics of their output and their performance in three tasks: cross validation, prediction of drug targets, and behavior with random input. Our work highlights strengths and weaknesses of each algorithm and results in a recommendation of algorithms best suited for performing different tasks.


Subject(s)
Algorithms , Gene Regulatory Networks , Models, Genetic , Benchmarking , Computational Biology , Databases, Genetic/statistics & numerical data , Databases, Protein/statistics & numerical data , Humans , Protein Interaction Maps/genetics
10.
Circulation ; 137(12): 1270-1277, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29222138

ABSTRACT

BACKGROUND: Emerging proteomic technologies using novel affinity-based reagents allow for efficient multiplexing with high-sample throughput. To identify early biomarkers of myocardial injury, we recently applied an aptamer-based proteomic profiling platform that measures 1129 proteins to samples from patients undergoing septal alcohol ablation for hypertrophic cardiomyopathy, a human model of planned myocardial injury. Here, we examined the scalability of this approach using a markedly expanded platform to study a far broader range of human proteins in the context of myocardial injury. METHODS: We applied a highly multiplexed, expanded proteomic technique that uses single-stranded DNA aptamers to assay 4783 human proteins (4137 distinct human gene targets) to derivation and validation cohorts of planned myocardial injury, individuals with spontaneous myocardial infarction, and at-risk controls. RESULTS: We found 376 target proteins that significantly changed in the blood after planned myocardial injury in a derivation cohort (n=20; P<1.05E-05, 1-way repeated measures analysis of variance, Bonferroni threshold). Two hundred forty-seven of these proteins were validated in an independent planned myocardial injury cohort (n=15; P<1.33E-04, 1-way repeated measures analysis of variance); >90% were directionally consistent and reached nominal significance in the validation cohort. Among the validated proteins that were increased within 1 hour after planned myocardial injury, 29 were also elevated in patients with spontaneous myocardial infarction (n=63; P<6.17E-04). Many of the novel markers identified in our study are intracellular proteins not previously identified in the peripheral circulation or have functional roles relevant to myocardial injury. For example, the cardiac LIM protein, cysteine- and glycine-rich protein 3, is thought to mediate cardiac mechanotransduction and stress responses, whereas the mitochondrial ATP synthase F0 subunit component is a vasoactive peptide on its release from cells. Last, we performed aptamer-affinity enrichment coupled with mass spectrometry to technically verify aptamer specificity for a subset of the new biomarkers. CONCLUSIONS: Our results demonstrate the feasibility of large-scale aptamer multiplexing at a level that has not previously been reported and with sample throughput that greatly exceeds other existing proteomic methods. The expanded aptamer-based proteomic platform provides a unique opportunity for biomarker and pathway discovery after myocardial injury.


Subject(s)
Aptamers, Nucleotide , Blood Proteins/metabolism , Cardiomyopathy, Hypertrophic/blood , Myocardium/metabolism , Proteomics/methods , ST Elevation Myocardial Infarction/blood , Ablation Techniques , Biomarkers/blood , Blood Proteins/genetics , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/surgery , Case-Control Studies , Feasibility Studies , High-Throughput Screening Assays , Humans , Myocardium/pathology , Predictive Value of Tests , Prognosis , Reproducibility of Results , ST Elevation Myocardial Infarction/genetics , ST Elevation Myocardial Infarction/pathology , Time Factors
11.
J Clin Invest ; 127(12): 4554-4568, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29130934

ABSTRACT

Transcriptional repression of ubiquitin B (UBB) is a cancer-subtype-specific alteration that occurs in a substantial population of patients with cancers of the female reproductive tract. UBB is 1 of 2 genes encoding for ubiquitin as a polyprotein consisting of multiple copies of ubiquitin monomers. Silencing of UBB reduces cellular UBB levels and results in an exquisite dependence on ubiquitin C (UBC), the second polyubiquitin gene. UBB is repressed in approximately 30% of high-grade serous ovarian cancer (HGSOC) patients and is a recurrent lesion in uterine carcinosarcoma and endometrial carcinoma. We identified ovarian tumor cell lines that retain UBB in a repressed state, used these cell lines to establish orthotopic ovarian tumors, and found that inducible expression of a UBC-targeting shRNA led to tumor regression, and substantial long-term survival benefit. Thus, we describe a recurrent cancer-specific lesion at the level of ubiquitin production. Moreover, these observations reveal the prognostic value of UBB repression and establish UBC as a promising therapeutic target for ovarian cancer patients with recurrent UBB silencing.


Subject(s)
Gene Silencing , Neoplasm Proteins/biosynthesis , Ovarian Neoplasms/metabolism , Ubiquitin C/biosynthesis , Ubiquitin/biosynthesis , Cell Line, Tumor , Female , Humans , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Ubiquitin/genetics , Ubiquitin C/genetics
12.
Sci Signal ; 8(376): ra44, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25969542

ABSTRACT

Vascular smooth muscle cells (VSMCs) undergo transcriptionally regulated reversible differentiation in growing and injured blood vessels. This dedifferentiation also contributes to VSMC hyperplasia after vascular injury, including that caused by angioplasty and stenting. Stents provide mechanical support and can contain and release rapamycin, an inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1). Rapamycin suppresses VSMC hyperplasia and promotes VSMC differentiation. We report that rapamycin-induced differentiation of VSMCs required the transcription factor GATA-6. Inhibition of mTORC1 stabilized GATA-6 and promoted the nuclear accumulation of GATA-6, its binding to DNA, its transactivation of promoters encoding contractile proteins, and its inhibition of proliferation. These effects were mediated by phosphorylation of GATA-6 at Ser(290), potentially by Akt2, a kinase that is activated in VSMCs when mTORC1 is inhibited. Rapamycin induced phosphorylation of GATA-6 in wild-type mice, but not in Akt2(-/-) mice. Intimal hyperplasia after arterial injury was greater in Akt2(-/-) mice than in wild-type mice, and the exacerbated response in Akt2(-/-) mice was rescued to a greater extent by local overexpression of the wild-type or phosphomimetic (S290D) mutant GATA-6 than by that of the phosphorylation-deficient (S290A) mutant. Our data indicated that GATA-6 and Akt2 are involved in the mTORC1-mediated regulation of VSMC proliferation and differentiation. Identifying the downstream transcriptional targets of mTORC1 may provide cell type-specific drug targets to combat cardiovascular diseases associated with excessive proliferation of VSMCs.


Subject(s)
Cell Differentiation/physiology , GATA6 Transcription Factor/metabolism , Multiprotein Complexes/metabolism , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Proliferation/physiology , GATA6 Transcription Factor/genetics , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics
13.
Circulation ; 129(15): 1598-609, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24474649

ABSTRACT

BACKGROUND: Platelet abnormalities are well-recognized complications of diabetes mellitus. Mitochondria play a central role in platelet metabolism and activation. Mitochondrial dysfunction is evident in diabetes mellitus. The molecular pathway for hyperglycemia-induced mitochondrial dysfunction in platelets in diabetes mellitus is unknown. METHODS AND RESULTS: Using both human and humanized mouse models, we report that hyperglycemia-induced aldose reductase activation and subsequent reactive oxygen species production lead to increased p53 phosphorylation (Ser15), which promotes mitochondrial dysfunction, damage, and rupture by sequestration of the antiapoptotic protein Bcl-xL. In a glucose dose-dependent manner, severe mitochondrial damage leads to loss of mitochondrial membrane potential and platelet apoptosis (cytochrome c release, caspase 3 activation, and phosphatidylserine exposure). Although platelet hyperactivation, mitochondrial dysfunction, aldose reductase activation, reactive oxygen species production, and p53 phosphorylation are all induced by hyperglycemia, we demonstrate that platelet apoptosis and hyperactivation are 2 distinct states that depend on the severity of the hyperglycemia and mitochondrial damage. Combined, both lead to increased thrombus formation in a mouse blood stasis model. CONCLUSIONS: Aldose reductase contributes to diabetes-mediated mitochondrial dysfunction and damage through the activation of p53. The degree of mitochondrial dysfunction and damage determines whether hyperactivity (mild damage) or apoptosis (severe damage) will ensue. These signaling components provide novel therapeutic targets for thrombotic complications in diabetes mellitus.


Subject(s)
Aldehyde Reductase/metabolism , Blood Platelets/metabolism , Diabetes Mellitus, Type 2/metabolism , Mitochondrial Diseases/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Animals , Apoptosis/physiology , Blood Platelets/pathology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitochondrial Diseases/pathology , Phosphorylation/physiology , Signal Transduction/physiology , Thrombosis/metabolism , Thrombosis/pathology , bcl-X Protein/metabolism
14.
PLoS One ; 8(6): e67314, 2013.
Article in English | MEDLINE | ID: mdl-23840660

ABSTRACT

Thromboxane and its receptor have emerged as key players in modulating vascular thrombotic events. Thus, a dysfunctional hTP genetic variant may protect against (hypoactivity) or promote (hyperactivity) vascular events, based upon its activity on platelets. After extensive in silico analysis, six hTP-α variants were selected (C(68)S, V(80)E, E(94)V, A(160)T, V(176)E, and V(217)I) for detailed biochemical studies based on structural proximity to key regions involved in receptor function and in silico predictions. Variant biochemical profiles ranged from severe instability (C(68)S) to normal (V(217)I), with most variants demonstrating functional alteration in binding, expression or activation (V(80)E, E(94)V, A(160)T, and V(176)E). In the absence of patient platelet samples, we developed and validated a novel megakaryocyte based system to evaluate human platelet function in the presence of detected dysfunctional genetic variants. Interestingly, variant V80E exhibited reduced platelet activation whereas A160T demonstrated platelet hyperactivity. This report provides the most comprehensive in silico, in vitro and "in platelet" evaluation of hTP variants to date and highlightscurrent inherent problems in evaluating genetic variants, with possible solutions. The study additionally provides clinical relevance to characterized dysfunctional hTP variants.


Subject(s)
Blood Platelets/metabolism , Polymorphism, Single Nucleotide , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Amino Acid Sequence , Amino Acid Substitution , Aspirin/pharmacology , Binding Sites , Binding, Competitive , Blood Platelets/drug effects , Cell Line , Cyclooxygenase Inhibitors/pharmacology , Genetic Association Studies , Humans , Models, Molecular , Molecular Sequence Data , Phosphoproteins/metabolism , Platelet Activation/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary , Proteome/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/chemistry , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Signal Transduction , Thromboxanes/physiology
15.
Circ Res ; 113(1): 22-31, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23603510

ABSTRACT

RATIONALE: The peptide ligand apelin and its receptor APJ constitute a signaling pathway with numerous effects on the cardiovascular system, including cardiovascular development in model organisms such as xenopus and zebrafish. OBJECTIVE: This study aimed to characterize the embryonic lethal phenotype of the Apj-/- mice and to define the involved downstream signaling targets. METHODS AND RESULTS: We report the first characterization of the embryonic lethality of the Apj-/- mice. More than half of the expected Apj-/- embryos died in utero because of cardiovascular developmental defects. Those succumbing to early embryonic death had markedly deformed vasculature of the yolk sac and the embryo, as well as poorly looped hearts with aberrantly formed right ventricles and defective atrioventricular cushion formation. Apj-/- embryos surviving to later stages demonstrated incomplete vascular maturation because of a deficiency of vascular smooth muscle cells and impaired myocardial trabeculation and ventricular wall development. The molecular mechanism implicates a novel, noncanonical signaling pathway downstream of apelin-APJ involving Gα13, which induces histone deacetylase (HDAC) 4 and HDAC5 phosphorylation and cytoplasmic translocation, resulting in activation of myocyte enhancer factor 2. Apj-/- mice have greater endocardial Hdac4 and Hdac5 nuclear localization and reduced expression of the myocyte enhancer factor 2 (MEF2) transcriptional target Krüppel-like factor 2. We identify a number of commonly shared transcriptional targets among apelin-APJ, Gα13, and MEF2 in endothelial cells, which are significantly decreased in the Apj-/- embryos and endothelial cells. CONCLUSIONS: Our results demonstrate a novel role for apelin-APJ signaling as a potent regulator of endothelial MEF2 function in the developing cardiovascular system.


Subject(s)
Cardiovascular Abnormalities/embryology , Cardiovascular System/embryology , Intercellular Signaling Peptides and Proteins/physiology , Myogenic Regulatory Factors/physiology , Receptors, G-Protein-Coupled/physiology , Active Transport, Cell Nucleus , Adipokines , Animals , Apelin , Apelin Receptors , Cardiovascular Abnormalities/genetics , Endocardium/embryology , Endocardium/metabolism , Endothelium, Vascular/metabolism , Female , Fetal Heart/abnormalities , GTP-Binding Protein alpha Subunits, G12-G13/physiology , Gene Expression Regulation, Developmental , Genes, Lethal , Histone Deacetylases/metabolism , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , MEF2 Transcription Factors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Processing, Post-Translational , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Transcription, Genetic
16.
J Lipid Res ; 54(6): 1678-1690, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23493750

ABSTRACT

Thromboxane A2 (TXA2) contributes to cardiovascular disease (CVD) by activating platelets and vascular constriction and proliferation. Despite their preclinical efficacy, pharmacological antagonists of the TXA2 receptor (TP), a G protein-coupled receptor, have not been clinically successful, raising interest in novel approaches to modifying TP function. We determined that disruption of a GxxxGxxxL helical interaction motif in the human TP's (α isoform) fifth transmembrane (TM) domain suppressed TP agonist-induced Gq signaling and TPα homodimerization, but not its cell surface expression, ligand affinity, or Gq association. Heterodimerization of TPα with the functionally opposing prostacyclin receptor (IP) shifts TPα to signal via the IP-Gs cascade contributing to prostacyclin's restraint of TXA2 function. Interestingly, disruption of the TPα-TM5 GxxxGxxxL motif did not modify either IP-TPα heterodimerization or its Gs-cAMP signaling. Our study indicates that distinct regions of the TPα receptor direct its homo- and heterodimerization and that homodimerization is necessary for normal TPα-Gq activation. Targeting the TPα-TM5 GxxxGxxxL domain may allow development of biased TPα homodimer antagonists that avoid suppression of IP-TPα heterodimer function. Such novel therapeutics may prove superior in CVD compared with nonselective suppression of all TP functions with TXA2 biosynthesis inhibitors or TP antagonists.


Subject(s)
Protein Multimerization/physiology , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane/metabolism , Second Messenger Systems/physiology , Amino Acid Motifs , Cyclic AMP/genetics , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Protein Structure, Tertiary , Receptors, Epoprostenol , Receptors, Prostaglandin/genetics , Receptors, Thromboxane/genetics
17.
Mol Cell Biol ; 33(2): 184-93, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23109431

ABSTRACT

G protein-coupled receptors (GPCRs) interact with heterotrimeric G proteins and initiate a wide variety of signaling pathways. The molecular nature of GPCR-G protein interactions in the clinically important thromboxane A2 (TxA(2)) receptor (TP) and prostacyclin (PGI(2)) receptor (IP) is poorly understood. The TP activates its cognate G protein (Gαq) in response to the binding of thromboxane, while the IP signals through Gαs in response to the binding of prostacyclin. Here, we utilized a combination of approaches consisting of chimeric receptors, molecular modeling, and site-directed mutagenesis to precisely study the specificity of G protein coupling. Multiple chimeric receptors were constructed by replacing the TP intracellular loops (ICLs) with the ICL regions of the IP. Our results demonstrate that both the sequences and lengths of ICL2 and ICL3 influenced G protein specificity. Importantly, we identified a precise ICL region on the prostanoid receptors TP and IP that can switch G protein specificities. The validities of the chimeric technique and the derived molecular model were confirmed by introducing clinically relevant naturally occurring mutations (R60L in the TP and R212C in the IP). Our findings provide new molecular insights into prostanoid receptor-G protein interactions, which are of general significance for understanding the structural basis of G protein activation by GPCRs in basic health and cardiovascular disease.


Subject(s)
GTP-Binding Proteins/genetics , Receptors, Epoprostenol/chemistry , Receptors, Thromboxane A2, Prostaglandin H2/chemistry , Amino Acid Sequence , Binding Sites , Calcium/analysis , Fluorescent Antibody Technique , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate/analysis , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nonlinear Dynamics , Protein Binding , Protein Conformation , Receptors, Epoprostenol/genetics , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Signal Transduction
18.
J Am Coll Cardiol ; 60(15): 1402-9, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22981548

ABSTRACT

OBJECTIVES: This study sought to determine hospital variation in the use of positive inotropic agents in patients with heart failure. BACKGROUND: Clinical guidelines recommend targeted use of positive inotropic agents in highly selected patients, but data are limited and the recommendations are not specific. METHODS: We analyzed data from 376 hospitals including 189,948 hospitalizations for heart failure from 2009 through 2010. We used hierarchical logistic regression models to estimate hospital-level risk-standardized rates of inotrope use and risk-standardized in-hospital mortality rates. RESULTS: The risk-standardized rates of inotrope use ranged across hospitals from 0.9% to 44.6% (median: 6.3%, interquartile range: 4.3% to 9.2%). We identified various hospital patterns based on the type of agents: dobutamine-predominant (29% of hospitals), dopamine-predominant (25%), milrinone-predominant (1%), mixed dobutamine and dopamine pattern (32%), and mixed pattern including all 3 agents (13%). When studying the factors associated with interhospital variation, the best model performance was with the hierarchical generalized linear models that adjusted for patient case mix and an individual hospital effect (receiver operating characteristic curves from 0.77 to 0.88). The intraclass correlation coefficients of the hierarchical generalized linear models (0.113 for any inotrope) indicated that a noteworthy proportion of the observed variation was related to an individual institutional effect. Hospital rates or patterns of use were not associated with differences in length of stay or risk-standardized mortality rates. CONCLUSIONS: We found marked differences in the use of inotropic agents for heart failure patients among a diverse group of hospitals. This variability, occurring in the context of little clinical evidence, indicates an urgent need to define the appropriate use of these medications.


Subject(s)
Cardiotonic Agents/therapeutic use , Heart Failure/drug therapy , Hospitals/standards , Cross-Sectional Studies , Heart Failure/mortality , Hospital Mortality/trends , Humans , Length of Stay/trends , Retrospective Studies , Survival Rate/trends , United States/epidemiology
19.
Cell Mol Life Sci ; 69(20): 3361-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22491820

ABSTRACT

Cardiovascular disease is the foremost cause of morbidity and mortality in the Western world. Atherosclerosis followed by thrombosis (atherothrombosis) is the pathological process underlying most myocardial, cerebral, and peripheral vascular events. Atherothrombosis is a complex and heterogeneous inflammatory process that involves interactions between many cell types (including vascular smooth muscle cells, endothelial cells, macrophages, and platelets) and processes (including migration, proliferation, and activation). Despite a wealth of knowledge from many recent studies using knockout mouse and human genetic studies (GWAS and candidate approach) identifying genes and proteins directly involved in these processes, traditional cardiovascular risk factors (hyperlipidemia, hypertension, smoking, diabetes mellitus, sex, and age) remain the most useful predictor of disease. Eicosanoids (20 carbon polyunsaturated fatty acid derivatives of arachidonic acid and other essential fatty acids) are emerging as important regulators of cardiovascular disease processes. Drugs indirectly modulating these signals, including COX-1/COX-2 inhibitors, have proven to play major roles in the atherothrombotic process. However, the complexity of their roles and regulation by opposing eicosanoid signaling, have contributed to the lack of therapies directed at the eicosanoid receptors themselves. This is likely to change, as our understanding of the structure, signaling, and function of the eicosanoid receptors improves. Indeed, a major advance is emerging from the characterization of dysfunctional naturally occurring mutations of the eicosanoid receptors. In light of the proven and continuing importance of risk factors, we have elected to focus on the relationship between eicosanoids and cardiovascular risk factors.


Subject(s)
Atherosclerosis/drug therapy , Eicosanoids/therapeutic use , Thrombosis/drug therapy , Animals , Humans , Mice , Risk Factors
20.
PLoS One ; 7(1): e29996, 2012.
Article in English | MEDLINE | ID: mdl-22272267

ABSTRACT

The human thromboxane A2 receptor (TP), belongs to the prostanoid subfamily of Class A GPCRs and mediates vasoconstriction and promotes thrombosis on binding to thromboxane (TXA2). In Class A GPCRs, transmembrane (TM) helix 4 appears to be a hot spot for non-synonymous single nucleotide polymorphic (nsSNP) variants. Interestingly, A160T is a novel nsSNP variant with unknown structure and function. Additionally, within this helix in TP, Ala160(4.53) is highly conserved as is Gly164(4.57). Here we target Ala160(4.53) and Gly164(4.57) in the TP for detailed structure-function analysis. Amino acid replacements with smaller residues, A160S and G164A mutants, were tolerated, while bulkier beta-branched replacements, A160T and A160V showed a significant decrease in receptor expression (Bmax). The nsSNP variant A160T displayed significant agonist-independent activity (constitutive activity). Guided by molecular modeling, a series of compensatory mutations were made on TM3, in order to accommodate the bulkier replacements on TM4. The A160V/F115A double mutant showed a moderate increase in expression level compared to either A160V or F115A single mutants. Thermal activity assays showed decrease in receptor stability in the order, wild type>A160S>A160V>A160T>G164A, with G164A being the least stable. Our study reveals that Ala160(4.53) and Gly164(4.57) in the TP play critical structural roles in packing of TM3 and TM4 helices. Naturally occurring mutations in conjunction with site-directed replacements can serve as powerful tools in assessing the importance of regional helix-helix interactions.


Subject(s)
Mutation, Missense , Protein Structure, Secondary , Receptors, Thromboxane A2, Prostaglandin H2/chemistry , Receptors, Thromboxane A2, Prostaglandin H2/genetics , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Binding, Competitive , Bridged Bicyclo Compounds, Heterocyclic , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Fatty Acids, Unsaturated , Glycine/chemistry , Glycine/genetics , Glycine/metabolism , HEK293 Cells , Humans , Hydrazines/metabolism , Hydrazines/pharmacology , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Polymorphism, Single Nucleotide , Radioligand Assay , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Structure-Activity Relationship , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...