Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Blood Adv ; 8(15): 3880-3892, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38739710

ABSTRACT

ABSTRACT: Provirus integration site for Moloney murine leukemia virus (PIM) family serine/threonine kinases perform protumorigenic functions in hematologic malignancies and solid tumors by phosphorylating substrates involved in tumor metabolism, cell survival, metastasis, inflammation, and immune cell invasion. However, a comprehensive understanding of PIM kinase functions is currently lacking. Multiple small-molecule PIM kinase inhibitors are currently being evaluated as cotherapeutics in patients with cancer. To further illuminate PIM kinase functions in cancer, we deeply profiled PIM1 substrates using the reverse in-gel kinase assay to identify downstream cellular processes targetable with small molecules. Pathway analyses of putative PIM substrates nominated RNA splicing and ribosomal RNA (rRNA) processing as PIM-regulated cellular processes. PIM inhibition elicited reproducible splicing changes in PIM-inhibitor-responsive acute myeloid leukemia (AML) cell lines. PIM inhibitors synergized with splicing modulators targeting splicing factor 3b subunit 1 (SF3B1) and serine-arginine protein kinase 1 (SRPK1) to kill AML cells. PIM inhibition also altered rRNA processing, and PIM inhibitors synergized with an RNA polymerase I inhibitor to kill AML cells and block AML tumor growth. These data demonstrate that deep kinase substrate knowledge can illuminate unappreciated kinase functions, nominating synergistic cotherapeutic strategies. This approach may expand the cotherapeutic armamentarium to overcome kinase inhibitor-resistant disease that limits durable responses in malignant disease.


Subject(s)
Leukemia, Myeloid, Acute , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-pim-1 , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Humans , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mice , Animals , Cell Line, Tumor , Substrate Specificity , RNA Splicing/drug effects , Xenograft Model Antitumor Assays
2.
Cardiovasc Res ; 119(3): 857-866, 2023 05 02.
Article in English | MEDLINE | ID: mdl-35727948

ABSTRACT

AIMS: The present study aims to characterize the genetic risk architecture of bicuspid aortic valve (BAV) disease, the most common congenital heart defect. METHODS AND RESULTS: We carried out a genome-wide association study (GWAS) including 2236 BAV patients and 11 604 controls. This led to the identification of a new risk locus for BAV on chromosome 3q29. The single nucleotide polymorphism rs2550262 was genome-wide significant BAV associated (P = 3.49 × 10-08) and was replicated in an independent case-control sample. The risk locus encodes a deleterious missense variant in MUC4 (p.Ala4821Ser), a gene that is involved in epithelial-to-mesenchymal transformation. Mechanistical studies in zebrafish revealed that loss of Muc4 led to a delay in cardiac valvular development suggesting that loss of MUC4 may also play a role in aortic valve malformation. The GWAS also confirmed previously reported BAV risk loci at PALMD (P = 3.97 × 10-16), GATA4 (P = 1.61 × 10-09), and TEX41 (P = 7.68 × 10-04). In addition, the genetic BAV architecture was examined beyond the single-marker level revealing that a substantial fraction of BAV heritability is polygenic and ∼20% of the observed heritability can be explained by our GWAS data. Furthermore, we used the largest human single-cell atlas for foetal gene expression and show that the transcriptome profile in endothelial cells is a major source contributing to BAV pathology. CONCLUSION: Our study provides a deeper understanding of the genetic risk architecture of BAV formation on the single marker and polygenic level.


Subject(s)
Bicuspid Aortic Valve Disease , Heart Valve Diseases , Animals , Humans , Bicuspid Aortic Valve Disease/metabolism , Bicuspid Aortic Valve Disease/pathology , Aortic Valve/pathology , Heart Valve Diseases/pathology , Genome-Wide Association Study , Zebrafish/genetics , Endothelial Cells/metabolism
3.
Br J Pharmacol ; 179(21): 4958-4973, 2022 11.
Article in English | MEDLINE | ID: mdl-35802072

ABSTRACT

BACKGROUND AND PURPOSE: Vascular tone is regulated by the relative contractile state of vascular smooth muscle cells (VSMCs). Several integrins directly modulate VSMC contraction by regulating calcium influx through L-type voltage-gated Ca2+ channels (VGCCs). Genetic variants in ITGA9, which encodes the α9 subunit of integrin α9ß1, and SVEP1, a ligand for integrin α9ß1, associate with elevated blood pressure; however, neither SVEP1 nor integrin α9ß1 has reported roles in vasoregulation. We determined whether SVEP1 and integrin α9ß1 can regulate VSMC contraction. EXPERIMENTAL APPROACH: SVEP1 and integrin binding were confirmed by immunoprecipitation and cell binding assays. Human induced pluripotent stem cell-derived VSMCs were used in in vitro [Ca2+ ]i studies, and aortas from a Svep1+/- knockout mouse model were used in wire myography to measure vessel contraction. KEY RESULTS: We confirmed the ligation of SVEP1 to integrin α9ß1 and additionally found SVEP1 to directly bind to integrin α4ß1. Inhibition of SVEP1, integrin α4ß1 or α9ß1 significantly enhanced [Ca2+ ]i levels in isolated VSMCs to Gαq/11 -vasoconstrictors. This response was confirmed in whole vessels where a greater contraction to U46619 was seen in vessels from Svep1+/- mice compared to littermate controls or when integrin α4ß1 or α9ß1 was inhibited. Inhibition studies suggested that this effect was mediated via VGCCs, PKC and Rho A/Rho kinase dependent mechanisms. CONCLUSIONS AND IMPLICATIONS: Our studies reveal a novel role for SVEP1 and the integrins α4ß1 and α9ß1 in reducing VSMC contractility. This could provide an explanation for the genetic associations with blood pressure risk at the SVEP1 and ITGA9 loci.


Subject(s)
Induced Pluripotent Stem Cells , Integrin alpha4beta1 , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Calcium/metabolism , Cell Adhesion Molecules , Humans , Integrins/genetics , Integrins/metabolism , Ligands , Mice , Vasoconstriction , Vasoconstrictor Agents , rho-Associated Kinases
4.
Eur J Heart Fail ; 24(6): 1009-1019, 2022 06.
Article in English | MEDLINE | ID: mdl-35570197

ABSTRACT

AIMS: Chronic heart failure (CHF) is a systemic syndrome with a poor prognosis and a need for novel therapies. We investigated whether whole blood transcriptomic profiling can provide new mechanistic insights into cardiovascular (CV) mortality in CHF. METHODS AND RESULTS: Transcriptome profiles were generated at baseline from 944 CHF patients from the BIOSTAT-CHF study, of whom 626 survived and 318 died from a CV cause during a follow-up of 21 months. Multivariable analysis, including adjustment for cell count, identified 1153 genes (6.5%) that were differentially expressed between those that survived or died and strongly related to a validated clinical risk score for adverse prognosis. The differentially expressed genes mainly belonged to five non-redundant pathways: adaptive immune response, proteasome-mediated ubiquitin-dependent protein catabolic process, T-cell co-stimulation, positive regulation of T-cell proliferation, and erythrocyte development. These five pathways were selectively related (RV coefficients >0.20) with seven circulating protein biomarkers of CV mortality (fibroblast growth factor 23, soluble ST2, adrenomedullin, hepcidin, pentraxin-3, WAP 4-disulfide core domain 2, and interleukin-6) revealing an intricate relationship between immune and iron homeostasis. The pattern of survival-associated gene expression matched with 29 perturbagen-induced transcriptome signatures in the iLINCS drug-repurposing database, identifying drugs, approved for other clinical indications, that were able to reverse in vitro the molecular changes associated with adverse prognosis in CHF. CONCLUSION: Systematic modelling of the whole blood protein-coding transcriptome defined molecular pathways that provide a link between clinical risk factors and adverse CV prognosis in CHF, identifying both established and new potential therapeutic targets.


Subject(s)
Heart Failure , Biomarkers , Chronic Disease , Humans , Prognosis , Transcriptome
5.
Proc Natl Acad Sci U S A ; 119(16): e2117857119, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35412907

ABSTRACT

The RB1 gene is frequently mutated in human cancers but its role in tumorigenesis remains incompletely defined. Using an induced pluripotent stem cell (iPSC) model of hereditary retinoblastoma (RB), we report that the spliceosome is an up-regulated target responding to oncogenic stress in RB1-mutant cells. By investigating transcriptomes and genome occupancies in RB iPSC­derived osteoblasts (OBs), we discover that both E2F3a, which mediates spliceosomal gene expression, and pRB, which antagonizes E2F3a, coregulate more than one-third of spliceosomal genes by cobinding to their promoters or enhancers. Pharmacological inhibition of the spliceosome in RB1-mutant cells leads to global intron retention, decreased cell proliferation, and impaired tumorigenesis. Tumor specimen studies and genome-wide TCGA (The Cancer Genome Atlas) expression profile analyses support the clinical relevance of pRB and E2F3a in modulating spliceosomal gene expression in multiple cancer types including osteosarcoma (OS). High levels of pRB/E2F3a­regulated spliceosomal genes are associated with poor OS patient survival. Collectively, these findings reveal an undiscovered connection between pRB, E2F3a, the spliceosome, and tumorigenesis, pointing to the spliceosomal machinery as a potentially widespread therapeutic vulnerability of pRB-deficient cancers.


Subject(s)
Bone Neoplasms , Carcinogenesis , E2F3 Transcription Factor , Gene Expression Regulation, Neoplastic , Induced Pluripotent Stem Cells , Osteosarcoma , Retinoblastoma Binding Proteins , Spliceosomes , Ubiquitin-Protein Ligases , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Carcinogenesis/genetics , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/metabolism , Genes, Retinoblastoma , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation , Osteosarcoma/genetics , Osteosarcoma/pathology , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
6.
Nucleic Acids Res ; 49(3): 1609-1618, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33469660

ABSTRACT

The endonuclease activity within the influenza virus cap-snatching process is a proven therapeutic target. The anti-influenza drug baloxavir is highly effective, but is associated with resistance mutations that threaten its clinical efficacy. The endonuclease resides within the N-terminal domain of the PA subunit (PAN) of the influenza RNA dependent RNA polymerase, and we report here complexes of PAN with RNA and DNA oligonucleotides to understand its specificity and the structural basis of baloxavir resistance mutations. The RNA and DNA oligonucleotides bind within the substrate binding groove of PAN in a similar fashion, explaining the ability of the enzyme to cleave both substrates. The individual nucleotides occupy adjacent conserved pockets that flank the two-metal active site. However, the 2' OH of the RNA ribose moieties engage in additional interactions that appear to optimize the binding and cleavage efficiency for the natural substrate. The major baloxavir resistance mutation at position 38 is at the core of the substrate binding site, but structural studies and modeling suggest that it maintains the necessary virus fitness via compensating interactions with RNA. These studies will facilitate the development of new influenza therapeutics that spatially match the substrate and are less likely to elicit resistance mutations.


Subject(s)
Endoribonucleases/chemistry , Influenza A Virus, H1N1 Subtype/enzymology , Viral Proteins/chemistry , Antiviral Agents/chemistry , DNA/chemistry , Dibenzothiepins/chemistry , Endoribonucleases/metabolism , Models, Molecular , Morpholines/chemistry , Pyridones/chemistry , RNA/chemistry , Substrate Specificity , Triazines/chemistry , Viral Proteins/metabolism
8.
PLoS One ; 15(5): e0233672, 2020.
Article in English | MEDLINE | ID: mdl-32469945

ABSTRACT

Agents that modulate pre-mRNA splicing are of interest in multiple therapeutic areas, including cancer. We report our recent screening results with the application of a cell-based Triple Exon Skipping Luciferase Reporter (TESLR) using a library that is composed of FDA approved drugs, clinical compounds, and mechanistically characterized tool compounds. Confirmatory assays showed that three clinical antitumor therapeutic candidates (milciclib, PF-3758309 and PF-562271) are potent splicing modulators and that these drugs are, in fact, nanomolar inhibitors of multiple kinases involved in the regulation the spliceosome. We also report the identification of new SF3B1 antagonists (sudemycinol C and E) and show that these antagonists can be used to develop a displacement assay for SF3B1 small molecule ligands. These results further support the broad potential for the development of agents that target the spliceosome for the treatment of cancer and other diseases, as well as new avenues for the discovery of new chemotherapeutic agents for a range of diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Exons/drug effects , RNA Precursors/genetics , RNA Splicing/drug effects , Cell Line, Tumor , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology
9.
J Am Heart Assoc ; 9(4): e014333, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32067586

ABSTRACT

Background Genome-wide association studies have shown an association between the single-nucleotide polymorphism rs17514846 on chromosome 15q26.1 and coronary artery disease susceptibility. The underlying biological mechanism is, however, not fully understood. rs17514846 is located in the FES Upstream Region (FURIN) gene, which is expressed in vascular endothelial cells (ECs). We investigated whether rs17514846 has an influence on FURIN expression in ECs and whether FURIN affects EC behavior. Methods and Results Quantitative reverse transcription-polymerase chain reaction analysis showed that cultured vascular ECs from individuals carrying the coronary artery disease risk allele of rs17514846 had higher FURIN expression than cells from noncarriers. In support, luciferase reporter analyses in ECs indicated that the risk allele had higher transcriptional activity than the nonrisk allele. Electrophoretic mobility shift assays using EC nuclear protein extracts detected a DNA-protein complex with allele-specific differential binding of a nuclear protein. Knockdown of FURIN in ECs reduced endothelin-1 secretion, nuclear factor-κB activity, vascular cell adhesion molecule-1, and MCP1 (monocyte chemotactic protein-1) expression and monocyte-endothelial adhesion and transmigration. A population-based study showed an association of the rs17514846 risk allele with higher circulating MCP1 levels and greater carotid intima-media thickness. Conclusions The coronary artery disease risk variant at the 15q26.1 locus modulates FURIN expression in vascular ECs. FURIN levels in ECs affect monocyte-endothelial adhesion and migration.


Subject(s)
Coronary Artery Disease/genetics , Endothelial Cells/metabolism , Furin/genetics , Monocytes/physiology , Polymorphism, Single Nucleotide/genetics , Transendothelial and Transepithelial Migration/physiology , Adult , Aged , Carotid Intima-Media Thickness , Chemokine CCL2/metabolism , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Endothelium, Vascular/pathology , Female , Furin/metabolism , Humans , Male , Middle Aged , Vascular Cell Adhesion Molecule-1/metabolism
10.
Circ Genom Precis Med ; 12(1): e002196, 2019 01.
Article in English | MEDLINE | ID: mdl-30645167

ABSTRACT

BACKGROUND: Although short-term trials have suggested that PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors are safe and reduce risk of cardiovascular diseases, their long-term safety is unclear. Genetic variants associated with lower activity of a gene can act as proxies to identify potential long-term side effects of drugs as recently exemplified by association of LDL (low-density lipoprotein)-lowering variants in the HMGCR (target for statins) and PCSK9 genes with increased risk of type 2 diabetes mellitus (T2DM). However, analyses of the full spectrum of potential side effects of PCSK9 inhibition using a genetic approach have not been undertaken. METHODS: We examined the association of an LDL-lowering variant in the PCSK9 gene (T allele of rs1159147), as well as 2 LDL-lowering HGCMR variants (G allele of rs17238484 and T allele of rs12916) with 80 diseases and traits in up to 479 522 individuals in UK Biobank. RESULTS: The PCSK9 T allele was significantly (Bonferroni P<6.25×10-4) associated with risk of T2DM, increased body mass index, waist circumference, waist-hip ratio, diastolic blood pressure, type 1 diabetes mellitus, and insulin use. The HMGCR variants were also associated with risk of T2DM, although their previously reported associations with anthropometric traits were found to be confounded. Mediation analysis suggested that the association of the PCSK9 T allele with risk of T2DM but not diastolic blood pressure was largely independent of its association with body mass index and central obesity. Nominally significant associations of the PCSK9 T allele were also seen with peptic ulcer disease, depression, asthma, chronic kidney disease, and venous thromboembolism. CONCLUSIONS: Our findings support previous genetic analyses suggesting that long-term use of PCSK9 inhibitors, like statins, may be associated with increased risk of T2DM. Some other potential side effects need to be looked for in future studies of PCSK9 inhibitors, although we did not find signals that raise substantial concerns about their long-term safety.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Anticholesteremic Agents/adverse effects , Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 2/etiology , Genetic Variation , PCSK9 Inhibitors , Proprotein Convertase 9/genetics , Antibodies, Monoclonal/adverse effects , Cardiovascular Diseases/pathology , Case-Control Studies , Cholesterol, LDL/blood , Diabetes Mellitus, Type 2/pathology , Female , Humans , Male , Middle Aged
11.
mBio ; 9(6)2018 11 06.
Article in English | MEDLINE | ID: mdl-30401776

ABSTRACT

The main obstacle to an HIV cure is the transcriptionally inert proviruses that persist in resting CD4 T cells and other reservoirs. None of the current approaches has significantly reduced the size of the viral reservoir. Hence, alternative approaches, such as permanent blocking of viral transcription, to achieve a sustained remission, need urgent attention. To identify cellular factors that may be important for this approach, we sought for host targets that when altered could block HIV transcription and reactivation. Here, we identified splicing factor 3B subunit 1 (SF3B1) as a critical HIV dependency factor required for viral replication. SF3B1 is a splicing factor involved in directing chromatin and nascent gene transcripts to appropriate splice sites. Inhibitors of SF3B1 are currently in development for cancer and have been found to be nontoxic to normal cells compared to malignant cells. Knockdown of SF3B1 abrogated HIV replication in all cell types tested. SF3B1 interacted with viral protein Tat in vitro and in vivo Genetic or pharmacologic inhibition of SF3B1 prevented Tat-mediated HIV transcription and RNA polymerase II association with the HIV promoter. In addition, an inhibitor of SF3B1 prevented HIV reactivation from latency irrespective of the latency-reversing agent used. The data show that SF3B1 is involved in viral transcription and reactivation from latency and may serve as a therapeutic target in the HIV cure efforts.IMPORTANCE The reason why HIV cannot be cured by current therapy is because of viral persistence in resting T cells. One approach to permanent HIV remission that has received less attention is the so-called "block and lock" approach. The idea behind this approach is that the virus could be permanently disabled in patients if viral genome or surrounding chromatin could be altered to silence the virus, thus enabling patients to stop therapy. In this work, we have identified splicing factor 3B subunit 1 (SF3B1) as a potential target for this approach. SF3B1 interacts with the viral protein Tat, which is critical for viral transcription. Inhibition of SF3B1 prevents HIV transcription and reactivation from latency. Since there are preclinical inhibitors for this protein, our findings could pave the way to silence HIV transcription, potentially leading to prolonged or permanent remission.


Subject(s)
HIV-1/physiology , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Virus Activation , Virus Latency , tat Gene Products, Human Immunodeficiency Virus/metabolism , Chromatin/genetics , Cyclohexylamines/pharmacology , HEK293 Cells , HIV-1/genetics , Humans , Jurkat Cells , Macrophages/drug effects , Macrophages/virology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , RNA Splicing Factors/antagonists & inhibitors , RNA Splicing Factors/metabolism , RNA, Small Interfering , Spiro Compounds/pharmacology , THP-1 Cells , Transcription, Genetic/drug effects , Virus Replication/drug effects , tat Gene Products, Human Immunodeficiency Virus/genetics
13.
J Am Coll Cardiol ; 72(16): 1883-1893, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30309464

ABSTRACT

BACKGROUND: Coronary artery disease (CAD) has substantial heritability and a polygenic architecture. However, the potential of genomic risk scores to help predict CAD outcomes has not been evaluated comprehensively, because available studies have involved limited genomic scope and limited sample sizes. OBJECTIVES: This study sought to construct a genomic risk score for CAD and to estimate its potential as a screening tool for primary prevention. METHODS: Using a meta-analytic approach to combine large-scale, genome-wide, and targeted genetic association data, we developed a new genomic risk score for CAD (metaGRS) consisting of 1.7 million genetic variants. We externally tested metaGRS, both by itself and in combination with available data on conventional risk factors, in 22,242 CAD cases and 460,387 noncases from the UK Biobank. RESULTS: The hazard ratio (HR) for CAD was 1.71 (95% confidence interval [CI]: 1.68 to 1.73) per SD increase in metaGRS, an association larger than any other externally tested genetic risk score previously published. The metaGRS stratified individuals into significantly different life course trajectories of CAD risk, with those in the top 20% of metaGRS distribution having an HR of 4.17 (95% CI: 3.97 to 4.38) compared with those in the bottom 20%. The corresponding HR was 2.83 (95% CI: 2.61 to 3.07) among individuals on lipid-lowering or antihypertensive medications. The metaGRS had a higher C-index (C = 0.623; 95% CI: 0.615 to 0.631) for incident CAD than any of 6 conventional factors (smoking, diabetes, hypertension, body mass index, self-reported high cholesterol, and family history). For men in the top 20% of metaGRS with >2 conventional factors, 10% cumulative risk of CAD was reached by 48 years of age. CONCLUSIONS: The genomic score developed and evaluated here substantially advances the concept of using genomic information to stratify individuals with different trajectories of CAD risk and highlights the potential for genomic screening in early life to complement conventional risk prediction.


Subject(s)
Coronary Artery Disease , Genomics , Primary Prevention/methods , Risk Assessment/methods , Coronary Artery Disease/diagnosis , Coronary Artery Disease/epidemiology , Coronary Artery Disease/genetics , Female , Genome-Wide Association Study , Genomics/methods , Genomics/statistics & numerical data , Humans , Male , Mass Screening/methods , Middle Aged , Multifactorial Inheritance , Predictive Value of Tests , Research Design , Risk Factors , United Kingdom/epidemiology
14.
RNA ; 24(8): 1056-1066, 2018 08.
Article in English | MEDLINE | ID: mdl-29844105

ABSTRACT

The recent identification of compounds that interact with the spliceosome (sudemycins, spliceostatin A, and meayamycin) indicates that these molecules modulate aberrant splicing via SF3B1 inhibition. Through whole transcriptome sequencing, we have demonstrated that treatment of Rh18 cells with sudemycin leads to exon skipping as the predominant aberrant splicing event. This was also observed following reanalysis of published RNA-seq data sets derived from HeLa cells after spliceostatin A exposure. These results are in contrast to previous reports that indicate that intron retention was the major consequence of SF3B1 inhibition. Analysis of the exon junctions up-regulated by these small molecules indicated that these sequences were absent in annotated human genes, suggesting that aberrant splicing events yielded novel RNA transcripts. Interestingly, the length of preferred downstream exons was significantly longer than the skipped exons, although there was no difference between the lengths of introns flanking skipped exons. The reading frame of the aberrantly skipped exons maintained a ratio of 2:1:1, close to that of the cassette exons (3:1:1) present in naturally occurring isoforms, suggesting negative selection by the nonsense-mediated decay (NMD) machinery for out-of-frame transcripts. Accordingly, genes involved in NMD and RNAs encoding proteins involved in the splicing process were enriched in both data sets. Our findings, therefore, further elucidate the mechanisms by which SF3B1 inhibition modulates pre-mRNA splicing.


Subject(s)
Epoxy Compounds/pharmacology , Exons/genetics , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Protein Biosynthesis/genetics , RNA Splicing Factors/antagonists & inhibitors , RNA Splicing Factors/genetics , RNA Splicing/genetics , Spiro Compounds/pharmacology , Spliceosomes/genetics , Base Sequence , Cell Line, Tumor , HCT116 Cells , HeLa Cells , Humans , Nonsense Mediated mRNA Decay/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reading Frames/genetics , Sequence Analysis, RNA , Transcriptome/genetics
15.
ACS Med Chem Lett ; 9(12): 1153-1155, 2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30613316

ABSTRACT

Improvements in the efficiency of the drug discovery process are needed in order to deliver life-saving medications to patients in a more cost-effective manner. While there are many reasons that the efficiency of this process has not gotten better, this Viewpoint proposes that the lack of integration of the three major disciplines (discovery, development, and clinical trials) plays a significant role in the ongoing high rate of failure in clinical trials for innovative drugs. Several specific proposals are made that may help to provide more integration, so that the gears of the human-driven drug discovery machine may mesh better in the future.

16.
Sci Rep ; 7(1): 17139, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215062

ABSTRACT

Influenza is a serious hazard to human health that causes hundreds of thousands of deaths annually. Though vaccines and current therapeutics can blunt some of the perilous impact of this viral infection, new treatments are needed due to the constantly evolving nature of this virus. Recently, our growing understanding of an essential influenza viral protein, PA, has led to the development of focused libraries of new small molecules that specifically target the active site of the PA influenza endonuclease, which we report here. Our overarching approach has been to proactively develop lead inhibitors that are less likely to rapidly develop clinical resistance by optimizing inhibitors that retain activity against induced resistant mutants. Here, we report details behind the discovery of new potent inhibitors of wild type and resistant mutant endonucleases along with their high-resolution co-crystal structure-activity relationships. These results add to our understanding of nuclease protein targets and potentially serve as starting points for a new therapeutic approach to the treatment of influenza.


Subject(s)
Antiviral Agents/pharmacology , Endoribonucleases/chemistry , Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/enzymology , Influenza, Human/drug therapy , RNA-Dependent RNA Polymerase/chemistry , Viral Proteins/chemistry , Amides/chemistry , Antiviral Agents/chemistry , Catalytic Domain , Cell Proliferation/drug effects , Crystallography, X-Ray , Endoribonucleases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/enzymology , Influenza, Human/virology , Molecular Structure , Protein Conformation , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Structure-Activity Relationship , Viral Proteins/antagonists & inhibitors
17.
Nat Commun ; 8(1): 1910, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29208933

ABSTRACT

Schistosomiasis is a debilitating tropical disease caused by infection with parasitic blood flukes. Approximately 260 million people are infected worldwide, underscoring the clinical and socioeconomic impact of this chronic infection. Schistosomiasis is treated with the drug praziquantel (PZQ), which has proved the therapeutic mainstay for over three decades of clinical use. However, the molecular target(s) of PZQ remain undefined. Here we identify a molecular target for the antischistosomal eutomer - (R)-PZQ - which functions as a partial agonist of the human serotoninergic 5HT2B receptor. (R)-PZQ modulation of serotoninergic signaling occurs over a concentration range sufficient to regulate vascular tone of the mesenteric blood vessels where the adult parasites reside within their host. These data establish (R)-PZQ as a G-protein-coupled receptor ligand and suggest that the efficacy of this clinically important anthelmintic is supported by a broad, cross species polypharmacology with PZQ modulating signaling events in both host and parasite.


Subject(s)
Anthelmintics/metabolism , Mesenteric Arteries/drug effects , Mesenteric Veins/drug effects , Praziquantel/metabolism , Schistosoma mansoni/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacokinetics , Vasoconstriction/drug effects , Animals , Anthelmintics/pharmacology , Cell Line , Computer Simulation , Drug Partial Agonism , Female , Humans , Mice , Myography , Praziquantel/pharmacology , Receptor, Serotonin, 5-HT2B/drug effects , Receptor, Serotonin, 5-HT2B/metabolism , Schistosomiasis mansoni/drug therapy , Serotonin 5-HT2 Receptor Agonists/pharmacology
18.
Biomark Insights ; 12: 1177271917730557, 2017.
Article in English | MEDLINE | ID: mdl-28932105

ABSTRACT

OBJECTIVE: The aim of the study was to define pharmacodynamic markers for sudemycin D6, an experimental cancer drug that changes alternative splicing in human blood. METHODS: Blood samples from 12 donors were incubated with sudemycin D6 for up to 24 hours, and at several time points total RNA from lymphocytes was prepared and the pre-messenger RNA (mRNA) splicing patterns were analyzed with reverse transcription-polymerase chain reaction. RESULTS: Similar to immortalized cells, blood lymphocytes change alternative splicing due to sudemycin D6 treatment. However, lymphocytes in blood respond slower than immortalized cultured cells. CONCLUSIONS: Exon skipping in the DUSP11 and SRRM1 pre-mRNAs are pharmacodynamic markers for sudemycin D6 treatment and show effects beginning at 9 hours after treatment.

19.
J Am Coll Cardiol ; 69(7): 823-836, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28209224

ABSTRACT

BACKGROUND: Genome-wide association studies have so far identified 56 loci associated with risk of coronary artery disease (CAD). Many CAD loci show pleiotropy; that is, they are also associated with other diseases or traits. OBJECTIVES: This study sought to systematically test if genetic variants identified for non-CAD diseases/traits also associate with CAD and to undertake a comprehensive analysis of the extent of pleiotropy of all CAD loci. METHODS: In discovery analyses involving 42,335 CAD cases and 78,240 control subjects we tested the association of 29,383 common (minor allele frequency >5%) single nucleotide polymorphisms available on the exome array, which included a substantial proportion of known or suspected single nucleotide polymorphisms associated with common diseases or traits as of 2011. Suggestive association signals were replicated in an additional 30,533 cases and 42,530 control subjects. To evaluate pleiotropy, we tested CAD loci for association with cardiovascular risk factors (lipid traits, blood pressure phenotypes, body mass index, diabetes, and smoking behavior), as well as with other diseases/traits through interrogation of currently available genome-wide association study catalogs. RESULTS: We identified 6 new loci associated with CAD at genome-wide significance: on 2q37 (KCNJ13-GIGYF2), 6p21 (C2), 11p15 (MRVI1-CTR9), 12q13 (LRP1), 12q24 (SCARB1), and 16q13 (CETP). Risk allele frequencies ranged from 0.15 to 0.86, and odds ratio per copy of the risk allele ranged from 1.04 to 1.09. Of 62 new and known CAD loci, 24 (38.7%) showed statistical association with a traditional cardiovascular risk factor, with some showing multiple associations, and 29 (47%) showed associations at p < 1 × 10-4 with a range of other diseases/traits. CONCLUSIONS: We identified 6 loci associated with CAD at genome-wide significance. Several CAD loci show substantial pleiotropy, which may help us understand the mechanisms by which these loci affect CAD risk.


Subject(s)
Coronary Artery Disease/genetics , Genetic Loci , Genetic Pleiotropy , Case-Control Studies , Coronary Artery Disease/epidemiology , Female , Gene Frequency , Genome-Wide Association Study , Humans , Male , Odds Ratio , Polymorphism, Single Nucleotide
20.
J Biol Chem ; 292(10): 4164-4175, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28154181

ABSTRACT

KRAS is the most frequently mutated oncogene in human cancer, but its therapeutic targeting remains challenging. Here, we report a synthetic lethal screen with a library of deubiquitinases and identify USP39, which encodes an essential splicing factor, as a critical gene for the viability of KRAS-dependent cells. We show that splicing fidelity inhibitors decrease preferentially the proliferation rate of KRAS-active cells. Moreover, depletion of DHX38, encoding an USP39-interacting splicing factor, also reduces the viability of these cells. In agreement with these results, USP39 depletion caused a significant reduction in pre-mRNA splicing efficiency, as demonstrated through RNA-seq experiments. Furthermore, we show that USP39 is up-regulated in lung and colon carcinomas and its expression correlates with KRAS levels and poor clinical outcome. Accordingly, our work provides critical information for the development of splicing-directed antitumor treatments and supports the potential of USP39-targeting strategies as the basis of new anticancer therapies.


Subject(s)
Colonic Neoplasms/pathology , Lung Neoplasms/pathology , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Ubiquitin-Specific Proteases/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Ubiquitin-Specific Proteases/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL