Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nucleic Acids Res ; 47(10): 5038-5048, 2019 06 04.
Article in English | MEDLINE | ID: mdl-30916320

ABSTRACT

PRMT5 is an arginine methyltransferase that accounts for the vast majority of the symmetric methylation in cells. PRMT5 exerts its function when complexed with MEP50/WDR77. This activity is often elevated in cancer cells and correlates with poor prognosis, making PRMT5 a therapeutic target. To investigate the PRMT5 signaling pathway and to identify genes whose loss-of-function sensitizes cancer cells to PRMT5 inhibition, we performed a CRISPR/Cas9 genetic screen in the presence of a PRMT5 inhibitor. We identified known components of the PRMT5 writer/reader pathway including PRMT5 itself, MEP50/WDR77, PPP4C, SMNDC1 and SRSF3. Interestingly, loss of PRMT1, the major asymmetric arginine methyltransferase, also sensitizes cells to PRMT5 inhibition. We investigated the interplay between PRMT5 and PRMT1, and found that combinatorial inhibitor treatment of small cell lung cancer and pancreatic cancer cell models have a synergistic effect. Furthermore, MTAP-deleted cells, which harbor an attenuated PRMT5-MEP50 signaling pathway, are generally more sensitive to PRMT1 inhibition. Together, these findings demonstrate that there is a degree of redundancy between the PRMT5 and PRMT1 pathways, even though these two enzymes deposit different types of arginine methylation marks. Targeting this redundancy provides a vulnerability for tumors carrying a co-deletion of MTAP and the adjacent CDKN2A tumor suppressor gene.


Subject(s)
Gene Deletion , Neoplasms/enzymology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , A549 Cells , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Ethylenediamines/pharmacology , Humans , Isoquinolines/pharmacology , MCF-7 Cells , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Pyrimidines/pharmacology , Pyrroles/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
2.
Biochem Biophys Res Commun ; 483(1): 203-208, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28034750

ABSTRACT

Computing the ligand-protein binding affinity (or the Gibbs free energy) with chemical accuracy has long been a challenge for which many methods/approaches have been developed and refined with various successful applications. False positives and, even more harmful, false negatives have been and still are a common occurrence in practical applications. Inevitable in all approaches are the errors in the force field parameters we obtain from quantum mechanical computation and/or empirical fittings for the intra- and inter-molecular interactions. These errors propagate to the final results of the computed binding affinities even if we were able to perfectly implement the statistical mechanics of all the processes relevant to a given problem. And they are actually amplified to various degrees even in the mature, sophisticated computational approaches. In particular, the free energy perturbation (alchemical) approaches amplify the errors in the force field parameters because they rely on extracting the small differences between similarly large numbers. In this paper, we develop a hybrid steered molecular dynamics (hSMD) approach to the difficult binding problems of a ligand buried deep inside a protein. Sampling the transition along a physical (not alchemical) dissociation path of opening up the binding cavity---pulling out the ligand---closing back the cavity, we can avoid the problem of error amplifications by not relying on small differences between similar numbers. We tested this new form of hSMD on retinol inside cellular retinol-binding protein 1 and three cases of a ligand (a benzylacetate, a 2-nitrothiophene, and a benzene) inside a T4 lysozyme L99A/M102Q(H) double mutant. In all cases, we obtained binding free energies in close agreement with the experimentally measured values. This indicates that the force field parameters we employed are accurate and that hSMD (a brute force, unsophisticated approach) is free from the problem of error amplification suffered by many sophisticated approaches in the literature.


Subject(s)
Molecular Dynamics Simulation , Proteins/chemistry , Proteins/metabolism , Benzene/chemistry , Benzene/metabolism , Benzyl Compounds/chemistry , Benzyl Compounds/metabolism , Binding Sites , Ligands , Muramidase/chemistry , Muramidase/genetics , Muramidase/metabolism , Mutation , Protein Conformation , Retinol-Binding Proteins, Cellular/chemistry , Retinol-Binding Proteins, Cellular/metabolism , Thiophenes/chemistry , Thiophenes/metabolism , Vitamin A/chemistry , Vitamin A/metabolism
3.
Phys Chem Chem Phys ; 17(5): 3680-8, 2015 Feb 07.
Article in English | MEDLINE | ID: mdl-25556346

ABSTRACT

In order to determine how functionalized gold nanoparticles (AuNPs) interact in a near-physiological environment, we performed all-atom molecular dynamics simulations on the icosahedral Au144 nanoparticles each coated with a homogeneous set of 60 thiolates selected from one of these five (5) types: 11-mercapto-1-undecanesulfonate -SC11H22(SO3(-)), 5-mercapto-1-pentanesulfonate -SC5H10(SO3(-)), 5-mercapto-1-pentaneamine -SC5H10(NH3(+)), 4-mercapto-benzoate -SPh(COO(-)), or 4-mercapto-benzamide -SPh(CONH3(+)). These thiolates were selected to elucidate how the aggregation behavior of AuNPs depends on ligand parameters, including the charge of the terminal group (anionic vs. cationic), and its length and conformational flexibility. For this purpose, each functionalized AuNP was paired with a copy of itself, placed in an aqueous cell, neutralized by 120 Na(+)/Cl(-) counter-ions and salinated with a 150 mM concentration of NaCl, to form five (5) systems of like-charged AuNPs pairs in a saline. We computed the potential of mean force (the reversible work of separation) as a function of the intra-pair distance and, based on which, the aggregation affinities. We found that the AuNPs coated with negatively charged, short ligands have very high affinities. Structurally, a significant number of Na(+) counter-ions reside on a plane between the AuNPs, mediating the interaction. Each such ion forms a "salt bridge" (or "ionic bonds") to both of the AuNPs when they are separated by its diameter plus 0.2-0.3 nm. The positively charged AuNPs have much weaker affinities, as Cl(-) counter-ions form fewer and weaker salt bridges between the AuNPs. In the case of Au144(SC11H22(SO3(-)))60 pair, the flexible ligands fluctuate much more than the other four cases. The large fluctuations disfavor the forming of salt bridges between two AuNPs, but enable hydrophobic contact between the exposed hydrocarbon chains of the two AuNPs, which are subject to an effective attraction at a separation much greater than the AuNP diameter and involve a higher concentration of counter ions in the inter-pair space.


Subject(s)
Gold/chemistry , Ligands , Metal Nanoparticles/chemistry , Hydrogen-Ion Concentration , Ions/chemistry , Molecular Dynamics Simulation , Sodium Chloride/chemistry , Sulfhydryl Compounds/chemistry , Thermodynamics
4.
Elife ; 122023 06 07.
Article in English | MEDLINE | ID: mdl-37285284

ABSTRACT

In skeletal muscle, muscle stem cells (MuSC) are the main cells responsible for regeneration upon injury. In diseased skeletal muscle, it would be therapeutically advantageous to replace defective MuSCs, or rejuvenate them with drugs to enhance their self-renewal and ensure long-term regenerative potential. One limitation of the replacement approach has been the inability to efficiently expand MuSCs ex vivo, while maintaining their stemness and engraftment abilities. Herein, we show that inhibition of type I protein arginine methyltransferases (PRMTs) with MS023 increases the proliferative capacity of ex vivo cultured MuSCs. Single cell RNA sequencing (scRNAseq) of ex vivo cultured MuSCs revealed the emergence of subpopulations in MS023-treated cells which are defined by elevated Pax7 expression and markers of MuSC quiescence, both features of enhanced self-renewal. Furthermore, the scRNAseq identified MS023-specific subpopulations to be metabolically altered with upregulated glycolysis and oxidative phosphorylation (OxPhos). Transplantation of MuSCs treated with MS023 had a better ability to repopulate the MuSC niche and contributed efficiently to muscle regeneration following injury. Interestingly, the preclinical mouse model of Duchenne muscular dystrophy had increased grip strength with MS023 treatment. Our findings show that inhibition of type I PRMTs increased the proliferation capabilities of MuSCs with altered cellular metabolism, while maintaining their stem-like properties such as self-renewal and engraftment potential.


Subject(s)
Muscular Dystrophy, Duchenne , Satellite Cells, Skeletal Muscle , Animals , Mice , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Cells, Cultured , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/metabolism , Protein-Arginine N-Methyltransferases/metabolism
5.
Cell Rep ; 38(13): 110582, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35354055

ABSTRACT

Despite the success of immune checkpoint inhibitor (ICI) therapy for cancer, resistance and relapse are frequent. Combination therapies are expected to enhance response rates and overcome this resistance. Herein, we report that combining PRMT7 inhibition with ICI therapy induces a strong anti-tumor T cell immunity and restrains tumor growth in vivo by increasing immune cell infiltration. PRMT7-deficient B16.F10 melanoma exhibits increased expression of genes in the interferon pathway, antigen presentation, and chemokine signaling. PRMT7 deficiency or inhibition with SGC3027 in B16.F10 melanoma results in reduced DNMT expression, loss of DNA methylation in the regulatory regions of endogenous retroviral elements (ERVs) causing their increased expression. PRMT7-deficient cells increase RIG-I and MDA5 expression with a reduction in the H4R3me2s repressive histone mark at their gene promoters. Our findings identify PRMT7 as a regulatory checkpoint for RIG-I, MDA5, and their ERV-double-stranded RNA (dsRNA) ligands, facilitating immune escape and anti-tumor T cell immunity to restrain tumor growth.


Subject(s)
Endogenous Retroviruses , Melanoma, Experimental , Animals , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Interferons/genetics , Melanoma, Experimental/genetics , Neoplasm Recurrence, Local/genetics
6.
Nat Commun ; 13(1): 474, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35078987

ABSTRACT

The specificity of CRISPR/Cas9 genome editing is largely determined by the sequences of guide RNA (gRNA) and the targeted DNA, yet the sequence-dependent rules underlying off-target effects are not fully understood. To systematically explore the sequence determinants governing CRISPR/Cas9 specificity, here we describe a dual-target system to measure the relative cleavage rate between off- and on-target sequences (off-on ratios) of 1902 gRNAs on 13,314 synthetic target sequences, and reveal a set of sequence rules involving 2 factors in off-targeting: 1) a guide-intrinsic mismatch tolerance (GMT) independent of the mismatch context; 2) an "epistasis-like" combinatorial effect of multiple mismatches, which are associated with the free-energy landscape in R-loop formation and are explainable by a multi-state kinetic model. These sequence rules lead to the development of MOFF, a model-based predictor of Cas9-mediated off-target effects. Moreover, the "epistasis-like" combinatorial effect suggests a strategy of allele-specific genome editing using mismatched guides. With the aid of MOFF prediction, this strategy significantly improves the selectivity and expands the application domain of Cas9-based allele-specific editing, as tested in a high-throughput allele-editing screen on 18 cancer hotspot mutations.


Subject(s)
Base Sequence/genetics , CRISPR-Cas Systems , Gene Editing/methods , Mutation , Neoplasms/therapy , RNA, Guide, Kinetoplastida/chemistry , Cell Line , Humans , Neoplasms/genetics , Neoplasms/pathology , RNA, Guide, Kinetoplastida/genetics
7.
Cell Rep ; 36(2): 109337, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260915

ABSTRACT

RNA-binding proteins play important roles in X-linked intellectual disability (XLID). In this study, we investigate the contribution of the XLID-associated RBMX in neuronal differentiation. We show that RBMX-depleted cells exhibit aberrant activation of the p53 pathway. Moreover, we identify that the RBMX RGG/RG motif is methylated by protein arginine methyltransferase 5 (PRMT5), and this regulates assembly with the SRSF1 splicing factor into higher-order complexes. Depletion of RBMX or disruption of the RBMX/SRSF1 complex in PRMT5-depleted cells reduces SRSF1 binding to the MDM4 precursor (pre-)mRNA, leading to exon 6 exclusion and lower MDM4 protein levels. Transcriptomic analysis of isogenic Shashi-XLID human-induced pluripotent stem cells (hiPSCs) generated using CRISPR-Cas9 reveals a dysregulation of MDM4 splicing and aberrant p53 upregulation. Shashi-XLID neural progenitor cells (NPCs) display differentiation and morphological abnormalities accompanied with excessive apoptosis. Our findings identify RBMX as a regulator of SRSF1 and the p53 pathway, suggesting that the loss of function of the RBMX RGG/RG motif is the cause of Shashi-XLID syndrome.


Subject(s)
Cell Differentiation , Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Mental Retardation, X-Linked/pathology , Neurons/metabolism , Neurons/pathology , Sequence Deletion , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Alternative Splicing/genetics , Amino Acid Motifs , Arginine/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Methylation , Neural Stem Cells/metabolism , Neurogenesis , Protein Binding , Protein Stability , Protein-Arginine N-Methyltransferases/metabolism , Proto-Oncogene Proteins/genetics , RNA/metabolism , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism
8.
Life Sci Alliance ; 3(10)2020 10.
Article in English | MEDLINE | ID: mdl-32747416

ABSTRACT

DDX5, XRN2, and PRMT5 have been shown to resolve DNA/RNA hybrids (R-loops) at RNA polymerase II transcription termination sites at few genomic loci. Herein, we perform genome-wide R-loop mapping using classical DNA/RNA immunoprecipitation and high-throughput sequencing (DRIP-seq) of loci regulated by DDX5, XRN2, and PRMT5. We observed hundreds to thousands of R-loop gains and losses at transcribed loci in DDX5-, XRN2-, and PRMT5-deficient U2OS cells. R-loop gains were characteristic of highly transcribed genes located at gene-rich regions, whereas R-loop losses were observed in low-density gene areas. DDX5, XRN2, and PRMT5 shared many R-loop gain loci at transcription termination sites, consistent with their coordinated role in RNA polymerase II transcription termination. DDX5-depleted cells had unique R-loop gain peaks near the transcription start site that did not overlap with those of siXRN2 and siPRMT5 cells, suggesting a role for DDX5 in transcription initiation independent of XRN2 and PRMT5. Moreover, we observed that the accumulated R-loops at certain loci in siDDX5, siXRN2, and siPRMT5 cells near the transcription start site of genes led to antisense intergenic transcription. Our findings define unique and shared roles of DDX5, XRN2, and PRMT5 in DNA/RNA hybrid regulation.


Subject(s)
DEAD-box RNA Helicases/metabolism , Exoribonucleases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , R-Loop Structures/genetics , Cell Line , DEAD-box RNA Helicases/genetics , DNA/genetics , Exoribonucleases/genetics , Genomics/methods , Humans , Immunoprecipitation/methods , Nucleic Acid Hybridization/genetics , Protein-Arginine N-Methyltransferases/genetics , R-Loop Structures/physiology , RNA/genetics , RNA Polymerase II/genetics , Transcription Termination, Genetic/physiology , Transcription, Genetic/genetics
9.
Leukemia ; 34(12): 3269-3285, 2020 12.
Article in English | MEDLINE | ID: mdl-32576962

ABSTRACT

Somatic mutations affecting CREBBP and EP300 are a hallmark of diffuse large B-cell lymphoma (DLBCL). These mutations are frequently monoallelic, within the histone acetyltransferase (HAT) domain and usually mutually exclusive, suggesting that they might affect a common pathway, and their residual WT expression is required for cell survival. Using in vitro and in vivo models, we found that inhibition of CARM1 activity (CARM1i) slows DLBCL growth, and that the levels of sensitivity are positively correlated with the CREBBP/EP300 mutation load. Conversely, treatment of DLBCLs that do not have CREBBP/EP300 mutations with CARM1i and a CBP/p300 inhibitor revealed a strong synergistic effect. Our mechanistic data show that CARM1i further reduces the HAT activity of CBP genome wide and downregulates CBP-target genes in DLBCL cells, resulting in a synthetic lethality that leverages the mutational status of CREBBP/EP300 as a biomarker for the use of small-molecule inhibitors of CARM1 in DLBCL and other cancers.


Subject(s)
CREB-Binding Protein/genetics , E1A-Associated p300 Protein/genetics , Histone Acetyltransferases/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Synthetic Lethal Mutations/genetics , Acetylation/drug effects , Animals , Cell Line , Down-Regulation/genetics , Mice , Mice, Inbred NOD , Mice, SCID
10.
Nat Commun ; 11(1): 1134, 2020 Feb 25.
Article in English | MEDLINE | ID: mdl-32098955

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Peptides ; 122: 170171, 2019 12.
Article in English | MEDLINE | ID: mdl-31614165

ABSTRACT

The current study investigated the angiotensin-converting enzyme (ACE) inhibitory activity of 4 synthetic tripeptides. All the peptides showed enzyme inhibitory activity, especially two promising ones, TTP (Thea-Thea-Pro) and gAgAP (GABA-GABA-Pro), with IC50 values of 0.92 and 3.4 µmol/L, respectively. Enzyme inhibition kinetics determined by Lineweaver-Burk plots revealed that TTP and gAgAP were competitive inhibitors with Ki values of 0.87 and 3.12 µmol/L, respectively. Molecular docking experiments confirmed that the higher inhibitory potency of TTP and gAgAP might be attributed to the formation of several critical hydrogen bonds with the active site residues in ACE. We further demonstrated that TTP and gAgAP initiated a rapid and significant decrease in systolic blood pressure (SBP) in spontaneously hypertensive rats (SHRs). TTP treatment lowered SBP to the same extent as captopril, although the duration of anti-hypertensive effect was shorter in TTP group than that observed in captopril group. Moreover, the transcription levels of angiotensin II receptor type 1 (agtr1) and miR-132/-212 were downregulated in SHRs after administration of TTP and gAgAP. In particular, TTP treatment caused a comparable reduction of agtr1 levels compared to captopril treatment, while miR-132/212 expression was significantly decreased. These results showed that compound TTP might be served as a potential antihypertensive candidate.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Hypertension/drug therapy , Peptides/pharmacology , Peptidyl-Dipeptidase A/genetics , Amino Acid Sequence/genetics , Angiotensin-Converting Enzyme Inhibitors/chemistry , Animals , Blood Pressure/drug effects , Humans , Hydrogen Bonding/drug effects , Kinetics , Peptides/chemistry , Peptides/genetics , Rats , Rats, Inbred SHR/genetics
12.
Nat Commun ; 10(1): 4541, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586052

ABSTRACT

High-throughput CRISPR-Cas9 knockout screens using a tiling-sgRNA design permit in situ evaluation of protein domain function. Here, to facilitate de novo identification of essential protein domains from such screens, we propose ProTiler, a computational method for the robust mapping of CRISPR knockout hyper-sensitive (CKHS) regions, which refer to the protein regions associated with a strong sgRNA dropout effect in the screens. Applied to a published CRISPR tiling screen dataset, ProTiler identifies 175 CKHS regions in 83 proteins. Of these CKHS regions, more than 80% overlap with annotated Pfam domains, including all of the 15 known drug targets in the dataset. ProTiler also reveals unannotated essential domains, including the N-terminus of the SWI/SNF subunit SMARCB1, which is validated experimentally. Surprisingly, the CKHS regions are negatively correlated with phosphorylation and acetylation sites, suggesting that protein domains and post-translational modification sites have distinct sensitivities to CRISPR-Cas9 mediated amino acids loss.


Subject(s)
Computational Biology/methods , Protein Domains/genetics , Algorithms , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Datasets as Topic , Gene Knockout Techniques , High-Throughput Screening Assays , Humans , Models, Genetic , Protein Processing, Post-Translational/genetics , RNA, Guide, Kinetoplastida/genetics , SMARCB1 Protein/genetics , Software
13.
J Syst Integr Neurosci ; 2(1): 91-98, 2016.
Article in English | MEDLINE | ID: mdl-27213050

ABSTRACT

Among the thirteen types of water channel proteins, aquaporins (AQPs), which play various essential roles in human physiology, AQP4 is richly expressed in cells of the central nervous system and implicated in pathological conditions such as brain edema. Therefore, researchers have been looking for ways to inhibit AQP4's water-conducting function. Many small molecules have been investigated for their interactions with the residues that form the AQP4 channel entry vestibule on the extracellular side and their interruption of waters entering into the conducting pore. Conducting all-atom simulations on the basis of CHARMM 36 force field, we study one such inhibitor, 5-acetamido-1,3,4-thiadiazole-2-sulfonamide (AZM), to achieve quantitative agreement between the computed and the experimentally measured values of AZM-AQP4 binding affinity. Using the same method, we examine the possibility of plugging up the AQP4 channel around the Asn-Pro-Ala motifs located near the channel center because a small molecule bound there would totally occlude water conduction through AQP4. We compute the binding affinities of 1,2-ethanediol (EDO) and 1,3-propanediol (PDO) inside the AQP4 conducting pore and identify the specificities of the interactions. The EDO-AQP4 interaction is weak with a dissociation constant of 80 mM. The PDO-AQP4 interaction is rather strong with a dissociation constant of 328 µM, which indicates that PDO is an efficacious AQP4 inhibitor with sufficiently high potency. Considering the fact that PDO is classified by the US Food and Drug Administration as generally safe, we predict that 1,3-propanediol could be an effective drug for brain edema and other AQP4-correlated neurological conditions.

14.
J Phys Chem B ; 119(50): 15502-8, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26581232

ABSTRACT

Nanometer-sized gold particles (AuNPs) are of peculiar interest because their behaviors in an aqueous solution are sensitive to changes in environmental factors including the size and shape of the solute ions. In order to determine these important characteristics, we performed all-atom molecular dynamics simulations on the icosahedral Au144 nanoparticles each coated with a homogeneous set of 60 thiolates (4-mercaptobenzoate, pMBA) in eight aqueous solutions having ions of varying sizes and shapes (Na(+), K(+), tetramethylamonium cation TMA(+), tris-ammonium cation TRS(+), Cl(-), and OH(-)). For each solution, we computed the reversible work (potential of mean of force) to bring two nanoparticles together as a function of their separation distance. We found that the behavior of pMBA protected Au144 nanoparticles can be readily modulated by tuning their aqueous environmental factors (pH and solute ion combinations). We examined the atomistic details on how the sizes and shapes of solute ions quantitatively factor in the definitive characteristics of nanoparticle-environment and nanoparticle-nanoparticle interactions. We predict that tuning the concentrations of nonspherical composite ions such as TRS(+) in an aqueous solution of AuNPs be an effective means to modulate the aggregation propensity desired in biomedical and other applications of small charged nanoparticles.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Solutions , Water
SELECTION OF CITATIONS
SEARCH DETAIL