Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37285284

RESUMO

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.


Assuntos
Distrofia Muscular de Duchenne , Células Satélites de Músculo Esquelético , Animais , Camundongos , Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Células Cultivadas , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo
2.
Cell Rep ; 38(13): 110582, 2022 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-35354055

RESUMO

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.


Assuntos
Retrovirus Endógenos , Melanoma Experimental , Animais , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Interferons/genética , Melanoma Experimental/genética , Recidiva Local de Neoplasia/genética
3.
Nat Commun ; 13(1): 474, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35078987

RESUMO

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.


Assuntos
Sequência de Bases/genética , Sistemas CRISPR-Cas , Edição de Genes/métodos , Mutação , Neoplasias/terapia , RNA Guia de Cinetoplastídeos/química , Linhagem Celular , Humanos , Neoplasias/genética , Neoplasias/patologia , RNA Guia de Cinetoplastídeos/genética
4.
Cell Rep ; 36(2): 109337, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34260915

RESUMO

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.


Assuntos
Diferenciação Celular , Ribonucleoproteínas Nucleares Heterogêneas/química , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Neurônios/metabolismo , Neurônios/patologia , Deleção de Sequência , Proteína Supressora de Tumor p53/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Processamento Alternativo/genética , Motivos de Aminoácidos , Arginina/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Metilação , Células-Tronco Neurais/metabolismo , Neurogênese , Ligação Proteica , Estabilidade Proteica , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas/genética , RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina/metabolismo
5.
Leukemia ; 34(12): 3269-3285, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32576962

RESUMO

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.


Assuntos
Proteína de Ligação a CREB/genética , Proteína p300 Associada a E1A/genética , Histona Acetiltransferases/metabolismo , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/metabolismo , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Mutações Sintéticas Letais/genética , Acetilação/efeitos dos fármacos , Animais , Linhagem Celular , Regulação para Baixo/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
6.
Peptides ; 122: 170171, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31614165

RESUMO

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.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Hipertensão/tratamento farmacológico , Peptídeos/farmacologia , Peptidil Dipeptidase A/genética , Sequência de Aminoácidos/genética , Inibidores da Enzima Conversora de Angiotensina/química , Animais , Pressão Sanguínea/efeitos dos fármacos , Humanos , Ligação de Hidrogênio/efeitos dos fármacos , Cinética , Peptídeos/química , Peptídeos/genética , Ratos , Ratos Endogâmicos SHR/genética
7.
Nat Commun ; 10(1): 4541, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31586052

RESUMO

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.


Assuntos
Biologia Computacional/métodos , Domínios Proteicos/genética , Algoritmos , Sistemas CRISPR-Cas/genética , Linhagem Celular Tumoral , Conjuntos de Dados como Assunto , Técnicas de Inativação de Genes , Ensaios de Triagem em Larga Escala , Humanos , Modelos Genéticos , Processamento de Proteína Pós-Traducional/genética , RNA Guia de Cinetoplastídeos/genética , Proteína SMARCB1/genética , Software
8.
Nucleic Acids Res ; 47(10): 5038-5048, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-30916320

RESUMO

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.


Assuntos
Deleção de Genes , Neoplasias/enzimologia , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Células A549 , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Etilenodiaminas/farmacologia , Humanos , Isoquinolinas/farmacologia , Células MCF-7 , Camundongos Knockout , Neoplasias/genética , Neoplasias/patologia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/genética , Pirimidinas/farmacologia , Pirróis/farmacologia , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA