Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 76(6): 938-952.e5, 2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31668930

RESUMO

High-resolution Cas9 structures have yet to reveal catalytic conformations due to HNH nuclease domain positioning away from the cleavage site. Nme1Cas9 and Nme2Cas9 are compact nucleases for in vivo genome editing. Here, we report structures of meningococcal Cas9 homologs in complex with sgRNA, dsDNA, or the AcrIIC3 anti-CRISPR protein. DNA-bound structures represent an early step of target recognition, a later HNH pre-catalytic state, the HNH catalytic state, and a cleaved-target-DNA-bound state. In the HNH catalytic state of Nme1Cas9, the active site is seen poised at the scissile phosphodiester linkage of the target strand, providing a high-resolution view of the active conformation. The HNH active conformation activates the RuvC domain. Our structures explain how Nme1Cas9 and Nme2Cas9 read distinct PAM sequences and how AcrIIC3 inhibits Nme1Cas9 activity. These structures provide insights into Cas9 domain rearrangements, guide-target engagement, cleavage mechanism, and anti-CRISPR inhibition, facilitating the optimization of these genome-editing platforms.


Assuntos
Bacteriófagos/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , DNA/metabolismo , Neisseria meningitidis/enzimologia , Proteínas Virais/metabolismo , Bacteriófagos/genética , Sítios de Ligação , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/ultraestrutura , Catálise , DNA/genética , DNA/ultraestrutura , Escherichia coli/enzimologia , Escherichia coli/genética , Neisseria meningitidis/genética , Ligação Proteica , Domínios Proteicos , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Relação Estrutura-Atividade , Proteínas Virais/genética , Proteínas Virais/ultraestrutura
2.
Mol Cell ; 73(4): 714-726.e4, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30581144

RESUMO

CRISPR-Cas9 genome editing has transformed biotechnology and therapeutics. However, in vivo applications of some Cas9s are hindered by large size (limiting delivery by adeno-associated virus [AAV] vectors), off-target editing, or complex protospacer-adjacent motifs (PAMs) that restrict the density of recognition sequences in target DNA. Here, we exploited natural variation in the PAM-interacting domains (PIDs) of closely related Cas9s to identify a compact ortholog from Neisseria meningitidis-Nme2Cas9-that recognizes a simple dinucleotide PAM (N4CC) that provides for high target site density. All-in-one AAV delivery of Nme2Cas9 with a guide RNA targeting Pcsk9 in adult mouse liver produces efficient genome editing and reduced serum cholesterol with exceptionally high specificity. We further expand our single-AAV platform to pre-implanted zygotes for streamlined generation of genome-edited mice. Nme2Cas9 combines all-in-one AAV compatibility, exceptional editing accuracy within cells, and high target site density for in vivo genome editing applications.


Assuntos
Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , DNA/genética , Edição de Genes/métodos , Fígado/enzimologia , Neisseria meningitidis/enzimologia , Pró-Proteína Convertase 9/genética , Animais , Proteína 9 Associada à CRISPR/metabolismo , DNA/metabolismo , Dependovirus/genética , Transferência Embrionária , Feminino , Vetores Genéticos , Células HEK293 , Humanos , Células K562 , Camundongos Endogâmicos C57BL , Motivos de Nucleotídeos , Pró-Proteína Convertase 9/metabolismo , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Especificidade por Substrato , Zigoto/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(11): e2117013119, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35259022

RESUMO

SignificanceThe study provided a long-sought molecular mechanism that could explain the link between fatty acid metabolism and cancer metastasis. Further understanding may lead to new strategies to inhibit cancer metastasis. The chemical proteomic approach developed here will be useful for discovering other regulatory mechanisms of protein function by small molecule metabolites.


Assuntos
Acil Coenzima A/metabolismo , Nucleosídeo NM23 Difosfato Quinases/antagonistas & inibidores , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias da Mama , Endocitose , Feminino , Humanos , Metástase Neoplásica , Neoplasias/etiologia , Ligação Proteica , Proteoma , Proteômica/métodos
4.
BMC Biol ; 21(1): 250, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37946200

RESUMO

BACKGROUND: Nme2ABE8e has been constructed and characterized as a compact, accurate adenine base editor with a less restrictive dinucleotide protospacer-adjacent motif (PAM: N4CC) but low editing efficiency at challenging loci in human cells. Here, we engineered a subset of domain-inlaid Nme2Cas9 base editors to bring the deaminase domain closer to the nontarget strand to improve editing efficiency. RESULTS: Our results demonstrated that Nme2ABE8e-797 with adenine deaminase inserted between amino acids 797 and 798 has a significantly increased editing efficiency with a wide editing window ranging from 4 to 18 bases in mammalian cells, especially at the sites that were difficult to edit by Nme2ABE8e. In addition, by swapping the PAM-interacting domain of Nme2ABE8e-797 with that of SmuCas9 or introducing point mutations of eNme2-C in Nme2ABE8e-797, we created Nme2ABE8e-797Smu and Nme2ABE8e-797-C, respectively, which exhibited robust activities at a wide range of sites with N4CN PAMs in human cells. Moreover, the modified domain-inlaid Nme2ABE8e can efficiently restore or install disease-related loci in Neuro-2a cells and mice. CONCLUSIONS: These novel Nme2ABE8es with increased on-target DNA editing and expanded PAM compatibility will expand the base editing toolset for efficient gene modification and therapeutic applications.


Assuntos
Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Animais , Humanos , Camundongos , Proteína 9 Associada à CRISPR/genética , Adenina/química , Edição de Genes/métodos , DNA/genética , Mamíferos/genética
5.
Mol Ther ; 30(1): 164-174, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33992807

RESUMO

Transthyretin (TTR) amyloidosis is a hereditary life-threatening disease characterized by deposition of amyloid fibrils. The main causes of TTR amyloidosis are mutations in the TTR gene that lead to the production of misfolded TTR protein. Reducing the production of toxic protein in the liver is a validated strategy to treat TTR amyloidosis. In this study, we established a humanized mouse model that expresses mutant human TTR (hTTR; V30M) protein in the liver to model TTR amyloidosis. Then, we compared the efficiency of reducing the expression of mutant hTTR by dual adeno-associated virus 8 (AAV8)-mediated split SpCas9 with that by single AAV8-mediated Nme2Cas9 in this model. With two gRNAs targeting different exons, dual AAV-mediated split SpCas9 system achieved efficiencies of 37% and 34% reduction of hTTR mRNA and reporter GFP expression, respectively, in the liver. Surprisingly, single AAV-mediated Nme2Cas9 treatment resulted in 65% and 71% reduction of hTTR mRNA and reporter GFP, respectively. No significant editing was identified in predicted off-target sites in the mouse and human genomes after Nme2Cas9 targeting. Thus, we provide proof of principle for using single AAV-mediated CRISPR-Nme2Cas9 to effectively reduce mutant hTTR expression in vivo, which may translate into gene therapy for TTR amyloidosis.


Assuntos
Neuropatias Amiloides Familiares , Amiloide , Neuropatias Amiloides Familiares/genética , Neuropatias Amiloides Familiares/terapia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Pré-Albumina/genética
6.
Proc Natl Acad Sci U S A ; 114(13): 3497-3502, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28292893

RESUMO

Stauprimide is a staurosporine analog that promotes embryonic stem cell (ESC) differentiation by inhibiting nuclear localization of the MYC transcription factor NME2, which in turn results in down-regulation of MYC transcription. Given the critical role the oncogene MYC plays in tumor initiation and maintenance, we explored the potential of stauprimide as an anticancer agent. Here we report that stauprimide suppresses MYC transcription in cancer cell lines derived from distinct tissues. Using renal cancer cells, we confirmed that stauprimide inhibits NME2 nuclear localization. Gene expression analysis also confirmed the selective down-regulation of MYC target genes by stauprimide. Consistent with this activity, administration of stauprimide inhibited tumor growth in rodent xenograft models. Our study provides a unique strategy for selectively targeting MYC transcription by pharmacological means as a potential treatment for MYC-dependent tumors.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/genética , Proteínas Proto-Oncogênicas c-myc/genética , Bibliotecas de Moléculas Pequenas/administração & dosagem , Animais , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Nucleosídeo NM23 Difosfato Quinases/genética , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo
7.
J Biol Chem ; 292(37): 15205-15215, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28717007

RESUMO

Transcriptional activation of the human telomerase reverse transcriptase (hTERT) gene, which remains repressed in adult somatic cells, is critical during tumorigenesis. Several transcription factors and the epigenetic state of the hTERT promoter are known to be important for tight control of hTERT in normal tissues, but the molecular mechanisms leading to hTERT reactivation in cancer are not well-understood. Surprisingly, here we found occupancy of the metastasis suppressor non-metastatic 2 (NME2) within the hTERT core promoter in HT1080 fibrosarcoma cells and HCT116 colon cancer cells and NME2-mediated transcriptional repression of hTERT in these cells. We also report that loss of NME2 results in up-regulated hTERT expression. Mechanistically, additional results indicated that the RE1-silencing transcription factor (REST)-lysine-specific histone demethylase 1 (LSD1) co-repressor complex associates with the hTERT promoter in an NME2-dependent way and that this assembly is required for maintaining repressive chromatin at the hTERT promoter. Interestingly, a G-quadruplex motif at the hTERT promoter was essential for occupancy of NME2 and the REST repressor complex on the hTERT promoter. In light of this mechanistic insight, we studied the effects of G-quadruplex-binding ligands on hTERT expression and observed that several of these ligands repressed hTERT expression. Together, our results support a mechanism of hTERT epigenetic control involving a G-quadruplex promoter motif, which potentially can be targeted by tailored small molecules.


Assuntos
Carcinoma/metabolismo , Repressão Epigenética , Fibrossarcoma/metabolismo , Quadruplex G , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Regiões Promotoras Genéticas , Telomerase/metabolismo , Substituição de Aminoácidos , Carcinoma/enzimologia , Carcinoma/patologia , Linhagem Celular Tumoral , Células Cultivadas , Imunoprecipitação da Cromatina , Fibrossarcoma/enzimologia , Fibrossarcoma/patologia , Genes Reporter , Histona Desmetilases/química , Histona Desmetilases/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Nucleosídeo NM23 Difosfato Quinases/antagonistas & inibidores , Nucleosídeo NM23 Difosfato Quinases/química , Nucleosídeo NM23 Difosfato Quinases/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Mutação Puntual , Multimerização Proteica , Interferência de RNA , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Telomerase/antagonistas & inibidores , Telomerase/genética
8.
Biochem Biophys Res Commun ; 472(4): 573-9, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26972251

RESUMO

In mouse testes, germ cell apoptosis can be caused by cigarette smoke and lead to declining quality of semen, but the exact molecular mechanisms remain unclear. To evaluate the effects of nicotine exposure on apoptosis during spermatogenesis, we first constructed a nicotine-treated mouse model and detected germ cell apoptosis activity in the testes using the TUNEL method. Then we analyzed the variation of telomere length and telomerase activity by real-time PCR and TRAP-real-time PCR, respectively. Further, we investigated a highly expressed gene, Nme2, in mouse testes after nicotine treatment from our previous results, which has close correlation with the apoptosis activity predicted by bioinformatics. We performed NME2 overexpression in Hela cells to confirm whether telomere length and telomerase activity were regulated by the Nme2 gene. Finally, we examined methylation of CpG islands in the Nme2 promoter with the Bisulfite Sequencing (BSP) method. The results showed that apoptosis had increased significantly, and then telomerase activity became weak. Further, telomere length was shortened in the germ cells among the nicotine-treated group. In Hela cells, both overexpression of the Nme2 gene and nicotine exposure can suppress the activity of telomerase activity and shorten telomere length. BSP results revealed that the Nme2 promoter appeared with low methylation in mouse testes after nicotine treatment. We assume that nicotine-induced apoptosis may be caused by telomerase activity decline, which is inhibited by the up expression of Nme2 because of its hypomethylation in mouse germ cells.


Assuntos
Apoptose/efeitos dos fármacos , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Nicotina/toxicidade , Agonistas Nicotínicos/farmacologia , Homeostase do Telômero/efeitos dos fármacos , Testículo/citologia , Testículo/efeitos dos fármacos , Animais , Metilação de DNA/efeitos dos fármacos , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nucleosídeo NM23 Difosfato Quinases/genética , Espermatogênese/efeitos dos fármacos , Telomerase/metabolismo , Testículo/metabolismo
9.
Biochem Biophys Res Commun ; 471(4): 522-7, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26896769

RESUMO

Chondroitin sulfate proteoglycans (CSPGs) are a major component of glial scars, inhibiting axonal growth in the central nervous system. Protein tyrosine phosphatase, receptor type S (PTPσ) has been identified as a receptor for CSPGs, whereas its downstream signaling pathway remains to be fully understood. Here, we report that nucleoside diphosphate kinase 2 (NME2) interacts with PTPσ. We screened proteins associated with PTPσ by mass spectrometry, and obtained NME2. Immunoprecipitation analysis revealed that NME2 associated with the PTPσ intracellular domain in HEK-293T cells. NME2 was expressed in the cytoplasm and nucleus of cortical neurons, and knockdown of NME2 in the cortical neurons completely rescued neurite outgrowth inhibition induced by CSPGs. These results demonstrate that NME2 associates with PTPσ to elicit neurite outgrowth inhibition in response to CSPGs.


Assuntos
Proteoglicanas de Sulfatos de Condroitina/metabolismo , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neuritos/fisiologia , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Animais , Núcleo Celular/enzimologia , Córtex Cerebral/citologia , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Citoplasma/enzimologia , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Espectrometria de Massas , Camundongos , Nucleosídeo NM23 Difosfato Quinases/genética , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/enzimologia , Neurônios/ultraestrutura , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética
10.
Arterioscler Thromb Vasc Biol ; 35(8): 1852-61, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26088577

RESUMO

OBJECTIVE: Vascular smooth muscle cells (VSMC) proliferation is a hallmark of atherosclerosis and vascular restenosis. The intermediate conductance Ca(2+)-activated K(+) (SK4) channel is required for pathological VSMC proliferation. In T lymphocytes, nucleoside diphosphate kinase B (NDPKB) has been implicated in SK4 channel activation. We thus investigated the role of NDPKB in the regulation of SK4 currents (ISK4) in proliferating VSMC and neointima formation. APPROACH AND RESULTS: Function and expression of SK4 channels in VSMC from injured mouse carotid arteries were assessed by patch-clamping and real-time polymerase chain reaction. ISK4 was detectable in VSMC from injured but not from uninjured arteries correlating with the occurrence of the proliferative phenotype. Direct application of NDPKB to the membrane of inside-out patches increased ISK4, whereas NDPKB did not alter currents in VSMC obtained from injured vessels of SK4-deficient mice. The NDPKB-induced increase in ISK4 was prevented by protein histidine phosphatase 1, but not an inactive protein histidine phosphatase 1 mutant indicating that ISK4 is regulated via histidine phosphorylation in proliferating VSMC; moreover, genetic NDPKB ablation reduced ISK4 by 50% suggesting a constitutive activation of ISK4 in proliferating VSMC. In line, neointima formation after wire injury of the carotid artery was substantially reduced in mice deficient in SK4 channels or NDPKB. CONCLUSIONS: NDPKB to SK4 signaling is required for neointima formation. Constitutive activation of SK4 by NDPKB in proliferating VSMC suggests that targeting this interaction via, for example, activation of protein histidine phosphatase 1 may provide clinically meaningful effects in vasculoproliferative diseases such as atherosclerosis and post angioplasty restenosis.


Assuntos
Lesões das Artérias Carótidas/enzimologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neointima , Animais , Artérias Carótidas/enzimologia , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/deficiência , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Potenciais da Membrana , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Nucleosídeo NM23 Difosfato Quinases/deficiência , Nucleosídeo NM23 Difosfato Quinases/genética , Transdução de Sinais
11.
Am J Respir Crit Care Med ; 192(1): 64-75, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25906011

RESUMO

RATIONALE: CARD-recruited membrane-associated protein 3 (CARMA3) is a novel scaffold protein that regulates nuclear factor (NF)-κB activation; however, the underlying mechanism of CARMA3 in lung cancer stemness and metastasis remains largely unknown. OBJECTIVES: To investigate the molecular mechanisms underlying the involvement of CARMA3 in non-small cell lung cancer progression. METHODS: The expression levels of CARMA3 and NME2 in a cohort of patients with lung cancer (n = 91) were examined by immunohistochemistry staining and assessed by Kaplan-Meier survival analysis. The effects of CARMA3, microRNA-182 (miR-182), and NME2 on cancer stemness and metastasis were measured in vitro and in vivo. Chromatin immunoprecipitation and luciferase reporter assays were performed to determine the mechanisms of NF-κB-driven miR-182 expression and NME2 regulation. MEASUREMENTS AND MAIN RESULTS: We observed that CARMA3 inversely correlated with NME2 expression in patients with lung cancer (Pearson correlation coefficient: R = -0.24; P = 0.022). NME2 levels were significantly decreased in tumor tissues compared with adjacent normal lung tissues (P < 0.001), and patients with lung cancer with higher levels of NME2 had longer survival outcomes (overall survival, P < 0.01; disease-free survival, P < 0.01). Mechanistically, CARMA3 promoted cell motility by reducing the level of NME2 through the NF-κB/miR-182 pathway and by increasing cancer stem cell properties and metastasis in lung cancer. CONCLUSIONS: We identified a novel mechanism of CARMA3 in lung cancer stemness and metastasis through the negative regulation of NME2 by NF-κB-dependent induction of miR-182. Our findings provide an attractive strategy for targeting the CARMA3/NF-κB/miR-182 pathway as a potential treatment for lung cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Estudos de Casos e Controles , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Metástase Neoplásica , Análise de Sobrevida
12.
Arterioscler Thromb Vasc Biol ; 34(10): 2292-300, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25147336

RESUMO

OBJECTIVE: Nucleoside diphosphate kinase B (NDPKB) participates in the activation of heterotrimeric and monomeric G proteins, which are pivotal mediators in angiogenic signaling. The role of NDPKB in angiogenesis has to date not been defined. Therefore, we analyzed the contribution of NDPKB to angiogenesis and its underlying mechanisms in well-characterized in vivo and in vitro models. APPROACH AND RESULTS: Zebrafish embryos were depleted of NDPKB by morpholino-mediated knockdown. These larvae displayed severe malformations specifically in vessels formed by angiogenesis. NDPKB-deficient (NDPKB(-/-)) mice were subjected to oxygen-induced retinopathy. In this model, the number of preretinal neovascularizations in NDPKB(-/-) mice was strongly reduced in comparison with wild-type littermates. In accordance, a delayed blood flow recovery was detected in the NDPKB(-/-) mice after hindlimb ligation. In in vitro studies, a small interfering RNA-mediated knockdown of NDPKB was performed in human umbilical endothelial cells. NDPKB depletion impaired vascular endothelial growth factor (VEGF)-induced sprouting and hampered the VEGF-induced spatial redistributions of the VEGF receptor type 2 and VE-cadherin at the plasma membrane. Concomitantly, NDPKB depletion increased the permeability of the human umbilical endothelial cell monolayer. CONCLUSIONS: This is the first report to show that NDPKB is required for VEGF-induced angiogenesis and contributes to the correct localization of VEGF receptor type 2 and VE-cadherin at the endothelial adherens junctions. Therefore, our data identify NDPKB as a novel molecular target to modulate VEGF-dependent angiogenesis.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Células Endoteliais/enzimologia , Músculo Esquelético/irrigação sanguínea , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neovascularização Fisiológica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Membro Posterior , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Isquemia/enzimologia , Isquemia/genética , Isquemia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nucleosídeo NM23 Difosfato Quinases/deficiência , Nucleosídeo NM23 Difosfato Quinases/genética , Interferência de RNA , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Neovascularização Retiniana/enzimologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/fisiopatologia , Transdução de Sinais , Fatores de Tempo , Transfecção , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
13.
Clin Transl Med ; 13(8): e1383, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37598400

RESUMO

BACKGROUND: Age-related macular degeneration (AMD), particularly wet AMD characterised by choroidal neovascularization (CNV), is a leading cause of vision loss in the elderly. The hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) pathway contributes to CNV pathogenesis. Previous gene editing research indicated that disrupting these genes in retinal pigment epithelial cells could have a preventive effect on CNV progression. However, no studies have yet been conducted using gene editing to disrupt VEGF signalling after CNV induction for therapeutic validation, which is critical to the clinical application of wet AMD gene editing therapies. METHOD: Here, we employed the single-adeno-associated virus-mediated Nme2 Cas9 to disrupt key molecules in VEGF signalling, Hif1α, Vegfa and Vegfr2 after inducing CNV and estimated their therapeutic effects. RESULTS: We found that Nme2 Cas9 made efficient editing in target genes up to 71.8% post 11 days in vivo. And only Nme2 Cas9-Vegfa treatment during the early stage of CNV development reduced the CNV lesion area by 49.5%, compared to the negative control, while Nme2 Cas9-Hif1α or Nme2 Cas9-Vegfr2 treatment did not show therapeutic effect. Besides, no off-target effects were observed in Nme2 Cas9-mediated gene editing in vivo. CONCLUSIONS: This study provides proof-of-concept possibility of employing Nme2 Cas9 for potential anti-angiogenesis therapy in wet AMD.


Assuntos
Degeneração Macular , Fator A de Crescimento do Endotélio Vascular , Idoso , Humanos , Fator A de Crescimento do Endotélio Vascular/genética , Sistemas CRISPR-Cas/genética , Degeneração Macular/genética , Degeneração Macular/terapia , Imunoterapia , Edição de Genes
14.
Vet Microbiol ; 252: 108926, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33223234

RESUMO

Our present study aimed to identify host cell proteins that may interact with avian reovirus (ARV) σA protein and their potential effect on ARV replication. The ARV structural protein σA has been demonstrated to suppress interferon production and confirmed to activate the PI3K/Akt pathway. However, host cell factors interacting with σA to affect ARV replication remain unknown. In current study, a cDNA library of chicken embryo fibroblasts (CEFs) was constructed, and host cell proteins interacting with σA were screened by a yeast two-hybrid system. We identified four candidate cellular proteins that interact with ARV σA protein. Among them, Gallus NME/NM23 nucleoside diphosphate kinase 2 (NME2) was further validated as a σA-binding protein through co-immunoprecipitation. The key interaction domain was identified at amino acids (aa) 121-416 in NME2 and at aa 71-139 in σA, respectively. We demonstrated that overexpression of NME2 substantially inhibited ARV replication. In addition silencing NME2 by small interfering RNAs (siRNAs) resulted in marked enhancement of ARV replication. Our work has demonstrated that NME2 is a σA-binding protein that may affect ARV replication in CEF cells.


Assuntos
Nucleosídeo NM23 Difosfato Quinases/metabolismo , Orthoreovirus Aviário/enzimologia , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral , Animais , Embrião de Galinha , Fibroblastos/fisiologia , Nucleosídeo NM23 Difosfato Quinases/genética , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/metabolismo , Orthoreovirus Aviário/genética , Orthoreovirus Aviário/fisiologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Mapeamento de Interação de Proteínas/veterinária , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Proteínas do Core Viral/genética
15.
Sci China Life Sci ; 64(8): 1355-1367, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33420918

RESUMO

Cytidine base editor (CBE), which is composed of a cytidine deaminase fused to Cas9 nickase, has been widely used to induce C-to-T conversions in a wide range of organisms. However, the targeting scope of current CBEs is largely restricted to protospacer adjacent motif (PAM) sequences containing G, T, or A bases. In this study, we developed a new base editor termed "nNme2-CBE" with excellent PAM compatibility for cytidine dinucleotide, significantly expanding the genome-targeting scope of CBEs. Using nNme2-CBE, targeted editing efficiencies of 29.0%-55.0% and 17.3%-52.5% were generated in human cells and rabbit embryos, respectively. In contrast to conventional nSp-CBE, the nNme2-CBE is a natural high-fidelity base editing platform with minimal DNA off-targeting detected in vivo. Significantly increased efficiency in GC context and precision were determined by combining nNme2Cas9 with rationally engineered cytidine deaminases. In addition, the Founder rabbits with accurate single-base substitutions at Fgf5 gene loci were successfully generated by using the nNme2-CBE system. These novel nNme2-CBEs with expanded PAM compatibility and high fidelity will expand the base editing toolset for efficient gene modification and therapeutic applications.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Citidina Desaminase/metabolismo , Citidina/metabolismo , Motivos de Nucleotídeos , Animais , Fluorescência , Humanos , Edição de RNA , Coelhos
16.
Methods Mol Biol ; 2077: 83-91, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31707653

RESUMO

Protein histidine phosphorylation has largely remained unexplored due to the challenges of analyzing relatively unstable phosphohistidine-containing proteins. We describe a procedure for determining the stoichiometry of histidine phosphorylation on the human histidine kinases NME1 and NME2 by intact mass spectrometry under conditions that retain this acid-labile protein modification. By characterizing these two model histidine protein kinases in the absence and presence of a suitable phosphate donor, the stoichiometry of histidine phosphorylation can be determined. The described method can be readily adapted for the analysis of other proteins containing phosphohistidine.


Assuntos
Histidina Quinase/química , Histidina/análogos & derivados , Histidina/química , Espectrometria de Massas , Cromatografia Líquida , Histidina Quinase/metabolismo , Humanos , Fosforilação
17.
Front Microbiol ; 11: 599205, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33281799

RESUMO

To date, a variety of Brucella effector proteins have been found to mediate host cell secretion, autophagy, inflammation, and other signal pathways, but nuclear effector proteins have not yet been reported. We identified the first Brucella nucleomodulin, BspJ, and we screened out the BspJ interaction host proteins NME/NM23 nucleoside diphosphate kinase 2 (NME2) and creatine kinase B (CKB) through yeast two-hybrid and co-immunoprecipitation assays. These proteins are related to the host cell energy synthesis, metabolism, and apoptosis pathways. Brucella nucleomodulin BspJ will decrease the expression level of NME2 and CKB. In addition, BspJ gene deletion strains promoted the apoptosis of macrophages and reduced the intracellular survival of Brucella in host cells. In short, we found nucleomodulin BspJ may directly or indirectly regulate host cell apoptosis through the interaction with NME2 and CKB by mediating energy metabolism pathways in response to the intracellular circulation of Brucella infection, but the mechanism needs further study.

18.
J Mol Histol ; 51(5): 573-581, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32860079

RESUMO

Histidine phosphorylation (pHis) was first reported in 1962. There are few studies on pHis because of the thermal and acidic instability of pHis and the lack of specific methods to detect it. pHis has two isomers of 1-phosphate histidine (1-pHis) and 3-phosphate histidine (3-pHis). pHis antibodies have been developed recently and have promoted research in this field. In this study, we established a CCl4-induced liver fibrosis model in C57 mice and a TGF-ß1-induced HSC activation model in LX-2 cells, to study the role of histidine phosphorylation. The expression of histidine kinases NME1 and NME2 was increased, histidine phosphatase PGAM5 and PHPT1 was unchanged, and 1-pHis and 3-pHis were increased in the in vivo and in vitro models. The expression of LHPP was decreased in the in vivo model but not in the in vitro model. To further study the role of NME1, NME2, and histidine phosphorylation in HSC activation, we silenced NME1 or NME2 and administered TGF-ß1 in LX-2 cells. The results showed silencing NME1 or NME2 decreased TGF-ß1-induced pHis levels and the expression of α-SMA and COL1A1, indicating the activation of HSC was suppressed. Then, we found the inhibitory effect on HSC activation is due to reduced phosphorylation of Smad2 and Smad3. In summary, our studies indicate that NME1 and NME2 are involved in TGF-ß1-induced HSC activation and CCl4-induced liver fibrosis, which may be mediated by histidine phosphorylation.


Assuntos
Tetracloreto de Carbono/efeitos adversos , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Apoptose , Biomarcadores , Linhagem Celular , Modelos Animais de Doenças , Suscetibilidade a Doenças , Inativação Gênica , Histidina/metabolismo , Humanos , Cirrose Hepática/patologia , Camundongos , Nucleosídeo NM23 Difosfato Quinases/genética , Fosforilação
19.
Methods Mol Biol ; 2077: 63-81, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31707652

RESUMO

Despite the discovery of protein histidine (His) phosphorylation nearly six decades ago, difficulties in measuring and quantifying this unstable post-translational modification (PTM) have limited its mechanistic analysis in prokaryotic and eukaryotic signaling. Here, we describe reliable procedures for affinity purification, cofactor-binding analysis and antibody-based detection of phosphohistidine (pHis), on the putative human His kinases NME1 (NDPK-A) and NME2 (NDPK-B) and the glycolytic phosphoglycerate mutase PGAM1. By exploiting isomer-specific monoclonal N1-pHis and N3-pHis antibodies, we describe robust protocols for immunological detection and isomer discrimination of site-specific pHis, including N3-pHis on His 11 of PGAM1.


Assuntos
Bactérias/genética , Bactérias/metabolismo , Histidina/análogos & derivados , Fosfoproteínas/metabolismo , Western Blotting , Varredura Diferencial de Calorimetria , Fluorometria , Histidina/metabolismo , Histidina Quinase/genética , Histidina Quinase/metabolismo , Humanos , Mutagênese , Fosfoproteínas/genética , Fosfoproteínas/isolamento & purificação , Fosforilação , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
20.
Cancer Biol Ther ; 19(7): 565-572, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29630434

RESUMO

Osteosarcoma (OS) is one of the most common primary bone tumors and has a high disablity rate and case-fatality rate. The protracted stagnancy of the chemotherapy program and surgical technology for OS treatment prompted us to focus on the mechanisms of cancer carcinogenesis progression in OS. Nucleoside diphosphate kinase B (NME2) is a type of nucleoside diphosphate kinase that plays an important role in cellular processes. In this study, we report overexpression of NME2 in OS cell lines and correlate this overexpression with the clinicopathologic features of osteosarcoma. We used si-NME2 to downregulate expression of NME2 in OS cell lines. The results of the CCK8 and clone forming assays show that NME2 promotes OS cell line proliferation. Western blot assays show that deregulation of NME2 results in enhanced the expression of c-Myc, which promotes OS proliferation.


Assuntos
Neoplasias Ósseas/patologia , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Osteossarcoma/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Adulto , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular , Humanos , Masculino , Nucleosídeo NM23 Difosfato Quinases/genética , Proto-Oncogenes , RNA Interferente Pequeno/metabolismo , Regulação para Cima , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA