Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Biol ; 216(12): 4165-4182, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29051265

RESUMO

Isoprenylcysteine carboxyl methyltransferase (ICMT) methylesterifies C-terminal prenylcysteine residues of CaaX proteins and some RAB GTPases. Deficiency of either ICMT or NOTCH1 accelerates pancreatic neoplasia in Pdx1-Cre;LSL-KrasG12D mice, suggesting that ICMT is required for NOTCH signaling. We used Drosophila melanogaster wing vein and scutellar bristle development to screen Rab proteins predicted to be substrates for ICMT (ste14 in flies). We identified Rab7 and Rab8 as ICMT substrates that when silenced phenocopy ste14 deficiency. ICMT, RAB7, and RAB8 were all required for efficient NOTCH1 signaling in mammalian cells. Overexpression of RAB8 rescued NOTCH activation after ICMT knockdown both in U2OS cells expressing NOTCH1 and in fly wing vein development. ICMT deficiency induced mislocalization of GFP-RAB7 and GFP-RAB8 from endomembrane to cytosol, enhanced binding to RABGDI, and decreased GTP loading of RAB7 and RAB8. Deficiency of ICMT, RAB7, or RAB8 led to mislocalization and diminished processing of NOTCH1-GFP. Thus, NOTCH signaling requires ICMT in part because it requires methylated RAB7 and RAB8.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , GTP Fosfo-Hidrolases/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Metiltransferases/genética , Receptor Notch1/genética , Proteínas rab de Ligação ao GTP/genética , Alquil e Aril Transferases/genética , Alquil e Aril Transferases/metabolismo , Animais , Linhagem Celular Tumoral , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Inibidores de Dissociação do Nucleotídeo Guanina/genética , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Metilação , Camundongos , Osteoblastos/metabolismo , Osteoblastos/patologia , Proteínas Metiltransferases/deficiência , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Asas de Animais , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
2.
J Neurosci ; 36(18): 5107-14, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27147662

RESUMO

UNLABELLED: Retinal neurons use multiple strategies to fine-tune visual signal transduction, including post-translational modifications of proteins, such as addition of an isoprenyl lipid to a carboxyl-terminal cysteine in proteins that terminate with a "CAAX motif." We previously showed that RAS converting enzyme 1 (RCE1)-mediated processing of isoprenylated proteins is required for photoreceptor maintenance and function. However, it is not yet known whether the requirement for the RCE1-mediated protein processing is related to the absence of the endoproteolytic processing step, the absence of the subsequent methylation step by isoprenylcysteine methyltransferase (ICMT), or both. To approach this issue and to understand the significance of protein methylation, we generated mice lacking Icmt expression in the retina. In the absence of Icmt expression, rod and cone light-mediated responses diminished progressively. Lack of ICMT-mediated methylation led to defective association of isoprenylated transducin and cone phosphodiesterase 6 (PDE6α') with photoreceptor membranes and resulted in decreased levels of transducin, PDE6α', and cone G-protein coupled receptor kinase-1 (GRK1). In contrast to our earlier findings with retina-specific Rce1 knock-out mice, rod PDE6 in Icmt-deficient mice trafficked normally to the photoreceptor outer segment, suggesting that the failure to remove the -AAX is responsible for blocking the movement of PDE6 to the outer segment. Our findings demonstrate that carboxyl methylation of isoprenylated proteins is crucial for maintenance of photoreceptor function. SIGNIFICANCE STATEMENT: In this report, we show that an absence of isoprenylcysteine methyltransferase-mediated protein methylation leads to progressive loss of vision. Photoreceptors also degenerate, although at a slower pace than the rate of visual loss. The reduction in photoresponses is due to defective association of crucial players in phototransduction cascade. Unlike the situation with RCE1 deficiency, where both methylation and removal of -AAX were affected, the transport of isoprenylated proteins in isoprenylcysteine methyltransferase-deficient retinas was not dependent on methylation. This finding implies that the retention of the -AAX in PDE6 catalytic subunits in Rce1(-/-) mice is responsible for impeding their transport to the rod photoreceptor outer segment. In conclusion, lack of methylation of isoprenylcysteines leads to age-dependent photoreceptor dysfunction.


Assuntos
Células Fotorreceptoras de Vertebrados , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Eletrorretinografia , Transdução de Sinal Luminoso , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Processamento de Proteína Pós-Traducional/genética , Retina/citologia , Retina/metabolismo , Retina/fisiopatologia , Segmento Externo das Células Fotorreceptoras da Retina , Transtornos da Visão/genética , Transtornos da Visão/fisiopatologia
3.
J Clin Invest ; 123(11): 4681-94, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24216479

RESUMO

RAS is the most frequently mutated oncogene in human cancers. Despite decades of effort, anti-RAS therapies have remained elusive. Isoprenylcysteine carboxylmethyltransferase (ICMT) methylates RAS and other CaaX-containing proteins, but its potential as a target for cancer therapy has not been fully evaluated. We crossed a Pdx1-Cre;LSL-KrasG12D mouse, which is a model of pancreatic ductal adenocarcinoma (PDA), with a mouse harboring a floxed allele of Icmt. Surprisingly, we found that ICMT deficiency dramatically accelerated the development and progression of neoplasia. ICMT-deficient pancreatic ductal epithelial cells had a slight growth advantage and were resistant to premature senescence by a mechanism that involved suppression of cyclin-dependent kinase inhibitor 2A (p16INK4A) expression. ICMT deficiency precisely phenocopied Notch1 deficiency in the Pdx1-Cre;LSL-KrasG12D model by exacerbating pancreatic intraepithelial neoplasias, promoting facial papillomas, and derepressing Wnt signaling. Silencing ICMT in human osteosarcoma cells decreased Notch1 signaling in response to stimulation with cell-surface ligands. Additionally, targeted silencing of Ste14, the Drosophila homolog of Icmt, resulted in defects in wing development, consistent with Notch loss of function. Our data suggest that ICMT behaves like a tumor suppressor in PDA because it is required for Notch1 signaling.


Assuntos
Genes ras , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Metiltransferases/deficiência , Receptor Notch1/metabolismo , Animais , Animais Geneticamente Modificados , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Feminino , Humanos , Masculino , Metaplasia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Mutação , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Proteínas Metiltransferases/genética , Transdução de Sinais
4.
Genes Dev ; 27(17): 1903-16, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24013503

RESUMO

The tight control of gene expression at the level of both transcription and post-transcriptional RNA processing is essential for mammalian development. We here investigate the role of protein arginine methyltransferase 5 (PRMT5), a putative splicing regulator and transcriptional cofactor, in mammalian development. We demonstrate that selective deletion of PRMT5 in neural stem/progenitor cells (NPCs) leads to postnatal death in mice. At the molecular level, the absence of PRMT5 results in reduced methylation of Sm proteins, aberrant constitutive splicing, and the alternative splicing of specific mRNAs with weak 5' donor sites. Intriguingly, the products of these mRNAs are, among others, several proteins regulating cell cycle progression. We identify Mdm4 as one of these key mRNAs that senses the defects in the spliceosomal machinery and transduces the signal to activate the p53 response, providing a mechanistic explanation of the phenotype observed in vivo. Our data demonstrate that PRMT5 is a master regulator of splicing in mammals and uncover a new role for the Mdm4 pre-mRNA, which could be exploited for anti-cancer therapy.


Assuntos
Processamento Alternativo/genética , Proteínas Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA/genética , Spliceossomos/patologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Sistema Nervoso Central/patologia , Genes p53/genética , Células HCT116 , Células HEK293 , Homeostase/genética , Humanos , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Ligação Proteica , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética , Proteína-Arginina N-Metiltransferases , Proteínas Proto-Oncogênicas/genética , Precursores de RNA/genética , Transdução de Sinais , Spliceossomos/genética , Spliceossomos/metabolismo , Ubiquitina-Proteína Ligases/genética
5.
Development ; 139(20): 3806-16, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22991445

RESUMO

In the developing brain, neural progenitor cells switch differentiation competency by changing gene expression profiles that are governed partly by epigenetic control, such as histone modification, although the precise mechanism is unknown. Here we found that ESET (Setdb1), a histone H3 Lys9 (H3K9) methyltransferase, is highly expressed at early stages of mouse brain development but downregulated over time, and that ablation of ESET leads to decreased H3K9 trimethylation and the misregulation of genes, resulting in severe brain defects and early lethality. In the mutant brain, endogenous retrotransposons were derepressed and non-neural gene expression was activated. Furthermore, early neurogenesis was severely impaired, whereas astrocyte formation was enhanced. We conclude that there is an epigenetic role of ESET in the temporal and tissue-specific gene expression that results in proper control of brain development.


Assuntos
Encéfalo/embriologia , Células-Tronco Neurais/metabolismo , Neurogênese , Proteínas Metiltransferases/metabolismo , Animais , Astrócitos/metabolismo , Sequência de Bases , Encéfalo/metabolismo , Diferenciação Celular , Proliferação de Células , Regulação para Baixo , Epigênese Genética , Neurônios GABAérgicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase , Camundongos , Camundongos Transgênicos , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética , Retroelementos , Análise de Sequência de RNA
6.
J Biol Chem ; 286(47): 41115-24, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21921037

RESUMO

Setdb1/Eset is a histone H3 lysine 9 (H3K9)-specific methyltransferase that associates with various transcription factors to regulate gene expression via chromatin remodeling. Here, we report that Setdb1 associates with promyelocytic leukemia (Pml) protein from the early stage of mouse development and is a constitutive member of promyelocytic leukemia (PML)-nuclear bodies (PML-NBs) that have been linked to many cellular processes such as apoptosis, DNA damage responses, and transcriptional regulation. Arsenic treatment, which induces Pml degradation, caused Setdb1 signals to disappear. Setdb1 knockdown resulted in dismantlement of PML-NBs. Immunoprecipitation results demonstrated physical interactions between Setdb1 and Pml. Chromatin immunoprecipitation revealed that, within the frame of PML-NBs, Setdb1 binds the promoter of Id2 and suppresses its expression through installing H3K9 methylation. Our findings suggest that Setdb1 performs dual, but inseparable, functions at PML-NBs to maintain the structural integrity of PML-NBs and to control PML-NB-associated genes transcriptionally.


Assuntos
Núcleo Celular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Metiltransferases/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Arsênio/farmacologia , Núcleo Celular/efeitos dos fármacos , Implantação do Embrião , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Histona-Lisina N-Metiltransferase , Proteína 2 Inibidora de Diferenciação/deficiência , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Masculino , Camundongos , Células NIH 3T3 , Gravidez , Proteína da Leucemia Promielocítica , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética
7.
Cell Stem Cell ; 8(6): 676-87, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21624812

RESUMO

DNA methylation and histone H3 lysine 9 trimethylation (H3K9me3) play important roles in silencing of genes and retroelements. However, a comprehensive comparison of genes and repetitive elements repressed by these pathways has not been reported. Here we show that in mouse embryonic stem cells (mESCs), the genes upregulated after deletion of the H3K9 methyltransferase Setdb1 are distinct from those derepressed in mESC deficient in the DNA methyltransferases Dnmt1, Dnmt3a, and Dnmt3b, with the exception of a small number of primarily germline-specific genes. Numerous endogenous retroviruses (ERVs) lose H3K9me3 and are concomitantly derepressed exclusively in SETDB1 knockout mESCs. Strikingly, ~15% of upregulated genes are induced in association with derepression of promoter-proximal ERVs, half in the context of "chimeric" transcripts that initiate within these retroelements and splice to genic exons. Thus, SETDB1 plays a previously unappreciated yet critical role in inhibiting aberrant gene transcription by suppressing the expression of proximal ERVs.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Retrovirus Endógenos/genética , Regulação da Expressão Gênica/genética , Proteínas Metiltransferases/metabolismo , Transcrição Gênica/genética , Animais , Células Cultivadas , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/deficiência , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/virologia , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Lisina/metabolismo , Camundongos , Camundongos Knockout , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética , Retroelementos/genética , DNA Metiltransferase 3B
8.
Nature ; 464(7290): 927-31, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20164836

RESUMO

Endogenous retroviruses (ERVs), retrovirus-like elements with long terminal repeats, are widely dispersed in the euchromatic compartment in mammalian cells, comprising approximately 10% of the mouse genome. These parasitic elements are responsible for >10% of spontaneous mutations. Whereas DNA methylation has an important role in proviral silencing in somatic and germ-lineage cells, an additional DNA-methylation-independent pathway also functions in embryonal carcinoma and embryonic stem (ES) cells to inhibit transcription of the exogenous gammaretrovirus murine leukaemia virus (MLV). Notably, a recent genome-wide study revealed that ERVs are also marked by histone H3 lysine 9 trimethylation (H3K9me3) and H4K20me3 in ES cells but not in mouse embryonic fibroblasts. However, the role that these marks have in proviral silencing remains unexplored. Here we show that the H3K9 methyltransferase ESET (also called SETDB1 or KMT1E) and the Krüppel-associated box (KRAB)-associated protein 1 (KAP1, also called TRIM28) are required for H3K9me3 and silencing of endogenous and introduced retroviruses specifically in mouse ES cells. Furthermore, whereas ESET enzymatic activity is crucial for HP1 binding and efficient proviral silencing, the H4K20 methyltransferases Suv420h1 and Suv420h2 are dispensable for silencing. Notably, in DNA methyltransferase triple knockout (Dnmt1(-/-)Dnmt3a(-/-)Dnmt3b(-/-)) mouse ES cells, ESET and KAP1 binding and ESET-mediated H3K9me3 are maintained and ERVs are minimally derepressed. We propose that a DNA-methylation-independent pathway involving KAP1 and ESET/ESET-mediated H3K9me3 is required for proviral silencing during the period early in embryogenesis when DNA methylation is dynamically reprogrammed.


Assuntos
Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/virologia , Retrovirus Endógenos/genética , Inativação Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas Metiltransferases/metabolismo , Provírus/genética , Animais , Linhagem Celular , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/deficiência , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/genética , DNA Metiltransferase 3A , Células-Tronco Embrionárias/metabolismo , Fibroblastos , Deleção de Genes , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/deficiência , Histona-Lisina N-Metiltransferase/genética , Camundongos , Proteínas Nucleares/metabolismo , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética , Proteínas Repressoras/metabolismo , Proteína 28 com Motivo Tripartido , DNA Metiltransferase 3B
9.
Blood ; 112(4): 1357-65, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18502828

RESUMO

Hyperactive signaling through the RAS proteins is involved in the pathogenesis of many forms of cancer. The RAS proteins and many other intracellular signaling proteins are either farnesylated or geranylgeranylated at a carboxyl-terminal cysteine. That isoprenylcysteine is then carboxyl methylated by isoprenylcysteine carboxyl methyltransferase (ICMT). We previously showed that inactivation of Icmt mislocalizes the RAS proteins away from the plasma membrane and blocks RAS transformation of mouse fibroblasts, suggesting that ICMT could be a therapeutic target. However, nothing is known about the impact of inhibiting ICMT on the development of malignancies in vivo. In the current study, we tested the hypothesis that inactivation of Icmt would inhibit the development or progression of a K-RAS-induced myeloproliferative disease in mice. We found that inactivating Icmt reduced splenomegaly, the number of immature myeloid cells in peripheral blood, and tissue infiltration by myeloid cells. Moreover, in the absence of Icmt, the ability of K-RAS-expressing hematopoietic cells to form colonies in methylcellulose without exogenous growth factors was reduced dramatically. Finally, inactivating Icmt reduced lung tumor development and myeloproliferation phenotypes in a mouse model of K-RAS-induced cancer. We conclude that inactivation of Icmt ameliorates phenotypes of K-RAS-induced malignancies in vivo.


Assuntos
Transtornos Mieloproliferativos/etiologia , Proteínas Metiltransferases/deficiência , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Animais , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/prevenção & controle , Camundongos , Camundongos Knockout , Células Mieloides/patologia , Transtornos Mieloproliferativos/patologia , Esplenomegalia
10.
Mol Cell Biol ; 26(19): 7224-35, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16980624

RESUMO

DNA methylation is vital for proper chromatin structure and function in mammalian cells. Genetic removal of the enzymes that catalyze DNA methylation results in defective imprinting, transposon silencing, X chromosome dosage compensation, and genome stability. This epigenetic modification is interpreted by methyl-DNA binding domain (MBD) proteins. MBD proteins respond to methylated DNA by recruiting histone deacetylases (HDAC) and other transcription repression factors to the chromatin. The MBD2 protein is dispensable for animal viability, but it is implicated in the genesis of colon tumors. Here we report that the MBD2 protein is controlled by arginine methylation. We identify the protein arginine methyltransferase enzymes that catalyze this modification and show that arginine methylation inhibits the function of MBD2. Arginine methylation of MBD2 reduces MBD2-methyl-DNA complex formation, reduces MBD2-HDAC repression complex formation, and impairs the transcription repression function of MBD2 in cells. Our report provides a molecular description of a potential regulatory mechanism for an MBD protein family member. It is the first to demonstrate that protein arginine methyltransferases participate in the DNA methylation system of chromatin control.


Assuntos
Arginina/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Sequência de Aminoácidos , Células Cultivadas , Ilhas de CpG/genética , DNA/metabolismo , Proteínas de Ligação a DNA/química , Inativação Gênica , Células HeLa , Histona Desacetilases/metabolismo , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Ligação Proteica , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/deficiência , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica
11.
Methods Enzymol ; 407: 144-59, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16757321

RESUMO

After isoprenylation, the Ras proteins and other proteins terminating with a so-called CAAX motif undergo two additional modifications: (1) endoproteolytic cleavage of the -AAX by Ras converting enzyme 1 (Rce1) and (2) carboxyl methylation of the isoprenylated cysteine residue by isoprenylcysteine carboxyl methyltransferase (Icmt). Although CAAX protein isoprenylation has been studied in great detail, until recently, very little was known about the biological role and functional importance of Icmt in mammalian cells. Studies over the past few years, however, have begun to fill in the blanks. Genetic experiments showed that Icmt-deficient embryos die at mid-gestation, whereas conditional inactivation of Icmt in the liver, spleen, and bone marrow is not associated with obvious pathology. One potential explanation for the embryonic lethality is that Icmt is the only enzyme in mouse cells capable of methylating isoprenylated CAAX proteins--including the Ras proteins. Furthermore, in addition to the CAAX proteins, Icmt methylates the CXC class of isoprenylated Rab proteins. In the absence of carboxyl methylation, the Ras proteins are mislocalized away from the plasma membrane and exhibit a shift in electrophoretic mobility. Given the important role of oncogenic Ras proteins in human tumorigenesis and the mislocalization of Ras proteins in Icmt-deficient cells, it has been hypothesized that inhibition of Icmt could be a strategy to block Ras-induced oncogenic transformation. Recent data provide strong support to that hypothesis: conditional inactivation of Icmt in mouse embryonic fibroblasts and treatment of cells with a novel selective inhibitor of Icmt, termed cysmethynil, results in a striking inhibition of Ras-induced oncogenic transformation.


Assuntos
Membrana Celular/metabolismo , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Proteínas ras/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Cisteína/análogos & derivados , Cisteína/farmacologia , Diterpenos/farmacologia , Inibidores Enzimáticos/farmacologia , Camundongos , Camundongos Knockout , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas rab3 de Ligação ao GTP/metabolismo
12.
Proc Natl Acad Sci U S A ; 102(12): 4336-41, 2005 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-15784746

RESUMO

Many key regulatory proteins, including members of the Ras family of GTPases, are modified at their C terminus by a process termed prenylation. This processing is initiated by the addition of an isoprenoid lipid, and the proteins are further modified by a proteolytic event and methylation of the C-terminal prenylcysteine. Although the biological consequences of prenylation have been characterized extensively, the contributions of prenylcysteine methylation to the functions of the modified proteins are not well understood. This reaction is catalyzed by the enzyme isoprenylcysteine carboxyl methyltransferase (Icmt). Recent genetic disruption studies have provided strong evidence that blocking Icmt activity has profound consequences on oncogenic transformation. Here, we report the identification of a selective small-molecule inhibitor of Icmt, 2-[5-(3-methylphenyl)-1-octyl-1H-indol-3-yl]acetamide (cysmethynil). Cysmethynil treatment results in inhibition of cell growth in an Icmt-dependent fashion, demonstrating mechanism-based activity of the compound. Treatment of cancer cells with cysmethynil results in mislocalization of Ras and impaired epidermal growth factor signaling. In a human colon cancer cell line, cysmethynil treatment blocks anchorage-independent growth, and this effect is reversed by overexpression of Icmt. These findings provide a compelling rationale for development of Icmt inhibitors as another approach to anticancer drug development.


Assuntos
Acetamidas/farmacologia , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Proteínas Metiltransferases/antagonistas & inibidores , Acetamidas/química , Animais , Antineoplásicos/química , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Cães , Inibidores Enzimáticos/química , Humanos , Camundongos , Fenótipo , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas ras/metabolismo
13.
Arch Biochem Biophys ; 381(2): 225-34, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11032409

RESUMO

We reviewed here that protein isomerization is enhanced in amyloid-beta peptides (Abeta) and paired helical filaments (PHFs) purified from Alzheimer's disease (AD) brains. Biochemical analyses revealed that Abeta purified from senile plaques and vascular amyloid are isomerized at Asp-1 and Asp-7. A specific antibody recognizing isoAsp-23 of Abeta further suggested the isomerization of Abeta at Asp-23 in vascular amyloid as well as in the core of senile plaques. Biochemical analyses of purified PHFs also revealed that heterogeneous molecular weight tau contains L-isoaspartate at Asp-193, Asn-381, and Asp-387, indicating a modification, other than phosphorylation, that differentiates between normal tau and PHF tau. Since protein isomerization as L-isoaspartate causes structural changes and functional inactivation, or enhances the aggregation process, this modification is proposed as one of the progression factors in AD. Protein L-isoaspartyl methyltransferase (PIMT) is suggested to play a role in the repair of isomerized proteins containing L-isoaspartate. We show here that PIMT is upregulated in neurodegenerative neurons and colocalizes in neurofibrillary tangles (NFTs) in AD. Taken together with the enhanced protein isomerization in AD brains, it is implicated that the upregulated PIMT may associate with increased protein isomerization in AD. We also reviewed studies on PIMT-deficient mice that confirmed that PIMT plays a physiological role in the repair of isomerized proteins containing L-isoaspartate. The knockout study also suggested that the brain of PIMT-deficient mice manifested neurodegenerative changes concomitant with accumulation of L-isoaspartate. We discuss the pathological implications of protein isomerization in the neurodegeneration found in model mice and AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Ácido Aspártico/biossíntese , Degeneração Neural/metabolismo , Doença de Alzheimer/etiologia , Sequência de Aminoácidos , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Circulação Cerebrovascular , Humanos , Técnicas In Vitro , Isomerismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Proteína D-Aspartato-L-Isoaspartato Metiltransferase , Proteínas Metiltransferases/deficiência , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
14.
J Biol Chem ; 275(23): 17605-10, 2000 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-10747846

RESUMO

After isoprenylation and endoproteolytic processing, the Ras proteins are methylated at the carboxyl-terminal isoprenylcysteine. The importance of isoprenylation for targeting of Ras proteins to the plasma membrane is well established, but the importance of carboxyl methylation, which is carried out by isoprenylcysteine carboxyl methyltransferase (Icmt), is less certain. We used gene targeting to produce homozygous Icmt knockout embryonic stem cells (Icmt-/-). Lysates from Icmt-/- cells lacked the ability to methylate farnesyl-K-Ras4B or small-molecule Icmt substrates such as N-acetyl-S-geranylgeranyl-L-cysteine. To assess the impact of absent Icmt activity on the localization of K-Ras within cells, wild-type and Icmt-/- cells were transfected with a green fluorescent protein (GFP)-K-Ras fusion construct. As expected, virtually all of the GFP-K-Ras fusion in wild-type cells was localized along the plasma membrane. In contrast, a large fraction of the fusion in Icmt-/- cells was trapped within the cytoplasm, and fluorescence at the plasma membrane was reduced. Also, cell fractionation/Western blot studies revealed that a smaller fraction of the K-Ras in Icmt-/- cells was associated with the membranes. We conclude that carboxyl methylation of the isoprenylcysteine is important for proper K-Ras localization in mammalian cells.


Assuntos
Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Proteínas de Fluorescência Verde , Homozigoto , Fígado/enzimologia , Proteínas Luminescentes/análise , Camundongos , Camundongos Knockout , Proteínas Metiltransferases/deficiência , Proteínas Recombinantes de Fusão/análise , Células-Tronco/citologia , Células-Tronco/enzimologia , Células-Tronco/fisiologia
16.
J Neurosci ; 18(6): 2063-74, 1998 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9482793

RESUMO

Protein L-isoaspartyl methyltransferase (PIMT) is suggested to play a role in the repair of aged protein spontaneously incorporated with isoaspartyl residues. We generated PIMT-deficient mice by targeted disruption of the PIMT gene to elucidate the biological role of the gene in vivo. PIMT-deficient mice died from progressive epileptic seizures with grand mal and myoclonus between 4 and 12 weeks of age. An anticonvulsive drug, dipropylacetic acid (DPA), improved their survival but failed to cure the fatal outcome. L-Isoaspartatate, the putative substrate for PIMT, was increased ninefold in the brains of PIMT-deficient mice. The brains of PIMT-deficient mice started to enlarge after 4 weeks of age when the apical dendrites of pyramidal neurons in cerebral cortices showed aberrant arborizations with disorganized microtubules. We conclude that methylation of modified proteins with isoaspartyl residues is essential for the maintenance of a mature CNS and that a deficiency in PIMT results in fatal progressive epilepsy in mice.


Assuntos
Epilepsia/etiologia , Epilepsia/fisiopatologia , Proteínas Metiltransferases/deficiência , Animais , Ácido Aspártico/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Progressão da Doença , Epilepsia/mortalidade , Epilepsia Tônico-Clônica/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Fenótipo , Proteína D-Aspartato-L-Isoaspartato Metiltransferase , Proteínas Metiltransferases/genética , Estereoisomerismo
17.
Proc Natl Acad Sci U S A ; 94(12): 6132-7, 1997 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-9177182

RESUMO

L-Asparaginyl and L-aspartyl residues in proteins are subject to spontaneous degradation reactions that generate isomerized and racemized aspartyl derivatives. Proteins containing L-isoaspartyl and D-aspartyl residues can have altered structures and diminished biological activity. These residues are recognized by a highly conserved cytosolic enzyme, the protein L-isoaspartate(D-aspartate) O-methyltransferase (EC 2.1.1.77). The enzymatic methyl esterification of these abnormal residues in vitro can lead to their conversion (i.e., repair) to normal L-aspartyl residues and should therefore prevent the accumulation of potentially dysfunctional proteins in vivo as cells and tissues age. Particularly high levels of the repair methyltransferase are present in the brain, although enyzme activity is present in all vertebrate tissues. To define the physiological relevance of this protein-repair pathway and to determine whether deficient protein repair would cause central nervous system dysfunction, we used gene targeting in mouse embryonic stem cells to generate protein L-isoaspartate(D-aspartate) O-methyltransferase-deficient mice. Analyses of tissues from methyltransferase knockout mice revealed a striking accumulation of protein substrates for this enzyme in the cytosolic fraction of brain, heart, liver, and erythrocytes. The knockout mice showed significant growth retardation and succumbed to fatal seizures at an average of 42 days after birth. These results suggest that the ability of mice to repair L-isoaspartyl- and D-aspartyl-containing proteins is essential for normal growth and for normal central nervous system function.


Assuntos
Transtornos do Crescimento/enzimologia , Proteínas Metiltransferases/deficiência , Proteínas/metabolismo , Convulsões/enzimologia , Animais , Asparagina/análise , Ácido Aspártico/análise , Peso Corporal , Primers do DNA , Morte Súbita , Feminino , Transtornos do Crescimento/genética , Heterozigoto , Humanos , Recém-Nascido , Masculino , Camundongos , Camundongos Knockout , Pentilenotetrazol , Reação em Cadeia da Polimerase , Proteína D-Aspartato-L-Isoaspartato Metiltransferase , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Proteínas Recombinantes/metabolismo , Mapeamento por Restrição , Convulsões/induzido quimicamente , Convulsões/genética
18.
N Engl J Med ; 306(14): 821-5, 1982 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-7062962

RESUMO

We studied protein-carboxyl methylase, an enzyme involved in the regulation of cellular locomotion in both bacteria and leukocytes, in semen from 22 normal fertile men, 10 vasectomized volunteers, and nine infertile patients with nonmotile spermatozoa. In normally motile spermatozoa protein-carboxyl methylase activity was 68.8 +/- 5.5 pmol per milligram of protein (mean +/- S.E.). On the other hand, the enzyme activity in nonmotile spermatozoa from the infertile patients was low (17.4 +/- 3.4 pmol per milligram of protein) and similar to that in the cellular debris from the vasectomized volunteers (10.4 +/- 1.3 pmol per milligram of protein). The low enzyme activity in the infertile patients was not caused by the presence of dead spermatozoa or spermatozoa with leaky plasma membranes, since mitochondrial protein synthesis and lactate dehydrogenase activity were normal in these patients. The deficiency of protein-carboxyl methylase activity in nonmotile sperm is probably not due to a primary genetic defect, since the enzyme activity is normal in the red cells of these patients and spontaneous recovery of motility is associated with the return of enzyme activity.


PIP: Protein-carboxyl methylase, and enzyme involved in the regulation of cellular locomotionn in both bacteria and leukocytes was studied in semen from 22 normal, fertile men, 10 vasectomized volunteers, and 9 infertile patients with nonmotile spermatozoa. In normally motile spermatozoa, the protein-carboxyl methylase activity was 68.8 +or- 5.5 pmol/mg of protein (mean +or- S.E.). On the other hand, the enzyme activity in nonmotile spermatozoa from the infertile patients was low (17.4 +or- 3.4 pmol/mg of protein) and similar to that in the cellular debris from the vasectomized volunteers (10.4 +or- 1.3 pmol/mg of protein). The low enzyme activity in the infertile group was not caused by the presence of dead spermatozoa or spermatosoa with leaky plasma membranes, since mitochondrial protein synthesis and lactate dehydrogenase activity were normal in these patients. The deficiency of protein-carboxyl methylase activity in nonmotile sperm is probably not due to a primary genetic defect since the enzyme activity is normal in the red cells of these patients and spontaneous recovery of motility is associated with the return of enzyme activity.


Assuntos
Infertilidade Masculina/enzimologia , Proteínas Metiltransferases/deficiência , Proteína O-Metiltransferase/deficiência , Motilidade dos Espermatozoides , Espermatozoides/enzimologia , Adulto , Humanos , Masculino , Sêmen/enzimologia , Espermatozoides/ultraestrutura , Vasectomia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...