Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Heart Rhythm ; 20(8): 1158-1166, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37164047

RESUMO

BACKGROUND: Truncating variants in filamin C (FLNC) can cause arrhythmogenic cardiomyopathy (ACM) through haploinsufficiency. Noncanonical splice-altering variants may contribute to this phenotype. OBJECTIVE: The purpose of this study was to investigate the clinical and functional consequences of a recurrent FLNC intronic variant of uncertain significance (VUS), c.970-4A>G. METHODS: Clinical data in 9 variant heterozygotes from 4 kindreds were obtained from 5 tertiary health care centers. We used in silico predictors and functional studies with peripheral blood and patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Isolated RNA was studied by reverse transcription polymerase chain reaction. iPSC-CMs were further characterized at baseline and after nonsense-mediated decay (NMD) inhibition, using quantitative polymerase chain reaction (qPCR), RNA-sequencing, and cellular electrophysiology. American College of Medical Genetics and Genomics (ACMG) criteria were used to adjudicate variant pathogenicity. RESULTS: Variant heterozygotes displayed a spectrum of disease phenotypes, spanning from mild ventricular dysfunction with palpitations to severe ventricular arrhythmias requiring device shocks or progressive cardiomyopathy requiring heart transplantation. Consistent with in silico predictors, the c.970-4A>G FLNC variant activated a cryptic splice acceptor site, introducing a 3-bp insertion containing a premature termination codon. NMD inhibition upregulated aberrantly spliced transcripts by qPCR and RNA-sequencing. Patch clamp studies revealed irregular spontaneous action potentials, increased action potential duration, and increased sodium late current in proband-derived iPSC-CMs. These findings fulfilled multiple ACMG criteria for pathogenicity. CONCLUSION: Clinical, in silico, and functional evidence support the prediction that the intronic c.970-4A>G VUS disrupts splicing and drives ACM, enabling reclassification from VUS to pathogenic.


Assuntos
Cardiomiopatias , Humanos , Cardiomiopatias/genética , Códon sem Sentido , Filaminas/genética , Mutação , Miócitos Cardíacos , RNA/genética
2.
Genet Med ; 25(7): 100838, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37057673

RESUMO

PURPOSE: Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) regulates cell growth in response to nutritional status. Central to the mTORC1 function is the Rag-GTPase heterodimer. One component of the Rag heterodimer is RagC (Ras-related GTP-binding protein C), which is encoded by the RRAGC gene. METHODS: Genetic testing via trio exome sequencing was applied to identify the underlying disease cause in 3 infants with dilated cardiomyopathy, hepatopathy, and brain abnormalities, including pachygyria, polymicrogyria, and septo-optic dysplasia. Studies in patient-derived skin fibroblasts and in a HEK293 cell model were performed to investigate the cellular consequences. RESULTS: We identified 3 de novo missense variants in RRAGC (NM_022157.4: c.269C>A, p.(Thr90Asn), c.353C>T, p.(Pro118Leu), and c.343T>C, p.(Trp115Arg)), which were previously reported as occurring somatically in follicular lymphoma. Studies of patient-derived fibroblasts carrying the p.(Thr90Asn) variant revealed increased cell size, as well as dysregulation of mTOR-related p70S6K (ribosomal protein S6 kinase 1) and transcription factor EB signaling. Moreover, subcellular localization of mTOR was decoupled from metabolic state. We confirmed the key findings for all RRAGC variants described in this study in a HEK293 cell model. CONCLUSION: The above results are in line with a constitutive overactivation of the mTORC1 pathway. Our study establishes de novo missense variants in RRAGC as cause of an early-onset mTORopathy with unfavorable prognosis.


Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Monoméricas de Ligação ao GTP , Serina-Treonina Quinases TOR , Humanos , Lactente , Fibroblastos/metabolismo , Doenças Genéticas Inatas/genética , Células HEK293 , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/genética , Mutação de Sentido Incorreto , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
3.
J Clin Invest ; 131(6)2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33497358

RESUMO

Hirschsprung disease (HSCR) is the most frequent developmental anomaly of the enteric nervous system, with an incidence of 1 in 5000 live births. Chronic intestinal pseudo-obstruction (CIPO) is less frequent and classified as neurogenic or myogenic. Isolated HSCR has an oligogenic inheritance with RET as the major disease-causing gene, while CIPO is genetically heterogeneous, caused by mutations in smooth muscle-specific genes. Here, we describe a series of patients with developmental disorders including gastrointestinal dysmotility, and investigate the underlying molecular bases. Trio-exome sequencing led to the identification of biallelic variants in ERBB3 and ERBB2 in 8 individuals variably associating HSCR, CIPO, peripheral neuropathy, and arthrogryposis. Thorough gut histology revealed aganglionosis, hypoganglionosis, and intestinal smooth muscle abnormalities. The cell type-specific ErbB3 and ErbB2 function was further analyzed in mouse single-cell RNA sequencing data and in a conditional ErbB3-deficient mouse model, revealing a primary role for ERBB3 in enteric progenitors. The consequences of the identified variants were evaluated using quantitative real-time PCR (RT-qPCR) on patient-derived fibroblasts or immunoblot assays on Neuro-2a cells overexpressing WT or mutant proteins, revealing either decreased expression or altered phosphorylation of the mutant receptors. Our results demonstrate that dysregulation of ERBB3 or ERBB2 leads to a broad spectrum of developmental anomalies, including intestinal dysmotility.


Assuntos
Deficiências do Desenvolvimento/genética , Pseudo-Obstrução Intestinal/genética , Mutação , Neuregulina-1/genética , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Adolescente , Animais , Pré-Escolar , Deficiências do Desenvolvimento/patologia , Modelos Animais de Doenças , Feminino , Motilidade Gastrointestinal/genética , Doença de Hirschsprung/genética , Doença de Hirschsprung/patologia , Humanos , Recém-Nascido , Pseudo-Obstrução Intestinal/patologia , Masculino , Camundongos , Modelos Moleculares , Linhagem , Fenótipo , Gravidez , Receptor ErbB-2/química , Receptor ErbB-3/química , Receptor ErbB-3/deficiência
4.
Eur J Med Genet ; 64(1): 104120, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33278652

RESUMO

Complex I deficiency is the most common pediatric mitochondrial disease. It can cause a wide range of clinical disorders, including Leigh syndrome. TIMMDC1 encodes an assembly protein of complex I and has been recently associated with early onset mitochondrial disease in three unrelated families. In all three families the same homozygous deep intronic variant was identified leading to inclusion of a new exon resulting in a frameshift and premature stop codon (c.596 + 2146A > G, p.Gly199_Thr200ins5*). Herein, we describe two brothers of Dutch descent, presenting in infancy with hypotonia and respiratory insufficiency and a rapidly progressive and fatal disease course. Laboratory findings and metabolic investigations revealed no specific abnormalities, notably no raised plasma lactate. MRI showed transient lesions in the basal ganglia of brother 1. A muscle biopsy demonstrated complex I deficiency in brother 2. Exome sequencing yielded a novel heterozygous TIMMDC1 variant: c.385C > T, p.(Arg129*). Targeted sequencing revealed the previously published deep intronic variant c.596 + 2146A > G, p.(Gly199_Thr200ins5*) on the second allele which is not detected by exome sequencing. In summary, we present the fourth family with TIMMDC1-related disease, with a novel nonsense variant. This report illustrates the importance of considering mitochondrial disease even when laboratory findings are normal, and the added value of targeted sequencing of introns.


Assuntos
Doenças Mitocondriais/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Fenótipo , Gânglios da Base/diagnóstico por imagem , Códon sem Sentido , Diagnóstico Tardio , Heterozigoto , Humanos , Lactente , Íntrons , Ácido Láctico/sangue , Masculino , Doenças Mitocondriais/diagnóstico , Proteínas de Transporte da Membrana Mitocondrial/deficiência , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Linhagem
5.
Hum Mutat ; 41(6): 1091-1111, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32112656

RESUMO

Filamin C (FLNC) variants are associated with cardiac and muscular phenotypes. Originally, FLNC variants were described in myofibrillar myopathy (MFM) patients. Later, high-throughput screening in cardiomyopathy cohorts determined a prominent role for FLNC in isolated hypertrophic and dilated cardiomyopathies (HCM and DCM). FLNC variants are now among the more prevalent causes of genetic DCM. FLNC-associated DCM is associated with a malignant clinical course and a high risk of sudden cardiac death. The clinical spectrum of FLNC suggests different pathomechanisms related to variant types and their location in the gene. The appropriate functioning of FLNC is crucial for structural integrity and cell signaling of the sarcomere. The secondary protein structure of FLNC is critical to ensure this function. Truncating variants with subsequent haploinsufficiency are associated with DCM and cardiac arrhythmias. Interference with the dimerization and folding of the protein leads to aggregate formation detrimental for muscle function, as found in HCM and MFM. Variants associated with HCM are predominantly missense variants, which cluster in the ROD2 domain. This domain is important for binding to the sarcomere and to ensure appropriate cell signaling. We here review FLNC genotype-phenotype correlations based on available evidence.


Assuntos
Cardiomiopatias/genética , Filaminas/genética , Doenças Musculares/genética , Animais , Arritmias Cardíacas/genética , Cardiomiopatia Dilatada/genética , Modelos Animais de Doenças , Estudos de Associação Genética , Humanos , Mutação , Miopatias Congênitas Estruturais/genética
6.
Neuromuscul Disord ; 29(9): 693-697, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31488384

RESUMO

We report a novel mitochondrial m.4414T>C variant in the mt-tRNAMet (MT-TM) gene in an adult patient with chronic progressive external ophthalmoplegia and myopathy whose muscle biopsy revealed focal cytochrome c oxidase (COX)-deficient and ragged red fibres. The m.4414T>C variant occurs at a strongly evolutionary conserved sequence position, disturbing a canonical base pair and disrupting the secondary and tertiary structure of the mt-tRNAMet. Definitive evidence of pathogenicity is provided by clear segregation of m.4414T>C mutant levels with COX deficiency in single muscle fibres. Interestingly, the variant is present in skeletal muscle at relatively low levels (30%) and undetectable in accessible, non-muscle tissues from the patient and her asymptomatic brother, emphasizing the continuing requirement for a diagnostic muscle biopsy as the preferred tissue for mtDNA genetic investigations of mt-tRNA variants leading to mitochondrial myopathy.


Assuntos
DNA Mitocondrial/genética , Músculo Esquelético/patologia , Oftalmoplegia Externa Progressiva Crônica/genética , RNA de Transferência de Metionina/genética , Idoso , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Humanos , Músculo Esquelético/metabolismo , Mutação , Índice de Gravidade de Doença
7.
J Med Genet ; 54(10): 693-697, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28668821

RESUMO

BACKGROUND: Preimplantation genetic diagnosis (PGD) is a reproductive strategy for mitochondrial DNA (mtDNA) mutation carriers, strongly reducing their risk of affected offspring. Embryos either without the mutation or with mutation load below the phenotypic threshold are transferred to the uterus. Because of incidental heteroplasmy deviations in single blastomere and the relatively limited data available, we so far preferred relying on two blastomeres rather than one. Considering the negative effect of a two-blastomere biopsy protocol compared with a single-blastomere biopsy protocol on live birth delivery rate, we re-evaluated the error rate in our current dataset. METHODS: For the m.3243A>G mutation, sufficient embryos/blastomeres were available for a powerful analysis. The diagnostic error rate, defined as a potential false-negative result, based on a threshold of 15%, was determined in 294 single blastomeres analysed in 73 embryos of 9 female m.3243A>G mutation carriers. RESULTS: Only one out of 294 single blastomeres (0.34%) would have resulted in a false-negative diagnosis. False-positive diagnoses were not detected. CONCLUSION: Our findings support a single-blastomere biopsy PGD protocol for the m.3243A>G mutation as the diagnostic error rate is very low. As in the early preimplantation embryo no mtDNA replication seems to occur and the mtDNA is divided randomly among the daughter cells, we conclude this result to be independent of the specific mutation and therefore applicable to all mtDNA mutations.


Assuntos
Blastômeros , DNA Mitocondrial/genética , Testes Genéticos/métodos , Diagnóstico Pré-Implantação/métodos , Biópsia , Blastocisto , Erros de Diagnóstico , Feminino , Heterozigoto , Humanos , Mutação , Gravidez
8.
Clin Cancer Res ; 15(12): 3990-7, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19509152

RESUMO

PURPOSE: The transcription factors GATA4 and GATA5 are involved in gastrointestinal development and are inactivated by promoter hypermethylation in colorectal cancer. Here, we evaluated GATA4/5 promoter methylation as potential biomarkers for noninvasive colorectal cancer detection, and investigated the role of GATA4/5 in colorectal cancer. EXPERIMENTAL DESIGN: Promoter methylation of GATA4/5 was analyzed in colorectal tissue and fecal DNA from colorectal cancer patients and healthy controls using methylation-specific PCR. The potential function of GATA4/5 as tumor suppressors was studied by inducing GATA4/5 overexpression in human colorectal cancer cell lines. RESULTS: GATA4/5 methylation was observed in 70% (63/90) and 79% (61/77) of colorectal carcinomas, respectively, and was independent of clinicopathologic features. Methylation frequencies in normal colon tissues from noncancerous controls were 6% (5 of 88, GATA4; P < 0.001) and 13% (13 of 100, GATA5; P < 0.001). GATA4/5 overexpression suppressed colony formation (P < 0.005), proliferation (P < 0.001), migration (P < 0.05), invasion (P < 0.05), and anchorage-independent growth (P < 0.0001) of colorectal cancer cells. Examination of GATA4 methylation in fecal DNA from two independent series of colorectal cancer patients and controls yielded a sensitivity of 71% [95% confidence interval (95% CI), 55-88%] and specificity of 84% (95% CI, 74-95%) for colorectal cancer detection in the training set, and a sensitivity of 51% (95% CI, 37-65%) and specificity of 93% (95% CI, 84-100%) in the validation set. CONCLUSIONS: Methylation of GATA4/5 is a common and specific event in colorectal carcinomas, and GATA4/5 exhibit tumor suppressive effects in colorectal cancer cells in vitro. GATA4 methylation in fecal DNA may be of interest for colorectal cancer detection.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma/diagnóstico , Neoplasias Colorretais/diagnóstico , Metilação de DNA , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA5/genética , Genes Supressores de Tumor , Carcinoma/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Ilhas de CpG/genética , Ilhas de CpG/fisiologia , Fezes/química , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Estudos Retrospectivos
9.
J Natl Cancer Inst ; 101(13): 916-27, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19535783

RESUMO

BACKGROUND: Identification of hypermethylated tumor suppressor genes in body fluids is an appealing strategy for the noninvasive detection of colorectal cancer. Here we examined the role of N-Myc downstream-regulated gene 4 (NDRG4) as a novel tumor suppressor and biomarker in colorectal cancer. METHODS: NDRG4 promoter methylation was analyzed in human colorectal cancer cell lines, colorectal tissue, and noncancerous colon mucosa by using methylation-specific polymerase chain reaction (PCR) and bisulfite sequencing. NDRG4 mRNA and protein expression were studied using real-time-PCR and immunohistochemistry, respectively. Tumor suppressor functions of NDRG4 were examined by colony formation, cell proliferation, and migration and invasion assays in colorectal cancer cell lines that were stably transfected with an NDRG4 expression construct. Quantitative methylation-specific PCR was used to examine the utility of NDRG4 promoter methylation as a biomarker in fecal DNA from 75 colorectal cancer patients and 75 control subjects. All P values are two-sided. RESULTS: The prevalence of NDRG4 promoter methylation in two independent series of colorectal cancers was 86% (71/83) and 70% (128/184) compared with 4% (2/48) in noncancerous colon mucosa (P < .001). NDRG4 mRNA and protein expression were decreased in colorectal cancer tissue compared with noncancerous colon mucosa. NDRG4 overexpression in colorectal cancer cell lines suppressed colony formation (P = .014), cell proliferation (P < .001), and invasion (P < .001). NDRG4 promoter methylation analysis in fecal DNA from a training set of colorectal cancer patients and control subjects yielded a sensitivity of 61% (95% confidence interval [CI] = 43% to 79%) and a specificity of 93% (95% CI = 90% to 97%). An independent test set of colorectal cancer patients and control subjects yielded a sensitivity of 53% (95% CI = 39% to 67%) and a specificity of 100% (95% CI = 86% to 100%). CONCLUSIONS: NDRG4 is a candidate tumor suppressor gene in colorectal cancer whose expression is frequently inactivated by promoter methylation. NDRG4 promoter methylation is a potential biomarker for the noninvasive detection of colorectal cancer in stool samples.


Assuntos
Adenoma/química , Biomarcadores Tumorais/análise , Carcinoma/química , Neoplasias Colorretais/química , Metilação de DNA , Fezes/química , Genes Supressores de Tumor , Mucosa Intestinal/química , Proteínas Musculares/análise , Proteínas Musculares/genética , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/genética , Adenoma/diagnóstico , Adenoma/genética , Adenoma/prevenção & controle , Adulto , Idoso , Biomarcadores Tumorais/genética , Carcinoma/diagnóstico , Carcinoma/genética , Carcinoma/prevenção & controle , Estudos de Casos e Controles , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/genética , Neoplasias Colorretais/prevenção & controle , Detecção Precoce de Câncer , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Reação em Cadeia da Polimerase , Prevalência , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Estudos Retrospectivos , Sensibilidade e Especificidade , Ensaio Tumoral de Célula-Tronco , Regulação para Cima
10.
Cancer Res ; 67(9): 4138-48, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17483324

RESUMO

Tumor angiogenesis requires intricate regulation of gene expression in endothelial cells. We recently showed that DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors directly repress endothelial cell growth and tumor angiogenesis, suggesting that epigenetic modifications mediated by DNMTs and HDAC are involved in regulation of endothelial cell gene expression during tumor angiogenesis. To understand the mechanisms behind the epigenetic regulation of tumor angiogenesis, we used microarray analysis to perform a comprehensive screen to identify genes down-regulated in tumor-conditioned versus quiescent endothelial cells, and reexpressed by 5-aza-2'-deoxycytidine (DAC) and trichostatin A (TSA). Among the 81 genes identified, 77% harbored a promoter CpG island. Validation of mRNA levels of a subset of genes confirmed significant down-regulation in tumor-conditioned endothelial cells and reactivation by treatment with a combination of DAC and TSA, as well as by both compounds separately. Silencing of these genes in tumor-conditioned endothelial cells correlated with promoter histone H3 deacetylation and loss of H3 lysine 4 methylation, but did not involve DNA methylation of promoter CpG islands. For six genes, down-regulation in microdissected human tumor endothelium was confirmed. Functional validation by RNA interference revealed that clusterin, fibrillin 1, and quiescin Q6 are negative regulators of endothelial cell growth and angiogenesis. In summary, our data identify novel angiogenesis-suppressing genes that become silenced in tumor-conditioned endothelial cells in association with promoter histone modifications and reactivated by DNMT and HDAC inhibitors through reversal of these epigenetic modifications, providing a mechanism for epigenetic regulation of tumor angiogenesis.


Assuntos
Neoplasias do Colo/irrigação sanguínea , Células Endoteliais/fisiologia , Inativação Gênica , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Células CACO-2 , Clusterina/genética , Neoplasias do Colo/genética , Metilação de DNA , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Epigênese Genética , Fibrilina-1 , Fibrilinas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteínas dos Microfilamentos/genética , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Oxirredutases atuantes sobre Doadores de Grupo Enxofre , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiorredoxinas/genética
11.
Biochim Biophys Acta ; 1775(1): 76-91, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16930846

RESUMO

Aberrant epigenetic silencing of tumor suppressor genes by promoter DNA hypermethylation and histone deacetylation plays an important role in the pathogenesis of cancer. The potential reversibility of epigenetic abnormalities encouraged the development of pharmacologic inhibitors of DNA methylation and histone deacetylation as anti-cancer therapeutics. (Pre)clinical studies of DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors have yielded encouraging results, especially against hematologic malignancies. Recently, several studies demonstrated that DNMT and HDAC inhibitors are also potent angiostatic agents, inhibiting (tumor) endothelial cells and angiogenesis in vitro and in vivo. By reactivation of epigenetically silenced tumor suppressor genes with angiogenesis inhibiting properties, DNMT and HDAC inhibitors might indirectly - via their effects on tumor cells - decrease tumor angiogenesis in vivo. However, this does not explain the direct angiostatic effects of these agents, which can be unraveled by gene expression studies and examination of epigenetic promoter modifications in endothelial cells treated with DNMT and HDAC inhibitors. Clearly, the dual targeting of epigenetic therapy on both tumor cells and tumor vasculature makes them attractive combinatorial anti-tumor therapeutics. Here we review the therapeutic potential of DNMT and HDAC inhibitors as anti-cancer drugs, as evaluated in clinical trials, and their angiostatic activities, apart from their inhibitory effects on tumor cells.


Assuntos
Antineoplásicos/uso terapêutico , Metilases de Modificação do DNA/antagonistas & inibidores , Inibidores Enzimáticos/uso terapêutico , Epigênese Genética/efeitos dos fármacos , Inibidores de Histona Desacetilases , Neoplasias/genética , Inibidores da Angiogênese/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico
12.
Cancer Res ; 66(22): 10770-7, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17108113

RESUMO

Tumors can escape from immunity by repressing leukocyte adhesion molecule expression on tumor endothelial cells and by rendering endothelial cells unresponsive to inflammatory activation. This endothelial cell anergy is induced by angiogenic growth factors and results in reduced leukocyte-vessel wall interactions, thereby attenuating infiltration of leukocytes into the tumor. This report describes a novel mechanism of endothelial cell anergy regulation. We recently reported that DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors have angiostatic activity. Here, we studied whether epigenetic mechanisms regulate this angiogenesis-mediated escape from immunity. We found that DNMT inhibitors 5-aza-2'-deoxycytidine and zebularine, as well as HDAC inhibitor trichostatin A, reexpressed intercellular adhesion molecule-1 (ICAM-1) on tumor-conditioned endothelial cells in vitro, resulting in restored leukocyte-endothelial cell adhesion. In addition, treatment with DNMT or HDAC inhibitors in vivo also restored ICAM-1 expression on tumor endothelial cells from two different mouse tumor models. Furthermore, leukocyte-vessel wall interactions in mouse tumors were increased by these compounds, as measured by intravital microscopy, resulting in enhanced leukocyte infiltration. We show that ICAM-1 down-regulation in tumor endothelial cells is associated with ICAM-1 promoter histone H3 deacetylation and loss of histone H3 Lys(4) methylation but not with DNA hypermethylation. In conclusion, our data show that ICAM-1 is epigenetically silenced in tumor endothelial cells by promoter histone modifications, which can be overcome by DNMT and HDAC inhibitors, suggesting a new molecular mechanism based on which novel therapeutic approaches for cancer can be pursued.


Assuntos
Células Endoteliais/fisiologia , Histonas/genética , Molécula 1 de Adesão Intercelular/genética , Acetilação , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Anergia Clonal , Citidina/análogos & derivados , Citidina/farmacologia , Metilases de Modificação do DNA/antagonistas & inibidores , Metilases de Modificação do DNA/metabolismo , Regulação para Baixo , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Epigênese Genética , Inativação Gênica , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Leucócitos/patologia , Melanoma Experimental/sangue , Melanoma Experimental/patologia , Metilação , Camundongos , Camundongos Endogâmicos C57BL
13.
J Natl Cancer Inst ; 98(13): 932-6, 2006 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-16818857

RESUMO

The inhibition of angiogenesis is a promising avenue for cancer treatment. Although some angiostatic compounds are in the process of development and testing, these often prove ineffective in vivo or have unwanted side effects. We have designed, synthesized, and evaluated a small library of nonpeptidic, calixarene-based protein surface topomimetics that display chemical substituents to approximate the molecular dimensions and amphipathic features (hydrophobic and positively charged residues) of the antiangiogenic peptide anginex, which, like many antiangiogenic proteins, consists primarily of an antiparallel beta-sheet structure as the functional unit. Two of the topomimetics (0118 and 1097) were potent angiogenesis inhibitors in vitro, as determined by endothelial cell proliferation, migration, and chick embryo chorioallantoic membrane assays. Moreover, both compounds were highly effective at inhibiting tumor angiogenesis and growth in two mouse models (MA148 human ovarian carcinoma and B16 murine melanoma). Our results demonstrate the feasibility of designing nonpeptidic protein surface topomimetics as novel pharmaceutical agents for clinical intervention against cancer through angiostatic or other mechanisms.


Assuntos
Inibidores da Angiogênese/farmacologia , Calixarenos/farmacologia , Melanoma Experimental/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Proteínas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Desenho de Fármacos , Estudos de Viabilidade , Feminino , Humanos , Melanoma Experimental/irrigação sanguínea , Camundongos , Neoplasias Ovarianas/irrigação sanguínea , Peptídeos
14.
Mol Cancer Ther ; 5(2): 467-75, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16505122

RESUMO

Inhibitors of DNA methyltransferases (DNMT) and histone deacetylases can reactivate epigenetically silenced tumor suppressor genes and thereby decrease tumor cell growth. Little, however, is known on the effects of these compounds in endothelial cell biology and tumor angiogenesis. Here, we show that the DNMT inhibitors 5-aza-2'-deoxycytidine and zebularine markedly decrease vessel formation in different tumor models. We show that DNMT inhibitors are antiproliferative for tumor-conditioned endothelial cells, without affecting endothelial cell apoptosis and migration. Furthermore, these compounds inhibit angiogenesis in vitro and in vivo as shown by inhibition of endothelial cells sprouting in a three-dimensional gel and inhibition of microvessel formation in the chorioallantoic membrane, respectively. 5-Aza-2'-deoxycytidine, as well as the histone deacetylase inhibitor trichostatin A, reactivates the growth-inhibiting genes TSP1, JUNB, and IGFBP3, which are suppressed in tumor-conditioned endothelial cells. Despite enhanced DNMT activity and increased overall genomic methylation levels in tumor-conditioned endothelial cells, silencing of these genes seemed not to be regulated by direct promoter hypermethylation. For IGFBP3, gene expression in endothelial cells correlated with histone H3 acetylation patterns. In conclusion, our data show that DNMT inhibitors have angiostatic activity in addition to their inhibitory effects on tumor cells. This dual action of these compounds makes them promising anticancer therapeutics.


Assuntos
Inibidores da Angiogênese/farmacologia , Azacitidina/farmacologia , Citidina/análogos & derivados , Metilases de Modificação do DNA/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Neovascularização Patológica/enzimologia , Neovascularização Fisiológica/efeitos dos fármacos , Acetilação , Inibidores da Angiogênese/uso terapêutico , Animais , Azacitidina/uso terapêutico , Movimento Celular/efeitos dos fármacos , Citidina/farmacologia , Citidina/uso terapêutico , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Inibidores Enzimáticos/uso terapêutico , Expressão Gênica/efeitos dos fármacos , Histonas/antagonistas & inibidores , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/enzimologia , Melanoma Experimental/genética , Camundongos , Camundongos Mutantes , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Fisiológica/genética , Trombospondina 1/genética
15.
Cell Oncol ; 26(5-6): 291-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15623939

RESUMO

Methylation-specific PCR (MSP) is a simple, quick and cost-effective method to analyze the DNA methylation status of virtually any group of CpG sites within a CpG island. The technique comprises two parts: (1) sodium bisulfite conversion of unmethylated cytosine's to uracil under conditions whereby methylated cytosines remains unchanged and (2) detection of the bisulfite induced sequence differences by PCR using specific primer sets for both unmethylated and methylated DNA. This review discusses the critical parameters of MSP and presents an overview of the available MSP variants and the (clinical) applications.


Assuntos
Metilação de DNA , Reação em Cadeia da Polimerase/métodos , Biomarcadores Tumorais , Cromatografia Líquida de Alta Pressão , Ilhas de CpG , Citosina/química , Primers do DNA/química , Inativação Gênica , Humanos , Software , Sulfitos/farmacologia , Uracila/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA