Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Cancer Treat Res ; 191: 217-244, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39133410

RESUMO

The scientific innovations have emphasized the importance of diet for one's health and wellbeing. The genetic revolution has enhanced our understanding about the effect of nutrients on genomic and transcriptomic profiles and gene-nutrition interactions (nutritional genomics). Furthermore, the contribution of micronutrient insufficiencies and macronutrient excess is evident in the development and progression of many diseases, especially cancer. It is speculated that nutrients have capacity to implicitly affect the physiological and pathophysiological processes via gene expression various regulatory processes. Moreover, the nutrients are known to affect the cellular networks involved in cancer progression and cancer inhibitory mechanisms targeting apoptosis or impaired angiogenesis. The interplay of regulatory processes in physiological systems and nutrients provides basis for the nutrigenomics. The functional genomics data further argue that cellular and molecular processes involved in the cancer progression are possibly programed genes during early development which may persist into adulthood and become detrimental. The incorporation of the functional interactions between nutrients and the genome has revolutionized the field of personalized medicine and provided the foundation for targeted cancer therapy through nutrients. There is growing evidence on the beneficial impacts of eating habits on lowering the risk of cancer, even if it can be difficult to pinpoint the precise role of nutrients. The nutrigenomic information may provide bases to develop disease prevention and treatment via nutrition, at the molecular level.


Assuntos
Neoplasias , Nutrigenômica , Humanos , Neoplasias/prevenção & controle , Neoplasias/genética , Neoplasias/etiologia , Nutrigenômica/métodos , Genômica/métodos
2.
RNA Biol ; 21(1): 1-15, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38372062

RESUMO

Although Argonaute (AGO) proteins have been the focus of microRNA (miRNA) studies, we observed AGO-free mature miRNAs directly interacting with RNA-binding proteins, implying the sophisticated nature of fine-tuning gene regulation by miRNAs. To investigate microRNA-binding proteins (miRBPs) globally, we analyzed PAR-CLIP data sets to identify RBP quaking (QKI) as a novel miRBP for let-7b. Potential existence of AGO-free miRNAs were further verified by measuring miRNA levels in genetically engineered AGO-depleted human and mouse cells. We have shown that QKI regulates miRNA-mediated gene silencing at multiple steps, and collectively serves as an auxiliary factor empowering AGO2/let-7b-mediated gene silencing. Depletion of QKI decreases interaction of AGO2 with let-7b and target mRNA, consequently controlling target mRNA decay. This finding indicates that QKI is a complementary factor in miRNA-mediated mRNA decay. QKI, however, also suppresses the dissociation of let-7b from AGO2, and slows the assembly of AGO2/miRNA/target mRNA complexes at the single-molecule level. We also revealed that QKI overexpression suppresses cMYC expression at post-transcriptional level, and decreases proliferation and migration of HeLa cells, demonstrating that QKI is a tumour suppressor gene by in part augmenting let-7b activity. Our data show that QKI is a new type of RBP implicated in the versatile regulation of miRNA-mediated gene silencing.


Assuntos
MicroRNAs , Humanos , Animais , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Células HeLa , Inativação Gênica , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , RNA Mensageiro/genética
3.
Respirology ; 29(9): 815-824, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38654512

RESUMO

BACKGROUND AND OBJECTIVE: Lymphangioleiomyomatosis (LAM) is a rare neoplastic disease associated with the functional tumour suppressor genes TSC1 and TSC2 and causes structural destruction in the lungs, which could potentially increase the risk of lung cancer. However, this relationship remains unclear because of the rarity of the disease. METHODS: We investigated the relative risk of developing lung cancer among patients diagnosed with LAM between 2001 and 2022 at a single high-volume centre in Japan, using data from the Japanese Cancer Registry as the reference population. Next-generation sequencing (NGS) was performed in cases where tumour samples were available. RESULTS: Among 642 patients diagnosed with LAM (sporadic LAM, n = 557; tuberous sclerosis complex-LAM, n = 80; unclassified, n = 5), 13 (2.2%) were diagnosed with lung cancer during a median follow-up period of 5.13 years. All patients were female, 61.5% were never smokers, and the median age at lung cancer diagnosis was 53 years. Eight patients developed lung cancer after LAM diagnosis. The estimated incidence of lung cancer was 301.4 cases per 100,000 person-years, and the standardized incidence ratio was 13.6 (95% confidence interval, 6.2-21.0; p = 0.0008). Actionable genetic alterations were identified in 38.5% of the patients (EGFR: 3, ALK: 1 and ERBB2: 1). No findings suggested loss of TSC gene function in the two patients analysed by NGS. CONCLUSION: Our study revealed that patients diagnosed with LAM had a significantly increased risk of lung cancer. Further research is warranted to clarify the carcinogenesis of lung cancer in patients with LAM.


Assuntos
Neoplasias Pulmonares , Linfangioleiomiomatose , Humanos , Linfangioleiomiomatose/genética , Linfangioleiomiomatose/epidemiologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/epidemiologia , Feminino , Japão/epidemiologia , Pessoa de Meia-Idade , Fatores de Risco , Adulto , Incidência , Idoso , Estudos de Coortes , Masculino , Sistema de Registros , População do Leste Asiático
4.
Int J Exp Pathol ; 103(3): 121-128, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35436013

RESUMO

Wilms' tumour is a solid tumour that frequently occurs in children. Genetic changes in WT1 and epigenetic aberrations that affect imprinted control region 1 in WT2 loci are implicated in its aetiology. Moreover, tumour suppressor genes are frequently silenced by methylation in this tumour. In the present study, we analysed the methylation statuses of promoter regions of 24 tumour suppressor genes using a methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA)-based approach in 6 Wilms' tumours. Methylation of RASSF1 was specific to all 6 Wilms' tumours and was not observed in normal tissues. Moreover, methylated HIC1 was identified in stromal-type Wilms' tumours and methylated BRCA1 was identified in epithelial-type Wilms' tumours. Unmethylated CASP8, RARB, MLH1_167, APC and CDKN2A were found only in blastemal predominant-type Wilms' tumour. Our results indicated that methylation of RASSF1 may be a vital event in the tumorigenesis of Wilms' tumour, which informs its clinical and therapeutic management. In addition, mixed-type Wilms' tumours may be classified according to epithelial, stromal and blastemal components via MS-MLPA-based approach.


Assuntos
Metilação de DNA , Neoplasias Renais , Tumor de Wilms , Criança , Genes Supressores de Tumor , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor , Tumor de Wilms/genética , Tumor de Wilms/patologia
5.
BMC Cancer ; 21(1): 433, 2021 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-33879096

RESUMO

BACKGROUND: Inositol polyphosphate 4-phosphatase type II (INPP4B) is a negative regulator of the PI3K-Akt signalling pathway and plays a contradictory role in different types of cancers. However, the its biological role played by INPP4B in human gallbladder cancer (GBC) has not been elucidated. In this study, we investigated the expression, clinical significance and biological function of INPP4B in GBC patients and cell lines. METHODS: The INPP4B protein expression levels in gallbladder cancer tissues and normal gallbladder tissues were detected by immunohistochemistry, and the clinical significance of INPP4B was analysed. Knockdown and overexpression of INPP4B in GBC-SD and SGC-996 cells followed by cell proliferation, clonogenic, apoptosis detection, scratch wound-healing and transwell assays were used to identify INPP4B function in vitro. RESULTS: INPP4B was up-regulated in human GBC tissues compared with normal gallbladder tissues and was related to histopathological differentiation (p = 0.026). Here, we observed that INPP4B was highly expressed in high-moderately differentiated tumours compared with low-undifferentiated tumours (p = 0.022). Additionally, we found that INPP4B expression was not associated with overall survival of GBC patients (p = 0.071) and was not an independent prognostic factor. Furthermore, when we stratified the relationship between INPP4B expression and the prognosis of GBC based on histopathological differentiation, we found that INPP4B played a contradictory role in GBC progression depending on the degree of differentiation. In addition, INPP4B knockdown inhibited the proliferation, colony formation, migration and invasion in GBC cells, while INPP4B overexpression had the opposite effects in vitro, which indicates its role as an oncoprotein. CONCLUSIONS: These findings suggested that INPP4B may play a dual role in the prognosis of GBC depending on the degree of differentiation and that INPP4B might act as an oncogene in gallbladder cancer cells.


Assuntos
Neoplasias da Vesícula Biliar/genética , Regulação Neoplásica da Expressão Gênica , Monoéster Fosfórico Hidrolases/genética , Idoso , Idoso de 80 Anos ou mais , Apoptose/genética , Biomarcadores , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Neoplasias da Vesícula Biliar/diagnóstico , Neoplasias da Vesícula Biliar/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Metástase Neoplásica , Estadiamento de Neoplasias , Monoéster Fosfórico Hidrolases/metabolismo , Carga Tumoral
6.
Cell Microbiol ; 21(12): e13099, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31414579

RESUMO

Several commensal and pathogenic Gram-negative bacteria produce DNA-damaging toxins that are considered bona fide carcinogenic agents. The microbiota of colorectal cancer (CRC) patients is enriched in genotoxin-producing bacteria, but their role in the pathogenesis of CRC is poorly understood. The adenomatous polyposis coli (APC) gene is mutated in familial adenomatous polyposis and in the majority of sporadic CRCs. We investigated whether the loss of APC alters the response of colonic epithelial cells to infection by Salmonella enterica, the only genotoxin-producing bacterium associated with cancer in humans. Using 2D and organotypic 3D cultures, we found that APC deficiency was associated with sustained activation of the DNA damage response, reduced capacity to repair different types of damage, including DNA breaks and oxidative damage, and failure to induce cell cycle arrest. The reduced DNA repair capacity and inability to activate adequate checkpoint responses was associated with increased genomic instability in APC-deficient cells exposed to the genotoxic bacterium. Inhibition of the checkpoint response was dependent on activation of the phosphatidylinositol 3-kinase pathway. These findings highlight the synergistic effect of the loss of APC and infection with genotoxin-producing bacteria in promoting a microenvironment conducive to malignant transformation.


Assuntos
Polipose Adenomatosa do Colo/genética , Colo/metabolismo , Células Epiteliais/metabolismo , Instabilidade Genômica/genética , Fosfatidilinositol 3-Quinases/metabolismo , Infecções por Salmonella/metabolismo , Salmonella enterica/metabolismo , Polipose Adenomatosa do Colo/microbiologia , Polipose Adenomatosa do Colo/patologia , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular , Colo/microbiologia , Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/patologia , Dano ao DNA/genética , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Genes Supressores de Tumor/fisiologia , Humanos , Camundongos , Mutagênicos/metabolismo , Infecções por Salmonella/genética , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia , Transdução de Sinais/genética , Microambiente Tumoral/genética
7.
Exp Mol Pathol ; 110: 104277, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31271729

RESUMO

Inflammation is linked to prostate cancer (PCa) and to other diseases of the prostate. The prostanoid thromboxane (TX)A2 is a pro-inflammatory mediator implicated in several prostatic diseases, including PCa. TXA2 signals through the TPα and TPß isoforms of the T Prostanoid receptor (TP) which exhibit several functional differences and transcriptionally regulated by distinct promoters Prm1 and Prm3, respectively, within the TBXA2R gene. This study examined the expression of TPα and TPß in inflammatory infiltrates within human prostate tissue. Strikingly, TPß expression was detected in 94% of infiltrates, including in B- and T-lymphocytes and macrophages. In contrast, TPα was more variably expressed and, where present, expression was mainly confined to macrophages. To gain molecular insight into these findings, expression of TPα and TPß was evaluated as a function of monocyte-to-macrophage differentiation in THP-1 cells. Expression of both TPα and TPß was upregulated following phorbol-12-myristate-13-acetate (PMA)-induced differentiation of monocytic THP-1 to their macrophage lineage. Furthermore, FOXP1, an essential transcriptional regulator down-regulated during monocyte-to-macrophage differentiation, was identified as a key trans-acting factor regulating TPß expression through Prm3 in THP-1 cells. Knockdown of FOXP1 increased TPß, but not TPα, expression in THP-1 cells, while genetic reporter and chromatin immunoprecipitation (ChIP) analyses established that FOXP1 exerts its repressive effect on TPß through binding to four cis-elements within Prm3. Collectively, FOXP1 functions as a transcriptional repressor of TPß in monocytes. This repression is lifted in differentiated macrophages, allowing for upregulation of TPß expression and possibly accounting for the prominent expression of TPß in prostate tissue-resident macrophages.


Assuntos
Diferenciação Celular/genética , Perfilação da Expressão Gênica , Inflamação/genética , Próstata/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Doença Crônica , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Inflamação/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Monócitos/citologia , Monócitos/metabolismo , Prostaglandinas/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Células THP-1
8.
J Pathol ; 245(1): 53-60, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29431189

RESUMO

PALB2 is established as the most clinically important moderate to high penetrance breast cancer predisposition gene after BRCA1 and BRCA2. Mutations in classical familial cancer predisposition genes are presumed to be recessive at the cellular level and therefore a second inactivating somatic mutation is required in the tumour tissue. However, from the limited data that exist, PALB2 may be an example of a cancer predisposition gene that does not conform to Knudson's 'two hit' paradigm. We conducted genome-wide copy number analysis and targeted sequencing of PALB2 and other breast cancer driver genes in 15 invasive breast cancers from individuals carrying pathogenic germline mutations in PALB2. The majority of cancers showed clear evidence of bi-allelic inactivation of PALB2 (10/15) either as loss of heterozygosity involving the wild-type allele (six tumours) or as somatic point mutations (four tumours). All PALB2-null cancers had high homologous recombination deficiency (HRD) scores consistent with a homologous recombination repair deficiency. Interestingly, all but one of the PALB2 heterozygous cancers also had high HRD scores, suggesting that alternative mechanisms of PALB2 functional loss might be operating in these cancers. Our findings demonstrate that PALB2 does undergo bi-allelic inactivation in the majority of breast cancers from PALB2 germline mutation carriers. This feature has implications for the discovery of new moderate to high penetrance breast cancer predisposition genes as it supports using the existence of a 'second hit' and mutation signatures as important search criteria. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Neoplasias da Mama/patologia , Proteína do Grupo de Complementação N da Anemia de Fanconi/genética , Predisposição Genética para Doença , Mutação em Linhagem Germinativa/genética , Mutação/genética , Proteína BRCA2/genética , Neoplasias da Mama/genética , Humanos , Reparo de DNA por Recombinação/genética , Proteínas Supressoras de Tumor/genética
9.
Medicina (Kaunas) ; 55(7)2019 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-31319555

RESUMO

Cervical cancer is the fourth most common cancer among women. Infection by high-risk human papillomavirus (HPV) is the main aetiology for the development of cervical cancer. Infection by high-risk human papillomavirus (HPV) and the integration of the HPV genome into the host chromosome of cervical epithelial cells are key early events in the neoplastic progression of cervical lesions. The viral oncoproteins, mainly E6 and E7, are responsible for the initial changes in epithelial cells. The viral proteins inactivate two main tumour suppressor proteins, p53, and retinoblastoma (pRb). Inactivation of these host proteins disrupts both the DNA repair mechanisms and apoptosis, leading to rapid cell proliferation. Multiple genes involved in DNA repair, cell proliferation, growth factor activity, angiogenesis, as well as mitogenesis genes become highly expressed in cervical intraepithelial neoplasia (CIN) and cancer. This genomic instability encourages HPV-infected cells to progress towards invasive carcinoma. The key molecular events involved in cervical carcinogenesis will be discussed in this review.


Assuntos
Células Epiteliais/patologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Neoplasias do Colo do Útero/virologia , Adulto , DNA Viral/efeitos adversos , Células Epiteliais/virologia , Feminino , Papillomavirus Humano 16/crescimento & desenvolvimento , Papillomavirus Humano 16/patogenicidade , Papillomavirus Humano 18/crescimento & desenvolvimento , Papillomavirus Humano 18/patogenicidade , Humanos , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/fisiopatologia , Neoplasias do Colo do Útero/etiologia , Neoplasias do Colo do Útero/fisiopatologia
10.
Exp Cell Res ; 358(2): 227-233, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28663058

RESUMO

The early preimplantation embryo is susceptible to a range of exogenous stresses which result in their reduced long-term developmental potential. The P53 tumour suppressor protein is normally held at low levels in the preimplantation embryo and we show that culture stress induces the expression of a range of canonical P53-response genes (Mdm2, Bax and Cdkn1a). Culture stress caused a P53-dependent loss of cells from resulting blastocysts, and this was most evident within the inner cell mass population. Culture stress increased the proportion of cells expressing active caspase-3 and undergoing apoptosis, while inhibition of caspase-3 increased the number of cells within the inner cell mass. The P53-dependent loss of cells from the inner cell mass was accompanied by a loss of NANOG-positive epiblast progenitors. Pharmacological activation of P53 by the MDM2 inhibitor, Nutlin-3, also caused increased P53-dependent transcription and the loss of cells from the inner cell mass. This loss of cells could be ameliorated by simultaneous treatment with the P53 inhibitor, Pifithrin-α. Culture stress causes reduced signalling via the phosphatidylinositol-3-kinase signalling pathway, and blocking this pathway caused P53-dependent loss of cells from the inner cell mass. These results point to P53 acting to limit the accumulation and survival of cells within the pluripotent lineage of the blastocyst and provide a molecular framework for the further investigation of the factors determining the effects of stressors on the embryo's developmental potential.


Assuntos
Blastocisto/citologia , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/efeitos dos fármacos , Benzotiazóis/metabolismo , Desenvolvimento Embrionário , Genes Homeobox , Imidazóis/metabolismo , Camundongos Transgênicos , Piperazinas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tolueno/análogos & derivados , Tolueno/metabolismo
11.
Int J Mol Sci ; 19(12)2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30513694

RESUMO

CCCTC-binding factor (CTCF) is a conserved transcription factor that performs diverse roles in transcriptional regulation and chromatin architecture. Cancer genome sequencing reveals diverse acquired mutations in CTCF, which we have shown functions as a tumour suppressor gene. While CTCF is essential for embryonic development, little is known of its absolute requirement in somatic cells and the consequences of CTCF haploinsufficiency. We examined the consequences of CTCF depletion in immortalised human and mouse cells using shRNA knockdown and CRISPR/Cas9 genome editing as well as examined the growth and development of heterozygous Ctcf (Ctcf+/-) mice. We also analysed the impact of CTCF haploinsufficiency by examining gene expression changes in CTCF-altered endometrial carcinoma. Knockdown and CRISPR/Cas9-mediated editing of CTCF reduced the cellular growth and colony-forming ability of K562 cells. CTCF knockdown also induced cell cycle arrest and a pro-survival response to apoptotic insult. However, in p53 shRNA-immortalised Ctcf+/- MEFs we observed the opposite: increased cellular proliferation, colony formation, cell cycle progression, and decreased survival after apoptotic insult compared to wild-type MEFs. CRISPR/Cas9-mediated targeting in Ctcf+/- MEFs revealed a predominance of in-frame microdeletions in Ctcf in surviving clones, however protein expression could not be ablated. Examination of CTCF mutations in endometrial cancers showed locus-specific alterations in gene expression due to CTCF haploinsufficiency, in concert with downregulation of tumour suppressor genes and upregulation of estrogen-responsive genes. Depletion of CTCF expression imparts a dramatic negative effect on normal cell function. However, CTCF haploinsufficiency can have growth-promoting effects consistent with known cancer hallmarks in the presence of additional genetic hits. Our results confirm the absolute requirement for CTCF expression in somatic cells and provide definitive evidence of CTCF's role as a haploinsufficient tumour suppressor gene. CTCF genetic alterations in endometrial cancer indicate that gene dysregulation is a likely consequence of CTCF loss, contributing to, but not solely driving cancer growth.


Assuntos
Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Sobrevivência Celular/fisiologia , Neoplasias do Endométrio/genética , Edição de Genes , Animais , Sistemas CRISPR-Cas , Proliferação de Células/genética , Proliferação de Células/fisiologia , Sobrevivência Celular/genética , Feminino , Haploinsuficiência/genética , Haploinsuficiência/fisiologia , Humanos , Células K562 , Camundongos , RNA Interferente Pequeno/genética
12.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3153-3169, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28890397

RESUMO

The prostanoid thromboxane (TX)A2 signals through the TPα and TPß isoforms of T Prostanoid receptor (TP) that are transcriptionally regulated by distinct promoters termed Prm1 and Prm3, respectively, within the TBXA2R gene. We recently demonstrated that expression of TPα and TPß is increased in PCa, differentially correlating with Gleason grade and with altered CpG methylation of the individual Prm1/Prm3 regions within the TBXA2R. The current study sought to localise the sites of CpG methylation within Prm1 and Prm3, and to identify the main transcription factors regulating TPß expression through Prm3 in the prostate adenocarcinoma PC-3 and LNCaP cell lines. Bisulfite sequencing revealed extensive differences in the pattern and status of CpG methylation of the individual Prm1 and Prm3 regions that regulate TPα and TPß expression, respectively, within the TBXA2R. More specifically, Prm1 is predominantly hypomethylated while Prm3 is hypermethylated across its entire sequence in PC-3 and LNCaP cells. Furthermore, the tumour suppressors FOXP1 and NKX3.1, strongly implicated in PCa development, were identified as key transcription factors regulating TPß expression through Prm3 in both PCa cell lines. Specific siRNA-disruption of FOXP1 and NKX3.1 each coincided with up-regulated TPß protein and mRNA expression, while genetic-reporter and chromatin immunoprecipitation (ChIP) analyses confirmed that both FOXP1 and NKX3.1 bind to cis­elements within Prm3 to transcriptionally repress TPß in the PCa lines. Collectively these data identify Prm3/TPß as a bona fide target of FOXP1 and NKX3.1 regulation, providing a mechanistic basis, at least in part, for the highly significant upregulation of TPß expression in PCa.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/metabolismo , Neoplasias da Próstata/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Proteínas Repressoras/metabolismo , Tromboxano A2/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG , Metilação de DNA , Regulação para Baixo , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Masculino , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Protaminas/genética , Isoformas de Proteínas , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Regulação para Cima
13.
J Cell Mol Med ; 20(1): 10-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26498914

RESUMO

Tongue squamous cell carcinoma (TSCC) is the most common type of oral squamous cell carcinomas and is well known for its high rate of lymph nodal metastasis. Despite the identification of many molecular mechanisms in TSCC, the number of deaths associated with TSCC increased during the past 5 years. MicroRNAs (miRNAs) are a family of small non-coding RNA molecules, which regulate gene expression by either translational inhibition or mRNA degradation. miRNAs have been proven to be key regulators of various biological and pathological processes including cell proliferation, development and tumourigenesis. Increasing evidence has demonstrated that the deregulated miRNAs are implicated in the diagnosis and treatment of TSCC. In this review, we summarized the expressions and roles of miRNAs in TSCC and comment on the potential roles of miRNAs in diagnosis, prognosis and treatment of this malignancy.


Assuntos
Carcinoma de Células Escamosas/genética , MicroRNAs/fisiologia , Neoplasias da Língua/genética , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundário , Proliferação de Células , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Prognóstico , Interferência de RNA , Neoplasias da Língua/metabolismo , Neoplasias da Língua/patologia
14.
Mol Cancer ; 15: 28, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27056547

RESUMO

BACKGROUND: The advantages and utility of microRNAs (miRNAs) as diagnostic and prognostic cancer markers is at the vanguard in recent years. In this study, we attempted to identify and validate the differential expression of miRNAs in oral squamous cell carcinoma (OSCC), to correlate their expression with the clinico-pathological profile of tumours and to identify the signaling pathways through which the aberrantly expressed miRNAs effect tumourigenesis. METHODS: miRCURY LNA™ array with probes specific to 1168 miRNAs and TaqMan assays specific for 10 miRNAs was employed to evaluate and validate miRNA expression in a discovery cohort (n = 29) and validation cohort (n = 61) of primary OSCC tissue specimens, respectively. A computational pipeline with sequential integration of data from miRTarBase, CytoScape, UniProtKB and DIANA-miRPath was utilized to map the target genes of deregulated miRNAs and associated molecular pathways. RESULTS: Microarray profiling identified 46 miRNAs that were differentially expressed in OSCC. Unsupervised clustering demonstrated a high degree of molecular heterogeneity across the tumour samples as the clusters did not represent any of their clinico-pathological characteristics. The differential expression of 10 miRNAs were validated by RT-qPCR (let-7a, let-7d, let-7f and miR-16 were downregulated while miR-29b, miR-142-3p, miR-144, miR-203, and miR-223 were upregulated in OSCC; the expression of miR-1275 was variable in tumours, with high levels associated to regional lymph node invasion; additionally, miR-223 exhibited an association with advanced tumour stage/size). In silico analyses of the experimentally confirmed target genes of miRNAs revamp the relationship of upregulated miRNAs with tumour suppressor genes and of downregulated miRNAs with oncogenes. Further, the differentially expressed miRNAs may play a role by simultaneously activating genes of PI3K/Akt signaling on one hand and by repressing genes of p53 signaling pathway on the other. CONCLUSIONS: The identified differentially expressed miRNAs and signaling pathways deregulated in OSCC have implications for the development of novel therapeutic strategies. To the best of our knowledge, this is the first report to show the association of miR-1275 with nodal invasion and the upregulation of miR-144 in OSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Bucais/genética , Carcinogênese/genética , Carcinogênese/patologia , Carcinoma de Células Escamosas/patologia , Diferenciação Celular/genética , Linhagem Celular Tumoral , Análise por Conglomerados , Regulação para Baixo/genética , Feminino , Heterogeneidade Genética , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Neoplasias Bucais/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Transdução de Sinais/genética , Regulação para Cima/genética
15.
Recent Results Cancer Res ; 205: 149-68, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27075353

RESUMO

Carcinoid tumours arise in cells of the diffuse neuroendocrine system and can develop in a number of anatomical sites including the lungs and the gastrointestinal tract. There has been a move away from the use of the term carcinoid tumour to the more appropriate use of neuroendocrine tumour (NET) to highlight the potential for invasion and metastasis associated with some NETs. Although most cases are sporadic, 15-20% of cases are related to a hereditary syndrome, the most common of these being multiple endocrine neoplasia 1 (MEN1). Other hereditary syndromes include the following: von Hippel-Lindau (VHL), neurofibromatosis 1 and tuberous sclerosis complex (TSC), which are all associated with a germline mutation of the associated tumour suppressor gene and an autosomal dominant inheritance pattern. Familial small intestinal NET (SI NET) is a recently described condition which is also inherited in an autosomal dominant manner. There appears to be more than one causative gene; thus far, only the IPMK gene has been identified as a causative germline mutation. This was identified by carrying out whole-exome sequencing of germline and tumour DNA in a family with multiple members diagnosed with SI NET. Identification of NET predisposition genes in other families via these methods will allow the development of dedicated NET gene panels which can be used to screen NET patients and at-risk relatives for hereditary mutations. Close surveillance of at-risk individuals is important to detect NETs early when curative surgery can be offered and the morbidity and mortality of metastatic NETs can be avoided.


Assuntos
Tumor Carcinoide/diagnóstico , Tumor Carcinoide/terapia , Síndromes Neoplásicas Hereditárias/diagnóstico , Síndromes Neoplásicas Hereditárias/terapia , Tumor Carcinoide/genética , Predisposição Genética para Doença , Humanos , Síndromes Neoplásicas Hereditárias/genética
16.
J Pathol ; 237(2): 203-14, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26011651

RESUMO

Malignant pleural mesothelioma (MPM) is a devastating malignancy characterized by invasive growth and rapid recurrence. The identification and inhibition of molecular components leading to this migratory and invasive phenotype are thus essential. Accordingly, a genome-wide expression array analysis was performed on MPM cell lines and a set of 139 genes was identified as differentially expressed in cells with high versus low migratory activity. Reduced expression of the novel tumour suppressor integrin α7 (ITGA7) was found in highly motile cells. A significant negative correlation was observed between ITGA7 transcript levels and average displacement of cells. Forced overexpression of ITGA7 in MPM cells with low endogenous ITGA7 expression inhibited cell motility, providing direct evidence for the regulatory role of ITGA7 in MPM cell migration. MPM cells showed decreased ITGA7 expressions at both transcription and protein levels when compared to non-malignant mesothelial cells. The majority of MPM cell cultures displayed hypermethylation of the ITGA7 promoter when compared to mesothelial cultures. A statistically significant negative correlation between ITGA7 methylation and ITGA7 expression was also observed in MPM cells. While normal human pleura samples unambiguously expressed ITGA7, a varying level of expression was found in a panel of 200 human MPM samples. In multivariate analysis, ITGA7 expression was found to be an independent prognostic factor. Although there was no correlation between histological subtypes and ITGA7 expression, importantly, patients with high tumour cell ITGA7 expression had an increased median overall survival compared to the low- or no-expression groups (463 versus 278 days). In conclusion, our data suggest that ITGA7 is an epigenetically regulated tumour suppressor gene and a prognostic factor in human MPM.


Assuntos
Antígenos CD/metabolismo , Movimento Celular , Epigênese Genética , Cadeias alfa de Integrinas/metabolismo , Neoplasias Pulmonares/metabolismo , Mesotelioma/metabolismo , Neoplasias Pleurais/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Antígenos CD/genética , Linhagem Celular Tumoral , Metilação de DNA , Regulação para Baixo , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Cadeias alfa de Integrinas/genética , Estimativa de Kaplan-Meier , Laminina/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Mesotelioma/genética , Mesotelioma/mortalidade , Mesotelioma/patologia , Mesotelioma Maligno , Análise Multivariada , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Pleurais/genética , Neoplasias Pleurais/mortalidade , Neoplasias Pleurais/patologia , Prognóstico , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , RNA Mensageiro/metabolismo , Fatores de Risco , Transdução de Sinais , Fatores de Tempo , Transfecção , Proteínas Supressoras de Tumor/genética
17.
Acta Paediatr ; 105(3): e132-41, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26607758

RESUMO

AIM: Protein tyrosine phosphatases receptor type D (PTPRD) is a tumour suppressor gene, and its epigenetic silencing is frequently found in glioblastoma. As aberrant deoxyribonucleic acid (DNA) methylation patterning has been shown to play a role in leukaemogenesis, we studied the promoter methylation, expression profiles and molecular functions of PTPRD in paediatric patients with acute myeloid leukaemia (AML). METHODS: Bone marrow specimens were obtained from 32 Chinese patients with a mean age of 7.2 years (range 1.1-16.5). PTPRD and methylation status were evaluated by real-time polymerase chain reaction (PCR) and methylation-specific PCR. Western blot and flow cytometry techniques were also used. RESULTS: PTPRD expression was decreased by promoter region methylation in six AML cells and methylated in 21 (65.6%) of the 32 samples. In addition, PTPRD expression could be induced by the DNA demethylating agent 5-aza-2'-deoxycytidine. Furthermore, functional studies showed that overexpression of PTPRD in AML cells inhibited cell proliferation and clonogenicity as well as inducing apoptosis. However, PTPRD knockdown increased cell proliferation. These effects were associated with downregulation of cyclin D1, c-myc and upregulation of Bax. CONCLUSION: The results of this study demonstrated that PTPRD was a potential tumour suppressor gene inactivated by DNA methylation in paediatric AML.


Assuntos
Leucemia Mieloide Aguda/enzimologia , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Adolescente , Apoptose , Proliferação de Células , Criança , Pré-Escolar , Metilação de DNA , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HL-60 , Humanos , Lactente , Leucemia Mieloide Aguda/genética , Masculino , Células U937
18.
J Viral Hepat ; 22(2): 201-11, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24910204

RESUMO

Several members of protocadherins have been found involved in human carcinogenesis, but little is known about PCDH20 in HCC. Here in this study, using quantitative real-time RT-PCR assay, we demonstrated the downregulation of PCDH20 expression in 6 of 7 HCC cell lines tested. Similarly, PCDH20 expression in primary HCC tissues was also significantly downregulated in comparison with that in either disease-free normal liver tissues or the adjacent nontumour liver tissues (P < 0.001, respectively). Among HCC tumour tissues studied, about 48% (51/107) of them showed reduced PCDH20 mRNA level. Further statistic analysis revealed that the reduced PCDH20 mRNA level in tumour tissues was much more common in younger patients group (aged <50 years) than that in older group (≥50 years) (60% vs 33%, P = 0.0303). Loss of heterozygosity (LOH) and promoter hypermethylation analysis revealed that deletion and/or aberrant epigenetic modulation of PCDH20 gene account for its downregulation, at least in a fraction of tumour specimens. Moreover, ectopic expression of PCDH20 in HCC cells significantly suppressed cell proliferation, clonogenicity, migration and tumour formation. Notably, we proved for the first time that, via activating GSK-3ß, PCDH20 could inhibit Wnt/ß-catenin signalling pathway. Furthermore, our data suggest that PCDH20 may conduct its Wnt/ß-catenin signalling antagonizing function through suppressing Akt and Erk activities and promoting GSK-3ß signalling activities. However, the detailed mechanism remained undiscovered. In conclusion, our data here strongly suggested that PCDH20 may act as a candidate tumour suppressor in HCC.


Assuntos
Caderinas/genética , Caderinas/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Genes Supressores de Tumor , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Via de Sinalização Wnt/fisiologia , Adulto , Idoso , Regulação para Baixo , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Protocaderinas
19.
J Pathol ; 232(5): 566-77, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24407904

RESUMO

Genomic changes affecting tumour suppressor genes are fundamental to cancer. We applied SNP array analysis to a panel of testicular germ cell tumours to search for novel tumour suppressor genes and identified a frequent small deletion on 6q25.3 affecting just one gene, ZDHHC14. The expression of ZDHHC14, a putative protein palmitoyltransferase with unknown cellular function, was decreased at both RNA and protein levels in testicular germ cell tumours. ZDHHC14 expression was also significantly decreased in a panel of prostate cancer samples and cell lines. In addition to our findings of genetic and protein expression changes in clinical samples, inducible overexpression of ZDHHC14 led to reduced cell viability and increased apoptosis through the classic caspase-dependent apoptotic pathway and heterozygous knockout of ZDHHC14 increased [CORRECTED] cell colony formation ability. Finally, we confirmed our in vitro findings of the tumour suppressor role of ZDHHC14 in a mouse xenograft model, showing that overexpression of ZDHHC14 inhibits tumourigenesis. Thus, we have identified a novel tumour suppressor gene that is commonly down-regulated in testicular germ cell tumours and prostate cancer, as well as given insight into the cellular functional role of ZDHHC14, a potential protein palmitoyltransferase that may play a key protective role in cancer.


Assuntos
Aciltransferases/genética , Genes Supressores de Tumor , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias da Próstata/genética , Neoplasias Testiculares/genética , Aciltransferases/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Regulação para Baixo , Deleção de Genes , Perfilação da Expressão Gênica/métodos , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Embrionárias de Células Germinativas/enzimologia , Neoplasias Embrionárias de Células Germinativas/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Polimorfismo de Nucleotídeo Único , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/prevenção & controle , Interferência de RNA , Neoplasias Testiculares/enzimologia , Neoplasias Testiculares/patologia , Fatores de Tempo , Transfecção , Carga Tumoral
20.
J Pathol ; 234(2): 142-5, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25074498

RESUMO

One hundred years have passed since the publication of Theodore Boveri's Zur Frage der Entstehung maligner Tumouren [Concerning the Origin of Malignant Tumours]. This prescient publication created the foundations for much of our understanding of the origins of cancer and in particular the genetic basis of some cancers. In his work, Boveri suggested that loss of key cellular attributes, now known as tumour suppressor genes, are a key driver event in the development of cancer and inheritance could play a role in cancer susceptibility. He also predicted that chromosomal (genomic) instability as a key hallmark of cancer. Whilst these key insights that still inform the practice of cancer genetics, they were not the main theme of Boveri's text, which was to describe the role of chromosomal abnormalities in the development of cancer. In making his case he also suggested that genetic information could be contained in distinct packages (genes) that are linearly arranged along chromosomes and that cancers arise from single cells. These remarkably accurate hypotheses add weight to the need to celebrate this landmark publication for its accurate prediction of so much that we take for granted. Here we focus on Boveri's contributions to our understanding of hereditary cancers, which, alongside the astute clinical observations of Paul Broca and Aldred Scott Warthin, were published decades before the field became respectable, yet could still inform anyone studying hereditary cancers.


Assuntos
Aberrações Cromossômicas , Predisposição Genética para Doença/história , Oncologia/história , Neoplasias/genética , Predisposição Genética para Doença/genética , Alemanha , História do Século XIX , História do Século XX , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa