Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Sci Rep ; 14(1): 2667, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38302662

RESUMO

Pediatric Crohn's disease (CD) is characterized by a severe disease course with frequent complications. We sought to apply machine learning-based models to predict risk of developing future complications in pediatric CD using ileal and colonic gene expression. Gene expression data was generated from 101 formalin-fixed, paraffin-embedded (FFPE) ileal and colonic biopsies obtained from treatment-naïve CD patients and controls. Clinical outcomes including development of strictures or fistulas and progression to surgery were analyzed using differential expression and modeled using machine learning. Differential expression analysis revealed downregulation of pathways related to inflammation and extra-cellular matrix production in patients with strictures. Machine learning-based models were able to incorporate colonic gene expression and clinical characteristics to predict outcomes with high accuracy. Models showed an area under the receiver operating characteristic curve (AUROC) of 0.84 for strictures, 0.83 for remission, and 0.75 for surgery. Genes with potential prognostic importance for strictures (REG1A, MMP3, and DUOX2) were not identified in single gene differential analysis but were found to have strong contributions to predictive models. Our findings in FFPE tissue support the importance of colonic gene expression and the potential for machine learning-based models in predicting outcomes for pediatric CD.


Assuntos
Doença de Crohn , Criança , Humanos , Constrição Patológica , Doença de Crohn/patologia , Expressão Gênica , Aprendizado de Máquina , Litostatina/genética
2.
Cell Commun Signal ; 22(1): 152, 2024 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-38414029

RESUMO

BACKGROUND: Germline mutations of E-cadherin contribute to hereditary diffuse gastric cancer (HDGC) and congenital malformations, such as oral facial clefts (OFC). However, the molecular mechanisms through which E-cadherin loss-of-function triggers distinct clinical outcomes remain unknown. We postulate that E-cadherin-mediated disorders result from abnormal interactions with the extracellular matrix and consequent aberrant intracellular signalling, affecting the coordination of cell migration. METHODS: Herein, we developed in vivo and in vitro models of E-cadherin mutants associated with either OFC or HDGC. Using a Drosophila approach, we addressed the impact of the different variants in cell morphology and migration ability. By combining gap closure migration assays and time-lapse microscopy, we further investigated the migration pattern of cells expressing OFC or HDGC variants. The adhesion profile of the variants was evaluated using high-throughput ECM arrays, whereas RNA sequencing technology was explored for identification of genes involved in aberrant cell motility. RESULTS: We have demonstrated that cells expressing OFC variants exhibit an excessive motility performance and irregular leading edges, which prevent the coordinated movement of the epithelial monolayer. Importantly, we found that OFC variants promote cell adhesion to a wider variety of extracellular matrices than HDGC variants, suggesting higher plasticity in response to different microenvironments. We unveiled a distinct transcriptomic profile in the OFC setting and pinpointed REG1A as a putative regulator of this outcome. Consistent with this, specific RNAi-mediated inhibition of REG1A shifted the migration pattern of OFC expressing cells, leading to slower wound closure with coordinated leading edges. CONCLUSIONS: We provide evidence that E-cadherin variants associated with OFC activate aberrant signalling pathways that support dynamic rearrangements of cells towards improved adaptability to the microenvironment. This proficiency results in abnormal tissue shaping and movement, possibly underlying the development of orofacial malformations.


Assuntos
Adenocarcinoma , Neoplasias Gástricas , Humanos , Caderinas/genética , Caderinas/metabolismo , Adesão Celular , Movimento Celular , Mutação em Linhagem Germinativa , Litostatina/genética , Neoplasias Gástricas/metabolismo , Microambiente Tumoral , Animais , Drosophila melanogaster
3.
Sci Rep ; 13(1): 12201, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37500741

RESUMO

Chronic pancreatitis (CP) is a disease characterized by the inflammation and destruction of pancreatic tissue, leading to the replacement of functional tissue with fibrotic tissue. The regenerating gene (Reg) family proteins have recently been implicated in the repair and regeneration of inflamed pancreatic tissue, though the exact mechanisms of their involvement in the pathogenesis of CP are not yet fully understood. To investigate the role of Reg family proteins in CP, we generated global knockout mice (Reg-/-) for Reg1-3 (Reg1,2,3a,3b,3d,3g) genes using the CRISPR/Cas9 system. We then investigated the effect of Reg family protein deficiency in a genetic model of CP (X-SPINK1) mice by knocking out Reg1-3 genes. We examined pancreatic morphology, inflammatory cytokines expression, and activation of pancreatic stellate cells (PSCs) at different ages. Reg-/- mice showed no abnormalities in general growth and pancreas development. Deficiency of Reg1-3 in CP mice led to a reduction in pancreatic parenchymal loss, decreased deposition of collagen, and reduced expression of proinflammatory cytokines. Additionally, Reg proteins were found to stimulate PSCs activation. Overall, our study suggests that Reg1-3 deficiency can lead to the remission of CP and Reg family proteins could be a potential therapeutic target for the treatment of CP.


Assuntos
Células Estreladas do Pâncreas , Pancreatite Crônica , Camundongos , Animais , Células Estreladas do Pâncreas/metabolismo , Pancreatite Crônica/metabolismo , Pâncreas/metabolismo , Inflamação/patologia , Camundongos Knockout , Colágeno/metabolismo , Citocinas/metabolismo , Regeneração , Fibrose , Litostatina/genética , Litostatina/metabolismo
4.
Cancer Invest ; 41(4): 394-404, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36797673

RESUMO

Identifying differentially expressed genes and co-expression modules lead to novel biomarkers. GO, pathway enrichment, network, and tumor stage analysis of 318 ovarian cancer samples from TCGA, categorised into primary and recurrent, pre-menopause and post-menopause, and early and late stage tumors was performed. Upregulated and downregulated genes in primary vs recurrent, early stage vs late-stage and pre-menopause vs post-menopause tumors were 84 and 62, 84 and 35, and 88 and 14, respectively. IRAK2 and CXCL8 had higher expression in recurrent tumors while REG1A had higher expression in post-menopause samples. In late stage tumors constant expression of IRAK2 and REG1A was observed, while that of CXCL8 and EGF decreased. These genes may be potential biomarkers for the diagnosis of the disease.


Assuntos
Redes Reguladoras de Genes , Neoplasias Ovarianas , Humanos , Feminino , Recidiva Local de Neoplasia , Biomarcadores , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Análise de Sequência de RNA , Litostatina/genética
5.
J Cell Mol Med ; 26(17): 4710-4720, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35946046

RESUMO

The pathophysiology of inflammatory bowel diseases (IBD) reflects a balance between mucosal injury and reparative mechanisms. Some regenerating gene (Reg) family members (REG Iα, REG Iß and REG IV) are expressed in Crohn's disease (CD) and ulcerative colitis (UC) and involved as proliferative mucosal factors in IBD. We revealed that REG Iα and REG Iß were induced in cell culture system by IL-6/IL-22. Although REG IV was upregulated in IBD biopsy samples, the upregulation of REG IV was not at all induced in cell culture by autoimmune-related cytokines such as IL-6, IL-22 and TNFα. Here, we analysed REG IV expression in LS-174 T and HT-29 human intestinal epithelial cells by real-time RT-PCR and elisa. REG IV expression was induced by lipopolysaccharide (LPS). However, LPS did not activate REG IV promoter activity. As the LPS-induced upregulation of REG IV was considered to be regulated post-transcriptionally, we searched targeted microRNA (miR), which revealed that REG IV mRNA has a potential target sequence for miR-24. We measured the miR-24 level of LPS-treated cells and found that the level was significantly lower. The LPS-induced increase of REG IV mRNA was abolished by the introduction of miR-24 mimic but not by non-specific control RNA.


Assuntos
Colite Ulcerativa , Doenças Inflamatórias Intestinais , MicroRNAs , Proteínas Associadas a Pancreatite/genética , Colite Ulcerativa/patologia , Regulação para Baixo/genética , Células Epiteliais/metabolismo , Humanos , Doenças Inflamatórias Intestinais/patologia , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Litostatina/genética , Litostatina/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro/metabolismo , Regulação para Cima/genética
6.
Int J Mol Sci ; 22(13)2021 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-34281249

RESUMO

Regenerating gene (REG) family proteins serve as multifunctional secretory molecules with trophic, antiapoptotic, anti-inflammatory, antimicrobial and probably immuno-regulatory effects. Since their discovery, accumulating evidence has clarified the potential roles of the REG family in the occurrence, progression and development of a wide range of inflammatory and inflammation-associated diseases of the gastrointestinal (GI) tract. However, significant gaps still exist due to the undefined nature of certain receptors, regulatory signaling pathways and possible interactions among distinct Reg members. In this narrative review, we first describe the structural features, distribution pattern and purported regulatory mechanisms of REG family proteins. Furthermore, we summarize the established and proposed roles of REG proteins in the pathogenesis of various inflammation-associated pathologies of the GI tract and the body as a whole, focusing particularly on carcinogenesis in the ulcerative colitis-colitic cancer sequence and gastric cancer. Finally, the clinical relevance of REG products in the context of diagnosis, treatment and prognostication are also discussed in detail. The current evidence suggests a need to better understanding the versatile roles of Reg family proteins in the pathogenesis of inflammatory-associated diseases, and their broadened future usage as therapeutic targets and prognostic biomarkers is anticipated.


Assuntos
Gastroenteropatias/metabolismo , Trato Gastrointestinal/metabolismo , Inflamação/imunologia , Animais , Apoptose/genética , Gastroenteropatias/imunologia , Trato Gastrointestinal/imunologia , Humanos , Inflamação/metabolismo , Doenças Inflamatórias Intestinais , Mucosa Intestinal/metabolismo , Lectinas Tipo C/metabolismo , Litostatina/genética , Litostatina/metabolismo , Proteínas Associadas a Pancreatite/genética , Proteínas Associadas a Pancreatite/metabolismo , Neoplasias Gástricas
7.
Sci Rep ; 11(1): 2077, 2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33483567

RESUMO

Organoids derived from epithelial tumors have recently been utilized as a preclinical model in basic and translational studies. This model is considered to represent the original tumor in terms of 3D structure, genetic and cellular heterogeneity, but not tumor microenvironment. In this study, we established organoids and paired cancer-associated fibroblasts (CAFs) from surgical specimens of colorectal carcinomas (CRCs), and evaluated gene expression profiles in organoids with and without co-culture with CAFs to assess interactions between tumor cells and CAFs in tumor tissues. We found that the expression levels of several genes, which are highly expressed in original CRC tissues, were downregulated in organoids but re-expressed in organoids by co-culturing with CAFs. They comprised immune response- and external stimulus-related genes, e.g., REG family and dual oxidases (DUOXs), which are known to have malignant functions, leading tumor cells to proliferative and/or anti-apoptotic states and drug resistant phenotypes. In addition, the degree of differential induction of REG1 and DUOX2 in the co-culture system varied depending on CAFs from each CRC case. In conclusion, the co-culture system of CRC organoids with paired CAFs was able to partially reproduce the tumor microenvironment.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Neoplasias Colorretais/genética , Oxidases Duais/genética , Regulação Neoplásica da Expressão Gênica , Litostatina/genética , Organoides/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Fibroblastos Associados a Câncer/citologia , Proliferação de Células , Neoplasias Colorretais/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Análise de Sequência com Séries de Oligonucleotídeos
8.
Int Immunopharmacol ; 92: 107304, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33513463

RESUMO

Regenerating islet-derived protein 1-alpha (REG1A) was abnormally upregulated in a series of gastrointestinal inflammatory disorders. However, the potential biological function and underlying regulatory mechanisms of the increased REG1A in inflammatory bowel disease (IBD) pathogenesis remain to be fully elucidated. In this study, we uncovered that REG1A was substantially increased in the inflamed colorectal tissues of IBD patients. And the aberrantly expressed REG1A in intestinal epithelial cells (IEC) prominently inhibited inflammatory responses, promoted cell proliferation and suppressed epithelial apoptosis. Mechanically, IL-6 and IL-22 markedly activated REG1A transcription through triggering JAK/STAT3 signaling pathway. In addition, overexpression of REG1A in mice by systematic delivery of REG1A lentivirus remarkably alleviated DSS-induced inflammatory injury and maintained the integrity of intestinal mucosal barrier. Taken together, our data demonstrated that the novel proliferative factor REG1A controlled by IL-6/IL-22-JAK-STAT3 signaling may provide a promising therapeutic target for patients with IBD.


Assuntos
Colite/prevenção & controle , Inflamação/prevenção & controle , Doenças Inflamatórias Intestinais/prevenção & controle , Janus Quinases/metabolismo , Litostatina/administração & dosagem , Substâncias Protetoras/administração & dosagem , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Colite/induzido quimicamente , Biologia Computacional/métodos , Bases de Dados Genéticas , Modelos Animais de Doenças , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/patologia , Janus Quinases/genética , Litostatina/genética , Litostatina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT3/genética , Transdução de Sinais
9.
Pancreatology ; 20(3): 301-304, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32037128

RESUMO

Pancreatic stone protein (PSP), discovered in the 1970ies, was first associated with stone formation during chronic pancreatitis. Later, the same protein was independently detected in islet preparations and named regenerating protein 1 (REG1). Additional isoforms of PSP, including pancreatitis-associated protein (PAP), belong to the same protein family. Although the names indicate a potential function in stone formation or islet regeneration, involvements in cellular processes were only suggestive and never unequivocally proven. We established an association between PSP levels in patient blood samples and the development of sepsis. In this review, written in connection with receiving the Lifetime Achievement Award of the European Pancreatic Club, the evolution of the sepsis aspect of PSP is described. We conclude that the true functional properties of this fascinating pancreatic protein still remain an enigma.


Assuntos
Cálculos/genética , Cálculos/patologia , Litostatina/genética , Pancreatite Crônica/genética , Pancreatite Crônica/patologia , Sepse/etiologia , Sepse/genética , Cálculos/complicações , Humanos , Isomerismo , Litostatina/metabolismo , Pancreatite Crônica/complicações
10.
Eur Arch Otorhinolaryngol ; 277(1): 221-226, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31541294

RESUMO

OBJECTIVE: Nasopharyngeal carcinoma (NPC) is a common malignancy in Southern China and Southeast Asia. Genetic susceptibility is a major contributing factor in determining the individual risk of NPC in these areas. To test the association between NPC and variants in regenerating gene 1A (REG1A), we conducted a hospital-based case-control study in a Cantonese-speaking population from Guangdong province. METHODS: We endeavored to determine whether genetic variants of the REG1A gene were associated with the risk of NPC amidst the Cantonese population in a hospital-based case-control study using polymerase chain reaction-restriction and direct sequencing analysis in 211 NPC patients and 150 healthy controls. The association between NPC risk and the 14C/T, 20C/T, 369G/T, 1201A/G, and 2922C/T polymorphisms was examined after adjustment for age and sex. RESULTS: We found an increased risk of developing NPC in individuals with REG1A 2922C/T variant genotype (p = 0.003, OR 0.419, 95% CI 0.235-0.746), and after adjustment for sex and age (p = 0.003, OR 0.406, 95% CI 0.226-0.732). No association between other polymorphisms (14C/T, 20C/T, 369G/T, and 1201A/G) and the risk of NPC was observed, before or after adjustment for age and sex. CONCLUSION: Our findings suggest that the REG1A 2922C/T polymorphism is associated with an increased risk of developing NPC in a Cantonese population from Guangdong province. Larger studies are required to confirm our findings and unravel the underlying mechanisms.


Assuntos
Predisposição Genética para Doença/genética , Litostatina/genética , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/genética , Adulto , Povo Asiático/genética , Estudos de Casos e Controles , China/epidemiologia , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/patologia , Polimorfismo de Nucleotídeo Único , Fatores de Risco
11.
Genes (Basel) ; 11(1)2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31877923

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is among the most lethal cancer types world-wide. Its high mortality is related to the difficulty in the diagnosis, which often occurs when the disease is already advanced. As of today, no early diagnostic tests are available, while only a limited number of prognostic tests have reached clinical practice. The main reason is the lack of reliable biomarkers that are able to capture the early development or the progression of the disease. Hence, the discovery of biomarkers for early diagnosis or prognosis of PDAC remains, de facto, an unmet need. An increasing number of studies has shown that cell-free DNA (cfDNA) methylation analysis represents a promising non-invasive approach for the discovery of biomarkers with diagnostic or prognostic potential. In particular, cfDNA methylation could be utilized for the identification of disease-specific signatures in pre-neoplastic lesions or chronic pancreatitis (CP), representing a sensitive and non-invasive method of early diagnosis of PDAC. In this review, we will discuss the advantages and pitfalls of cfDNA methylation studies. Further, we will present the current advances in the discovery of pancreatic cancer biomarkers with early diagnostic or prognostic potential, focusing on pancreas-specific (e.g., CUX2 or REG1A) or abnormal (e.g., ADAMTS1 or BNC1) cfDNA methylation signatures in high risk pre-neoplastic conditions and PDAC.


Assuntos
Carcinoma Ductal Pancreático/genética , Ácidos Nucleicos Livres/genética , Proteína ADAMTS1/genética , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/metabolismo , Ácidos Nucleicos Livres/metabolismo , Metilação de DNA/genética , Proteínas de Ligação a DNA/genética , Epigênese Genética/genética , Proteínas de Homeodomínio/genética , Humanos , Litostatina/genética , Estadiamento de Neoplasias , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Pancreatite/genética , Pancreatite/metabolismo , Prognóstico , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Neoplasias Pancreáticas
12.
Am J Physiol Cell Physiol ; 317(2): C366-C374, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31166710

RESUMO

Regenerating islet-derived (Reg) proteins, which were first discovered in the pancreas, are associated with increased proliferation, prevention of apoptosis, and enhanced differentiation in normal and disease states, but very little is known about the regulation of their expression. We hypothesized that Reg expression is influenced by microRNAs. Bioinformatic analysis predicted Reg1 to be a target of microRNA-7 (miR-7), which influences pancreatic ß-cell function. To this end, we investigated the effects of miR-7 on Reg1 expression in pancreatic acinar and islet ß-cells. High levels of Reg1 were noted by immunostaining and Western blotting in acinar cells in contrast to islet cells. A reciprocal expression pattern was observed for miR-7. Overexpression of miR-7 resulted in Reg1 mRNA suppression and reduction of secreted Reg1 protein. Conversely, miR-7 knockdown led to increases in Reg1. Targeting of Reg1 by miR-7 was confirmed via luciferase activity assays. In contrast, miR-7 did not directly repress the human ortholog of Reg1 REG1A as well as REG1B indicating species differences in the regulation of Reg expression. This is the first account of microRNA modulation of any Reg member warranting studies to fill gaps in our knowledge of Reg protein biology, particularly in disease contexts.


Assuntos
Células Secretoras de Insulina/metabolismo , Litostatina/metabolismo , MicroRNAs/metabolismo , Pâncreas Exócrino/metabolismo , Regiões 3' não Traduzidas , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HEK293 , Humanos , Litostatina/genética , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Pâncreas Exócrino/citologia , Especificidade da Espécie
13.
Mol Cancer Res ; 17(7): 1459-1467, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30988165

RESUMO

Tumor protein p53 (TP53) mutation is a well-known occurrence at the late phase of carcinogenesis during the adenoma-carcinoma sequence of a sporadic colon cancer. Although numerous reports about clinical information of the patients with colon cancer have suggested that TP53 mutation might be related to various types of malignant potential, the direct effects of this mutation on the malignant potential of colon cancer remain unknown. Notably, no previous report has described a relationship between TP53 mutation and cancer stemness. We therefore aimed to assess the function of a TP53 mutant induced by the CRISPR-Cas9 system in colon cancer cells. In this study, two TP53 mutations, corresponding to exon 3 (TP53E3) and 10 (TP53E10), were generated in LS174T cells derived from a wild-type TP53 human colon cancer via a lentiviral CRISPR-Cas9 system. The loss of function of TP53 resulting from both mutations manifested as resistance to Nutlin3a-induced apoptosis and the downregulation of target genes of TP53. TP53 mutants exhibited an enhanced malignant potential, characterized by accelerated cell growth, invasiveness, chemoresistance, and cancer stemness. Interestingly, TP53E10 but not TP53E3 cells exhibited aberrant transcriptional activity of regenerating family member 1-α (REG1A) and expression of REG1A, resulting in the acquisition of enhanced malignant potential. In conclusion, we demonstrated for the first time that TP53 genomic mutation into human colon cancer cells affects the malignant potential. IMPLICATIONS: These findings suggest that both a loss of function and an aberrant gain of function of TP53 might promote high malignant potentials at the late phase of carcinogenesis in colon cancer.


Assuntos
Carcinogênese/genética , Neoplasias do Colo/genética , Litostatina/genética , Proteína Supressora de Tumor p53/genética , Sistemas CRISPR-Cas/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Éxons/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Mutação/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia
14.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2138-2148, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31029827

RESUMO

Defects in the development, maintenance or expansion of ß-cell mass can result in impaired glucose metabolism and diabetes. N6-methyladenosine affects mRNA stability and translation efficiency, and impacts cell differentiation and stress response. To determine if there is a role for m6A in ß-cells, we investigated the effect of Mettl14, a key component of the m6A methyltransferase complex, on ß-cell survival and function using rat insulin-2 promoter-Cre-mediated deletion of Mettl14 mouse line (ßKO). We found that ßKO mice with normal chow exhibited glucose intolerance, lower levels of glucose-stimulated insulin secretion, increased ß-cell death and decreased ß-cell mass. In addition, HFD-fed ßKO mice developed glucose intolerance, decreased ß-cell mass and proliferation, exhibited lower body weight, increased adipose tissue mass, and enhanced insulin sensitivity due to enhanced AKT signaling and decreased gluconeogenesis in the liver. HFD-fed ßKO mice also showed a decrease in de novo lipogenesis, and an increase in lipolysis in the liver. RNA sequencing in islets revealed that Mettl14 deficiency in ß-cells altered mRNA expression levels of some genes related to cell death and inflammation. Together, we showed that Mettl14 in ß-cells plays a key role in ß-cell survival, insulin secretion and glucose homeostasis.


Assuntos
Secreção de Insulina , Metiltransferases/metabolismo , Animais , Sobrevivência Celular , Dieta Hiperlipídica , Regulação da Expressão Gênica , Gluconeogênese , Intolerância à Glucose , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Lipogênese , Litostatina/genética , Litostatina/metabolismo , Fígado/metabolismo , Fígado/patologia , Masculino , Metiltransferases/antagonistas & inibidores , Metiltransferases/genética , Camundongos , Camundongos Knockout , Proteínas Associadas a Pancreatite/genética , Proteínas Associadas a Pancreatite/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo
15.
Cancer Biother Radiopharm ; 34(6): 362-370, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30973271

RESUMO

Objective: The reverse of chemoresistance and the improvement of sensitivity to chemotherapeutic agents of colorectal cancer cells have great clinical significance and the mechanism underlying the drug resistance is still unclear. REG Iα was reported to be upregulated in colorectal cancer tissues, but the roles of chemoresistance are still unclear. Materials and Methods: The expression of REG Iα in colorectal cancer cell lines was assessed by quantitative real-time polymerase chain reaction (Q-PCR). The expression of REG Iα in HCT116 and LOVO cells was knockdown by siRNA. The cell viability and IC50 (half maximal inhibitory concentration) values were analyzed by the CCK8 assay. The proportion of apoptosis and cell cycles were analyzed by flow cytometry. The migration potency of HCT116 and LOVO cells was analyzed by cell migration assay. The protein level of Cyclin D1, CDK4 (cyclin-dependent kinase 4), Bax and Bcl-2 were analyzed by western blot. Results: Knockdown of REG Iα enhances the sensitivity to 5-Fu of colorectal cancer cells. REG Iα knockdown promoted the cell apoptosis of HCT116 and LOVO under the 5-Fu treatment. The cell migration and cycle of colorectal cancer cells was also inhibited by REG Iα knockdown. We also found that REG Iα knockdown induced cell cycle arrest and cell apoptosis by Cyclin D1/CDK4 pathway and BAX/BCL-2 pathways. Conclusions: Knockdown of REG Iα enhances the sensitivity to 5-Fu of colorectal cancer cells via cyclin D1/CDK4 pathway and BAX/BCL-2 pathways.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Fluoruracila/farmacologia , Litostatina/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Apoptose , Ciclo Celular , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Ciclina D1/genética , Quinase 4 Dependente de Ciclina/genética , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Humanos , Litostatina/genética , Litostatina/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Células Tumorais Cultivadas , Proteína X Associada a bcl-2/genética
16.
Hepatology ; 69(6): 2546-2561, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30791110

RESUMO

Chronic infection of hepatitis B virus (HBV) is associated with an increased incidence of hepatocellular carcinoma (HCC). HBV encodes an oncoprotein, hepatitis B x protein (HBx), that is crucial for viral replication and interferes with multiple cellular activities including gene expression, histone modifications, and genomic stability. To date, it remains unclear how disruption of these activities contributes to hepatocarcinogenesis. Here, we report that HBV exhibits antiresection activity by disrupting DNA end resection, thus impairing the initial steps of homologous recombination (HR). This antiresection activity occurs in primary human hepatocytes undergoing a natural viral infection-replication cycle as well as in cells with integrated HBV genomes. Among the seven HBV-encoded proteins, we identified HBx as the sole viral factor that inhibits resection. By disrupting an evolutionarily conserved Cullin4A-damage-specific DNA binding protein 1-RING type of E3 ligase, CRL4WDR70 , through its H-box, we show that HBx inhibits H2B monoubiquitylation at lysine 120 at double-strand breaks, thus reducing the efficiency of long-range resection. We further show that directly impairing H2B monoubiquitylation elicited tumorigenesis upon engraftment of deficient cells in athymic mice, confirming that the impairment of CRL4WDR70 function by HBx is sufficient to promote carcinogenesis. Finally, we demonstrate that lack of H2B monoubiquitylation is manifest in human HBV-associated HCC when compared with HBV-free HCC, implying corresponding defects of epigenetic regulation and end resection. Conclusion: The antiresection activity of HBx induces an HR defect and genomic instability and contributes to tumorigenesis of host hepatocytes.


Assuntos
Carcinoma Hepatocelular/virologia , Instabilidade Genômica/genética , Hepatite B/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virologia , Transativadores/genética , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Células Cultivadas , Proteínas de Ligação a DNA/genética , Epigênese Genética , Hepatite B/patologia , Vírus da Hepatite B/genética , Hepatócitos/citologia , Hepatócitos/fisiologia , Humanos , Litostatina/genética , Neoplasias Hepáticas/patologia , Camundongos , Sensibilidade e Especificidade , Proteínas Virais Reguladoras e Acessórias , Replicação Viral/genética
17.
Neurobiol Dis ; 119: 136-148, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30092268

RESUMO

REG-1α, a secreted protein containing a C-type lectin domain, is expressed in various organs and plays different roles in digestive system cells in physiological and pathological conditions. Like other members of the Reg family, REG-1α is expressed also in the brain where it has different functions. For instance, we previously reported that REG-1α regulates neurite outgrowth and is overexpressed during the very early stages of Alzheimer's disease (AD). However, REG-1α function in neural cells during neural degeneration remains unknown. First, REG-1α and phosphorylated tau expression were assessed in tissue sections from the hippocampus, representing neurofibrillary tangles (NFTs), from patients with AD, and from basal ganglia, representing subcortical NFTs, from patients with progressive supranuclear palsy (PSP). We found an association between REG-1α expression, tau hyperphosphorylation and NFTs in human brain samples from patients with these neurodegenerative diseases. Then, the effects of REG-1α overexpression on tau phosphorylation and axonal morphology were investigated i) in primary cultures of rat neurons that express human tau P301L and ii) in a transgenic zebrafish model of tauopathy that expresses human tau P301L. In the tau P301L cell model, REG-1α overexpression increased tau phosphorylation at the S202/T205 and S396 residues (early and late stages of abnormal phosphorylation, respectively) through the AKT/GSK3-ß pathway. This effect was associated with axonal defects both in tau P301L-expressing rat neurons and zebrafish embryos. Our findings suggest a functional role for REG-1α during tauopathy development and progression and, specifically, its involvement in the modification of tau phosphorylation temporal sequence.


Assuntos
Modelos Animais de Doenças , Litostatina/biossíntese , Tauopatias/metabolismo , Proteínas tau/biossíntese , Idoso , Idoso de 80 Anos ou mais , Animais , Animais Geneticamente Modificados , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Feminino , Humanos , Litostatina/genética , Masculino , Pessoa de Meia-Idade , Fosforilação/fisiologia , Ratos , Tauopatias/genética , Tauopatias/patologia , Peixe-Zebra , Proteínas tau/genética
18.
Biochem Biophys Res Commun ; 503(2): 963-969, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29935186

RESUMO

Recently, we reported the presence of distinct cell clusters named acinar-like cell clusters touching Langerhans islets with thin interstitial surrounding (ATLANTIS) in human pancreas. A morphological study in humans demonstrated that ATLANTIS and islet cell clusters are found together in the microenvironment enclosed by a common basement membrane, and ATLANTIS releases vesicles containing Regenerating gene protein (REG Iα) to islet cell clusters. We examined 1) the presence or absence of ATLANTIS in homozygous Reg I (mouse homologue of human REG Iα) deficient (Reg I-/-) and wild-type mice, and 2) the possible role of ATLANTIS in the regeneration of beta cell clusters after encephalomyocarditis (EMC) virus (D-variant) infection in Reg I-/- and wild-type mice. ATLANTIS was found in both wild-type and Reg I-/- mice. In both groups, mean blood glucose increased transiently to greater than 14.0 mmol/L at 5 days after EMC virus infection and recovered to baseline at 12 days. At 12 days after EMC virus infection, lower BrdU labeling indices were observed in islet beta cells of Reg I-/- mice compared to wild-type mice. Beta cell volume 12 days after EMC virus infection in Reg I-/- mice did not differ from that of wild-type mice. These results suggest that Reg I, which is released from ATLANTIS to islet beta cell clusters, has a crucial role in beta cell regeneration in EMC virus-induced diabetes. The presence of mechanism(s) other than that mediated by Reg I in beta cell restoration after destruction by EMC virus was also suggested.


Assuntos
Infecções por Cardiovirus/complicações , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/virologia , Células Secretoras de Insulina/citologia , Litostatina/metabolismo , Pâncreas/citologia , Animais , Contagem de Células , Proliferação de Células , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Vírus da Encefalomiocardite/isolamento & purificação , Deleção de Genes , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/virologia , Litostatina/genética , Masculino , Camundongos , Mitose , Pâncreas/metabolismo , Pâncreas/patologia , Pâncreas/virologia
19.
Med Sci Monit ; 23: 5834-5843, 2017 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-29222406

RESUMO

BACKGROUND Gastric cancer (GC) is one of the most common cause of cancer-related deaths. The clinical trials still lack the effective methods to treat or monitor the disease progression. In this research, the biological function and the underlying molecular mechanism of regenerating protein 1 alpha (REG1A) in GC were investigated. MATERIAL AND METHODS Gene expression omnibus (GEO), KMplot datasets and GC tissue microarray (n=164) were used to analyze the expression of REG1A and related patient prognoses in GC. Transwell matrigel assay, flow cytometry analysis and CCK8 cell viability assay were performed to detect the biological functions of REG1A. Western blotting and real-time PCR were used to detect the REG1A expression and PI3K/Akt related signaling. RESULTS It was found that the expression of REG1A was significantly downregulated in GC and closely related with clinicopathological findings or patient prognoses. REG1A overexpression could suppress the invasion, cell viability and promote the apoptosis of GC cells. Moreover, we found that the epigenetic methylation suppressed the expression level of REG1A in GC, and REG1A overexpression could suppress the phosphorylation of Akt or GSK3ß signaling. CONCLUSIONS Taken together, REG1A regulates cell invasion, apoptosis and viability in GC through activating PI3K/Akt-GSK3ß signaling. REG1A may serve as a promising therapeutic strategy for GC.


Assuntos
Litostatina/genética , Neoplasias Gástricas/genética , Apoptose/genética , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Sobrevivência Celular/genética , Metilação de DNA/genética , Regulação para Baixo , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Litostatina/metabolismo , Prognóstico , Transdução de Sinais
20.
Biol Res ; 50(1): 37, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29162157

RESUMO

BACKGROUND: Regenerating gene IA (REGIA) plays an important role in tissue regeneration and tumors prognosis of epithelium origin. However, the role of REGIA in nasopharyngeal carcinoma (NPC) is unclear. This study aims to investigate the expression and function of REG1A in NPC. RESULTS: We have found that there was 63 patients with REGIA positive expression of 155 patients in this study (40.65%). The positive expression rate of REGIA was 30.50, 44.44 and 47.83% in stage T2, T3 and T4 patients, respectively. The REGIA expression was significantly difference in T2 and T4 stage tumors or T2 and T3-T4 stage. The positive expression rate of REGIA was found to be higher in patients with cervical lymph node persistence than those with cervical lymph node complete regression. Patients with negative REGIA expression had a better overall survival and free survival than those with REGIA positive expression. In addition, according to the univariate and multivariate analysis, the REGIA expression was an independent adverse prognostic factor for NPC patients. CONCLUSION: REGIA expression was a useful biomarker in NPC patients for assessing T stage and survival.


Assuntos
Carcinoma/genética , Carcinoma/patologia , Litostatina/genética , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Biópsia , Carcinoma/mortalidade , Carcinoma/terapia , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Litostatina/fisiologia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/mortalidade , Neoplasias Nasofaríngeas/terapia , Invasividade Neoplásica/patologia , Prognóstico , Estatísticas não Paramétricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...