Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Environ Anal Health Toxicol ; 39(1): e2024007-0, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38631399

RESUMO

Di(2-ethylhexyl)phthalate (DEHP) is commonly used to increase the flexibility of plastics. In our previous study, DEHP may increase hepatic lipid accumulation through modulating of acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) expression. Nevertheless, it is hard to understand the association between DEHP and DGAT1 in the liver because only one dosage of DEHP was used. Thus, this study performed to investigate the role of DGAT1 on hepatic lipid metabolism after various dosages of DEHP exposure. Four-week-old male Sprague-Dawley rats (n = 5/group) were administered corn oil (vehicle) or DEHP (0.75, 7.5, 15, or 150 mg/kg/day) once daily for seven days. DEHP 150 mg/kg/day treated group increased body weight gain and relative liver weight compared to the control (P = 0.044 and P = 0.049, respectively). In histological observation, elevation of hepatic lipid accumulation was observed in all DEHP-treated groups, except DEHP 150 mg/kg/day, compared to that in the control (all P < 0.001). Portal inflammatory infiltration and acidophilic bodies were observed in the liver at DEHP 7.5 mg/kg/day and above treated groups. In addition, malondiadehyde levels, a marker of lipid peroxidation, in the liver were increased in DEHP 7.5, 15 and 150 mg/kg/day compared to the control (P = 0.017, P = 0.001, and P = 0.002, respectively). The expression of Dgat1 in the liver was significantly increased in DEHP 7.5, 15 and 150 mg/kg/day compared to the control group (P = 0.019, P = 0.002, and P < 0.001, respectively); however, there were no significant changes in the protein levels. Therefore, excessive oxidative stress caused by DEHP may induce liver damage such as inflammation rather than hepatic lipid accumulation by regulating DGAT1 transcription.

2.
Cell Commun Signal ; 22(1): 48, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-38233853

RESUMO

BACKGROUND: Interferon Regulatory Factor 3 (IRF3) is a transcription factor that plays a crucial role in the innate immune response by recognizing and responding to foreign antigens. Recently, its roles in sterile conditions are being studied, as in metabolic and fibrotic diseases. However, the search on the upstream regulator for efficient pharmacological targeting is yet to be fully explored. Here, we show that G protein-coupled receptors (GPCRs) can regulate IRF3 phosphorylation through of GPCR-Gα protein interaction. RESULTS: IRF3 and target genes were strongly associated with fibrosis markers in liver fibrosis patients and models. Conditioned media from MIHA hepatocytes overexpressing IRF3 induced fibrogenic activation of LX-2 hepatic stellate cells (HSCs). In an overexpression library screening using active mutant Gα subunits and Phos-tag immunoblotting, Gαs was found out to strongly phosphorylate IRF3. Stimulation of Gαs by glucagon or epinephrine or by Gαs-specific designed GPCR phosphorylated IRF3. Protein kinase A (PKA) signaling was primarily responsible for IRF3 phosphorylation and Interleukin 33 (IL-33) expression downstream of Gαs. PKA phosphorylated IRF3 on a previously unrecognized residue and did not require reported upstream kinases such as TANK-binding kinase 1 (TBK1). Activation of Gαs signaling by glucagon induced IL-33 production in hepatocytes. Conditioned media from the hepatocytes activated HSCs, as indicated by α-SMA and COL1A1 expression, and this was reversed by pre-treatment of the media with IL-33 neutralizing antibody. CONCLUSIONS: Gαs-coupled GPCR signaling increases IRF3 phosphorylation through cAMP-mediated activation of PKA. This leads to an increase of IL-33 expression, which further contributes to HSC activation. Our findings that hepatocyte GPCR signaling regulates IRF3 to control hepatic stellate cell transdifferentiation provides an insight for understanding the complex intercellular communication during liver fibrosis progression and suggests therapeutic opportunities for the disease. Video Abstract.


Assuntos
Células Estreladas do Fígado , Interleucina-33 , Humanos , Interleucina-33/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Transdiferenciação Celular , Meios de Cultivo Condicionados , Glucagon/metabolismo , Hepatócitos/metabolismo , Cirrose Hepática/metabolismo , Fibrose
3.
Arch Pathol Lab Med ; 147(9): 1060-1074, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-36445719

RESUMO

CONTEXT.­: The tumor-host interaction in the tumor microenvironment (TME) affects the prognosis of patients with malignant tumors. TME assessed via tumor budding (BD) and tumor-infiltrating lymphocyte (TIL) had a prognostic impact in patients with nonampullary small intestinal and colorectal carcinomas. In ampullary carcinoma (AC), MUC5AC was recently revealed as a significant prognosticator, but studies about the TME have not been conducted. OBJECTIVE.­: To assess TME-based prognostic risk in AC. DESIGN.­: We generated a collective TME risk index based on high-grade BD at the invasive front (BD3) and high density of stromal-TIL (>5%) in 64 surgically resected ACs. We evaluated its predictive values for overall survival (OS) and recurrence-free survival (RFS). We also investigated the relationship of TME to MUC5AC expression. RESULTS.­: TME prognostic risk index was classified into low-risk (BDLow/TILHigh; 26 of 64; 41%), intermediate-risk (BDLow/TILLow or BDHigh/TILHigh; 23; 36%), and high-risk (BDHigh/TILLow; 15; 23%) groups. Higher TME prognostic risk was associated with higher tumor grade (P = .03), lymphovascular invasion (P = .05), and MUC5AC immunopositivity (P = .02). TME prognostic risk index displayed better predictive ability for both OS (53.9 versus 46.1 versus 42.2) and RFS (24.8 versus 16.9 versus 15.3) than BD or TIL alone. In multivariate analysis, TME prognostic risk index was an independent prognosticator for OS (P = .003) and RFS (P = .03). CONCLUSIONS.­: TME risk index in combination with BD and TIL was a stronger predictor of prognostic risk stratification than either BD or TIL alone for both OS and RFS in patients with AC. MUC5AC may modulate the interaction between tumor cells and immunity toward enhancing invasiveness in TME.


Assuntos
Ampola Hepatopancreática , Neoplasias Colorretais , Humanos , Prognóstico , Microambiente Tumoral , Ampola Hepatopancreática/metabolismo , Ampola Hepatopancreática/patologia , Neoplasias Colorretais/patologia , Linfócitos do Interstício Tumoral , Mucina-5AC/metabolismo
4.
Mol Neurobiol ; 60(1): 145-159, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36242734

RESUMO

Although a couple of studies have reported that mutant superoxide dismutase 1 (SOD1), one of the causative genes of familial amyotrophic lateral, interacts physically with lysyl-tRNA synthetase (KARS1) by a gain of function, there is limited evidence regarding the detailed mechanism about how the interaction leads to neuronal cell death. Our results indicated that the aminoacyl-tRNA synthetase-interacting multi-functional protein 2 (AIMP2) mediated cell death upon the interplay between mutant SOD1 and KARS1 in ALS. Binding of mutant SOD1 with KARS1 led to the release of AIMP2 from its original binding partner KARS1, and the free form of AIMP2 induced TRAF2 degradation followed by TNF-α-induced cell death. We also suggest a therapeutic application that overexpression of DX2, the exon 2-deleted antagonistic splicing variant of AIMP2 (AIMP2-DX2), reduced neuronal cell death in the ALS mouse model. Expression of DX2 suppressed TRAF2 degradation and TNF-α-induced cell death by competing mode of action against full-length AIMP2. Motor neuron differentiated form iPSC showed a resistance in neuronal cell death after DX2 administration. Further, intrathecal administration of DX2-coding adeno-associated virus (AAV) improved locomotive activity and survival in a mutant SOD1-induced ALS mouse model. Taken together, these results indicated that DX2 could prolong life span and delay the ALS symptoms through compensation in neuronal inflammation.


Assuntos
Esclerose Amiotrófica Lateral , Proteínas Nucleares , Animais , Camundongos , Morte Celular , Linhagem Celular Tumoral , Mutação , Proteínas Nucleares/metabolismo , Superóxido Dismutase-1/metabolismo , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Isoformas de Proteínas
5.
Am J Surg Pathol ; 45(7): 917-929, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33443865

RESUMO

The tumor microenvironment (TME) has become an important area of investigation with respect to improving prognosis in malignancies. Here we evaluated TME prognostic risk in small intestinal adenocarcinomas based on histologic assessment of tumor budding at the peritumoral-invasive front (pTB) and stromal tumor-infiltrating lymphocytes (sTILs). pTB and sTILs were analyzed in 230 surgically resected small intestinal adenocarcinomas, as recommended by the International Tumor Budding Consensus Conference (ITBCC) and the International TILs Working Group (ITWG). On the basis of high levels of pTB count (≥10) and sTIL density (≥20%), we combined pTB and sTIL to produce a collective TME-based prognostic risk index: low-risk (pTBLow/sTILHigh; n=39, 17.0%), intermediate-risk (pTBLow/sTILLow or pTBHigh/sTILHigh; n=99, 43.0%), and high-risk groups (pTBHigh/sTILLow; n=92, 40.0%). TME risk index provided better prognostic stratification than the individual pTB and sTIL (14.9 vs. 6.7 vs. 10.3). Tumors with higher TME prognostic risk were associated with an infiltrative growth pattern and nonintestinal immunophenotype (both P=0.001), pancreatic invasion (P=0.010), lymphovascular (P<0.001) or perineural invasion (P=0.006), higher T-category (P<0.001), N-category (P=0.004), and stage grouping (P=0.002), and KRAS mutation (P=0.008). In multivariate analysis, higher TME prognostic risk index (P<0.001), distal tumor location and nonintestinal immunophenotype (both P=0.001), higher N-category (P<0.001), and microsatellite stable (P=0.015) were worse-independent prognosticators. TME prognostic risk index consistently stratified patient survival regardless of tumor location (P<0.001 in proximal; P=0.002 in distal), stages (P<0.001 in lower stages I to II; P=0.028 in stage III), and DNA mismatch repair gene status (P<0.001 in microsatellite stable; P=0.001 in microsatellite instability). TME risk index is a powerful prognostic predictor for risk stratification of patients with small intestinal adenocarcinoma.


Assuntos
Adenocarcinoma/patologia , Movimento Celular , Técnicas de Apoio para a Decisão , Neoplasias Intestinais/patologia , Linfócitos do Interstício Tumoral/patologia , Células Estromais/patologia , Microambiente Tumoral , Adenocarcinoma/mortalidade , Adenocarcinoma/cirurgia , Idoso , Procedimentos Cirúrgicos do Sistema Digestório/efeitos adversos , Procedimentos Cirúrgicos do Sistema Digestório/mortalidade , Neoplasias Duodenais/mortalidade , Neoplasias Duodenais/patologia , Neoplasias Duodenais/cirurgia , Feminino , Humanos , Neoplasias do Íleo/mortalidade , Neoplasias do Íleo/patologia , Neoplasias do Íleo/cirurgia , Neoplasias Intestinais/mortalidade , Neoplasias Intestinais/cirurgia , Neoplasias do Jejuno/mortalidade , Neoplasias do Jejuno/patologia , Neoplasias do Jejuno/cirurgia , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
6.
Cancer Genomics Proteomics ; 17(6): 813-826, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33099482

RESUMO

BACKGROUND/AIM: Mesonephric carcinoma (MNC) is a rare but notable entity of the female genital tract. While many researchers have acknowledged and studied MNC, much remains unknown on the characteristics of mesonephric remnant (MNR) or hyperplasia (MNH). There has not been any study examining the molecular features of MNR and MNH so far. The aim of this study was to investigate the clinicopathological and molecular characteristics of ten uterine mesonephric lesions, including two MNRs without atypia, four MNHs without atypia, and three MNHs with atypia. MATERIALS AND METHODS: We reviewed the electronic medical records and all available slides of ten cases from multiple institutions. Targeted sequencing and array comparative genomic hybridization were performed. RESULTS: Three atypical MNHs displayed nuclear enlargement, mild-to-moderate nuclear pleomorphism, and nuclear membrane irregularity, and harbored pathogenic Kirsten rat sarcoma 2 viral oncogene homolograt sarcoma 2 viral oncogene homolog (KRAS) mutation. Two of those that co-existed with MNC harbored the same sequence alterations as each of their adjacent MNC. One of the three atypical MNHs harbored chromosome 1q gain. CONCLUSION: Atypical MNH is a potential premalignant lesion in which KRAS mutation and chromosome 1q gain play an important role in the early stage of mesonephric carcinogenesis.


Assuntos
Cromossomos Humanos Par 1/genética , Mutação com Ganho de Função , Hiperplasia/patologia , Mesonefroma/patologia , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Neoplasias do Colo do Útero/patologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Biomarcadores Tumorais/genética , Feminino , Humanos , Hiperplasia/genética , Mesonefroma/genética , Pessoa de Meia-Idade , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Prognóstico , Neoplasias do Colo do Útero/genética
7.
Cancer Genomics Proteomics ; 17(6): 803-812, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33099481

RESUMO

BACKGROUND/AIM: Glioblastoma is the most malignant form of astrocytoma. The purpose of this study was to analyze the genetic characteristics of primary and recurrent glioblastomas using targeted sequencing and investigate the differences in mutational profiles between the locations of tumor recurrence. MATERIALS AND METHODS: Fourteen glioblastoma patients who developed local (n=10) or distal (n=4) recurrence were included in the study. Targeted sequencing analysis was performed using the primary (n=14) and corresponding recurrent (n=14) tumor tissue samples. RESULTS: The local and distal recurrence groups showed different genetic evolutionary patterns. Most of the locally recurrent glioblastomas demonstrated concordant mutational profiles between the primary and recurrent tumors, suggesting a linear evolution. In contrast, all cases of distally recurrent glioblastomas showed changes in mutational profiles with newly acquired mutations when compared to the corresponding primary tumors, suggesting a branching evolution. CONCLUSION: Locally and distally recurrent glioblastomas exhibit different evolutionary patterns.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação , Recidiva Local de Neoplasia/patologia , Sequenciamento Completo do Genoma/métodos , Neoplasias Encefálicas/genética , Evolução Molecular , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Humanos , Recidiva Local de Neoplasia/genética , Prognóstico
9.
Cancer Genomics Proteomics ; 15(4): 291-298, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29976634

RESUMO

BACKGROUND/AIM: Combined hepatocellular-cholangiocarcinoma (cHCC-CC) is a very rare type of tumor, comprising these two different components in a single mass. Although several studies have determined the genetic characteristics of cHCC-CC, next-generation sequencing (NGS) data for comparing clonality of cHCC-CC are currently unavailable. MATERIALS AND METHODS: Four cHCC-CC cases were selected and HCC, CC and normal components from each case were separately micro-dissected. DNA and RNA were isolated from each sample and sequenced by Oncomine Comprehensive Panel interrogating 143 cancer genes using the Ion S5 XL sequence platform. Genetic features of HCC and CC from each patient were compared. RESULTS: All cases successfully produced NGS data. Two cases demonstrated different mutations in their HCC and CC components (biclone), while two cases shared the same mutations in the two components (monoclone). Single nucleotide polymorphisms (SNPs) of TP53 (4/4) and phosphatase and tensin homolog (PTEN) (1/4), and gene amplifications of mesenchymal-epithelial transition factor (MET) (1/4), c-MYC (1/4), and cyclin-dependent kinase 6 (CDK6) (1/4) were found in the CC component. In the HCC component, SNPs of TP53 (3/4), PTEN (1/4) and catenin beta 1 (CTNNB1) (1/4) and cyclin D1 (CCND1) amplification (1/4) were detected. Two biclonal cases showed a histologically distinct border between HCC and CC components with or without intermediate cell foci. Two monoclonal cases showed a histologically ambiguous border between HCC and CC components with more intermingled pattern than biclonal cases. CONCLUSION: Based on our study, cHCC-CC can be genetically divided into biclonal and monoclonal forms. Therefore, separate sequencing of each component of cHCC-CC is recommended for exact molecular classification and targeted therapy.


Assuntos
Neoplasias dos Ductos Biliares/diagnóstico , Carcinoma Hepatocelular/diagnóstico , Colangiocarcinoma/diagnóstico , Evolução Clonal , Neoplasias Hepáticas/diagnóstico , Idoso , Neoplasias dos Ductos Biliares/genética , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Linhagem da Célula , Colangiocarcinoma/genética , Diagnóstico Diferencial , Feminino , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Neoplasias Hepáticas/genética , Masculino , Pessoa de Meia-Idade , Mutação , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Polimorfismo de Nucleotídeo Único
10.
Oncotarget ; 8(13): 21483-21500, 2017 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-28206961

RESUMO

Lynch syndrome is an autosomal-dominant disorder caused by defective DNA mismatch repair (MMR) genes and is associated with increased risk of malignancies in multiple organs. Small-intestinal adenocarcinomas are common initial manifestations of Lynch syndrome. To define the incidence and characteristics of Lynch syndrome-related small-intestinal adenocarcinomas, meticulous familial and clinical histories were obtained from 195 patients with small-intestinal adenocarcinoma, and MMR protein immunohistochemistry, microsatellite instability, MLH1 methylation, and germline mutational analyses were performed. Lynch syndrome was confirmed in eight patients (4%), all of whom had synchronous/metachronous malignancies without noticeable familial histories. Small-intestinal adenocarcinomas were the first clinical manifestation in 37% (3/8) of Lynch syndrome patients, and second malignancies developed within 5 years in 63% (5/8). The patients with accompanying Lynch syndrome were younger (≤50 years; P=0.04) and more likely to have mucinous adenocarcinomas (P=0.003), and tended to survive longer (P=0.11) than those with sporadic cases. A meticulous patient history taking, MMR protein immunolabeling, and germline MMR gene mutational analysis are important for the diagnosis of Lynch syndrome-related small-intestinal adenocarcinomas. Identifying Lynch syndrome in patients with small-intestinal adenocarcinoma can be beneficial for the early detection and treatment of additional Lynch syndrome-related cancers, especially in patients who are young or have mucinous adenocarcinomas.


Assuntos
Adenocarcinoma/epidemiologia , Neoplasias Colorretais Hereditárias sem Polipose/epidemiologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adulto , Idoso , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais Hereditárias sem Polipose/patologia , Reparo de Erro de Pareamento de DNA/genética , Análise Mutacional de DNA , Proteínas de Ligação a DNA/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Incidência , Masculino , Microdissecção , Pessoa de Meia-Idade , Endonuclease PMS2 de Reparo de Erro de Pareamento/genética , Proteína 1 Homóloga a MutL/genética , Proteína 2 Homóloga a MutS/genética , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...