Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38673741

RESUMO

A widely used organophosphate flame retardant (OPFR), triphenyl phosphate (TPP), is frequently detected in various environmental media and humans. However, there is little known on the human corneal epithelium of health risk when exposed to TPP. In this study, human normal corneal epithelial cells (HCECs) were used to investigate the cell viability, morphology, apoptosis, and mitochondrial membrane potential after they were exposed to TPP, as well as their underlying molecular mechanisms. We found that TPP decreased cell viability in a concentration-dependent manner, with a half maximal inhibitory concentration (IC50) of 220 µM. Furthermore, TPP significantly induced HCEC apoptosis, decreased mitochondrial membrane potential in a dose-dependent manner, and changed the mRNA levels of the apoptosis biomarker genes (Cyt c, Caspase-9, Caspase-3, Bcl-2, and Bax). The results showed that TPP induced cytotoxicity in HCECs, eventually leading to apoptosis and changes in mitochondrial membrane potential. In addition, the caspase-dependent mitochondrial pathways may be involved in TPP-induced HCEC apoptosis. This study provides a reference for the human corneal toxicity of TPP, indicating that the risks of OPFR to human health cannot be ignored.


Assuntos
Apoptose , Sobrevivência Celular , Epitélio Corneano , Retardadores de Chama , Potencial da Membrana Mitocondrial , Mitocôndrias , Humanos , Apoptose/efeitos dos fármacos , Retardadores de Chama/toxicidade , Retardadores de Chama/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/metabolismo , Epitélio Corneano/citologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Caspases/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Organofosfatos/farmacologia , Organofosfatos/toxicidade , Células Cultivadas
2.
J Mater Chem B ; 12(17): 4097-4117, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38587869

RESUMO

Single phototherapy and immunotherapy have individually made great achievements in tumor treatment. However, monotherapy has difficulty in balancing accuracy and efficiency. Combining phototherapy with immunotherapy can realize the growth inhibition of distal metastatic tumors and enable the remote monitoring of tumor treatment. The development of nanomaterials with photo-responsiveness and anti-tumor immunity activation ability is crucial for achieving photo-immunotherapy. As immune adjuvants, photosensitizers and photothermal agents, manganese-based nanoparticles (Mn-based NPs) have become a research hotspot owing to their multiple ways of anti-tumor immunity regulation, photothermal conversion and multimodal imaging. However, systematic studies on the synergistic photo-immunotherapy applications of Mn-based NPs are still limited; especially, the green synthesis and mechanism of Mn-based NPs applied in immunotherapy are rarely comprehensively discussed. In this review, the synthesis strategies and function of Mn-based NPs in immunotherapy are first introduced. Next, the different mechanisms and leading applications of Mn-based NPs in immunotherapy are reviewed. In addition, the advantages of Mn-based NPs in synergistic photo-immunotherapy are highlighted. Finally, the challenges and research focus of Mn-based NPs in combination therapy are discussed, which might provide guidance for future personalized cancer therapy.


Assuntos
Imunoterapia , Manganês , Humanos , Manganês/química , Manganês/farmacologia , Imunoterapia/métodos , Fototerapia/métodos , Química Verde , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Animais , Nanoestruturas/química , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/síntese química , Tamanho da Partícula
3.
Int J Mol Sci ; 23(22)2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36430782

RESUMO

Tris (1-chloro-2-propyl) phosphate (TCPP) is one of the most frequently detected organophosphorus flames in the environment. Continuous daily exposure to TCPP may harm human skin. However, little is known about the adverse effects of TCPP on human skin. In this study, we first evaluated the detrimental effects and tried to uncover the underlying mechanisms of TCPP on human skin keratinocytes (HaCaT) after 24 h exposure. We found that TCPP caused a concentration-dependent decrease in HaCaT cell viability after exposure to 1.56-400 µg/mL for 24 h, with an IC50 of 275 µg/mL. TCPP also promoted the generation of intracellular reactive oxygen species (ROS) and triggered DNA damage, evidenced by an increase of phosphorylated histone H2A.X (γH2A.X) in the nucleus. Furthermore, the cell cycle was arrested at the G1 phase at 100 µg/mL by upregulation of the mRNA expression of p53 and p21 and downregulation of cyclin D1 and CDK4 expression. Additionally, both the senescence-associated-ß-galactosidase activity and related proinflammatory cytokine IL-1ß and IL-6 were elevated, indicating that TCPP exposure caused cellular senescence may be through the p53-dependent DNA damage signal pathway in HaCaT cells. Taken together, our data suggest that flame-retardant exposure may be a key precipitating factor for human skin aging.


Assuntos
Retardadores de Chama , Envelhecimento da Pele , Humanos , Senescência Celular , Retardadores de Chama/toxicidade , Queratinócitos/metabolismo , Compostos Organofosforados/toxicidade , Compostos Organofosforados/metabolismo , Proteína Supressora de Tumor p53/metabolismo
4.
Transl Oncol ; 18: 101367, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35176624

RESUMO

BACKGROUND: Immune cells, vital components of tumor microenvironment, regulate tumor survival and progression. Lung adenocarcinoma (LUAD), the tumor with the highest mortality rate worldwide, reconstitutes tumor immune microenvironment (TIME) to avoid immune destruction. Data have shown that TIME influences LUAD prognosis and predicts immunotherapeutic efficacy. The related information about the role of TIME's characteristics in LUAD is limited. METHODS: We performed unsupervised consensus clustering via machine-learning techniques to identify TIME clusters among 1906 patients and gathered survival data. The characteristics of TIME clusters of LUAD were visualized by multi-omics analysis, pseudo-time dynamic analysis, and enrichment analysis. TIME score model was constructed by principal component analysis. Comprehensive analysis and validation were conducted to test the prognostic efficacy and immunotherapeutic response of TIME score. RESULTS: TIME clusters (A, B and C) were constructed and exhibited different immune infiltration states. Multi-omics analyses included significant mutated genes (SMG), copy number variation (CNV) and cancer stemness that were significantly different among the three clusters. TIME cluster A had a lower SMG, lower CNV, and lower stemness but a higher immune infiltration level compared to TIME clusters B and C. TIME score showed that patients in low TIME score group had higher overall survival rates, higher immune infiltration level and high expression of immune checkpoints. In validation cohorts, low TIME score subgroup had better drug sensitivity and favorable immunotherapeutic response. CONCLUSION: We constructed a stable model of LUAD immune microenvironment characteristics that may improve the prognostic accuracy of patients, provide improved explanations of LUAD responses to immunotherapy, and provide new strategies for LUAD treatment.

5.
Clin Transl Med ; 11(9): e517, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34586745

RESUMO

BACKGROUND: Platinum-based chemotherapy is effective in inducing shrinkage of primary lung cancer lesions; however, it shows finite therapeutic efficacy in patients suffering from brain metastasis (BM). The intrinsic changes of BM cells, which contribute to the poor results remain unknown. METHODS: Platinum drug-sensitivity was assessed by utilizing a preclinical BM model of PC9 lung adenocarcinoma cells in vitro and in vivo. High consumption of glutathione (GSH) and two associated upregulated proteins (GPX4 and GSTM1) in BM were identified by integrated metabolomics and proteomics in cell lines and verified by clinical serum sample. Gain-of-function and rescue experiments were implemented to reveal the impact and mechanism of GPX4 and GSTM1 on the chemosensitivity in BM. The interaction between GPX4 and GSTM1 was examined by immunoblotting and immunoprecipitation. The mechanism of upregulation of GPX4 was further uncovered by luciferase reporter assay, immunoprecipitation, and electrophoretic mobility shift assay. RESULTS: The derivative brain metastatic subpopulations (PC9-BrMs) of parental cells PC9 developed obvious resistance to platinum. Radically altered profiles of BM metabolism and protein expression compared with primary lung cancer cells were described and GPX4 and GSTM1 were identified as being responsible for the high consumption of GSH, leading to decreased chemosensitivity by negatively regulating ferroptosis. Besides, GSTM1 was found regulated by GPX4, which was transcriptionally activated by the Wnt/NR2F2 signaling axis in BM. CONCLUSIONS: Collectively, our findings demonstrated that Wnt/NR2F2/GPX4 promoted acquired chemoresistance by suppressing ferroptosis with high consumption of GSH. GPX4 inhibitor was found to augment the anticancer effect of platinum drugs in lung cancer BM, providing novel strategies for lung cancer patients with BM.


Assuntos
Neoplasias Encefálicas , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Platina/farmacologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Ferroptose/genética , Glutationa/metabolismo , Glutationa Transferase/química , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/antagonistas & inibidores , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/química , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo
6.
Environ Health Prev Med ; 26(1): 80, 2021 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-34388980

RESUMO

High levels (> 100 ug/L) of arsenic are known to cause lung cancer; however, whether low (≤ 10 ug/L) and medium (10 to 100 ug/L) doses of arsenic will cause lung cancer or other lung diseases, and whether arsenic has dose-dependent or threshold effects, remains unknown. Summarizing the results of previous studies, we infer that low- and medium-concentration arsenic cause lung diseases in a dose-dependent manner. Arsenic trioxide (ATO) is recognized as a chemotherapeutic drug for acute promyelocytic leukemia (APL), also having a significant effect on lung cancer. The anti-lung cancer mechanisms of ATO include inhibition of proliferation, promotion of apoptosis, anti-angiogenesis, and inhibition of tumor metastasis. In this review, we summarized the role of arsenic in lung disease from both pathogenic and therapeutic perspectives. Understanding the paradoxical effects of arsenic in the lungs may provide some ideas for further research on the occurrence and treatment of lung diseases.


Assuntos
Arsênio , Neoplasias Pulmonares , Pulmão , Animais , Arsênio/efeitos adversos , Arsênio/uso terapêutico , Arsênio/toxicidade , Relação Dose-Resposta a Droga , Humanos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/tratamento farmacológico , Camundongos
7.
Aging (Albany NY) ; 13(12): 16577-16599, 2021 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-34175839

RESUMO

Since the imbalance of gene expression has been demonstrated to tightly related to breast cancer (BRCA) genesis and growth, common genes expressed of BRCA were screened to explore the essence in-between. In current work, most common differentially expressed genes (DEGs) in various subtypes of BRCA were identified. Functional enrichment analysis illustrated the driving factor of deactivation of the cell cycle and the oocyte meiosis, which critically triggers the development of BRCA. Herein, we constructed a 12-gene prognostic risk model relative to differential gene expression. Subsequently, the K-M curves, analysis on time-ROC curve and Cox regression were performed to assess this risk model by determining the respective prognostic value, and the prediction performance were ascertained for both training and validation cohorts. In addition, multivariate Cox regression was analysed to reveal the independence between risk score and prognostic stage, and the accuracy and sensitivity of prognosis are particularly improved after clinical indicators are included into the analysis. In summary, this study offers novel insights into the imbalance of gene expression within BRCA, and highlights 12 selected genes associated with patient prognosis. The risk model can help individualize treatment for patients at different risks, and propose precise strategies and treatments for BRCA therapy.


Assuntos
Neoplasias da Mama/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Modelos Biológicos , Bases de Dados Genéticas , Feminino , Ontologia Genética , Humanos , Análise Multivariada , Prognóstico , Modelos de Riscos Proporcionais , Mapas de Interação de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco , Análise de Sobrevida
8.
Cancer Biol Med ; 17(2): 458-467, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32587781

RESUMO

Objective: Lynch syndrome (LS) predisposes patients to early onset endometrioid endometrial cancer (EEC). However, little is known about LS-related EEC in the Chinese population. The aim of this study was to investigate the prevalence of LS and to identify the specific variants of LS in Chinese patients with EEC. Methods: We applied universal immunohistochemistry screening to detect the expression of mismatch repair (MMR) proteins, which was followed by MLH1 methylation analysis to identify suspected LS cases, next-generation sequencing (NGS) to confirm LS, and microsatellite instability (MSI) analysis to verify LS. Results: We collected 211 samples with EEC. Twenty-seven (27/211, 12.8%) EEC cases had a loss of MMR protein expression. After MLH1 methylation analysis, 16 EEC cases were suggested to be associated with LS. Finally, through NGS and MSI analysis, we determined that 10 EEC (10/209, 4.78%) cases were associated with LS. Among those cases, 3 unreported mutations (1 frameshift and 2 nonsense) were identified. MSH6 c.597_597delC, found in 4 patients, is likely to be a founder mutation in China. Conclusions: We demonstrated the feasibility of a process for LS screening in Chinese patients with EEC, by using universal immunohistochemistry screening followed by MLH1 methylation analysis and confirmation through NGS and MSI analysis. The novel mutations identified in this study expand knowledge of LS.


Assuntos
Neoplasias Colorretais Hereditárias sem Polipose/genética , Reparo de Erro de Pareamento de DNA , Neoplasias do Endométrio/genética , Proteína 1 Homóloga a MutL/genética , Adulto , Idoso , Povo Asiático/genética , Biomarcadores Tumorais/genética , China , Neoplasias Colorretais Hereditárias sem Polipose/diagnóstico , Neoplasias Colorretais Hereditárias sem Polipose/patologia , Metilação de DNA , Neoplasias do Endométrio/diagnóstico , Neoplasias do Endométrio/patologia , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Instabilidade de Microssatélites , Pessoa de Meia-Idade
9.
Int J Gynecol Pathol ; 36(1): 58-63, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27171541

RESUMO

In order to explore whether NF-κB activation correlates to the prognosis, chemoresistance, and sex hormone receptors status in ovarian serous carcinoma, we analyzed the expression of NF-κB, ER, and PR by immunohistochemistry in 72 cases of ovarian serous carcinoma, investigated the association among these markers, and evaluated their relations to clinicopathologic factors and prognosis. The positive rates were 88.9% for NF-κB cytoplasmic expression, 45.8% for NF-κB nuclear expression, 41.7% for ER, and 29.2% for PR. NF-κB nuclear expression was positively correlated with the 4th edition WHO grade (P=0.045) and tumor stage (P=0.001). NF-κB cytoplasmic expression was associated with preoperative serum CA125 level (P=0.015) and ascites (P=0.042). Neither cytoplasmic nor nuclear staining of NF-κB showed any association with survival. PR expression was correlated with tumor stage (P=0.023) and omental metastasis (P=0.022). Omental metastasis occurred more frequently in ER-/PR- tumors (P=0.009). No correlation between NF-κB expression and ER, PR expression was observed. In conclusion, in ovarian serous carcinoma, NF-κB nuclear expression correlated with the 4th edition WHO grade and PR was a favorable prognostic factor for ovarian serous carcinoma.


Assuntos
Cistadenocarcinoma Seroso/metabolismo , NF-kappa B/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Cistadenocarcinoma Seroso/diagnóstico , Cistadenocarcinoma Seroso/patologia , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/patologia , Prognóstico
10.
J Clin Pathol ; 69(2): 122-7, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26254281

RESUMO

AIM: Tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that preferentially induces apoptosis in cancer cells while exhibiting little or no toxicity in normal cells. In this study, we evaluated the clinicopathological significance of TRAIL signalling members' expression profiles in cervical squamous cell carcinoma (CSCC). METHODS: TRAIL, DR5, caspase-8 and cellular FLICE-inhibitory protein (c-FLIP) protein expression was investigated in 72 stage IA2-IIIA CSCC patients using immunohistochemistry. Correlation between protein expression and clinicopathological features, radiotherapy response and survival was statistically analysed. RESULTS: Positive c-FLIP expression was an independent negative indicator for disease-free survival (DFS) (p=0.015) in multivariate Cox regression analysis. The DR5 nuclear positive group (p=0.069 by log rank test) showed some advantage of radiotherapy for overall survival (OS) compared with the DR5 nuclear negative cohort (p=0.568 by log rank test). In addition, loss of TRAIL expression was associated with worse differentiation (p=0.004), while absence of caspase-8 staining was more frequently observed in cases with lymphovascular invasion (p=0.035). CONCLUSIONS: High c-FLIP expression is shown to be an independent prognostic variable, DR5 nuclear expression may serve as a predictive biomarker for radiotherapy, and TRAIL as well as caspase-8 loss may be associated with malignant progression.


Assuntos
Biomarcadores Tumorais/análise , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/análise , Carcinoma de Células Escamosas/química , Caspase 8/análise , Receptores do Fator de Necrose Tumoral/análise , Ligante Indutor de Apoptose Relacionado a TNF/análise , Neoplasias do Colo do Útero/química , Adulto , Idoso , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/cirurgia , Distribuição de Qui-Quadrado , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Análise Multivariada , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Radioterapia Adjuvante , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Neoplasias do Colo do Útero/mortalidade , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/cirurgia , Adulto Jovem
11.
Mol Med Rep ; 13(1): 837-44, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26648170

RESUMO

Protein 4.1N (4.1N) is a member of the protein 4.1 family and is essential for the regulation of cell adhesion, motility and signaling. Previous studies have suggested that 4.1N may serve a tumor suppressor role. However, the molecular mechanisms remain unclear. In the current study, the role of 4.1N in the downregulation of hypoxia­induced factor 1α (HIF­1α) under hypoxic conditions and therefore the suppression of hypoxia induced epithelial­mesenchymal transition (EMT) was investigated. The data were obtained from overexpressed and knockdown 4.1N epithelial ovarian cancer (EOC) cell lines. It was identified that 4.1N was capable of regulating the sub­cellular localization and expression levels of HIF­1α, by which 4.1N served a dominant role in the suppression of hypoxia­induced EMT and associated genes. Collectively, the data of the current study identified 4.1N as an inhibitor of hypoxia­induced tumor progression in EOC cells and highlighted its potential role in EOC therapy.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Transição Epitelial-Mesenquimal , Proteínas de Membrana/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neuropeptídeos/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Caderinas/metabolismo , Carcinoma Epitelial do Ovário , Hipóxia Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular , Feminino , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Modelos Biológicos
12.
J Clin Pathol ; 68(7): 522-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25827135

RESUMO

AIMS: Our aim was to analyse the utility of the algorithm combining PAX8 with clinicopathological characteristics (tumour size, laterality and patient age) in differentiating primary ovarian mucinous tumours (POMTs) from extragenital metastatic mucinous carcinomas involving the ovary (eMOMCs). METHODS AND RESULTS: Immunohistochemical staining for PAX8 was performed on formalin fixed, paraffin embedded tissues from 47 POMTs, 18 eMOMCs and 70 extragenital primary mucinous carcinomas (ePMCs) using anti-PAX8 rabbit polyclonal antibody (pAb) and anti-PAX8 rabbit monoclonal antibody (mAb). PAX8 (pAb) positive signals were found in 3/18 eMOMCs and in 32/70 ePMCs. PAX8 (mAb) demonstrated superior specificity, with 0% positivity in both eMOMCs and ePMCs, but unfavourable sensitivity, with 60.9% in ovarian mucinous borderline tumours and 45.8% in POMCs. Although PAX8 (mAb) immunostaining status (66.2%), tumour size (75.4%) and laterality (84.6%) demonstrated unsatisfactory accuracy when they were evaluated individually in differentiating POMTs from eMOMCs, a combination of PAX8 (mAb) immunostaining status, tumour size and laterality markedly increased accuracy (86.2%), with a satisfactory Youden Index (63.7%). CONCLUSIONS: PAX8 (mAb) was a specific marker in differentiating POMTs from eMOMCs. As a simple, convenient and high performance to price ratio algorithm, a combination of PAX8 (mAb) immunostaining with tumour size and laterality will improve the diagnostic criteria of ovarian mucinous metastasis.


Assuntos
Adenocarcinoma Mucinoso/química , Adenocarcinoma Mucinoso/patologia , Algoritmos , Biomarcadores Tumorais/análise , Neoplasias Ovarianas/química , Neoplasias Ovarianas/patologia , Fatores de Transcrição Box Pareados/análise , Carga Tumoral , Adenocarcinoma Mucinoso/secundário , Fatores Etários , Área Sob a Curva , Biópsia , Diagnóstico Diferencial , Feminino , Humanos , Imuno-Histoquímica , Estadiamento de Neoplasias , Neoplasias Ovarianas/secundário , Fator de Transcrição PAX8 , Valor Preditivo dos Testes , Curva ROC
14.
Sci China Life Sci ; 57(7): 690-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24907935

RESUMO

Kindlin-2 functions in the maintenance of homeostasis and in human diseases. This study investigated the interrelationship between Kindlin-2 expression in tissues and the corresponding germ layers from which these tissues originated. Kindlin-2 expression was examined in normal adult human organs and human cancer tissues by immunohistochemical analyses. Analysis of Kindlin-2 mRNA levels in adult human organs in the Oncomine dataset revealed Kindlin-2 is highly expressed in mesoderm-derived organs. However, Kindlin-2 was negative or weakly expressed in endoderm/ectoderm-derived organs. Interestingly, the abnormal expression of Kindlin-2 was observed in a variety of human cancers. In agreement with its expression profile in humans, Kindlin-2 was also highly expressed in mesoderm-derived organs in mouse embryos with the exception of strong Kindlin-2 expression in ectoderm-derived spinal cord and ganglia, tissues that are highly mobile during embryonic development. Importantly, we demonstrated the expression level of Kindlin-2 in adult organs correlated with their embryonic dermal origins and deregulation of Kindlin-2 in tissues is associated with tumor progression. This finding will help us understand the dual role of Kindlin-2 in the regulation of tumor progression and embryonic development.


Assuntos
Perfilação da Expressão Gênica , Camadas Germinativas/metabolismo , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Adulto , Animais , Ectoderma/metabolismo , Embrião de Mamíferos/metabolismo , Endoderma/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/metabolismo , Mesoderma/metabolismo , Camundongos Endogâmicos ICR , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Biochem Biophys Res Commun ; 446(1): 187-94, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24583125

RESUMO

Kindlin-2 has been known to promote most cancer progression through regulation of multiple signaling pathways. However, a novel tumor suppressive role of Kindlin-2 was identified in serous epithelial ovarian cancer progression, which sharply contrasts to the tumor promoting roles for Kindlin-2 in most other cancers. While we demonstrated that Kindlin-2 was highly expressed in control tissues, a drastic low expression of Kindlin-2 was found in the tumor tissues of serous epithelial ovarian cancer, especially in the high-grade serous epithelial ovarian cancer. Importantly, Kindlin-2 inhibited serous epithelial ovarian cancer cell peritoneal dissemination in a mouse model. For clinical relevance, low Kindlin-2 expression correlated with higher tumor grade and older patients. Intriguingly, decreased Kindlin-2 expression predicts poor overall and progression-free survivals in serous epithelial ovarian cancer patients. Mechanistically, Kindlin-2 induced a mesenchymal to epithelial transition in serous epithelial ovarian cancer cells, at least in part, by up-regulation of estrogen receptor α which was recruited to the promoter of E-cadherin and thereby enhanced the transcription of E-cadherin. Collectively, we concluded that inadequate Kindlin-2 is an independent risk factor for serous epithelial ovarian cancer patients.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Animais , Caderinas/genética , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Movimento Celular , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Cistadenoma Seroso/genética , Cistadenoma Seroso/metabolismo , Cistadenoma Seroso/patologia , Progressão da Doença , Transição Epitelial-Mesenquimal , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Expressão Gênica , Xenoenxertos , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Prognóstico , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
16.
Reprod Sci ; 21(6): 696-703, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24336678

RESUMO

Hyperandrogenemia is the leading defect in patients with polycystic ovary syndrome (PCOS) and considered to be involved in the ovulation dysfunction of PCOS. During the process of ovulation, granulosa cells (GCs) undergo epithelial-mesenchymal transition (EMT), and integrin-interacting protein kindlin 2 is a well-known regulator in EMT. Therefore, our objective here was to compare the expression levels of kindlin 2 in luteinized GCs between patients with PCOS and control women and the relationship between kindlin 2 and PCOS pathogenesis. In this study, kindlin 2 expression was significantly increased in luteinized GCs from patients with PCOS, and kindlin 2 could be induced by testosterone both in vitro and in vivo. Meanwhile, kindlin 2 was positively correlated with androgen receptor (AR) in PCOS GCs. Taken together, kindlin 2 may play a role in luteinized GCs, especially in the case of excess androgen. Further studies are required to assess the specific role of kindlin 2 in follicular development and PCOS pathogenesis.


Assuntos
Células da Granulosa/metabolismo , Hiperandrogenismo/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Neoplasias/biossíntese , Síndrome do Ovário Policístico/metabolismo , Receptores Androgênicos/biossíntese , Adulto , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Humanos , Hiperandrogenismo/epidemiologia , Camundongos , Camundongos Endogâmicos ICR , Síndrome do Ovário Policístico/epidemiologia
17.
Zhonghua Bing Li Xue Za Zhi ; 42(8): 509-14, 2013 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-24246914

RESUMO

OBJECTIVE: To compare the efficiency of three different estrogen receptor (ER) immunostaining scoring systems by analyzing the correlation between ER status and clinicopathologic features for prediction of prognosis of patients with endometrial carcinoma (EC). METHODS: ER immunostaining (EnVision method) was performed in 160 type I EC and 39 type II EC paraffin samples and was scored by ASCO/CAP criterion, H-Score and Allred scoring system. Correlation between ER status and clinicopathologic features was statistically analyzed. RESULTS: ASCO/CAP criterion, H-Score and Allred (cutoff point: 4-8) scoring system showed high concordance in the following aspects. In EC patients, ER status was significantly associated with presurgical CA125 levels (P = 0.015, P = 0.007, P = 0.023), histologic grades (all P < 0.01) and PR status (all P < 0.01). In type I EC cohort, ER status was significantly correlated with PR status (P = 0.008, P < 0.01, P < 0.01) and p53 status (P = 0.042, P = 0.001, P < 0.01). As of the predictive value of ER status for type I EC patient age, ASCO/CAP (P = 0.027) and H-Score criteria (P = 0.035) were both superior to Allred score system (P = 0.064). Among well-known predictive clinicopathologic parameters, including FIGO stage, lympho-vascular involvement, lymph node metastasis, depth of myometrial invasion and omental involvement, ASCO/CAP scoring offered a better correlation (P = 0.005, P = 0.002, P = 0.021, P = 0.067, and P = 0.067, respectively) than H-Score (P > 0.05) and Allred scoring system (P > 0.05). CONCLUSIONS: Compared with H-Score and Allred scoring system, ASCO/CAP criterion is more closely correlated with predictive clinicopathologic parameters. Therefore it may be used as a simple, highly efficient prognostic indicator for EC patients in routine practice.


Assuntos
Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Imuno-Histoquímica/métodos , Receptores de Estrogênio/metabolismo , Antígeno Ca-125/metabolismo , Neoplasias do Endométrio/classificação , Feminino , Humanos , Metástase Linfática , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Gradação de Tumores , Invasividade Neoplásica , Estadiamento de Neoplasias , Receptores de Progesterona/metabolismo , Proteína Supressora de Tumor p53/metabolismo
18.
Gynecol Oncol ; 131(3): 764-71, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23994105

RESUMO

OBJECTIVE: Protein 4.1N (4.1N) is a member of the Protein 4.1 family that is involved in cellular processes such as cell adhesion, migration and signaling. In this study, we evaluated the expression of 4.1N protein and its potential roles in epithelial ovarian cancer (EOC) tumorigenesis and progression. METHODS: 4.1N protein expression was investigated in a total of 280 samples including 74 normal tissues, 35 benign, 30 borderline and 141 malignant epithelial ovarian tumors by immunohistochemistry. Correlation between 4.1N expression levels and clinicopathologic features was statistically analyzed. The expression of 4.1N in EOC cell lines was examined by western blotting. RESULTS: Immunohistochemistry analysis revealed that, although there was no loss of 4.1N expression in normal tissues and benign tumors, absence of Protein 4.1N was significantly more common in EOCs (44.0%) than in borderline tumors (3.3%) (p<0.001). Furthermore, loss or decreased expression of 4.1N protein expression was correlated with malignant potential of the tumors (14.3% in benign tumors, 56.7% in borderline tumors and 92.9% in malignancy) (p<0.001). In EOC samples, loss of 4.1N protein was significantly associated with advanced-stage (p=0.004), ascites (p=0.009), omental metastasis (p=0.018), suboptimal debulking (p=0.024), poorly histological differentiation (p=0.009), high-grade serous carcinoma (p=0.001), short progression-free-survival (p=0.018) and poor chemosensitivity to first-line chemotherapy (p=0.029). Moreover, western blotting analysis revealed that expression of 4.1N protein was lost in 4/8 (50%) EOC cell lines. CONCLUSIONS: 4.1N protein expression level was significantly decreased during malignant transformation of epithelial ovarian tumors and that loss of 4.1N expression was closely correlated to poorly differentiated and biologically aggressive EOCs.


Assuntos
Proteínas do Citoesqueleto/biossíntese , Proteínas de Membrana/biossíntese , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neuropeptídeos/biossíntese , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/deficiência , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Imuno-Histoquímica , Proteínas de Membrana/deficiência , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neuropeptídeos/deficiência , Neoplasias Ovarianas/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA