Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
J Pathol ; 261(1): 105-119, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37550813

RESUMEN

Granulomatous slack skin (GSS) is an extremely rare subtype of cutaneous T-cell lymphoma accompanied by an abundant number of macrophages and is clinically characterized by the development of pendulous skin folds. However, the characteristics of these macrophages in GSS remain unclear. Here, we conducted a spatial transcriptomic study on one frozen GSS sample and drew transcriptomic maps of GSS for the first time. Gene expression analysis revealed the enrichment of three clusters with macrophage transcripts, each exhibiting distinct characteristics suggesting that their primary composition consists of different subpopulations of macrophages. The CD163+ /CD206+ cluster showed a tumor-associated macrophage (TAM) M2-like phenotype and highly expressed ZFP36, CCL2, TNFAIP6, and KLF2, which are known to be involved in T-cell interaction and tumor progression. The APOC1+ /APOE+ cluster presented a non-M1 or -M2 phenotype and may be related to lipid metabolism. The CD11c+ /LYZ+ cluster exhibited an M1-like phenotype. Notably, these cells strongly expressed MMP9, MMP12, CHI3L1, CHIT1, COL1A1, TIMP1, and SPP1, which are responsible for extracellular matrix (ECM) degradation and tissue remodeling. This may partially explain the symptoms of cutaneous relaxation in GSS. Further immunohistochemistry on four GSS cases demonstrated that CD11c predominantly marked granulomas and multinucleated giant cells, whereas CD163 was mainly expressed on scattered macrophages, appearing as a mutually exclusive pattern. The expression pattern of MMP9 overlapped with that of CD11c, implying that CD11c+ macrophages may be a source of MMP9. Our data shed light on the characteristics of macrophages in the GSS microenvironment and provide a theoretical basis for the application of MMP9 inhibitors to prevent cutaneous relaxation of GSS. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Linfoma Cutáneo de Células T , Neoplasias Cutáneas , Humanos , Metaloproteinasa 9 de la Matriz , Neoplasias Cutáneas/genética , Microambiente Tumoral , Transcriptoma , Linfoma Cutáneo de Células T/complicaciones , Linfoma Cutáneo de Células T/diagnóstico , Linfoma Cutáneo de Células T/patología , Macrófagos/patología , Perfilación de la Expresión Génica
2.
Adv Sci (Weinh) ; 10(24): e2302613, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37345586

RESUMEN

Liver metastasis is the main cause of death in patients with colorectal cancer (CRC); thus, necessitating effective biomarkers and therapeutic targets for colorectal cancer liver metastasis (CRLM). Fibroblast growth factor 19 (FGF19) is a protumorigenic gene in numerous human malignancies. In this study, it is shown that FGF19 plays an indispensable role in CRLM. FGF19 expression and secretion are markedly correlated with liver metastasis and lower overall survival rates of patients with CRC. An in vivo metastasis model shows that FGF19 overexpression confers stronger liver-metastatic potential in CRC cells. Mechanistically, FGF19 exerts an immunomodulatory function that creates an environment conducive for metastasis in CRLM. FGF19 mediates the polarization of hepatic stellate cells to inflammatory cancer-associated fibroblasts (iCAFs) by activating the autocrine effect of IL-1α via the FGFR4-JAK2-STAT3 pathway. FGF19-induced iCAFs promote neutrophil infiltration and mediate neutrophil extracellular trap (NET) formation in liver metastatic niches via the production of complement C5a and IL-1ß, which in turn accelerates the liver colonization of CRC cells. Importantly, targeting FGF19 signaling with fisogatinib efficiently suppresses FGF19-induced liver metastasis in a mouse model. In summary, this study describes the mechanism by which FGF19 regulates CRLM, thereby providing a novel target for CRLM intervention.


Asunto(s)
Neoplasias Colorrectales , Trampas Extracelulares , Neoplasias Hepáticas , Animales , Ratones , Humanos , Trampas Extracelulares/metabolismo , Línea Celular Tumoral , Neoplasias Hepáticas/metabolismo , Transducción de Señal , Neoplasias Colorrectales/genética , Factores de Crecimiento de Fibroblastos/metabolismo
3.
Front Immunol ; 12: 779325, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925357

RESUMEN

Background: Macrophage extracellular traps (METs) and tumor-infiltrating macrophages contribute to the progression of several diseases. But the role of METs and tumor-infiltrating macrophages in colon cancer (CC) has not been illuminated. In this study, we aimed to clarify the prognostic value of METs for CC patients and to explore the interaction between CC cells and METs in vitro and in vivo. Methods: A training cohort consisting of 116 patients and a validation cohort of 94 patients were enrolled in this study. Immunofluorescence (IF) staining was conducted to determine METs formation in CC patients. Cox regression was used to perform prognostic analysis and screen out the best prognostic model. A nomogram was established to predict 5-year overall survival (OS). The correlation between METs with clinicopathological features and inflammatory markers was analyzed. The formation of METs in vitro was detected by SYTOX® green and IF staining, and the effect of METs on CC cells was detected by transwell assays. PAD2-IN-1, a selective inhibitor of peptidylarginine deiminase 2 (PAD2), was introduced to destroy the crosstalk between CC cells and METs in vitro and in vivo. Results: METs levels were higher in CC tissues and were an independent prognostic factor for CC patients. The prognostic model consisting of age, tumors local invasion, lymph node metastasis and METs were confirmed to be consistent and accurate for predicting the 5-year OS of CC patients. Besides, METs were correlated with distant metastasis and inflammation. Through in vitro experiments, we confirmed that there was a positive feedback loop between CC cells and METs, in that METs promoted the invasion of CC cells and CC cells enhanced the production of METs, in turn. This interaction could be blocked by PAD2-IN-1 inhibitors. More importantly, animal experiments revealed that PAD2-IN-1 inhibited METs formation and CC liver metastasis in vivo. Conclusions: METs were the potential biomarker of CC patient prognosis. PAD2-IN-1 inhibited the crosstalk between CC cells and METs in vitro and in vivo, which should be emphasized in CC therapy.


Asunto(s)
Comunicación Celular , Neoplasias del Colon/patología , Trampas Extracelulares/fisiología , Macrófagos/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/mortalidad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico , Modelos de Riesgos Proporcionales , Desiminasas de la Arginina Proteica/antagonistas & inhibidores
4.
BMC Cancer ; 21(1): 747, 2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34187415

RESUMEN

BACKGROUND: The molecular pathogenesis of endometrial cancer is not completely understood. CypB upregulated in many cancers, however, its role in endometrial carcinoma has not been studied. Here, we determine the effect of CypB on the growth of endometrial cancer. METHODS: In this study, we examined the expression of CypB in endometrial cancer tissues using immunohistochemistry. CypB silenced in HEC-1-B cell line by shRNA. CCK-8, colony formation assays, wound healing assays, and transwell analysis were performed to assess its effect on tumor cell proliferation and metastasis. Furthermore, microarray analysis was carried out to compare the global mRNA expression profile between the HEC-1-B and CypB-silenced HEC-1-B cells. Gene ontology and KEGG pathway enrichment analysis were performed to determine the potential function of differentially expressed genes related to CypB. RESULTS: We found that CypB was upregulated in endometrial cancer, inhibit CypB expression could significantly suppress cell proliferation, metastasis, and migration. We identified 1536 differentially expressed genes related to CypB (onefold change, p < 0.05), among which 652 genes were upregulated and 884 genes were downregulated. The genes with significant difference in top were mainly enriched in the cell cycle, glycosphingolipid biosynthesis, adherens junctions, and metabolism pathways. CONCLUSION: The results of our study suggest that CypB may serve as a novel regulator of endometrial cell proliferation and metastasis, thus representing a novel target for gene-targeted endometrial therapy. TRIAL REGISTRATION: YLYLLS [2018] 008. Registered 27 November 2017.


Asunto(s)
Neoplasias Endometriales/genética , Proliferación Celular , Ciclofilinas/genética , Femenino , Humanos , Metástasis de la Neoplasia
5.
J Pathol Clin Res ; 7(2): 154-164, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33416213

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) has previously been reported to be upregulated in many malignant tumors. This study investigated the significance of PRMT5 in endometrial carcinoma (EC) and explored its function in tumorigenesis. Immunohistochemistry was performed to evaluate PRMT5 expression in 62 EC and 66 endometrial hyperplasia samples. The functions of PRMT5 were investigated by cell counting kit-8, plate colony formation, wound healing, and transwell and flow cytometry assays. Quantitative reverse transcription-polymerase chain reaction and western blotting were used to measure the expression of PRMT5, changes in estrogen receptor α (ERα), and related functional proteins. Coimmunoprecipitation was performed to examine the interaction of PRMT5 with ERα and its coactivator steroid receptor coactivator-1 (SRC1). Compared to endometrial hyperplasia tissue, PRMT5 was overexpressed in endometrioid adenocarcinoma (EAC) but not overexpressed in mucinous EC. The main expression pattern of PRMT5 in EAC was cytoplasmic. However, the positive cases of endometrial hyperplasia showed both cytoplasmic and nuclear positivity in the endometrial glands or were mainly positive in stromal cells. Knockdown of PRMT5 significantly inhibited the growth and migration ability of EAC cells and promoted their apoptosis by regulating cyclin D1, c-myc, p53, and Bcl2 proteins. Furthermore, PRMT5 could form a complex with ERα and SRC1 to promote the expression of ERα. In conclusion, PRMT5 plays a significant role in the progression of EAC by interacting with ERα and impacting the cell cycle signaling pathways.


Asunto(s)
Carcinoma Endometrioide/genética , Neoplasias Endometriales/genética , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína-Arginina N-Metiltransferasas/metabolismo , Transducción de Señal , Adulto , Anciano , Apoptosis , Carcinogénesis , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patología , Ciclo Celular , Línea Celular Tumoral , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Proteína-Arginina N-Metiltransferasas/genética , Regulación hacia Arriba
6.
Diagn Pathol ; 15(1): 104, 2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32843061

RESUMEN

PURPOSE: Squamous cell carcinomas and adenocarcinomas are the most common types of cervical cancer. Compared to squamous cell carcinomas, adenocarcinomas are more common in younger women and have a poorer prognosis. Yet, so far, no useful biomarkers have been developed for these two types of cancer. In the following study, we examined the combination of cytokeratin 5/6, p63, p40 and MUC5AC for distinguishing squamous cell carcinoma (SCC) from adenocarcinoma of the cervix (AEC). MATERIALS AND METHODS: A total of 101 SCC and 108 AEC were collected. Immunohistochemical analyses were conducted to determine the expression of CK5/6, p63, p40, CK7 and MUC5AC. One pathologist who was blinded to the patient's clinical and pathological data interpreted the staining results. RESULTS: MUC5AC and CK7 were detected in 81.48 and 82.41% of AEC cases compared to 9.9 and 49.50% of SCC cases (P < 0.05); the specificity of MUC5AC was higher than that of CK7 in AEC (P < 0.05). The sensitivity of MUC5AC combined with p40 or p63 was similar to that of CK7, but the specificity was slightly higher than that of CK7 in AEC. Moreover, the expression of MUC5AC was correlated with the degree of tumor differentiation in adenocarcinomas (P = 0.036) and was not related to the prognosis of cervical adenocarcinoma and subtypes. CONCLUSIONS: MUC5AC may be useful as a biomarker for differential diagnoses between squamous carcinoma and adenocarcinoma of the cervix.


Asunto(s)
Adenocarcinoma/diagnóstico , Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/diagnóstico , Neoplasias del Cuello Uterino/diagnóstico , Adulto , Anciano , Femenino , Humanos , Queratina-5/análisis , Queratina-6/análisis , Proteínas de la Membrana/análisis , Persona de Mediana Edad , Mucina 5AC/análisis
7.
Oncol Lett ; 19(5): 3602-3608, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32269635

RESUMEN

Homeobox D 10 (HOXD10) is important in cell differentiation and morphogenesis and serves as a tumor suppressor gene (TSG) in a number of malignancies. The present study investigated its promoter methylation status and association with the clinicopathological features of endometrial cancer (EC), and measured HOXD10 protein expression levels. EC samples (n=62), including 50 endometroid adenocarcinoma (EA) and 12 mucinous endometrial carcinoma samples (EC) and 70 non-cancerous samples were collected. All samples were evaluated for the methylation status of several TSGs, including HOXD10, using methylation-specific PCR. HOXD10 expression level was evaluated using immunohistochemistry. 5-Aza-2-deoxycytidine treatment was performed in the EC cell line Ishikawa to observe the change in HOXD10 expression levels. HOXD10 promoter methylation was more frequent in cancer samples (P<0.001). Downregulation of HOXD10 in EC samples was confirmed at the protein level using immunohistochemistry (P<0.001) and immunohistochemical staining was negatively associated with methylation status (P<0.05). Less HOXD10 protein was expressed in MEC compared with EA samples (P<0.001). The HOXD10 promoter was hypermethylated in both EA and MEC, causing decreased HOXD10 protein expression levels in EC cells. HOXD10 expression levels were partially reversed by 5-Aza-2-deoxycytidine treatment. The results of the present study demonstrated that epigenetic silencing of HOXD10 putatively contributed to the tumorigenesis of EA. Although there was no significant difference in HOXD10 methylation between EA and MEC, HOXD10 protein expression levels differed between these two diseases, indicating that it may be a useful protein biomarker for distinguishing between these two lesions.

8.
Transl Cancer Res ; 9(5): 3499-3506, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-35117715

RESUMEN

BACKGROUND: The occurrence and development of gastric cancer is a multi-factor, multi-stage, multi-gene abnormal accumulation process. Both genetic and epigenetic mechanisms play an important role in the molecular mechanism of gastric cancer. DNA methylation is one of the most studied epigenetic expression mechanisms. To study the correlation between gene promoter methylation status and protein expression of vascular endothelial growth factor receptor 3 (VEGFR3), as well as their association with clinicopathological features in early gastric cancer (EGC) cases. METHODS: Immunohistochemical analysis and methylation-specific PCR (MSP) were used to detect the expression of VEGFR3 protein and methylation status of the VEGFR3 promoter in 50 cases of EGC and their paired normal gastric mucosa tissues. The level of DNA methylation of the VEGFR3 promoter, in situ VEGFR3 protein expression, and the clinicopathological characteristics of EGC patients were statistically analyzed. RESULTS: The positive rate of VEGFR3 protein expression in EGC tumor tissue (60%) was significantly higher than that in the normal gastric mucosa (10%). The detectable methylation frequency of VEGFR3 promoter in EGC tumor tissue (48%) was significantly lower than that in the normal gastric mucosa (85%). As anticipated, the methylation level of the VEGFR3 gene promoter was negatively associated with the overexpression of VEGFR3 protein. In addition, methylation status of the VEGFR3 gene promoter was positively correlated with lymph node metastasis in EGC patients (P<0.05), but was not linked to patients' gender, age, tumor size, degree of differentiation, or tumor invasion depth (P>0.05). CONCLUSIONS: Hypomethylation of the VEGFR3 gene promoter is one of the major mechanisms underlying VEGFR3 gene overexpression in EGC tumor tissues and is related to lymph node metastasis in EGC patients. DNA methylation of VEGFR3 is expected to become a molecular diagnostic and prognostic biomarker for EGC.

9.
Transl Cancer Res ; 9(10): 6624, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35129553

RESUMEN

[This corrects the article DOI: 10.21037/tcr.2020.03.74.].

11.
Int Immunopharmacol ; 77: 105928, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31711940

RESUMEN

Osteoarthritis (OA), a common and severe disease, is predominantly characterized by cartilage destruction, which results in the degeneration of joint surfaces. Nowadays, it is accepted that TNFα plays a critical role in OA. Scutellarin, the main bioactive flavonoid glycoside extracted form Erigeron breviscapus, has been reported to exert positive effects on anti-inflammatory reactions. However, the effect of scutellarin in OA is still unknown. In this study, we isolated and cultured primary murine chondrocytes, stimulating TNF-α, in the presence or absence of scutellarin treatment. We found that the inflammatory response stimulated by TNF-α was significantly inhibited by the addition of scutellarin. Moreover, we established OA mouse models induced by surgery. In this mouse model, both inflammatory reaction and cartilage degeneration were markedly inhibited by oral administration of scutellarin. Furthermore, the cellular mechanism underlying the protective effect of scutellarin in OA was clearly associated with the NF-κB and PI3K/AKT signaling pathways. Collectively, this study proposes scutellarin as a potential therapeutic to treat joint degenerative diseases, including OA.


Asunto(s)
Antiinflamatorios/uso terapéutico , Apigenina/uso terapéutico , Cartílago/efectos de los fármacos , Glucuronatos/uso terapéutico , FN-kappa B/metabolismo , Osteoartritis/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Antiinflamatorios/farmacología , Apigenina/farmacología , Cartílago/metabolismo , Cartílago/patología , Glucuronatos/farmacología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Articulación de la Rodilla/efectos de los fármacos , Articulación de la Rodilla/metabolismo , Articulación de la Rodilla/patología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/genética , Osteoartritis/metabolismo , Osteoartritis/patología , Transducción de Señal/efectos de los fármacos
12.
J Cell Mol Med ; 23(11): 7331-7341, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31507089

RESUMEN

Persistently activated IL-6/STAT3 pathway promotes acquired resistance to targeted therapy with epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) in non-small-cell lung cancer (NSCLC) treatment. miR-206 has been verified to be dysregulated and plays as a negative regulator in lung cancer. However, whether miR-206 may overcome IL6-induced gefitinib resistance in EGFR-mutant lung cancer remains elusive. In this study, we investigated the role of miR-206 in IL6-induced gefitinib-resistant EGFR-mutated lung cancer cell lines. We showed that forced miR-206 expression restored gefitinib sensitivity in IL6-induced gefitinib-resistant EGFR-mutant lung cancer cells by inhibiting IL6/JAK1/STAT3 pathway. Specifically, mechanistic investigations revealed that miR-206 blocked IL-6/STAT3 signalling via directly targeting the 3'-UTR of intracellular IL-6 messenger RNA. Moreover, IL-6 induced miR-206 down-regulation by reducing the cropping process of primary miR-206 (pri-miR-206) into the Drosha/DGCR8 complex. Taken together, our findings reveal a direct role of miR-206 in regulating IL-6/STAT3 pathway and contrarily activated IL-6/STAT3 signalling mediates the miR-206 maturation process in gefitinib-resistant EGFR-mutant lung cancer cells.


Asunto(s)
Resistencia a Antineoplásicos/genética , Interleucina-6/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Regiones no Traducidas 3'/genética , Anciano , Línea Celular Tumoral , Regulación hacia Abajo/genética , Receptores ErbB/genética , Femenino , Gefitinib/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Persona de Mediana Edad
13.
Oncol Lett ; 18(3): 2845-2852, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31452763

RESUMEN

Cyclin-dependent kinase subunit (CKS) 2 is a member of the CKS family, which plays an important role in the regulation of meiosis and mitosis. Overexpression of CKS2 has been reported in several types of tumors. However, few studies have investigated its role in uterine leiomyosarcoma (ULMS). In the present study, the expression of CKS2 in 38 cases of ULMS and 38 cases of uterine leiomyoma (ULM) was analyzed by immunohistochemistry. Moreover, the functional analysis of CKS2 was performed in ULMS cell lines. A significantly higher expression of CKS2 was found in ULMS tissues than in ULM tissues (P<0.01) and high CKS2 expression was associated with increased tumor size, low progesterone receptor expression and poor prognosis in patients with ULMS. Multivariate Cox regression analysis revealed that CKS2 expression status was an independent predictor of overall survival for ULMS. Furthermore, silencing of CKS2 in ULMS cells inhibited cell proliferation, colony formation, migration and invasion, and resulted in cell cycle arrest. In conclusion, the present study demonstrated that CKS2 may serve as a marker for the differential diagnosis of ULMS and ULM. In addition, it may act as an independent prognostic factor in patients with ULMS, and serve as a novel target for ULMS therapy.

14.
Apoptosis ; 24(11-12): 837-848, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31352641

RESUMEN

Renal cell carcinoma (RCC) is the third most common urological cancer with highly metastatic potential. MAGI1 plays an important role in stabilization of the adherens junctions and has been confirmed to suppress invasiveness and metastasis in multiple cancers in clinic. However, its expression and anti-metastatic ability in RCC are still unclear. In this study, we demonstrated that MAGI1 was markedly decreased in the RCC and indicated poor survival. Furthermore, we found that MAGI1 suppressed the invasion and migration of human RCC cells. Mechanistic investigations revealed that MAGI1 stabilized the PTEN/MAGI1/ß-catenin complex to inhibit ß-catenin signaling pathway. Moreover, MAGI1 was targeted by miR-520h which was transcriptionally activated by c-Myb. Collectively, our findings suggested that MAGI1mediated tumor metastasis through c-Myb/miR-520h/MAGI1 signaling pathway in RCC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Células Renales/metabolismo , Moléculas de Adhesión Celular/metabolismo , Guanilato-Quinasas/metabolismo , Neoplasias Renales/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/secundario , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Guanilato-Quinasas/genética , Humanos , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Masculino , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica/genética , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myb/genética , Transducción de Señal/genética , beta Catenina/genética , beta Catenina/metabolismo
15.
J BUON ; 23(1): 134-142, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29552773

RESUMEN

PURPOSE: To investigate the differential gene expression pattern between invasive multifocal/multicentric (MMBC) and unifocal breast cancer (UFBC) with cDNA array and to discover the potential outlier genes associated with the incidence of MMBC and also to provide a guidance for clinical treatment and prognosis prediction. METHODS: This retrospective study analyzed the gene expression pattern alteration in breast cancer. We collected 156 MMBC (136 cases with 2 foci, 20 cases with 3 foci) and 130 UFBC samples from patients hospitalized in Yuhuangding Hospital, Yantai, from January 2005 to December 2015. The outlier genes were screened by cDNA expression microarray and validated by RT-PCR. RESULTS: 18 overexpressed and 22 underexpressed genes were identified in the differential analysis, including family genes ABCC11, ABCB5 and PRODH, PROL1. Noteworthily, ABCC11 was significantly upregulated, while ABCB3 was downregulated, which were confirmed by RT-PCR results. CONCLUSION: The differential expression pattern of ABCC11 and ABCB5 genes may serve as outliers, potentially associated with incidence of MMBS.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Transportadoras de Casetes de Unión a ATP , Neoplasias de la Mama , Invasividad Neoplásica , Estadificación de Neoplasias , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica/genética , Pronóstico , Estudios Retrospectivos
16.
Pathol Res Pract ; 214(2): 233-239, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29274810

RESUMEN

OBJECTIVE: Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified oncoprotein that stabilizes c-Myc and promotes cell proliferation and transformation. Here, we investigated the clinical significance and biological function of CIP2A in endometrial cancer. METHOD: CIP2A expression was assessed in normal endometrium, endometrial hyperplasia, endometrial atypical hyperplasia, and endometrioid adenocarcinoma tissues using immunohistochemistry, western blot, and RT-PCR. The effect of reduced CIP2A expression was assessed by siRNA knockdown in Ishikawa and An3ca endometrial cell lines. The roles of CIP2A in proliferation, apoptosis, and the cell cycle were assessed using CCK-8 assays, colony formation assays, and flow cytometry, respectively. RESULTS: Our results show that CIP2A expression was higher in endometrioid adenocarcinoma tissues and cell lines. Furthermore, CIP2A siRNA significantly reduced the proliferation rate and invasion of Ishikawa and An3ca cells, and induced a significant level of apoptosis in Ishikawa cells. Moreover, CIP2A depletion resulted in reduced c-Myc and cyclin D1 protein levels, and increased caspase-3 expression. CONCLUSIONS: CIP2A is overexpressed in endometrioid adenocarcinoma and CIP2A promotes the malignant growth and invasion,decrease apoptosis in entometrioid adenocarcinoma cell lines. These results validate that CIP2A plays an important role in the carcinogenesis of endometrioid adenocarcinoma and establishes CIP2A as a clinically relevant oncoprotein and may presents a promising therapeutic target for cancer treatment.


Asunto(s)
Adenocarcinoma de Células Claras/metabolismo , Autoantígenos/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Endometriales/patología , Proteínas de la Membrana/metabolismo , Adenocarcinoma de Células Claras/patología , Adulto , Anciano , Apoptosis/fisiología , Ciclo Celular/fisiología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Persona de Mediana Edad , Clasificación del Tumor
17.
Mol Med Rep ; 14(3): 2739-45, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27431680

RESUMEN

Aberrant epigenetic modification is associated with the development and progression of cancer. Hypermethylation of tumor suppressor gene promoters and cooperative histone modification have been considered to be the primary mechanisms of epigenetic modification. Ovary granulosa cell tumors (GCTs) are relatively rare, accounting for ~3% of all ovarian malignancies. The present study assessed hypermethylation of the cadherin 13 (CDH13), dickkopf WNT signaling pathway inhibitor 3 (DKK3) and forkhead box L2 (FOXL2) promoters in 30 GCT tissues and 30 healthy control tissues using methylation-specific polymerase chain reaction analysis. The data showed that the frequencies of CDH13, DKK3 and FOXL2 promoter methylation were significantly higher in the GCT tissues, compared with the healthy control tissues (86.67, vs. 23.33%; 80, vs. 26.67% and 66.67, vs. 20%, respectively; P<0.001). Immunostaining of enhancer of zeste homolog 2 (EZH2), a histone H3K27 methyltransferase, showed that the EZH2 protein was expressed in 11 of the 30 GCT tissue samples, whereas no EZH2 protein was expressed in the 30 healthy control tissues (P<0.01). These data suggested that hypermethylation of the CDH13, DKK3 and FOXL2 gene promoters, and overexpression of the EZH2 protein were involved in the development of GCT.


Asunto(s)
Cadherinas/genética , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2/genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Tumor de Células de la Granulosa/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Regiones Promotoras Genéticas , Proteínas Adaptadoras Transductoras de Señales , Adulto , Biomarcadores , Cadherinas/metabolismo , Quimiocinas , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/metabolismo , Predisposición Genética a la Enfermedad , Tumor de Células de la Granulosa/diagnóstico , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Recurrencia , Carga Tumoral , Adulto Joven
18.
J BUON ; 21(1): 35-41, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27061528

RESUMEN

PURPOSE: In this study, we examined the expression of human mammaglobin (hMAM) mRNA and the protein levels in patients with breast cancer and their relationship with prognostic clinicopathological parameters. METHODS: hMAM mRNA expression in leucocytes from peripheral blood samples from patients diagnosed with primary invasive breast cancer (IC), carcinoma in situ (CIS), or benign breast diseases was analyzed using RT-PCR. The hMAM protein levels and expression patterns in tissue from 3 patient groups were evaluated by immunohistochemical staining, and several non-breast neoplasms were selected as negative controls, undergoing the same examination. RESULTS: The expression of hMAM mRNA was significantly higher in patients with IC or CIS compared to those with benign tumors (both<0.01). Immunohistochemical staining revealed similar results, where patients with IC or CIS had higher levels of hMAM protein (p<0.01 and p<0.01, respectively), while none of the negative controls expressed hMAM. Further analyses showed a strong correlation between hMAM protein/mRNA expression and clinicopathological factors, such as histological grade, clinical stage, and lymph node status, in patients with IC. CONCLUSION: The hMAM mRNA and protein expression profiles validate the potential of hMAM as a specific marker for breast cancer diagnosis and target treatment delivery.


Asunto(s)
Neoplasias de la Mama/química , Mamoglobina A/genética , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/patología , Carcinoma in Situ/química , Femenino , Humanos , Inmunohistoquímica , Mamoglobina A/análisis , Pronóstico , ARN Mensajero/análisis
19.
Pathol Res Pract ; 212(6): 555-9, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27067809

RESUMEN

Scavenger receptor class B type 1 (SR-B1) is an integral membrane protein that is expressed in numerous cells and tissue types. The primary role of SR-B1 is to facilitate uptake of cholesteryl esters from high-density lipoproteins (HDL) in the liver. Altered SR-B1 expression contributes to human diseases. This study assessed association of SR-B1 expression in breast tissue specimens with breast cancer development and prognosis. Tissue specimens from 30 cases of adjacent normal breast tissues, ductal carcinoma in situ (DCIS) and invasive ductal breast cancer (IDCA) were subjected to Western blot analysis, and 135 cases of DCIS and IDCA were used for quantitative immunohistochemical analysis of SR-B1 expression. The data showed that SR-B1 was significantly overexpressed in IDCA tissues compared to normal breast and DCIS tissues. SR-B1 expression was associated with pre-menopausal status, tumor size, and worse overall survival of patients. The data from this ex vivo study suggests that up-regulated SR-B1 protein expression is associated with malignant behaviors of breast cancer and that SR-B1 is an independent predictor for poor survival in breast cancer patients.


Asunto(s)
Neoplasias de la Mama/genética , Antígenos CD36/genética , Carcinoma Ductal de Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Regulación Neoplásica de la Expresión Génica , Regulación hacia Arriba , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/mortalidad , Carcinoma Intraductal no Infiltrante/patología , Femenino , Humanos , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia
20.
Gynecol Oncol ; 140(1): 145-51, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26597461

RESUMEN

OBJECTIVE: Epigenetic changes in cancer and precancerous lesions could be utilized as biomarkers for cancer early detection. This study aims to investigate the novel biomarkers in endometrial carcinoma, and explore their epigenetic regulation. METHODS: Methylation of six tumor suppressor genes (CDH13, SHP1, HIN1, DKK3, CTNNA1 and PCDH8) was evaluated in 155 endometrium samples. Changes of methylation and mRNA expression after treatment with 5-Aza-2'-deoxycytidine (5-Aza-CdR) or/and trichostatin A (TSA) were investigated by MSP and qRT-PCR respectively. Co-immunoprecipitation was used to detect the interactions between UHRF1 and PRMT5 proteins. RESULTS: CDH13 and SHP1 promoters were highly methylated (81.36% and 86.44%, respectively) in endometrial carcinoma, while CDH13 promoter methylation was also present in complex hyperplasia and atypical hyperplasia (51.72% and 50.00%, respectively). Methylation of CDH13 and SHP1 promoters was associated with age and tumor differentiation or muscular infiltration depth. CDH13 and SHP1 promoters were completely methylated in endometrial carcinoma cell lines and were partially reversed by 5-Aza-CdR or TSA to induce mRNA levels (P<0.01). After combined treatment with these two agents, methylation of CDH13 and SHP1 promoters was completely reversed and expression of their mRNA was significantly increased (P<0.01). Moreover, PRMT5 could bind to UHRF1 and down-regulated by 5-Aza-CdR and/or TSA treatment (P<0.05). CONCLUSIONS: Our data demonstrate for the first time that SHP1 methylation has high specificity for diagnosis of endometrial carcinoma, while CDH13 promoter methylation plays a role in the earlier stage. Furthermore, UHRF1 could form a complex with PRMT5 to contribute to the endometrial carcinogenesis.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Cadherinas/genética , Metilación de ADN , Neoplasias Endometriales/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína-Arginina N-Metiltransferasas/genética , Azacitidina/análogos & derivados , Azacitidina/farmacología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Cadherinas/biosíntesis , Cadherinas/metabolismo , Línea Celular Tumoral , Decitabina , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Genes Supresores de Tumor , Humanos , Ácidos Hidroxámicos/farmacología , Persona de Mediana Edad , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 6/biosíntesis , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ubiquitina-Proteína Ligasas , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...