Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
BMC Cancer ; 24(1): 567, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711020

RESUMEN

BACKGROUND: Pyroptosis is a type of programmed cell death mediated by the gasdermin family. Gasdermin B (GSDMB), as a member of gasdermin family, can promote the occurrence of cell pyroptosis. However, the correlations of the GSDMB expression in colorectal cancer with clinicopathological predictors, immune microenvironment, and prognosis are unclear. METHODS: Specimens from 267 colorectal cancer cases were analyzed by immunohistochemistry to determine GSDMB expression, CD3+, CD4+, and CD8+ T lymphocytes, CD20+ B lymphocytes, CD68+ macrophages, and S100A8+ immune cells. GSDMB expression in cancer cells was scored in the membrane, cytoplasm, and nucleus respectively. GSDMB+ immune cell density was calculated. Univariate and multivariate survival analyses were performed. The association of GSDMB expression with other clinicopathological variables and immune cells were also analyzed. Double immunofluorescence was used to identify the nature of GSDMB+ immune cells. Cytotoxicity assays and sensitivity assays were performed to detect the sensitivity of cells to 5-fluorouracil. RESULTS: Multivariate survival analysis showed that cytoplasmic GSDMB expression was an independent favorable prognostic indicator. Patients with positive cytoplasmic or nuclear GSDMB expression would benefit from 5-fluorouracil based chemotherapy. The assays in vitro showed that high GSDMB expression enhanced the sensitivity of colorectal cancer cells to 5-fluorouracil. Patients with positive membranous or nuclear GSDMB expression had more abundant S100A8+ immune cells in the tumor invasive front. Positive nuclear GSDMB expression indicated more CD68+ macrophages in the tumor microenvironment. Moreover, GSDMB+ immune cell density in the stroma was associated with a higher neutrophil percentage but a lower lymphocyte counts and monocyte percentage in peripheral blood. Furthermore, the results of double immunofluorescence showed that GSDMB co-expressed with CD68 or S100A8 in stroma cells. CONCLUSION: The GSDMB staining patterns are linked to its role in cancer progression, the immune microenvironment, systemic inflammatory response, chemotherapeutic efficacy, and prognosis. Colorectal cancer cells with high GSDMB expression are more sensitive to 5-fluorouracil. However, GSDMB expression in immune cells has different effects on cancer progression from that in cancer cells.


Asunto(s)
Neoplasias Colorrectales , Progresión de la Enfermedad , Gasderminas , Microambiente Tumoral , Humanos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/mortalidad , Masculino , Pronóstico , Femenino , Persona de Mediana Edad , Microambiente Tumoral/inmunología , Anciano , Biomarcadores de Tumor/metabolismo , Fluorouracilo/uso terapéutico , Fluorouracilo/farmacología , Proteínas de Neoplasias/metabolismo , Inmunohistoquímica , Adulto , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Piroptosis
2.
Phytomedicine ; 121: 155111, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37804819

RESUMEN

BACKGROUND: Current evidence indicates a rising global prevalence of Non-Alcoholic Fatty Liver Disease (NAFLD), which is closely associated to conditions such as obesity, dyslipidemia, insulin resistance, and metabolic syndrome. The relationship between the gut microbiome and metabolites in NAFLD is gaining attention understanding the pathogenesis and progression of dysregulated lipid metabolism and inflammation. The Xie Zhuo Tiao Zhi (XZTZ) decoction has been employed in clinical practice for alleviating hyperlipidemia and symptoms related to metabolic disorders. However, the pharmacological mechanisms underlying the effects of XZTZ remain to be elucidated. PURPOSE: The objective of this study was to examine the pharmacological mechanisms underlying the hypolipidemic and anti-inflammatory effects of XZTZ decoction in a mouse model of NAFLD, as well as the effects of supplementing exogenous metabolites on PO induced cell damage and lipid accumulation in cultured hepatocytes. METHODS: A high-fat diet (HFD) mouse model was established to examine the effects of XZTZ through oral gavage. The general condition of mice and the protective effect of XZTZ on liver injury were evaluated using histological and biochemical methods. Hematoxylin and eosin staining (H&E) staining and oil red O staining were performed to assess inflammatory and lipid accumulation detection, and cytokine levels were quantitatively analyzed. Additionally, the study included full-length 16S rRNA sequencing, liver transcriptome analysis, and non-targeted metabolomics analysis to investigate the relationship among intestinal microbiome, liver metabolic function, and XZTZ decoction. RESULTS: XZTZ had a significant impact on the microbial community structure in NAFLD mice. Notably, the abundance of Ileibacterium valens, which was significantly enriched by XZTZ, exhibited a negative correlation with liver injury biomarkers such as, alanine transaminase (ALT) and aspartate transaminase (AST) activity. Moreover, treatment with XZTZ led to a significant enrichment of the purine metabolism pathway in liver tissue metabolites, with inosine, a purine metabolite, showing a significant positive correlation with the abundance of I. valens. XZTZ and inosine also significantly enhanced fatty acid ß-oxidation, which led to a reduction in the expression of pro-inflammatory cytokines and the inhibition of liver pyroptosis. These effects contributed to the mitigation of liver injury and hepatocyte damage, both in vivo and vitro. Furthermore, the utilization of HPLC fingerprints and UPLC-Q-TOF-MS elucidated the principal constituents within the XZTZ decoction, including naringin, neohesperidin, atractylenolide III, 23-o-Acetylalisol B, pachymic acid, and ursolic acid which are likely responsible for its therapeutic efficacy. Further investigations are imperative to fully uncover and validate the pharmacodynamic mechanisms underlying these observations. CONCLUSION: The administration of XZTZ decoction demonstrates a protective effect on the livers of NAFLD mice by inhibiting lipid accumulation and reducing hepatocyte inflammatory damage. This protective effect is mediated by the upregulation of I.valens abundance in the intestine, highlighting the importance of the gut-liver axis. Furthermore, the presesnce of inosine, adenosine, and their derivatives are important in promoting the protective effects of XZTZ. Furthermore, the in vitro approaching, we provide hitherto undocumented evidence indicating that the inosine significantly improves lipid accumulation, inflammatory damage, and pyroptosis in AML12 cells incubated with free fatty acids.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Piroptosis , ARN Ribosómico 16S , Hígado , Metabolismo de los Lípidos , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos no Esterificados/metabolismo , Purinas/farmacología , Inosina/metabolismo , Inosina/farmacología , Inosina/uso terapéutico , Ratones Endogámicos C57BL
3.
Pharmaceuticals (Basel) ; 16(6)2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37375731

RESUMEN

The transcription factor nuclear factor-κB (NF-κB) plays a complicated role in multiple tumors. Mounting evidence demonstrates that NF-κB activation supports tumorigenesis and development by enhancing cell proliferation, invasion, and metastasis, preventing cell death, facilitating angiogenesis, regulating tumor immune microenvironment and metabolism, and inducing therapeutic resistance. Notably, NF-κB functions as a double-edged sword exerting positive or negative influences on cancers. In this review, we summarize and discuss recent research on the regulation of NF-κB in cancer cell deaths, therapy resistance, and NF-κB-based nano delivery systems.

4.
Front Public Health ; 10: 808873, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35570909

RESUMEN

Background: The outbreak of coronavirus disease 2019 (COVID-19) has endangered human health and life. This pandemic has changed people's lifestyle and affected the regular delivery of standard cancer treatment. In the present study, we aimed to explore the influencing factors of delayed treatment in patients with breast cancer during COVID-19 pandemic. Methods: This study was a cross-sectional investigation, and the subjects were patients who were discharged from the department of burn and plastic surgery after February 2020. All participants completed this study's online questionnaire based on the WeChat and Wenjuanxing platforms. Levels of anxiety and depression were measured by the Hospital Anxiety and Depression Scale (HADS). Patients were divided into a delay group and non-delay group according to the occurrence of delayed treatment. Univariate analysis was performed by using the t test or chi-square test. A logistic regression model was employed to determine factors associated with delayed treatment. Results: The present study included a total of 397 patients with breast cancer, among whom delayed treatment occurred in 76 patients, accounting for 19.1%. Scores on both the anxiety subscale and depression subscale in delay group were significantly higher than those in non-delay group. Compared with non-delay group, we found that patients in delay group usually had a higher level of education (P = 0.020), worse self-feeling (P = 0.030), poor compliance of medical order (P = 0.042), and a higher prevalence of anxiety (P = 0.004) and depression (P = 0.012). Traffic inconvenience was also an important relevant factor for delayed treatment (P = 0.001). The prevalence of recurrence in delay group was higher than that in non-delay group (P = 0.018). By using logistic multivariate regression analysis, the results revealed that level of education and traffic inconvenience were independent factors influencing delayed treatment in patients with breast cancer during COVID-19 pandemic. Conclusion: The prevalence of delayed treatment in patients with breast cancer during COVID-19 pandemic is relatively high. Our findings reveal several influencing factors closely associated with delayed treatment, which is useful information that will be beneficial for patients to receive standardized therapy by taking targeted measures.


Asunto(s)
Neoplasias de la Mama , COVID-19 , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/terapia , COVID-19/epidemiología , Estudios Transversales , Femenino , Humanos , Pandemias , SARS-CoV-2 , Tiempo de Tratamiento
5.
J Inflamm Res ; 14: 6223-6235, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858044

RESUMEN

BACKGROUND: Pyroptosis is a type of cell death that causes an immune reaction. Gasdermin D (GSDMD), as an executor of pyroptosis, has become an attractive target in cancer research. However, the clinical significance of GSDMD expression in different subcellular locations remains unclear. METHODS: GSDMD was detected by immunohistochemistry in 178 cases of colorectal cancer with follow-up information. General data and information on systemic inflammatory indicators were collected from case records, and the clinicopathological parameters were reviewed by microscopy. CD3+, CD4+, and CD8+ T lymphocytes, CD20+ B lymphocytes, and CD68+ macrophages were detected by immunohistochemistry. Univariate survival analysis (Kaplan-Meier method, Log rank test) and a multivariate Cox proportional hazard model were used to analyze the impact of GSDMD on overall survival. RESULTS: Survival analysis showed that high expression of cytoplasmic GSDMD was an independent favorable indicator for prognosis (P=0.027) and improved the efficacy of chemotherapy (P=0.012). Positive cytoplasmic GSDMD expression indicated lower probability of distant metastasis (P=0.024), yet nuclear GSDMD expression predicted deeper infiltration depth (P=0.007). Membranous GSDMD expression positively correlated with CD68+ macrophages in tumor center (P=0.002) and CD8+ lymphocytes in tumor invasive front (P=0.007). However, nuclear GSDMD was negatively related to CD68+ macrophages in tumor invasive front (P<0.001) and CD8+ lymphocytes in tumor center (P=0.069). Cytoplasmic GSDMD was associated with more CD3+ lymphocytes both in tumor center (P=0.066) and tumor invasive front (P=0.008). Moreover, positive membranous GSDMD indicated a lower neutrophil-to-lymphocyte ratio (P=0.013). CONCLUSION: GSDMD subcellular localization patterns are related to CRC progression and immune reaction, and should be investigated in future studies.

6.
Cancer Metab ; 9(1): 23, 2021 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980323

RESUMEN

BACKGROUND: Cancer cell is generally characterized by enhanced glycolysis. Inflammasome activation is interaction with glycolysis. The concentration of lipopolysaccharide (LPS), a classic inflammasome activator, is significantly higher in colorectal cancer tissue than in normal intestinal mucosa. However, the mechanism of LPS on glycolysis and metastasis has not been fully elucidated. This study aimed to investigate the roles of LPS on inflammasome activation, glycolysis, and metastasis, and unravel metformin's potential in treatment of CRC. METHODS: We detected inflammasome activation and cell motility following LPS exposure in CRC cell lines. Glycolysis analysis was performed, and the key glycolytic rate-limiting enzymes were detected. Dual-luciferase reporter gene assay, co-immunoprecipitation, chromatin immunoprecipitation (ChIP) analysis, and ChIP-reChIP assay were performed to identify the specific mechanisms of LPS on glycolysis. Mouse metastasis models were used to determine the effects of LPS and metformin on metastasis. Correlation analysis of the expression of various molecules was performed in 635 CRC samples from The Cancer Genome Atlas and 83 CRC samples from our lab. RESULTS: LPS activates caspase-1 through NF-κB and upregulates the expression of Snail and HK3 depending on caspase-1 activation. LPS potentiates migration and invasion depending on accelerated glycolysis, which could be reversed by knockdown of glycolytic rate-limiting enzyme HK3. Nuclear Snail is upregulated by NF-κB under LPS treatment and then forms a complex with NF-κB, then directly binds to the HK3 promoter region to upregulate the expression of HK3. Metformin suppresses the NF-κB/Snail/HK3 signaling axis that is activated by LPS and then inhibits LPS-induced metastasis. In vivo, LPS-treated cells form more metastasis in the lungs of mice, and metformin completely reverses this effect of LPS. CONCLUSION: LPS activates inflammasomes in cancer cells through NF-κB and promotes metastasis through glycolysis enhanced by the NF-κB/Snail/HK3 signaling pathway in CRC. Metformin could prevent this effect of LPS.

7.
Microsc Res Tech ; 84(7): 1484-1497, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33470484

RESUMEN

The spruce bark beetle, Ips typographus, is an invasive forest pest that occurs across Eurasia. To further understand the mechanism of insect-host chemical communication and the feeding choices of adult I. typographus, we examined the sensilla on the antennae and mouthparts of I. typographus using scanning electron microscopy. No sexual dimorphism in sensilla type or distribution was observed. Ten subtypes of antennal sensilla were identified: sensilla trichodea Types I-III (TR1 , TR2 , and TR3 ); sensilla chaetica Types I and II (CH1 and CH2 ); sensilla basiconica Types I-III (BA1 , BA2 , and BA3 ); sensilla coeloconica (CO); and Böhm sensilla (BS). BA2 were the most abundant among the antennal sensilla in both sexes. Thirteen mouthpart sensilla subtypes were observed: sensilla trichodea Types I-IV (TR1 , TR2 , TR3 , and TR4 ); sensilla chaetica Types I-III (CH1 , CH2 , and CH3 ); sensilla digitiformia (DI); sensilla basiconica Types I-II (BA1 and BA2 ); and sensilla styloconica Types I-III (ST1 , ST2 , and ST3 ). Among these, TR4 were the most abundant in both sexes. The apex of each maxillary and labial palp carried the same sensilla (BA2 , ST1 , ST2 , and ST3 ), although the apex of the maxillary palp contained more total sensilla. The functional roles of each sensilla type are discussed based on their external structure and distribution. The results provide a theoretical basis for further studies on the olfactory and feeding behaviors and electrophysiology of adult I. typographus.


Asunto(s)
Escarabajos , Gorgojos , Animales , Antenas de Artrópodos , Femenino , Masculino , Microscopía Electrónica de Rastreo , Corteza de la Planta , Sensilos
8.
Cancer Commun (Lond) ; 41(2): 154-170, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33389821

RESUMEN

BACKGROUND: The transforming growth factor-ß (TGF-ß) pathway plays a pivotal role in inducing epithelial-mesenchymal transition (EMT), which is a key step in cancer invasion and metastasis. However, the regulatory mechanism of TGF-ß in inducing EMT in colorectal cancer (CRC) has not been fully elucidated. In previous studies, it was found that S100A8 may regulate EMT. This study aimed to clarify the role of S100A8 in TGF-ß-induced EMT and explore the underlying mechanism in CRC. METHODS: S100A8 and upstream transcription factor 2 (USF2) expression was detected by immunohistochemistry in 412 CRC tissues. Kaplan-Meier survival analysis was performed. In vitro, Western blot, and migration and invasion assays were performed to investigate the effects of S100A8 and USF2 on TGF-ß-induced EMT. Mouse metastasis models were used to determine in vivo metastasis ability. Luciferase reporter and chromatin immunoprecipitation assay were used to explore the role of USF2 on S100A8 transcription. RESULTS: During TGF-ß-induced EMT in CRC cells, S100A8 and the transcription factor USF2 were upregulated. S100A8 promoted cell migration and invasion and EMT. USF2 transcriptionally regulated S100A8 expression by directly binding to its promoter region. Furthermore, TGF-ß enhanced the USF2/S100A8 signaling axis of CRC cells whereas extracellular S100A8 inhibited the USF2/S100A8 axis of CRC cells. S100A8 expression in tumor cells was associated with poor overall survival in CRC. USF2 expression was positively related to S100A8 expression in tumor cells but negatively related to S100A8-positive stromal cells. CONCLUSIONS: TGF-ß was found to promote EMT and metastasis through the USF2/S100A8 axis in CRC while extracellular S100A8 suppressed the USF2/S100A8 axis. USF2 was identified as an important switch on the intracellular and extracellular S100A8 feedback loop.


Asunto(s)
Neoplasias Colorrectales , Transición Epitelial-Mesenquimal , Animales , Calgranulina A/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Factor de Crecimiento Transformador beta/metabolismo , Factores Estimuladores hacia 5'
9.
Clin Colorectal Cancer ; 17(3): e579-e592, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29921496

RESUMEN

BACKGROUND: Tumor tissues consist of heterogeneous cancer cells and stroma cells, including cancer stem cells and immune cells. Epithelial-mesenchymal transition (EMT) programs closely associate with acquisition of stemness. We investigated for the first time the clinical significance of combining cancer stem cells, immune cells, and EMT traits. MATERIALS AND METHODS: In 419 colorectal carcinomas, stem-cell markers (Nanog, Lgr5, CD44v6, ALDH1A1), EMT markers (E-cadherin, Snail), and immune-cell markers (CD3+, CD4+ or CD8+ T lymphocytes, CD20+ B lymphocytes, CD68+ macrophages) were detected in tumor center (TC) and tumor invasive front by an immunohistochemical method. Unsupervised hierarchical clustering analysis was performed to group the data according to correlation analyses. Survival analysis and chi-square test were performed to explore the significance of this clustering. RESULTS: There were correlations among the expression of Nanog, Lgr5, CD44v6, and immune cell counts (P < .05). Nanog, Lgr5, CD44v6, and ALDH1A1 positively related to E-cadherin or Snail (P < .05). A cluster (termed cluster SIE) based on cancer stemness markers (Nanog, Lgr5, CD44v6, ALDH1A1 in TC), EMT markers (E-cadherin, Snail in TC), and immune-cell markers (CD4+ and CD8+ T-lymphocyte counts in TC, and CD68+ macrophages in tumor invasive front) could significantly predict 5-year survival (P = .040). Multivariate Cox proportional hazard model showed that only tumor, node, metastasis classification system stage and cluster SIE were independent prognostic predictors (hazard ratio = 1.920; 95% confidence interval, 1.082-3.407; P = .026). CONCLUSION: Cancer stemness, immune state, and EMT programs should be considered as a whole. Cluster SIE was an independent predictor for 5-year survival of patients with colorectal cancer.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal , Células Madre Neoplásicas/patología , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , China , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/cirugía , Femenino , Estudios de Seguimiento , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia , Análisis de Matrices Tisulares , Resultado del Tratamiento , Microambiente Tumoral/inmunología
10.
Oncoimmunology ; 7(2): e1380765, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29416940

RESUMEN

The immune contexture, a composition of the tumor microenvironment, plays multiple important roles in cancer stem cell (CSC) and epithelial-mesenchymal transition (EMT), and hence critically influences tumor initiation, progression and patient outcome. Tumor-associated macrophages (TAMs) are abundant in immune contexture, however their roles in CSC, EMT and prognosis of colorectal cancer (CRC) have not been elucidated. In 419 colorectal carcinomas, immune cell types (CD68+ macrophages, CD3+, CD4+ or CD8+ T lymphocytes, CD20+ B lymphocytes), EMT markers (E-cadherin and Snail) as well as the stem cell marker (CD44v6) were detected in tumor center (TC) and tumor invasive front (TF) respectively by immunohistochemistry. Tumor buds, that represent EMT phenotype, were also counted. It was found CD68+ macrophages were the most infiltrating immune cells in CRC. By correlation analysis, more CD68+TF macrophages were associated with more CD44v6 expression (p < 0.001), lower SnailTF expression (p = 0.08) and fewer tumor buds (p < 0.001). More CD68+TF macrophages were significantly related to more CD3+TF T lymphocytes (p = 0.002), CD8+TF T lymphocytes (p < 0.001) and CD20+TF B lymphocytes counts (p = 0.004). Strong CD68+TF macrophages infiltration also predicted long term overall survival. CRC patients with more tumor buds had worse survival. However, strong CD68+TF macrophages infiltration could reverse the unfavorable results since patients with more tumor buds but increasing CD68+TF macrophages infiltration had the favorable outcome, similar to lower tumor buds groups. This study provided direct morphological evidence that tumor-associated macrophages in the invasive front play critical roles in fighting with the unfavorable results of tumor buds, thus resulting favorable outcomes for CRC patients.

11.
Carcinogenesis ; 38(11): 1057-1062, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-28968818

RESUMEN

Copy number variations (CNVs) contribute to the development of colorectal cancer (CRC). We conducted a two-stage association study to identify CNV risk loci for CRC. We performed a gene-based rare CNV study on 694 sporadic CRC and 1641 controls using Illumina Human-OmniExpress-12v1.0 BeadChips, and further replicated in 934 CRC cases and 2680 controls for risk CNVs by using TaqMan Copy Number Assay. Tumor buddings, cancer cells in the center of primary tumor and normal intestinal epithelial cells were captured using laser capture microdissection (LCM) and were assayed using AffymetrixGeneChip® Human Genome U133 Plus 2.0 Array. In addition, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus data were assessed for the effects of risk CNVs. We found that germline deletions affecting the last six exons of SLC18A1 significantly associated with CRC with a combined P value of 6.4 × 10-5 by a two-stage analysis. Both in TCGA CRC RNA seq dataset and GDS4382, SLC18A1 was significantly down regulated in CRC tissues than in paired normal tissues (N = 32 and 17 pairs, P = 0.004 and 0.009, respectively). In LCM samples, similar observations were obtained that the expression levels of SLC18A1 in the tumor buddings, cancer cells in the center of primary tumor, and stroma of both tumor budding and cancer cells were lower than normal intestinal epithelial and stromal cells (fold change = 0.17-0.62, 0.12-0.57 and 0.37-0.68, respectively). In summary, the germline deletions at SLC18A1 contributed to the development of CRC. The role of SLC18A1 required further exploration.


Asunto(s)
Neoplasias Colorrectales/genética , Eliminación de Secuencia/genética , Proteínas de Transporte Vesicular de Monoaminas/genética , Adulto , Estudios de Casos y Controles , China , Variaciones en el Número de Copia de ADN/genética , Regulación hacia Abajo/genética , Células Epiteliales/metabolismo , Exones/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Genoma Humano/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Mucosa Intestinal/metabolismo , Masculino , Persona de Mediana Edad , Factores de Riesgo , Células del Estroma/metabolismo
12.
Sci Rep ; 7(1): 4915, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28687755

RESUMEN

Substantial evidence has shown that epithelial-mesenchymal transition (EMT) plays critical roles in colorectal cancer (CRC) development and prognosis. To uncover the pivotal regulators that function in the cooperative interactions between cancer cells and their microenvironment and consequently affect the EMT process, we carried out a systematic analysis and evaluated prognosis in CRC specimens. Tumor buds and their surrounding stroma were captured using laser microdissection. We used gene expression profiling, bioinformatics analysis and regulatory network construction for molecular selection. The clinical significance of potential biomarkers was investigated. We identified potential EMT biomarkers, including BGN, MMP1, LGALS1, SERPINB5, and TM4SF4, all of which participated in the integrated pathway of TGFß/Snail with TNFα/NFκB. We also found that BGN, MMP1, LGALS1, SERPINB5 and TM4SF4 were related to CRC patient prognosis. Patients with higher expression of these individual potential biomarkers had poorer prognosis. Among the identified biomarkers, BGN and TM4SF4 are reported, for the first time, to probably be involved in the EMT process and to predict CRC prognosis. Our results strongly suggest that the integrated pathway of TGFß/Snail with TNFα/NFκB may be the principal axis that links cancer cells to their microenvironment during the EMT process and results in poor prognosis in CRC patients.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinogénesis/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Biglicano/genética , Biglicano/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Pronóstico , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Análisis de Supervivencia , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
13.
Oncoimmunology ; 6(1): e1260213, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28197382

RESUMEN

Gene microarray and bioinformatic analysis showed that S100A8 was more abundant in the stroma surrounding tumor buddings (TBs) than in the stroma surrounding primary tumor cells in colorectal carcinomas. Here, S100A8+ cells in 419 colorectal carcinoma samples were stained by immunohistochemistry and counted using Image-pro plus 6.0. TBs were also counted and biomarkers associated with the epithelial-mesenchymal transition and apoptosis were assessed by immunohistochemistry. We evaluated the association between S100A8+ cells and clinico-pathological variables as well as survival. Migration and invasion as well as biomarkers of the epithelial-mesenchymal transition and apoptosis were tested in CRC cells, treated with graded concentrations of recombinant human S100A8 protein. We found that the density of S100A8+ cells in the tumor invasive front (S100A8+TIF) clearly distinguished patients with 5-y survival from those who did not survive (p = 0.01). The S100A8+-associated tumor budding (SATB) index determined by the S100A8+TIF and TB was an independent predictor of overall survival (p = 0.001) other than the S100A8+TIF or TB alone. Migration and invasion properties of CRC cells were inhibited by recombinant human S100A8 treatment. The particular S100A8+ cells in the stroma were associated with important biomarkers of the epithelial-mesenchymal transition (E-cadherin and SNAIL) and apoptosis (BCL2). In conclusion, S100A8+ cells in the stroma predict a good prognosis in colorectal carcinoma. An index combining S100A8+ cells and TB independently predicts survival. Recombinant human S100A8 inhibited CRC cell migration and invasion, which was involved in epithelial-mesenchymal transition (E-cadherin and SNAIL) and apoptosis (BCL2).

14.
Mol Cancer ; 15(1): 77, 2016 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-27912767

RESUMEN

BACKGROUND: Lipocalin2 (LCN2) is a secretory protein that is aberrantly expressed in several types of cancer and has been involved in metastatic progression. However, neither mechanisms nor the role that LCN2 plays in the metastasis of colorectal cancer are clear. METHODS: LCN2 expression in colorectal cancer was detected by immunohistochemistry in 400 tissue specimens and Kaplan-Meier survival analysis was performed. In vitro, real-time PCR, western blot, colony formation assay, immunofluorescence assay, wound healing assay, migration and invasion experiment were performed to investigate the effects of LCN2 in epithelial mesenchymal transition (EMT), migration and invasion, respectively. In vivo mouse xenograft and metastasis models were utilized to determine tumorigenicity and metastasis ability, and immunohistochemistry, real-time PCR, western blot were used to evaluate the related protein expression. Luciferase reporter assay was used to explore the role of LCN2 on NF-ĸB promoter. RESULTS: LCN2 was highly expressed in 66.5% of the specimens, and significantly correlated with positive E-cadherin in the membrane and negative nuclear ß-catenin. Higher expression of LCN2 together with negative NF-κB expression was negatively related to nuclear accumulation of snail and predicted favorable prognosis. LCN2 blocked cell proliferation, migration and invasion in vitro and in vivo, and inhibited translocation of NF-κB into nucleus. NF-κB could reverse the effect of LCN2 on EMT and promote snail expression. Rescued snail expression had similar effect without influencing NF-κB activity. CONCLUSION: LCN2 may be an important negative regulator in EMT, invasion and metastasis of CRC via acting as upstream of NF-κB/snail signaling pathway. Thereby combinative manipulation of LCN2 and NF-κB/snail pathway may represent a novel and promising therapeutic approach for the patients with CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Lipocalina 2/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Regulación hacia Arriba , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Lipocalina 2/genética , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Trasplante de Neoplasias , Transducción de Señal , Análisis de Supervivencia
15.
Oncotarget ; 7(32): 51408-51422, 2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27285982

RESUMEN

Continued use of trastuzumab in PTEN-deficient HER2+ breast cancer induces the epithelial-to-mesenchymal transition (EMT), transforms HER2+ to triple negative breast cancer, and expands breast cancer stem cells (BCSCs). Using cancer cell lines with two distinct states, epithelial and mesenchymal, we identified novel targets during EMT in PTEN-deficient trastuzumab-resistant breast cancer. Differential gene expression and distinct responses to a small molecule in BT474 (HER2+ trastuzumab-sensitive) and the PTEN-deficient trastuzumab-resistant derivative (BT474-PTEN-LTT) provided the selection tools to identify targets during EMT. siRNA knockdown and small molecule inhibition confirmed MEOX1 as one of the critical molecular targets to regulate both BCSCs and mesenchymal-like cell proliferation. MEOX1 was associated with poor survival, lymph node metastasis, and stage of breast cancer patients. These findings suggest that MEOX1 is a clinically relevant novel target in BCSCs and mesenchymal-like cancer cells in PTEN-deficient trastuzumab resistant breast cancer and may serve as target for future drug development.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/fisiología , Trastuzumab/uso terapéutico , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/fisiología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio , Humanos , Persona de Mediana Edad , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Fosfohidrolasa PTEN/genética , ARN Interferente Pequeño/farmacología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
16.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(1): 24-30, 2016 01.
Artículo en Chino | MEDLINE | ID: mdl-27045237

RESUMEN

OBJECTIVE: To investigate the association of neuroendocrine differentiation with progression and prognosis of gastric adenocarcinoma. METHODS: Clinicopathological data of 240 patients with gastric adenocarcinomas were retrospectively analyzed. The expression of chromogranin A, synaptophysin and secrectagogin in cancer tissue was assessed by immunohistochemistry. The association of neuoroendocrine differentiation parameters with disease progression and survival of patients was analyzed. RESULTS: The expression of synaptophysin was positively correlated with depth of invasion and secretagogin more often expressed in cases with lymph node metastasis. In Lauren diffuse type of cancer, expression of chromogranin A and secretagogin was unfavorable prognostic predictor. In TNM stage II adenocarcinoma, expression of chromogranin A and synaptophysin related to poor survival, and multivariate Cox proportional hazard model showed that synaptophysin was an independent predictor for poor survival. CONCLUSION: Neuroendocrine differentiation predicts deeper depth of invasion, more possibility of lymph node metastasis and poor survival in gastric adenocarcinoma.


Asunto(s)
Adenocarcinoma/patología , Tumores Neuroendocrinos/patología , Neoplasias Gástricas/patología , Adenocarcinoma/diagnóstico , Biomarcadores de Tumor/metabolismo , Cromogranina A/metabolismo , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Metástasis Linfática , Estadificación de Neoplasias , Tumores Neuroendocrinos/diagnóstico , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Secretagoginas/metabolismo , Neoplasias Gástricas/diagnóstico , Sinaptofisina/metabolismo
17.
Cell Adh Migr ; 10(4): 434-46, 2016 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-26743180

RESUMEN

Tumor budding occurs at the invasive front of cancer; the tumor cells involved have metastatic and stemness features, indicating a poor prognosis. Tumor budding is partly responsible for cancer metastasis, and its initiation is based on the epithelial-mesenchymal transition (EMT) process. The EMT process involves the conversion of epithelial cells into migratory and invasive cells, and is a profound event in tumorigenesis. The EMT, associated with the formation of cancer stem cells (CSCs) and resistance to therapy, results from a combination of gene mutation, epigenetic regulation, and microenvironmental control. Tumor budding can be taken to represent the EMT in vivo. The EMT process is under the influence of the tumor microenvironment as well as tumor cells themselves. Here, we demonstrate that the tumor microenvironment dominates EMT development and impacts cancer metastasis, as well as promotes CSC formation and mediates drug resistance. In this review, we mainly discuss components of the microenvironment, such as the extracellular matrix (ECM), inflammatory cytokines, metabolic products, and hypoxia, that are involved in and impact on the acquisition of tumor-cell motility and dissemination, the EMT, metastatic tumor-cell formation, tumor budding and CSCs, and cancer metastasis, including subsequent chemo-resistance. From our point of view, the tumor microenvironment now constitutes a promising target for cancer therapy.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias/patología , Microambiente Tumoral , Animales , Matriz Extracelular/metabolismo , Humanos , Inflamación/patología , Metástasis de la Neoplasia
18.
Clin Chim Acta ; 431: 23-32, 2014 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-24513543

RESUMEN

Insulin-like growth factor binding protein-related protein 1 (IGFBP-rP1) belongs to the IGFBP family whose members have a conserved structural homology. It has a low affinity for IGFs and a high affinity for insulin, suggesting that IGFBP-rP1 may have a biological function distinct from other members of the family. IGFBP-rP1 is ubiquitously expressed in normal human tissues and has diverse biological functions, regulating cell proliferation, apoptosis and senescence; it may also have a key role in vascular biology. Increasing evidence suggests that IGFBP-rP1 acts as a tumor suppressor. It elicits its biological effects by both insulin/IGF-dependent and -independent mechanisms. This paper provides a brief overview of the structure and regulation of IGFBP-rP1 and its various biological functions in cancer, as well as the underlying molecular mechanisms.


Asunto(s)
Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Neoplasias/metabolismo , Animales , Fibrosis/patología , Humanos , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/química , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/fisiología
19.
Mutagenesis ; 28(1): 25-37, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23188737

RESUMEN

Recently, many new loci associated with type 2 diabetes have been uncovered by genetic association studies and genome-wide association studies. As more reports are made, particularly with respect to varying ethnicities, there is a need to determine more precisely the effect sizes in each major racial group. In addition, some reports have claimed ethnic-specific associations with alternative single-nucleotide polymorphisms (SNPs), and to that end there has been a degree of confusion. We conducted a meta-analysis using an additive genetic model. Eight polymorphisms in 155 studies with 121174 subjects (53385 cases and 67789 controls) were addressed in this meta-analysis. Significant associations were found between type 2 diabetes and rs7903146, rs12255372, rs11196205, rs7901695, rs7895340 and rs4506565, with summary odds ratios (ORs) (95% confidence interval) of 1.39 (1.34-1.45), 1.33 (1.27-1.40), 1.20 (1.14-1.26), 1.32 (1.25-1.39), 1.21 (1.13-1.29) and 1.39 (1.29-1.49), respectively. In addition, no significant associations were found between the two polymorphisms (rs290487 and rs11196218) and type 2 diabetes. The summary ORs for the six statistically significant associations (P < 0.05) were further evaluated by estimating the false-positive report probability, with results indicating that all of the six significant associations were considered noteworthy, and may plausibly be true associations. Significant associations were found between the six polymorphisms (rs7903146, rs12255372, rs11196205, rs7901695, rs7895340 and rs4506565) in the TCF7L2 gene and type 2 diabetes risk, and the other two polymorphisms (rs11196218 and rs290487) were not found to be significantly associated with type 2 diabetes. Subgroups analyses show that significant associations are not found between the six SNPs (rs7903146, rs12255372, rs11196205, rs7901695, rs7895340, and rs4506565) and the type 2 diabetes in some ethnic populations.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Proteína 2 Similar al Factor de Transcripción 7/genética , Humanos , Desequilibrio de Ligamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...