Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
BMC Pregnancy Childbirth ; 22(1): 916, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36482358

ABSTRACT

PURPOSE: This review aimed to investigate the association of insulin resistance (IR) in women with recurrent pregnancy loss compared to women with normal pregnancy history. METHODS: PubMed, EMBASE, the Web of Science and Google Scholar databases were accessed to collect published observational studies that compared IR of recurrent pregnancy loss women with healthy women until the 6th of October 2022. Outcomes assessed in this review and meta-analysis included fasting blood glucose, fasting plasma insulin, homeostasis model assessment for IR, glucose to insulin ratio. Mean differences, odds ratios with 95% confidence interval were pooled using the fixed or random effect models. Sensitivity analyses were performed to validate the robustness of the results. Review Manager version 5.4.1 and Stata version 8.0 were used. RESULTS: A total of nineteen studies involving 4453 individuals were included. Recurrent pregnancy loss patients presented significantly higher fasting blood glucose, fasting plasma insulin, homeostasis model assessment for IR, and lower glucose to insulin ratios. Additionally, recurrent pregnancy loss patients had higher rates of IR as defined by abnormal fasting plasma insulin, homeostasis model assessment for IR, and glucose to insulin ratio. Sensitivity analyses validated the robustness of the results. CONCLUSION: In the current review, we show that recurrent pregnancy loss is associated with a higher degree of IR and highlight the importance of screening and treatment of IR.


Subject(s)
Abortion, Habitual , Insulin Resistance , Humans , Female , Blood Glucose , Insulin
2.
J Ovarian Res ; 15(1): 109, 2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36180908

ABSTRACT

OBJECTIVE: This review aimed to investigate the metabolic profile of women with premature ovarian insufficiency (POI) compared relative to women with normal ovarian functioning. METHODS: A systematic search of PubMed, EMBASE, and the Web of Science for observational studies published up until the 6th of July 2021 that compared the metabolic profile of POI women with a healthy control group were assessed. Mean differences (MD) and 95% confidence interval (CI) were pooled using the fixed or random effect models. RESULTS: A total of 21 studies involving 1573 women with POI and 1762 control women were included. POI patients presented significantly higher waist circumference, total cholesterol, low-density lipoprotein, high-density lipoprotein, triglycerides, and fasting glucose. Additionally, POI patients had marginally higher insulin level. However, the differences in systolic, and diastolic blood pressure were non-significant relative to the control group. CONCLUSIONS: POI is associated with alterations in certain metabolic parameters compared to control women. This finding highlights the importance of early screening and the lifelong management of metabolic health for women with POI.


Subject(s)
Insulins , Menopause, Premature , Primary Ovarian Insufficiency , Cholesterol , Female , Glucose , Humans , Lipoproteins , Lipoproteins, HDL , Primary Ovarian Insufficiency/metabolism , Triglycerides
3.
Reprod Biomed Online ; 45(4): 805-814, 2022 10.
Article in English | MEDLINE | ID: mdl-35902299

ABSTRACT

RESEARCH QUESTION: What is the association between preconception serum lipid concentrations and reproductive outcomes after ovulation induction in women with polycystic ovary syndrome (PCOS)? DESIGN: A secondary analysis of a randomized controlled trial with 1000 PCOS women undergoing ovulation induction with clomiphene with or without acupuncture. Preconception serum total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides, apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB) were measured. Outcomes were ovulation, conception, pregnancy, live birth and miscarriage. Logistic regression was used to calculate odds ratios (OR) with 95% confidence intervals (CI). RESULTS: In total, 780 women ovulated; 320 women achieved conception, 218 had a clinical pregnancy, 205 had a live birth and 115 had a miscarriage. Serum lipid concentrations per one unit increment were independently associated with reproductive outcomes after controlling for confounders. Increasing LDL-C (OR 0.79, 95% CI 0.63-0.99) was independently associated with a lower chance of ovulation. Increasing total cholesterol (OR 0.76, 95% CI 0.62-0.92), LDL-C (OR 0.73, 95% CI 0.57-0.93), triglycerides (OR 0.74, 95% CI 0.58-0.95) and ApoB (OR 0.34, 95% CI 0.16-0.74) were independently associated with a lower chance of clinical pregnancy. Increased total cholesterol (OR 0.78, 95% CI 0.64-0.96), LDL-C (OR 0.77, 95% CI 0.60-0.99), triglycerides (OR 0.76, 95% CI 0.59-0.96) and ApoB (OR 0.39, 95% CI 0.18-0.86) were independently associated with a lower chance of live birth. Furthermore, increased total cholesterol (OR 1.43, 95% CI 1.06-1.93), LDL-C (OR 1.51, 95% CI 1.04-2.19) and ApoB (OR 3.82, 95% CI 1.17-12.41) were independently associated with a higher chance of miscarriage. CONCLUSIONS: Increased serum lipids were negatively associated with the reproductive outcomes of PCOS women undergoing ovulation induction with clomiphene with or without acupuncture.


Subject(s)
Abortion, Spontaneous , Infertility, Female , Polycystic Ovary Syndrome , Abortion, Spontaneous/drug therapy , Apolipoprotein A-I , Apolipoproteins B/therapeutic use , Birth Rate , Cholesterol, LDL/therapeutic use , Clomiphene/therapeutic use , Female , Humans , Infertility, Female/complications , Lipoproteins, HDL/therapeutic use , Ovulation Induction , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/drug therapy , Pregnancy , Treatment Outcome , Triglycerides
4.
Front Endocrinol (Lausanne) ; 12: 683040, 2021.
Article in English | MEDLINE | ID: mdl-34248844

ABSTRACT

Objective: This article aimed to investigate whether serum magnesium is associated with insulin resistance index and testosterone level in women with polycystic ovary syndrome (PCOS). Materials and Methods: Overall 1000 women with PCOS were enrolled in a randomized controlled trial and a cross-sectional analysis of the association of serum magnesium with glucose metabolism markers and testosterone was performed. Serum magnesium, glucose metabolism markers and testosterone were measured. Insulin resistance was evaluated by homeostatic model assessment of insulin resistance (HOMA-IR) and quantitative insulin-sensitivity check index (QUICKI). Multivariable linear regression and logistic regression models were used to estimate the association between serum magnesium, insulin resistance and testosterone. Results: In comparative analyses, women with higher quartile of serum magnesium had significantly lower fasting glucose, HOMA-IR and testosterone. Multiple linear regression showed serum magnesium was independently negatively associated with insulin, glucose, HOMA-IR, testosterone and positively associated with QUICKI (P for trend <0.05) after adjusting confounding covariates. Logistic regression showed serum magnesium in quartile 1 and 2 were independently associated with insulin resistance status (Quartile 1: OR: 2.15, 95%CI: 1.35-3.40, P = 0.001; Quartile 2: OR: 1.90, 95%CI: 1.20-3.02, P = 0.006), while quartile 1 was marginally associated with hyperandrogenemia status (Quartile 1: OR: 1.45, 95%CI: 0.99-2.11, P = 0.055) after adjusting confounding covariates. Conclusion: The current findings suggest that lower serum magnesium was associated with aggravated insulin resistance and higher testosterone levels among women with PCOS.


Subject(s)
Glucose/metabolism , Insulin Resistance , Magnesium/blood , Polycystic Ovary Syndrome/blood , Testosterone/blood , Adult , Blood Glucose/analysis , Female , Humans , Randomized Controlled Trials as Topic , Young Adult
5.
Biomark Med ; 15(12): 1043-1053, 2021 08.
Article in English | MEDLINE | ID: mdl-34289735

ABSTRACT

Aim: To detect predictive value of preconception or early pregnancy sex hormone-binding globulin (SHBG) for subsequent gestational diabetes mellitus (GDM). Materials & methods: We searched Embase, Medline, PubMed, Web of Science and Cochrane library up to January 2020. Studies assessing diagnostic performance of SHBG for GDM diagnosed by well-defined diagnostic criteria using oral glucose tolerance test. Results: Totally seven studies with 1947 women were included and 247 were diagnosed as GDM. SHBG had a combined diagnostic odds ratio of 6.68 (95% CI: 4.58-9.74), sensitivity of 0.70 (95% CI: 0.51-0.84), specificity of 0.74 (95% CI: 0.52-0.88), positive likelihood ratio of 2.49 (95% CI: 1.73-3.57) and negative likelihood ratio of 0.37 (95% CI: 0.23-0.61). The area under the summary receiver operating characteristic curve was 0.78 (95% CI: 0.74-0.82). Conclusion: SHBG had a predictive value for GDM and might improve GDM screening. However, heterogeneity between studies warrants more research into this topic.


Subject(s)
Biomarkers/blood , Diabetes, Gestational/blood , Mass Screening/methods , Sex Hormone-Binding Globulin/analysis , Adult , Diabetes, Gestational/diagnosis , Female , Humans , Predictive Value of Tests , Pregnancy , ROC Curve , Reproducibility of Results
6.
Front Endocrinol (Lausanne) ; 12: 633766, 2021.
Article in English | MEDLINE | ID: mdl-33763032

ABSTRACT

Objective: To evaluate associations between serum lipid levels and treatment outcomes in women undergoing assisted reproduction. Materials and Methods: The study included 2011 women who underwent in vitro fertilization/intracytoplasmic sperm injection with fresh embryo transfer. Serum lipid evaluation included total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides (TG). Ovarian stimulation outcomes included endometrial thickness and the number of oocytes retrieved, and reproductive outcomes included live birth, clinical pregnancy, and miscarriage. Results: Higher HDL-C quartiles were associated with more oocytes retrieved. Lower TC (quartile 1 odds ratio [OR] 1.59 [1.21-2.08], quartile 3 OR 1.36 [1.04-1.77]), LDL-C (quartile 1 OR 1.41 [1.07-1.86]), and TG (quartile 2 OR 1.39 [1.06-1.84]) were independently associated with clinical pregnancy after adjusting for potential confounders. Lower LDL-C (quartile 1 OR 2.22 [1.58-3.13], quartile 2 OR 1.78 [1.27-2.50], quartile 3 OR 1.51 [1.07-2.13]), TC (quartile 1 OR 1.39 [1.00-1.93]), TG (quartile 1 OR 1.44 [1.03-2.03], quartile 2 OR 1.46 [1.04-2.04], quartile 3 OR 1.44 [1.04-1.99]), and higher HDL-C (quartile 2 OR 0.71 [0.51-0.99]) were independently associated with live birth. Higher LDL-C (quartile 1 OR 0.44 [0.30-0.66], quartile 2 OR 0.49 [0.33-0.73], quartile 3 OR 0.63 [0.43-0.94]) and lower HDL-C (quartile 1 OR 1.60 [1.07-2.39]) were independently associated with miscarriage. Conclusions: Serum lipid levels were associated with treatment outcomes in women undergoing assisted reproduction.


Subject(s)
Infertility, Female/therapy , Lipids/blood , Reproductive Techniques, Assisted , Abortion, Spontaneous/metabolism , Adolescent , Adult , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Female , Fertilization in Vitro , Humans , Live Birth , Middle Aged , Pregnancy , Pregnancy Outcome , Retrospective Studies , Sperm Injections, Intracytoplasmic/methods , Treatment Outcome , Triglycerides/blood , Young Adult
7.
Front Med (Lausanne) ; 8: 736320, 2021.
Article in English | MEDLINE | ID: mdl-35071255

ABSTRACT

Objective: To evaluate the effect of hyperinsulinemia (HI) and insulin resistance (IR) on endocrine, metabolic, and reproductive outcomes in women without polycystic ovary syndrome (PCOS) undergoing assisted reproduction. Materials and Methods: The study included 1,104 non-PCOS women undergoing in vitro fertilization/intracytoplasmic sperm injection-fresh embryo transfer. HI was evaluated by serum fasting insulin (FIN), and IR was evaluated by homeostatic model assessment of insulin resistance index (HOMA-IR). In addition, biometric, sex hormone, and metabolic parameters were measured. Independent t-test, linear, and logistic regression examined associations between HI, IR, and endocrine, metabolic, ovarian stimulation characteristics, and reproductive outcomes. Results: Women with HI and IR had lower levels of progesterone, luteinizing hormone, follicle-stimulating hormone, estradiol, high-density lipoproteins, and increased levels of triglycerides low-density lipoproteins. For ovarian stimulation characteristics, those with HI and IR had a longer duration of stimulation, a higher total gonadotropin dose, and a lower peak estradiol level. Linear regression confirmed these associations. For reproductive outcomes, HI and IR were not associated with clinical pregnancy, live birth, and miscarriage. Conclusions: HI and IR did not impair reproductive outcomes in non-PCOS women undergoing assisted reproduction.

8.
Front Cardiovasc Med ; 8: 790454, 2021.
Article in English | MEDLINE | ID: mdl-34977197

ABSTRACT

Objective: To investigate the prevalence, pattern and risk predictors for dyslipidemia among Chinese women with polycystic ovary syndrome (PCOS). Study Design and Methods: A total of 1,000 women diagnosed as PCOS by modified Rotterdam criteria were enrolled in 27 hospitals across China in a randomized controlled trial. Anthropometric, metabolic parameters, sex hormone, and lipid levels were measured at the baseline visit. Dyslipidemia was defined according to total cholesterol (TC), low density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides (TG) level. Independent t-test and logistic regression were used to identify predictors for dyslipidemia. Area under the receiver operating characteristic curve (AUC) was calculated. Results: A total of 41.3% of the women had dyslipidemia, and the prevalence of abnormal TC, LDL-C, HDL-C, and TG were 8.6, 9.1, 26.9, and 17.5%, respectively. Logistic regression found that age, waist circumference, insulin, follicle-stimulating hormone, and sex hormone-binding globulin were independent predictors for dyslipidemia. When combining these predictors, the AUC was 0.744. The cut-off points were age >28.5 years, waist circumference >86.5 cm, insulin >96.0 pmol/L, follicle-stimulating hormone <5.6 mIU/mL, and sex hormone-binding hormone <31.0 nmol/L, respectively. Conclusion: Dyslipidemia was common in Chinese women with PCOS, and low HDL-C level was the predominant lipid abnormality. Age, waist circumference, follicle-stimulating hormone, insulin and sex hormone-binding globulin were predictive for dyslipidemia among Chinese women with PCOS.

9.
Int J Endocrinol ; 2020: 7580218, 2020.
Article in English | MEDLINE | ID: mdl-33101409

ABSTRACT

OBJECTIVE: To investigate the relationships between sex hormone-binding globulin (SHBG) and comprehensive metabolic parameters including biometric, glycemic, lipid, liver, and renal functions of women with polycystic ovary syndrome (PCOS). Study Design and Methods. A total of 1000 women diagnosed as PCOS by modified Rotterdam criteria were enrolled in a randomized controlled trial. SHBG and comprehensive metabolic parameters were measured at the baseline visit. Metabolic parameters included biometric parameters, glucose and lipid panels, and liver and renal function parameters. An independent t-test and linear regression were performed to investigate the associations between SHBG and metabolic parameters. Logistic regression was used to detect the relationship between SHBG and the presence of metabolic syndrome. RESULTS: In comparative analyses, PCOS women with lower SHBG levels had higher body mass index, waist circumference, insulin, homeostatic model assessment-insulin resistance (HOMA-IR) index, systolic and diastolic blood pressure, triglycerides, apolipoprotein B (APOB), low-density lipoprotein (LDL), aspartate transferase (AST), alanine transferase (ALT), and blood urea nitrogen (BUN), but lower high-density lipoprotein (HDL) and apolipoprotein A1 (APOA1). In linear regression, SHBG was inversely associated with waist circumference, systolic blood pressure, triglyceride, LDL, APOB, ALT, AST, and BUN but positively associated with HDL and APOA1 after adjusting the BMI. In logistic regression, SHBG is a protective predictor for metabolic syndrome (odds ratio = 0.96; 95% confidence interval: 0.95-0.97). The area under the receiver-operator characteristic curve is 0.732 with a 95% confidence interval of 0.695-0.770. SHBG <26.75 mmol/L is the cutoff point with the best Youden index, which has a sensitivity of 0.656 and specificity of 0.698. CONCLUSIONS: Lower SHBG was associated with worsening biometric, lipid, liver, and renal functions but not glycemic parameters among women with PCOS. SHBG can be used as a tool to screen metabolic syndrome. This trial is registered with NCT01573858 and ChiCTR-TRC-12002081.

10.
Theranostics ; 10(19): 8633-8647, 2020.
Article in English | MEDLINE | ID: mdl-32754268

ABSTRACT

Rationale: The prognosis of gastric cancer (GC) patients is poor, and there is limited therapeutic efficacy due to genetic heterogeneity and difficulty in early-stage screening. Here, we developed and validated an individualized gene set-based prognostic signature for gastric cancer (GPSGC) and further explored survival-related regulatory mechanisms as well as therapeutic targets in GC. Methods: By implementing machine learning, a prognostic model was established based on gastric cancer gene expression datasets from 1699 patients from five independent cohorts with reported full clinical annotations. Analysis of the tumor microenvironment, including stromal and immune subcomponents, cell types, panimmune gene sets, and immunomodulatory genes, was carried out in 834 GC patients from three independent cohorts to explore regulatory survival mechanisms and therapeutic targets related to the GPSGC. To prove the stability and reliability of the GPSGC model and therapeutic targets, multiplex fluorescent immunohistochemistry was conducted with tissue microarrays representing 186 GC patients. Based on multivariate Cox analysis, a nomogram that integrated the GPSGC and other clinical risk factors was constructed with two training cohorts and was verified by two validation cohorts. Results: Through machine learning, we obtained an optimal risk assessment model, the GPSGC, which showed higher accuracy in predicting survival than individual prognostic factors. The impact of the GPSGC score on poor survival of GC patients was probably correlated with the remodeling of stromal components in the tumor microenvironment. Specifically, TGFß and angiogenesis-related gene sets were significantly associated with the GPSGC risk score and poor outcome. Immunomodulatory gene analysis combined with experimental verification further revealed that TGFß1 and VEGFB may be developed as potential therapeutic targets of GC patients with poor prognosis according to the GPSGC. Furthermore, we developed a nomogram based on the GPSGC and other clinical variables to predict the 3-year and 5-year overall survival for GC patients, which showed improved prognostic accuracy than clinical characteristics only. Conclusion: As a tumor microenvironment-relevant gene set-based prognostic signature, the GPSGC model provides an effective approach to evaluate GC patient survival outcomes and may prolong overall survival by enabling the selection of individualized targeted therapy.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling/methods , Stomach Neoplasms/mortality , Transforming Growth Factor beta1/genetics , Vascular Endothelial Growth Factor B/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Machine Learning , Male , Middle Aged , Nomograms , Precision Medicine , Prognosis , Proportional Hazards Models , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Analysis , Tissue Array Analysis , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment , Vascular Endothelial Growth Factor B/metabolism , Young Adult
11.
Cell Commun Signal ; 17(1): 72, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31288844

ABSTRACT

BACKGROUND: Chronic gastritis has been demonstrated to be a key cause of gastric cancer (GC), and control of gastric inflammation is regarded as an effective treatment for the clinical prevention of gastric carcinogenesis. However, there remains an unmet need to identify the dominant regulators of gastric oncogenesis-associated inflammation in vivo. METHODS: The mouse model for the study of inflammation-associated GC was induced by Benzo[a]pyrene (BaP) intragastric administration in Bcl6b-/- and wildtype mice on a C57BL/6 background. 5-Aza-2'-deoxycytidine (5-Aza), the demethylation drug, was intraperitoneally injected to restore Bcl6b expression. Human GC tissue array was used to analyse patient survival based on BCL6B and CD3 protein expression. RESULTS: Bcl6b was gradually downregulated by its own promoter hypermethylation in parallel to an increasing inflammatory response during the progression of BaP-induced gastric carcinogenesis in mice. Moreover, knockout of Bcl6b dramatically worsened the severity of gastric cancer and aggravated the inflammatory response in the BaP-induced mice GC model. Re-activation of Bcl6b by 5-Aza impeded inflammatory amplification and BaP-induced GC development, prolonging survival time in wildtype mice, whereas no notable curative effect occurred in Bcl6b-/- mice with 5-Aza treatment. Finally, significant negative correlations were detected between the mRNA levels of BCL6B and inflammatory cytokines in human GC tissues; patients harbouring BCL6B-negetive and severe-inflammation GC tumours were found to exhibit the shortest survival time. CONCLUSIONS: Epigenetic inactivation of Bcl6b promotes gastric cancer through amplification of the gastric inflammatory response in vivo and offers a new approach for GC treatment and regenerative medicine.


Subject(s)
Carcinogenesis/genetics , Gene Knockout Techniques , Repressor Proteins/deficiency , Repressor Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Animals , Carcinogenesis/drug effects , Decitabine/pharmacology , Disease Progression , Down-Regulation/drug effects , Epigenesis, Genetic , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Stomach Neoplasms/metabolism , Survival Analysis
12.
J Exp Clin Cancer Res ; 37(1): 104, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29764469

ABSTRACT

BACKGROUND: Great progress has been achieved in the study of the aerobic glycolysis or the so-called Warburg effect in a variety of cancers; however, the regulation of the Warburg effect in Nasopharyngeal carcinoma (NPC) has not been completely defined. METHODS: Gene expression pattern of NPC cells were used to test associations between Chibby and ß-catenin expression. Chibby siRNAs and over-expression vector were transfected into NPC cells to down-regulate or up-regulate Chibby expression. Loss- and gain-of function assays were performed to investigate the role of Chibby in NPC cells. Western blot, cell proliferation, Glucose uptake, Lactate release, ATP level, and O2 consumption assays were used to determine the mechanism of Chibby regulation of underlying targets. Finally, immunohistochemistry assay of fresh NPC and nasopharyngeal normal tissue sample were used to detect the expression of Chibby, ß-Catenin, and PDK1 by immunostaining. RESULTS: We observed that Chibby, a ß-catenin-associated antagonist, is down-regulated in nasopharyngeal carcinoma cell lines and inhibits Wnt/ß-Catenin signaling induced Warburg effect. Mechanism study revealed that Chibby regulates aerobic glycolysis in NPC cells through pyruvate dehydrogenase kinase 1(PDK1), an important enzyme involved in glucose metabolism. Moreover, Chibby suppresses aerobic glycolysis of NPC via Wnt/ß-Catenin-Lin28/let7-PDK1 cascade. Chibby and PDK1 are critical for Wnt/ß-Catenin signaling induced NPC cell proliferation both in vitro and in vivo. Finally, immunostaining assay of tissue samples provides an important clinical relevance among Chibby, Wnt/ß-Catenin signaling and PDK1. CONCLUSIONS: Our study reveals an association between Chibby expression and cancer aerobic glycolysis, which highlights the importance of Wnt/ß-catenin pathway in regulation of energy metabolism of NPC. These results indicate that Chibby and PDK1 are the potential target for NPC treatment.


Subject(s)
Carrier Proteins/metabolism , MicroRNAs/genetics , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Aerobiosis , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Glycolysis , Heterografts , Humans , Immunohistochemistry , Mice , Nasopharyngeal Carcinoma/pathology , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA-Binding Proteins/genetics , Signal Transduction , Wnt Proteins/genetics , beta Catenin/genetics
13.
Mol Cancer ; 17(1): 84, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29690888

ABSTRACT

Conventional tumor markers for non-invasive diagnosis of gastric cancer (GC) exhibit insufficient sensitivity and specificity to facilitate detection of early gastric cancer (EGC). We aimed to identify EGC-specific exosomal lncRNA biomarkers that are highly sensitive and stable for the non-invasive diagnosis of EGC. Hence, in the present study, exosomes from the plasma of five healthy individuals and ten stage I GC patients and from culture media of four human primary stomach epithelial cells and four gastric cancer cells (GCCs) were isolated. Exosomal RNA profiling was performed using RNA sequencing to identify EGC-specific exosomal lncRNAs. A total of 79 and 285 exosomal RNAs were expressed at significantly higher levels in stage I GC patients and GCCs, respectively, than that in normal controls. Through combinational analysis of the RNA sequencing results, we found two EGC-specific exosomal lncRNAs, lncUEGC1 and lncUEGC2, which were further confirmed to be remarkably up-regulated in exosomes derived from EGC patients and GCCs. Furthermore, stability testing demonstrates that almost all the plasma lncUEGC1 was encapsulated within exosomes and thus protected from RNase degradation. The diagnostic accuracy of exosomal lncUEGC1 was evaluated, and lncUEGC1 exhibited AUC values of 0.8760 and 0.8406 in discriminating EGC patients from healthy individuals and those with premalignant chronic atrophic gastritis, respectively, which was higher than the diagnostic accuracy of carcinoembryonic antigen. Consequently, exosomal lncUEGC1 may be promising in the development of highly sensitive, stable, and non-invasive biomarkers for EGC diagnosis.


Subject(s)
Biomarkers, Tumor/blood , Exosomes/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/pathology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Early Detection of Cancer , Female , Humans , Male , Neoplasm Staging , RNA, Long Noncoding/blood , Stomach Neoplasms/blood , Stomach Neoplasms/genetics
14.
Oncotarget ; 8(40): 66987-67000, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978011

ABSTRACT

Marked up-regulation of aldose reductase (AR) is reportedly associated with the development of hepatocellular carcinoma (HCC). We investigated how aberrantly overexpressed AR might promote oncogenic transformation in liver cells and tissues. We found that overexpressed AR interacted with the kinase domain of AKT1 to increase AKT/mTOR signaling. In both cultured liver cancer cells and liver tissues in DEN-induced transgenic HCC model mice, we observed that AR overexpression-induced AKT/mTOR signaling tended to enhance lactate formation and hepatic inflammation to enhance hepatocarcinogenesis. Conversely, AR knockdown suppressed lactate formation and inflammation. Using cultured liver cancer cells, we also demonstrated that AKT1 was essential for AR-induced dysregulation of AKT/mTOR signaling, metabolic reprogramming, antioxidant defense, and inflammatory responses. These findings suggest that aberrantly overexpressed/over-activated hepatic AR promotes HCC development at least in part by interacting with oncogenic AKT1 to augment AKT/mTOR signaling. Inhibition of AR and/or AKT1 might serve as an effective strategy for the prevention and therapy of liver cancer.

15.
Hepatology ; 65(4): 1206-1221, 2017 04.
Article in English | MEDLINE | ID: mdl-27809333

ABSTRACT

Great progress has been achieved in the study of Hippo signaling in regulating tumorigenesis; however, the downstream molecular events that mediate this process have not been completely defined. Moreover, regulation of Hippo signaling during tumorigenesis in hepatocellular carcinoma (HCC) remains largely unknown. In the present study, we systematically investigated the relationship between Yes-associated protein/TEA domain family member (YAP-TEAD) and hepatocyte nuclear factor 4-alpha (HNF4α) in the hepatocarcinogenesis of HCC cells. Our results indicated that HNF4α expression was negatively regulated by YAP1 in HCC cells by a ubiquitin proteasome pathway. By contrast, HNF4α was found to directly associate with TEAD4 to compete with YAP1 for binding to TEAD4, thus inhibiting the transcriptional activity of YAP-TEAD and expression of their target genes. Moreover, overexpression of HNF4α was found to significantly compromise YAP-TEAD-induced HCC cell proliferation and stem cell expansion. Finally, we documented the regulatory mechanism between YAP-TEAD and HNF4α in rat and mouse tumor models, which confirmed our in vitro results. CONCLUSION: There is a double-negative feedback mechanism that controls TEAD-YAP and HNF4α expression in vitro and in vivo, thereby regulating cellular proliferation and differentiation. Given that YAP acts as a dominant oncogene in HCC and plays a crucial role in stem cell homeostasis and tissue regeneration, manipulating the interaction between YAP, TEADs, and HNF4α may provide a new approach for HCC treatment and regenerative medicine. (Hepatology 2017;65:1206-1221).


Subject(s)
Carcinoma, Hepatocellular/genetics , Hepatocyte Nuclear Factor 4/genetics , Liver Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Biopsy, Needle , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Neoplastic , Immunohistochemistry , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/genetics , Random Allocation , Rats , Rats, Wistar , Sensitivity and Specificity , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/genetics , YAP-Signaling Proteins
16.
Gastroenterology ; 150(3): 659-671.e16, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26619963

ABSTRACT

BACKGROUND & AIMS: Activation of WNT signaling promotes the invasive activities of several types of cancer cells, but it is not clear if it regulates the same processes in colorectal cancer (CRC) cells, or what mechanisms are involved. We studied the expression and function of OVOL2, a member of the Ovo family of conserved zinc-finger transcription factors regulated by the WNT signaling pathway, in intestinal tumors of mice and human beings. METHODS: We analyzed the expression of OVOL2 protein and messenger RNA in CRC cell lines and tissue arrays, as well as CRC samples from patients who underwent surgery at Xiamen University in China from 2009 to 2012; clinical information also was collected. CRC cell lines (SW620) were infected with lentivirus expressing OVOL2, analyzed in migration and invasion assays, and injected into nude mice to assess tumor growth and metastasis. Tandem affinity purification was used to purify the OVOL2-containing complex from CRC cells; the complex was analyzed by liquid chromatography, tandem mass spectrometry, and immunoprecipitation experiments. Gene promoter activities were measured in luciferase reporter assays. We analyzed mice with an intestine-specific disruption of Ovol2 (Ovol2(flox/+) transgenic mice), as well as Apc(min/+) mice; these mice were crossed and analyzed. RESULTS: Analysis of data from patients indicated that the levels of OVOL2 messenger RNA were significantly lower in colon carcinomas than adenomas, and decreased significantly as carcinomas progressed from grades 2 to 4. Immunohistochemical analysis of a tissue array of 275 CRC samples showed a negative association between tumor stage and OVOL2 level. Overexpression of OVOL2 in SW620 cells decreased their migration and invasion, reduced markers of the epithelial-to-mesenchymal transition, and suppressed their metastasis as xenograft tumors in nude mice; knockdown of OVOL2 caused LS174T cells to transition from epithelial to mesenchymal phenotypes. OVOL2 bound T-cell factor (TCF)4 and ß-catenin, facilitating recruitment of histone deacetylase 1 to the TCF4-ß-catenin complex; this inhibited expression of epithelial-to-mesenchymal transition-related genes regulated by WNT, such as SLUG, in CRC cell lines. OVOL2 was a downstream target of WNT signaling in LS174T and SW480 cells. The OVOL2 promoter was hypermethylated in late-stage CRC specimens from patients and in SW620 cells; hypermethylation resulted in OVOL2 down-regulation and an inability to inhibit WNT signaling. Disruption of Ovol2 in Apc(min/+) mice increased WNT activity in intestinal tissues and the formation of invasive intestinal tumors. CONCLUSIONS: OVOL2 is a colorectal tumor suppressor that blocks WNT signaling by facilitating the recruitment of histone deacetylase 1 to the TCF4-ß-catenin complex. Strategies to increase levels of OVOL2 might be developed to reduce colorectal tumor progression and metastasis.


Subject(s)
Cell Movement , Colorectal Neoplasms/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Caco-2 Cells , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Down-Regulation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Genotype , HCT116 Cells , HEK293 Cells , Histone Deacetylase 1/metabolism , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Promoter Regions, Genetic , RNA, Messenger/metabolism , Time Factors , Transcription Factor 4 , Transcription Factors/genetics , Transfection , Tumor Burden , beta Catenin/metabolism
17.
PLoS One ; 10(5): e0126890, 2015.
Article in English | MEDLINE | ID: mdl-26010149

ABSTRACT

MicroRNAs (miRs) play important roles in modulating gene expression during the processes of tumorigenesis and tumor development. Previous studies have found that miR-145 is down-regulated in the stomach neoplasm and is related to tumor migration and invasion. However, both the molecular mechanism and function of miR-145 in gastric cancer remain unclear. The present study is the first demonstration of the significant down-regulation of miR-145 expression in infiltrative gastric cancer compared to expanding gastric cancer. Additionally, correlation analyses revealed strong inverse correlations between miR-145 and FSCN1 expression levels in infiltrative gastric cancer. Furthermore, we demonstrated that miR-145 directly targets FSCN1 and suppresses cell migration and invasion in gastric cancer. Knocking down the expression of FSCN1 led to the suppression of migration and invasion in gastric cancer cells, and re-expressing FSCN1 in miR-145-overexpressing cells reversed their migration and invasion defects. Thus, we concluded that miR-145 regulates cell migration and invasion in gastric cancer primarily by directly targeting FSCN1.


Subject(s)
Carrier Proteins/metabolism , Cell Movement , MicroRNAs/metabolism , Microfilament Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , 3' Untranslated Regions , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MicroRNAs/genetics , Microfilament Proteins/genetics , Neoplasm Invasiveness , RNA, Small Interfering/metabolism
18.
J Cell Sci ; 126(Pt 24): 5692-703, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24101726

ABSTRACT

Wnt-ß-catenin signaling participates in the epithelial-mesenchymal transition (EMT) in a variety of cancers; however, its involvement in hepatocellular carcinoma (HCC) and downstream molecular events is largely undefined. HNF4α is the most prominent and specific factor maintaining the differentiation of hepatic lineage cells and a potential EMT regulator in HCC cells. However, the molecular mechanisms by which HNF4α maintains the differentiated liver epithelium and inhibits EMT have not been completely defined. In this study, we systematically explored the relationship between Wnt-ß-catenin signaling and HNF4α in the EMT process of HCC cells. Our results indicated that HNF4α expression was negatively regulated during Wnt-ß-catenin signaling-induced EMT through Snail and Slug in HCC cells. In contrast, HNF4α was found to directly associate with TCF4 to compete with ß-catenin but facilitate transcription co-repressor activities, thus inhibiting expression of EMT-related Wnt-ß-catenin targets. Moreover, HNF4α may control the switch between the transcriptional and adhesion functions of ß-catenin. Overexpression of HNF4α was found to completely compromise the Wnt-ß-catenin-signaling-induced EMT phenotype. Finally, we determined the regulation pattern between Wnt-ß-catenin signaling and HNF4α in rat tumor models. Our studies have identified a double-negative feedback mechanism controlling Wnt-ß-catenin signaling and HNF4α expression in vitro and in vivo, which sheds new light on the regulation of EMT in HCC. The modulation of these molecular processes may be a method of inhibiting HCC invasion by blocking Wnt-ß-catenin signaling or restoring HNF4α expression to prevent EMT.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Liver Neoplasms, Experimental/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Feedback, Physiological , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Liver Neoplasms, Experimental/pathology , Male , Protein Binding , Rats , Rats, Wistar , Snail Family Transcription Factors , Transcription Factor 4 , Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
19.
J Cell Sci ; 126(Pt 13): 2877-89, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23613467

ABSTRACT

Wnt signalling through ß-catenin and the lymphoid-enhancing factor 1/T-cell factor (LEF1/TCF) family of transcription factors maintains stem cell properties in both normal and malignant tissues; however, the underlying molecular pathway involved in this process has not been completely defined. Using a microRNA microarray screening assay, we identified let-7 miRNAs as downstream targets of the Wnt-ß-catenin pathway. Expression studies indicated that the Wnt-ß-catenin pathway suppresses mature let-7 miRNAs but not the primary transcripts, which suggests a post-transcriptional regulation of repression. Furthermore, we identified Lin28, a negative let-7 biogenesis regulator, as a novel direct downstream target of the Wnt-ß-catenin pathway. Loss of function of Lin28 impairs Wnt-ß-catenin-pathway-mediated let-7 inhibition and breast cancer stem cell expansion; enforced expression of let-7 blocks the Wnt-ß-catenin pathway-stimulated breast cancer stem cell phenotype. Finally, we demonstrated that the Wnt-ß-catenin pathway induces Lin28 upregulation and let-7 downregulation in both cancer samples and mouse tumour models. Moreover, the delivery of a modified lin28 siRNA or a let-7a agomir into the premalignant mammary tissues of MMTV-wnt-1 mice resulted in a complete rescue of the stem cell phenotype driven by the Wnt-ß-catenin pathway. These findings highlight a pivotal role for Lin28/let-7 in Wnt-ß-catenin-pathway-mediated cellular phenotypes. Thus, the Wnt-ß-catenin pathway, Lin28 and let-7 miRNAs, three of the most crucial stem cell regulators, connect in one signal cascade.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/genetics , Wnt1 Protein/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , RNA-Binding Proteins/genetics , Transcriptional Activation , Wnt1 Protein/genetics , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...