Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Oral Oncol ; 152: 106760, 2024 May.
Article in English | MEDLINE | ID: mdl-38520758

ABSTRACT

This study explored the relationship between betel-nut chewing and programmed death-ligand 1 (PD-L1) expression in recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC) patients in Taiwan. A total 280 R/M HNSCC patients, predominantly male, were evaluated; 75.4 % of whom chewed betel-nut. The prevalence of PD-L1 expression (combined positive score ≥1) was 94.3 % with similar PD-L1 expression rates between betel-nut-exposed and non-exposed groups. PD-L1 prevalence did not differ in those who received prior first-or second-line systemic therapy. In summary, betel-nut exposure did not notably affect PD-L1 expression rates in R/M HNSCC patients in Taiwan.


Subject(s)
Areca , B7-H1 Antigen , Head and Neck Neoplasms , Neoplasm Recurrence, Local , Squamous Cell Carcinoma of Head and Neck , Humans , B7-H1 Antigen/metabolism , Male , Areca/adverse effects , Female , Middle Aged , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Prospective Studies , Aged , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/epidemiology , Biomarkers, Tumor/metabolism , Adult , Taiwan/epidemiology , Mastication , Prevalence , Neoplasm Metastasis
2.
Gynecol Oncol ; 155(2): 275-279, 2019 11.
Article in English | MEDLINE | ID: mdl-31481248

ABSTRACT

OBJECTIVE: Clinical genetic testing to diagnose germline mutations often requires blood sample or saliva smear from a cancer-affected individual. This rules out testing in families when cancer-affected individuals are deceased. We explored the use of a next-generation sequencing (NGS) platform to diagnose germline pathogenic mutations from tumors. METHODS: Archival tumors (ovarian = 26, breast = 25, others = 9) were retrieved from 60 cancer patients who have undergone multi-gene panel blood testing. Genomic DNA was extracted and sequenced for BRCA1/2 using a NGS platform. 41/60 specimens were sequenced for 5 other genes (APC, ATM, PALB2, PTEN, TP53). Tumor testing and results interpretation were performed blinded to the blood test result. RESULTS: All 38 patients with no BRCA1/2 mutations on blood testing were correctly tested negative on tumor. Tumor testing correctly diagnosed BRCA1/2 pathogenic mutations in 15/22 (68%) patients while in 7/22 (32%) patients, the mutation was either detected but incorrectly classified as VUS (n = 3) or not detected at all (n = 4). Overall concordance rate for tumor and blood testing for BRCA1/2 mutations was 88%, with 0% false positive and 32% false negative rate for pathogenic mutations. Tumor testing correctly diagnosed 1/2 pathogenic germline ATM mutation, 1/1 pathogenic germline PALB2 mutation and 2/2 pathogenic germline TP53 mutations. False positive germline mutations were diagnosed in 4 genes at a rate of 2.4%-10.3% (APC = 2.4%, PALB2 = 2.4%, PTEN = 4.9%, TP53 = 10.3%). CONCLUSION: Tumor testing for BRCA1/2 germline mutations using an NGS platform is fairly reliable with no false positive findings, and correctly diagnosed more than two-thirds of pathogenic germline BRCA1/2 mutations. However, it is not reliable to diagnose pathogenic germline mutations in genes frequently mutated in sporadic cancers, such as PTEN and TP53.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1/physiology , Genes, BRCA2/physiology , Germ-Line Mutation/genetics , Ovarian Neoplasms/genetics , Adult , Aged , Breast Neoplasms/diagnosis , DNA, Neoplasm/genetics , False Positive Reactions , Female , Humans , Middle Aged , Ovarian Neoplasms/diagnosis , Prospective Studies , Sequence Analysis, DNA , Tumor Suppressor Protein p53/metabolism
3.
Sci Transl Med ; 9(412)2017 Oct 18.
Article in English | MEDLINE | ID: mdl-29046434

ABSTRACT

Many traditional pharmacopeias include Aristolochia and related plants, which contain nephrotoxins and mutagens in the form of aristolochic acids and similar compounds (collectively, AA). AA is implicated in multiple cancer types, sometimes with very high mutational burdens, especially in upper tract urothelial cancers (UTUCs). AA-associated kidney failure and UTUCs are prevalent in Taiwan, but AA's role in hepatocellular carcinomas (HCCs) there remains unexplored. Therefore, we sequenced the whole exomes of 98 HCCs from two hospitals in Taiwan and found that 78% showed the distinctive mutational signature of AA exposure, accounting for most of the nonsilent mutations in known cancer driver genes. We then searched for the AA signature in 1400 HCCs from diverse geographic regions. Consistent with exposure through known herbal medicines, 47% of Chinese HCCs showed the signature, albeit with lower mutation loads than in Taiwan. In addition, 29% of HCCs from Southeast Asia showed the signature. The AA signature was also detected in 13 and 2.7% of HCCs from Korea and Japan as well as in 4.8 and 1.7% of HCCs from North America and Europe, respectively, excluding one U.S. hospital where 22% of 87 "Asian" HCCs had the signature. Thus, AA exposure is geographically widespread. Asia, especially Taiwan, appears to be much more extensively affected, which is consistent with other evidence of patterns of AA exposure. We propose that additional measures aimed at primary prevention through avoidance of AA exposure and investigation of possible approaches to secondary prevention are warranted.


Subject(s)
Aristolochic Acids/therapeutic use , Liver Neoplasms/drug therapy , Asia , Geography , Humans , Immunotherapy , Liver Neoplasms/genetics , Mutagenesis/genetics , Mutation/genetics , Taiwan
4.
Genome Res ; 27(9): 1475-1486, 2017 09.
Article in English | MEDLINE | ID: mdl-28739859

ABSTRACT

Aflatoxin B1 (AFB1) is a mutagen and IARC (International Agency for Research on Cancer) Group 1 carcinogen that causes hepatocellular carcinoma (HCC). Here, we present the first whole-genome data on the mutational signatures of AFB1 exposure from a total of >40,000 mutations in four experimental systems: two different human cell lines, in liver tumors in wild-type mice, and in mice that carried a hepatitis B surface antigen transgene-this to model the multiplicative effects of aflatoxin exposure and hepatitis B in causing HCC. AFB1 mutational signatures from all four experimental systems were remarkably similar. We integrated the experimental mutational signatures with data from newly sequenced HCCs from Qidong County, China, a region of well-studied aflatoxin exposure. This indicated that COSMIC mutational signature 24, previously hypothesized to stem from aflatoxin exposure, indeed likely represents AFB1 exposure, possibly combined with other exposures. Among published somatic mutation data, we found evidence of AFB1 exposure in 0.7% of HCCs treated in North America, 1% of HCCs from Japan, but 16% of HCCs from Hong Kong. Thus, aflatoxin exposure apparently remains a substantial public health issue in some areas. This aspect of our study exemplifies the promise of future widespread resequencing of tumor genomes in providing new insights into the contribution of mutagenic exposures to cancer incidence.


Subject(s)
Aflatoxin B1/toxicity , Carcinogens/toxicity , DNA Mutational Analysis , Mutation/drug effects , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , China , Hepatitis B Surface Antigens/genetics , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mutation/genetics
5.
Sci Transl Med ; 9(378)2017 02 22.
Article in English | MEDLINE | ID: mdl-28228601

ABSTRACT

Trithorax-like group complex containing KDM6A acts antagonistically to Polycomb-repressive complex 2 (PRC2) containing EZH2 in maintaining the dynamics of the repression and activation of gene expression through H3K27 methylation. In urothelial bladder carcinoma, KDM6A (a H3K27 demethylase) is frequently mutated, but its functional consequences and therapeutic targetability remain unknown. About 70% of KDM6A mutations resulted in a total loss of expression and a consequent loss of demethylase function in this cancer type. Further transcriptome analysis found multiple deregulated pathways, especially PRC2/EZH2, in KDM6A-mutated urothelial bladder carcinoma. Chromatin immunoprecipitation sequencing analysis revealed enrichment of H3K27me3 at specific loci in KDM6A-null cells, including PRC2/EZH2 and their downstream targets. Consequently, we targeted EZH2 (an H3K27 methylase) and demonstrated that KDM6A-null urothelial bladder carcinoma cell lines were sensitive to EZH2 inhibition. Loss- and gain-of-function assays confirmed that cells with loss of KDM6A are vulnerable to EZH2. IGFBP3, a direct KDM6A/EZH2/H3K27me3 target, was up-regulated by EZH2 inhibition and contributed to the observed EZH2-dependent growth suppression in KDM6A-null cell lines. EZH2 inhibition delayed tumor onset in KDM6A-null cells and caused regression of KDM6A-null bladder tumors in both patient-derived and cell line xenograft models. In summary, our study demonstrates that inactivating mutations of KDM6A, which are common in urothelial bladder carcinoma, are potentially targetable by inhibiting EZH2.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone Demethylases/metabolism , Nuclear Proteins/metabolism , Polycomb Repressive Complex 2/metabolism , Transcription, Genetic , Urinary Bladder Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mice, Nude , Models, Biological , Neoplasm Invasiveness , Urinary Bladder Neoplasms/pathology , Urothelium/pathology
6.
Anticancer Res ; 35(12): 6639-53, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26637880

ABSTRACT

BACKGROUND/AIM: Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is a rare autosomal dominant disorder characterized by fumarate hydratase (FH) gene mutation. It is associated with the development of very aggressive kidney tumors, characterized by early onset and high metastatic potential, and has no effective therapy. The aim of the study was to establish a new preclinical platform for investigating morphogenetic and metabolic features, and alternative therapy of metastatic hereditary papillary renal cell carcinoma type 2 (PRCC2). MATERIALS AND METHODS: Fresh cells were collected from pleural fluid of a patient with metastatic hereditary PRCC2. Morphogenetic and functional characteristics were evaluated via microscopy, FH gene sequencing analysis, real-time polymerase chaine reaction and enzymatic activity measurement. We performed bioenergetic analysis, gene-expression profiling, and cell viability assay with 19 anti-neoplastic drugs. RESULTS: We established a new in vitro model of hereditary PRCC2 - the NCCFH1 cell line. The cell line possesses a c.1162 delA - p.Thr375fs frameshift mutation in the FH gene. Our findings indicate severe attenuation of oxidative phosphorylation and glucose-dependent growth of NCCFH1 cells that is consistent with the Warburg effect. Furthermore, gene-expression profiling identified that the most prominent molecular features reflected a high level of apoptosis, cell adhesion, and cell signaling. Drug screening revealed a marked sensitivity of FH(-/-) cells to mitoxantrone, epirubicin, topotecan and a high sensitivity to bortezomib. CONCLUSION: We demonstrated that the NCCFH1 cell line is a very interesting preclinical model for studying the metabolic features and testing new therapies for hereditary PRCC2, while bortezomib may be a potential efficient therapeutic option.


Subject(s)
Carcinoma, Renal Cell/genetics , Fumarate Hydratase/metabolism , Adolescent , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Humans , In Vitro Techniques , Male
7.
Genome Med ; 7(1): 38, 2015.
Article in English | MEDLINE | ID: mdl-26015808

ABSTRACT

BACKGROUND: Aristolochic acid (AA) is a natural compound found in many plants of the Aristolochia genus, and these plants are widely used in traditional medicines for numerous conditions and for weight loss. Previous work has connected AA-mutagenesis to upper-tract urothelial cell carcinomas and hepatocellular carcinomas. We hypothesize that AA may also contribute to bladder cancer. METHODS: Here, we investigated the involvement of AA-mutagenesis in bladder cancer by sequencing bladder tumor genomes from two patients with known exposure to AA. After detecting strong mutational signatures of AA exposure in these tumors, we exome-sequenced and analyzed an additional 11 bladder tumors and analyzed publicly available somatic mutation data from a further 336 bladder tumors. RESULTS: The somatic mutations in the bladder tumors from the two patients with known AA exposure showed overwhelming AA signatures. We also detected evidence of AA exposure in 1 out of 11 bladder tumors from Singapore and in 3 out of 99 bladder tumors from China. In addition, 1 out of 194 bladder tumors from North America showed a pattern of mutations that might have resulted from exposure to an unknown mutagen with a heretofore undescribed pattern of A > T mutations. Besides the signature of AA exposure, the bladder tumors also showed the CpG > TpG and activated-APOBEC signatures, which have been previously reported in bladder cancer. CONCLUSIONS: This study demonstrates the utility of inferring mutagenic exposures from somatic mutation spectra. Moreover, AA exposure in bladder cancer appears to be more pervasive in the East, where traditional herbal medicine is more widely used. More broadly, our results suggest that AA exposure is more extensive than previously thought both in terms of populations at risk and in terms of types of cancers involved. This appears to be an important public health issue that should be addressed by further investigation and by primary prevention through regulation and education. In addition to opportunities for primary prevention, knowledge of AA exposure would provide opportunities for secondary prevention in the form of intensified screening of patients with known or suspected AA exposure.

8.
J Clin Endocrinol Metab ; 100(2): E360-4, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25387265

ABSTRACT

CONTEXT: Cell division cycle 73 (CDC73), encoding the protein parafibromin, is the most prevalent mutated gene in familial and sporadic parathyroid carcinoma (PC). OBJECTIVE: To identify additional genetic abnormalities in PCs. DESIGN: Whole-exome sequencing was performed using DNA from seven pairs of matched PCs and one triplet containing double primary tumor and normal leukocyte. Somatic variants were confirmed using Sanger sequencing and recurrently mutated genes were assessed in 13 additional PCs as well as 40 parathyroid adenomas (PA). RESULTS: PC had an average of 51 somatic variants/tumor (range 3-176) with approximately 58% of variants occurring as nonsynonymous single nucleotide variants. The importance of CDC73 in PC is reinforced with a remarkable preferential amplification of the mutant CDC73 allele. Furthermore, recurrent germ line and somatic mutations in prune homolog 2 [Drosophila] (PRUNE2) were found in PC and computationally predicted to be deleterious; in addition, recurrent mutations in kinase genes related to cell migration and invasion were found. PRUNE2 showed recurrent mutations in 18% (4/22) of PCs with additional screening in 40 PAs revealing only one rare missense polymorphism (Asp1677Asn). For the first time, the mutational signature associated with apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC)-catalyzed cytosine-to-uracil deamination is found in a subset of PC. CONCLUSION: This study outlines the genetic landscape of PC and attempts to characterize the mutational processes shaping the PC genome.


Subject(s)
Carcinoma/genetics , Cell Movement/genetics , Mutation , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Parathyroid Neoplasms/genetics , Adolescent , Adult , Aged , Carcinoma/metabolism , Carcinoma/pathology , DNA Mutational Analysis , Exome , Female , Humans , Male , Middle Aged , Mutagenesis , Neoplasm Invasiveness/pathology , Neoplasm Proteins/metabolism , Parathyroid Neoplasms/metabolism , Parathyroid Neoplasms/pathology , Young Adult
9.
Genome Med ; 6(3): 24, 2014.
Article in English | MEDLINE | ID: mdl-25031618

ABSTRACT

Exposure to environmental mutagens is an important cause of human cancer, and measures to reduce mutagenic and carcinogenic exposures have been highly successful at controlling cancer. Until recently, it has been possible to connect the chemical characteristics of mutagens to actual mutations observed in human tumors only indirectly. Now, next-generation sequencing technology enables us to observe in detail the DNA-sequence-level effects of well-known mutagens, such as ultraviolet radiation and tobacco smoke, as well as endogenous mutagenic processes, such as those involving activated DNA cytidine deaminases (APOBECs). We can also observe the effects of less well-known but potent mutagens, including those recently found to be present in some herbal remedies. Crucially, we can now tease apart the superimposed effects of several mutational exposures and processes and determine which ones occurred during the development of individual tumors. Here, we review advances in detecting these mutation signatures and discuss the implications for surveillance and prevention of cancer. The number of sequenced tumors from diverse cancer types and multiple geographic regions is growing explosively, and the genomes of these tumors will bear the signatures of even more diverse mutagenic exposures. Thus, we envision development of wide-ranging compendia of mutation signatures from tumors and a concerted effort to experimentally elucidate the signatures of a large number of mutagens. This information will be used to link signatures observed in tumors to the exposures responsible for them, which will offer unprecedented opportunities for prevention.

10.
Sci Transl Med ; 5(197): 197ra101, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23926199

ABSTRACT

Aristolochic acid (AA), a natural product of Aristolochia plants found in herbal remedies and health supplements, is a group 1 carcinogen that can cause nephrotoxicity and upper urinary tract urothelial cell carcinoma (UTUC). Whole-genome and exome analysis of nine AA-associated UTUCs revealed a strikingly high somatic mutation rate (150 mutations/Mb), exceeding smoking-associated lung cancer (8 mutations/Mb) and ultraviolet radiation-associated melanoma (111 mutations/Mb). The AA-UTUC mutational signature was characterized by A:T to T:A transversions at the sequence motif A[C|T]AGG, located primarily on nontranscribed strands. AA-induced mutations were also significantly enriched at splice sites, suggesting a role for splice-site mutations in UTUC pathogenesis. RNA sequencing of AA-UTUC confirmed a general up-regulation of nonsense-mediated decay machinery components and aberrant splicing events associated with splice-site mutations. We observed a high frequency of somatic mutations in chromatin modifiers, particularly KDM6A, in AA-UTUC, demonstrated the sufficiency of AA to induce renal dysplasia in mice, and reproduced the AA mutational signature in experimentally treated human renal tubular cells. Finally, exploring other malignancies that were not known to be associated with AA, we screened 93 hepatocellular carcinoma genomes/exomes and identified AA-like mutational signatures in 11. Our study highlights an unusual genome-wide AA mutational signature and the potential use of mutation signatures as "molecular fingerprints" for interrogating high-throughput cancer genome data to infer previous carcinogen exposures.


Subject(s)
Aristolochic Acids/adverse effects , Carcinogens/analysis , Genome, Human/genetics , Mutation/drug effects , Mutation/genetics , Animals , Aristolochic Acids/analysis , Carcinogens/toxicity , Cell Line, Tumor , Humans , Kidney Diseases/chemically induced , Mice , Mice, Inbred C57BL , Mutagens/analysis , Mutagens/toxicity , Neoplasms/genetics , RNA Splicing/genetics , Urologic Neoplasms/genetics , Urothelium/pathology
11.
Cancer Discov ; 2(7): 591-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22705984

ABSTRACT

UNLABELLED: The molecular pathogenesis of natural killer/T-cell lymphoma (NKTCL) is not well understood. We conducted whole-exome sequencing and identified Janus kinase 3 (JAK3) somatic-activating mutations (A572V and A573V) in 2 of 4 patients with NKTCLs. Further validation of the prevalence of JAK3 mutations was determined by Sanger sequencing and high-resolution melt (HRM) analysis in an additional 61 cases. In total, 23 of 65 (35.4%) cases harbored JAK3 mutations. Functional characterization of the JAK3 mutations support its involvement in cytokine-independent JAK/STAT constitutive activation leading to increased cell growth. Moreover, treatment of both JAK3-mutant and wild-type NKTCL cell lines with a novel pan-JAK inhibitor, CP-690550, resulted in dose-dependent reduction of phosphorylated STAT5, reduced cell viability, and increased apoptosis. Hence, targeting the deregulated JAK/STAT pathway could be a promising therapy for patients with NKTCLs. SIGNIFICANCE: Gene mutations causing NKTCL have not been fully identified. Through exome sequencing, we identified activating mutations of JAK3 that may play a significant role in the pathogenesis of NKTCLs. Our findings have important implications for the management of patients with NKTCLs.


Subject(s)
Janus Kinase 3/genetics , Lymphoma, T-Cell/genetics , Mutation , Natural Killer T-Cells/metabolism , Adult , Aged , Aged, 80 and over , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation , DNA Mutational Analysis , Enzyme Activation/genetics , Female , Humans , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Male , Middle Aged , Natural Killer T-Cells/pathology , Phosphorylation , Piperidines , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA Interference , STAT5 Transcription Factor/metabolism
12.
Nat Genet ; 44(5): 570-4, 2012 May.
Article in English | MEDLINE | ID: mdl-22484628

ABSTRACT

Gastric cancer is a major cause of global cancer mortality. We surveyed the spectrum of somatic alterations in gastric cancer by sequencing the exomes of 15 gastric adenocarcinomas and their matched normal DNAs. Frequently mutated genes in the adenocarcinomas included TP53 (11/15 tumors), PIK3CA (3/15) and ARID1A (3/15). Cell adhesion was the most enriched biological pathway among the frequently mutated genes. A prevalence screening confirmed mutations in FAT4, a cadherin family gene, in 5% of gastric cancers (6/110) and FAT4 genomic deletions in 4% (3/83) of gastric tumors. Frequent mutations in chromatin remodeling genes (ARID1A, MLL3 and MLL) also occurred in 47% of the gastric cancers. We detected ARID1A mutations in 8% of tumors (9/110), which were associated with concurrent PIK3CA mutations and microsatellite instability. In functional assays, we observed both FAT4 and ARID1A to exert tumor-suppressor activity. Somatic inactivation of FAT4 and ARID1A may thus be key tumorigenic events in a subset of gastric cancers.


Subject(s)
Adenocarcinoma/genetics , Cell Adhesion/genetics , Chromatin Assembly and Disassembly/genetics , Exome/genetics , Genes, Tumor Suppressor , Mutation/genetics , Stomach Neoplasms/genetics , Case-Control Studies , DNA/genetics , Gastric Mucosa/metabolism , Humans , Microsatellite Instability , Polymerase Chain Reaction , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA
13.
Biosci Biotechnol Biochem ; 75(4): 723-31, 2011.
Article in English | MEDLINE | ID: mdl-21512251

ABSTRACT

Cordycepin, a pure compound of Cordyceps sinensis (CS), is known as an adenosine analog. We have found that CS stimulated Leydig cell steroidogenesis. Here we investigated the in vivo and in vitro effects of cordycepin in primary mouse Leydig cell steroidogenesis. The results indicate that cordycepin increased the plasma testosterone concentration. Cordycepin also stimulated in vitro mouse Leydig cell testosterone production in dose- and time-dependent manners. We further observed that cordycepin regulated the mRNA expression of the A1, A2a, A2b, and A3 adenosine receptors in the mouse Leydig cells, and that antagonists of A1, A2a, and A3 suppressed testosterone production 20-50% testosterone production. Furthermore, Rp-cAMPS (cAMP antagonist) and Protein Kinase A (PKA) inhibitors (H89 and PKI) significantly decreased cordycepin-induced testosterone production, indicating that the PKA-cAMP signal pathway was activated by cordycepin through adenosine receptors. Moreover, cordycepin induced StAR protein expression, and H89 suppressed cordycepin-induced steroidogenic acute regulatory (StAR) protein expression. Conclusively, cordycepin associated with adenosine receptors to activate cAMP-PKA-StAR pathway and steroidogenesis in the mouse Leydig cells.


Subject(s)
Deoxyadenosines/pharmacology , Leydig Cells/drug effects , Leydig Cells/metabolism , Steroids/biosynthesis , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Leydig Cells/cytology , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Purinergic P1 Receptor Antagonists/pharmacology , Receptors, Purinergic P1/genetics , Signal Transduction/drug effects , Testosterone/blood
14.
Mol Endocrinol ; 25(2): 327-38, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21193558

ABSTRACT

GnRH-II enhances ovarian cancer cell invasion in an autocrine manner. We have now found that GnRH-II increases 37-kDa laminin receptor precursor (LRP) production in GnRH receptor (GnRHR)-positive OVCAR-3 and CaOV-3 ovarian cancer cells, while small interfering RNA (siRNA)-mediated depletion of GnRH-II or GnRHR mRNA abrogates this. The invasiveness of ovarian cancer cells is also reduced >85% by siRNA-mediated knockdown of LRP levels and >50% by pretreatment of Matrigel with a synthetic peptide that blocks interactions between laminin and the 67-kDa nonintegrin laminin receptor which comprises two LRP subunits. Conversely, overexpressing LRP in CaOV-3 cells increases their invasiveness 5-fold, while overexpressing LRP with a nonfunctional laminin-binding site does not. Depletion of LRP by siRNA treatment reduces CaOV-3 cell attachment to laminin-coated plates by ∼80% but only reduces their binding to Matrigel by ∼20%. Thus, while LRP influences CaOV-3 cell adhesion to laminin, LRP must act in other ways to enhance invasion. Matrix metalloproteinases (MMPs) are key mediators of invasion, and LRP siRNA treatment of OVCAR-3 and CaOV-3 cells inhibits MMP-2 but not MMP-9 mRNA levels. Overexpressing LRP in these cells increases MMP-2 production specifically, while a laminin-binding deficient LRP does not. Importantly, LRP siRNA treatment abolishes GnRH-II-induced MMP-2 production, and invasion in OVCAR-3 and CaOV-3 cells, which was also seen after MMP-2 siRNA treatment. These results suggest that GnRH-II-induced LRP expression increases the amount of the 67-kDa nonintegrin laminin receptor, which appears to interact with laminin in the extracellular matrix to promote MMP-2 expression and enhance ovarian cancer cell invasion.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Matrix Metalloproteinase 2/genetics , Ovarian Neoplasms/metabolism , Receptors, Laminin/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Collagen/metabolism , Drug Combinations , Female , Gene Expression , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Humans , Laminin/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Precursors/genetics , Protein Precursors/metabolism , Proteoglycans/metabolism , RNA, Messenger , RNA, Small Interfering , Receptors, Laminin/genetics
15.
Reprod Biol Endocrinol ; 7: 115, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19849856

ABSTRACT

BACKGROUND: Obesity has been linked to an increased risk of female infertility. Leptin, an adipocytokine which is elevated during obesity, may influence gonadal function through modulating steroidogenesis in granulosa cells. METHODS: The effect of leptin on progesterone production in simian virus 40 immortalized granulosa (SVOG) cells was examined by Enzyme linked immunosorbent assay (ELISA). The effect of leptin on the expression of the steroidogenic enzymes (StAR, P450scc, 3betaHSD) in SVOG cells was examined by real-time PCR and Western blotting. The mRNA expression of leptin receptor isoforms in SVOG cells were examined by using PCR. SVOG cells were co-treated with leptin and specific pharmacological inhibitors to identify the signaling pathways involved in leptin-reduced progesterone production. Silencing RNA against leptin receptor was used to determine that the inhibition of leptin on cAMP-induced steroidogenesis acts in a leptin receptor-dependent manner. RESULTS AND CONCLUSION: In the present study, we investigated the cellular mechanisms underlying leptin-regulated steroidogenesis in human granulosa cells. We show that leptin inhibits 8-bromo cAMP-stimulated progesterone production in a concentration-dependent manner. Furthermore, we show that leptin inhibits expression of the cAMP-stimulated steroidogenic acute regulatory (StAR) protein, the rate limiting de novo protein in progesterone synthesis. Leptin induces the activation of ERK1/2, p38 and JNK but only the ERK1/2 (PD98059) and p38 (SB203580) inhibitors attenuate the leptin-induced inhibition of cAMP-stimulated StAR protein expression and progesterone production. These data suggest that the leptin-induced MAPK signal transduction pathway interferes with cAMP/PKA-stimulated steroidogenesis in human granulosa cells. Moreover, siRNA mediated knock-down of the endogenous leptin receptor attenuates the effect of leptin on cAMP-induced StAR protein expression and progesterone production, suggesting that the effect of leptin on steroidogenesis in granulosa cells is receptor dependent. In summary, leptin acts through the MAPK pathway to downregulate cAMP-induced StAR protein expression and progesterone production in immortalized human granulosa cells. These results suggest a possible mechanism by which gonadal steroidogenesis could be suppressed in obese women.


Subject(s)
Cyclic AMP/pharmacology , Granulosa Cells/drug effects , Signal Transduction/drug effects , Steroids/biosynthesis , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cells, Cultured , Cyclic AMP/metabolism , Female , Granulosa Cells/metabolism , Humans , Leptin/metabolism , Leptin/pharmacology , Leptin/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoproteins/metabolism , Progesterone/metabolism , Receptors, Leptin/metabolism , Receptors, Leptin/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Mol Endocrinol ; 23(10): 1646-56, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19608641

ABSTRACT

GnRH-II modulates ovarian cancer cells invasion and is expressed in normal ovary and ovarian epithelial cancer cells; however, the upstream regulator(s) of GnRH-II expression in these cells remains unclear. We now demonstrate that epidermal growth factor (EGF) increases GnRH-II mRNA levels in several human ovarian carcinoma cell lines and up-regulates GnRH-II promoter activity in OVCAR-3 cells in a dose-dependent manner, whereas an EGF receptor inhibitor (AG148) abolishes EGF-induced increases in GnRH-II promoter activity and GnRH-II mRNA levels. EGF increases the phosphorylation of cAMP-responsive element-binding protein (p-CREB) and its association with the coregulator, CCAAT/enhancer binding protein beta, whereas blocking the EGF-induced ERK1/2 phosphorylation with MAPK inhibitors (PD98059/U0126) markedly reduced these effects. Moreover, depletion of CREB using small interfering RNA attenuated EGF-induced GnRH-II promoter activity. Chromatin immunoprecipitation assays demonstrated that EGF induces p-CREB binding to a cAMP responsive-element within the GnRH-II promoter, likely in association with CCAAT/enhancer binding protein beta, and mutagenesis of this cAMP responsive-element prevented EGF-induced GnRH-II promoter activity in OVCAR-3 cells. Importantly, GnRH-II acts additively with EGF to promote invasion of OVCAR-3 and CaOV-3 cells, but not SKOV-3 cells that express low levels of GnRH receptor (GnRHR). Treatment with GnRHR small interfering RNA also partially inhibited the EGF-induced invasion of OVCAR-3 and CaOV-3 cells. Furthermore, EGF treatment transiently increases GnRHR levels in OVCAR-3 and CaOV-3, which likely accentuates the effects of increase GnRH-II production on cell invasion. These results provide evidence that EGF is an upstream regulator of the autocrine actions of GnRH-II on the invasive properties of ovarian cancer cells.


Subject(s)
Epidermal Growth Factor/pharmacology , Gonadotropin-Releasing Hormone/analogs & derivatives , Ovarian Neoplasms/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropin-Releasing Hormone/genetics , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Neoplasm Invasiveness , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Phosphorylation/drug effects , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics
17.
Biol Reprod ; 81(2): 243-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19357364

ABSTRACT

Gonadotropin-releasing hormone (GNRH) activates the progesterone receptor (PGR) in pituitary cells and accentuates gonadotropin expression. We show that GNRH1 increases Fshb mRNA levels in LbetaT2 mouse pituitary cells within 8 h and is three times more effective than GNRH2. By contrast, GNRH1 and GNRH2 do not affect Lhb gene expression in these cells. Within the same time frame, small interfering RNA (siRNA) knockdown of the PGR in LbetaT2 cells reduced GNRH1 activation of a PGR response element (PRE)-driven luciferase reporter gene and Fshb mRNA levels by >50%. Chromatin immunoprecipitation (ChIP) assays also demonstrated that PGR loading on the PRE within the Fshb gene promoter in LbetaT2 cells occurred within 8 h after GNRH1 treatment and was lost by 24 h. While the GNRH1-induced upregulation of the PRE reporter gene and Fshb mRNA levels was attenuated by cotreatment with protein kinase A (H-89) and protein kinase C (GF109203X) inhibitors, only GF109203X inhibited PGR phosphorylation at Ser249 in LbetaT2 cells. Immunoprecipitation assays also showed a progressive increase in the interaction between the PGR and its coactivator NCOA3 that peaked at 8 h coincident with the increase in Fshb mRNA after GNRH1 treatment. The siRNA-mediated knockdown of NCOA3 in LbetaT2 cells also reduced Fshb mRNA levels after GNRH1 treatment and loading of NCOA3 on the Fshb promoter PRE in a ChIP assay. We conclude that the rapid effect of GNRH1 on Fshb expression in LbetaT2 cells is mediated by PGR phosphorylation and loading at the PRE within the Fshb promoter together with NCOA3.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/genetics , Gonadotropin-Releasing Hormone/metabolism , Pituitary Gland/metabolism , Receptors, Progesterone/physiology , AMP-Activated Protein Kinases/antagonists & inhibitors , Analysis of Variance , Animals , Cell Line , Chromatin Immunoprecipitation , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Genes, Reporter , Gonadotropin-Releasing Hormone/administration & dosage , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Mice , Nuclear Receptor Coactivator 3/genetics , Nuclear Receptor Coactivator 3/metabolism , Phosphorylation , Pituitary Gland/cytology , Progesterone/administration & dosage , Protein Binding , Protein Isoforms , Protein Kinase C/antagonists & inhibitors , Protein Processing, Post-Translational , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Response Elements , Time Factors , Up-Regulation
18.
Endocr Relat Cancer ; 16(1): 179-88, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19022848

ABSTRACT

In addition to their critical roles in folliculogenesis and ovarian granulosa cell steroidogenesis, gonadotropins have been implicated as potential risk factors in ovarian epithelial carcinomas, most of which are derived from ovarian surface epithelium (OSE). However, the molecular mechanism underlying the effects of FSH and LH in OSE and its neoplastic counterpart is not well understood. We previously demonstrated that gonadotropins promote the growth of OSE cells by regulating the levels of epidermal growth factor receptor (EGFR) via the activation of ERK1/2 and PI3K pathways in immortalized human OSE (IOSE) cells. In this study, we investigated whether cAMP and its novel binding target, named exchange protein activated by cAMP (Epac), are involved in the gonadotropin-induced EGFR expression in OSE cells. Gonadotropins elevated intracellular cAMP levels in both IOSE and granulosa cells, and this increase was attenuated by SQ22536, an inhibitor of adenylyl cyclase (AC). The activation of the ERK1/2 and Akt pathways as well as the expression of EGFR was stimulated by reagents that elevate intracellular cAMP levels, via cAMP analog 8-bromo-cAMP and AC activator forskolin. A similar increase was observed when the cells were treated with a novel cAMP analog, 8-(4-chlorophenylthio)-2'-O-methyl adenosine-3',5'-cyclic monophosphate (8-CPT-2ME-cAMP), which activates Epac specifically but not PKA. Moreover, the gonadotropin-induced EGFR expression and ERK1/2 and Akt activation were abolished by overexpression of dominant negative Epac. Taken together, these results indicate that the AC/cAMP/Epac signaling pathway may mediate the up-regulation of EGFR by gonadotropins via ERK1/2 and Akt activation.


Subject(s)
Epithelial Cells/cytology , ErbB Receptors/metabolism , Follicle Stimulating Hormone/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Luteinizing Hormone/metabolism , Ovarian Follicle/cytology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Epithelial Cells/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Humans , Luteinizing Hormone/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
19.
FEBS J ; 275(22): 5496-511, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18959739

ABSTRACT

The hypothalamic decapeptide gonadotropin-releasing hormone (GnRH) is well known for its role in the control of pituitary gonadotropin secretion, but the hormone and receptor are also expressed in extrapituitary tissues and tumor cells, including epithelial ovarian cancers. It is hypothesized that they may function as a local autocrine regulatory system in nonpituitary contexts. Numerous studies have demonstrated a direct antiproliferative effect on ovarian cancer cell lines of GnRH and its synthetic analogs. This effect appears to be attributable to multiple steps in the GnRH signaling cascade, such as cell cycle arrest at G(0)/G(1). In contrast to GnRH signaling in pituitary gonadotropes, the involvement of G(alpha q), protein kinase C and mitogen-activated protein kinases is less apparent in neoplastic cells. Instead, in ovarian cancer cells, GnRH receptors appear to couple to the pertussis toxin-sensitive protein G(alpha i), leading to the activation of protein phosphatase, which in turn interferes with growth factor-induced mitogenic signals. Apoptotic involvement is still controversial, although GnRH analogs have been shown to protect cancer cells from doxorubicin-induced apoptosis. Recently, data supporting a regulatory role of GnRH analogs in ovarian cancer cell migration/invasion have started to emerge. In this minireview, we summarize the current understanding of the antiproliferative actions of GnRH analogs, as well as the recent observations of GnRH effects on ovarian cancer cell apoptosis and motogenesis. The molecular mechanisms that mediate GnRH actions and the clinical applications of GnRH analogs in ovarian cancer patients are also discussed.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Ovarian Neoplasms/pathology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , Female , Gonadotropin-Releasing Hormone/physiology , Humans , Ovarian Neoplasms/drug therapy
20.
Asian J Androl ; 10(6): 929-36, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18958357

ABSTRACT

AIM: To study the effect and mechanism of gonadotrophin-releasing hormone (GnRH) on murine Leydig cell steroidogenesis. METHODS: Purified murine Leydig cells were treated with GnRH-I and -II agonists, and testosterone production and steroidogenic enzyme expressions were determined. RESULTS: GnRH-I and -II agonists significantly stimulated murine Leydig cell steroidogenesis 60%-80% in a dose- and time-dependent manner (P < 0.05). The mRNA expressions of steroidogenic acute regulatory (StAR) protein, P450scc, 3beta-hydroxysteroid dehydrogenase (HSD), but not 17alpha-hydroxylase or 17beta-HSD, were significantly stimulated by both GnRH agonists with a 1.5- to 3-fold increase (P < 0.05). However, only 3beta-HSD protein expression was induced by both GnRH agonists, with a 1.6- to 2-fold increase (P < 0.05). CONCLUSION: GnRH directly stimulated murine Leydig cell steroidogenesis by activating 3b-HSD enzyme expression.


Subject(s)
Gonadotropin-Releasing Hormone/pharmacology , Leydig Cells/metabolism , Sexual Maturation/physiology , Steroids/biosynthesis , 3-Hydroxysteroid Dehydrogenases/biosynthesis , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Blotting, Western , Cell Separation , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Dose-Response Relationship, Drug , Gonadotropin-Releasing Hormone/agonists , Leydig Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...