1
|
Beevors LI, Sundar S, Foster PA. Steroid metabolism and hormonal dynamics in normal and malignant ovaries. Essays Biochem 2024:EBC20240028. [PMID: 38994724 DOI: 10.1042/ebc20240028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
The ovaries are key steroid hormone production sites in post-pubertal females. However, current research on steroidogenic enzymes, endogenous hormone concentrations and their effects on healthy ovarian function and malignant development is limited. Here, we discuss the importance of steroid enzymes in normal and malignant ovaries, alongside hormone concentrations, receptor expression and action. Key enzymes include STS, 3β-HSD2, HSD17B1, ARK1C3, and aromatase, which influence ovarian steroidal action. Both androgen and oestrogen action, via their facilitating enzyme, drives ovarian follicle activation, development and maturation in healthy ovarian tissue. In ovarian cancer, some data suggest STS and oestrogen receptor α may be linked to aggressive forms, while various oestrogen-responsive factors may be involved in ovarian cancer metastasis. In contrast, androgen receptor expression and action vary across ovarian cancer subtypes. For future studies investigating steroidogenesis and steroidal activity in ovarian cancer, it is necessary to differentiate between disease subtypes for a comprehensive understanding.
Collapse
Affiliation(s)
- Lucy I Beevors
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, U.K
| | - Sudha Sundar
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, U.K
| | - Paul A Foster
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, U.K
- Centre for Diabetes, Endocrinology, and Metabolism, Birmingham Health Partners, Birmingham, U.K
| |
Collapse
|
2
|
Tan Y, Li Y, Ren L, Fu H, Li Q, Liu S. Integrative proteome and metabolome analyses reveal molecular basis underlying growth and nutrient composition in the Pacific oyster, Crassostrea gigas. J Proteomics 2024; 290:105021. [PMID: 37838097 DOI: 10.1016/j.jprot.2023.105021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
In order to comprehend the molecular basis of growth, nutrient composition, and color pigmentation in oysters, comparative proteome and metabolome analyses of two selectively bred oyster strains with contrasting growth rate and shell color were used in this study. A total of 289 proteins and 224 metabolites were identified differentially expressed between the two strains. We identified a series of specifically enriched functional clusters implicated in protein biosynthesis (RPL4, MRPS7, and CARS), fatty acid metabolism (ACSL5, PEX3, ACOXI, CPTIA, FABP6, and HSD17B12), energy metabolism (FH, PPP1R7, CLAM2, and RGN), cell proliferation (MYB, NFYC, DOHH, TOP2a, SMARCA5, and SMARCC2), material transport (ABCB1, ABCB8, VPS16, and VPS33a), and pigmentation (RDH7, RDH13, Retsat, COX15, and Cyp3a9). Integrated proteome and metabolome analyses indicate that fast-growing strain utilize energy-efficient mechanisms of ATP generation while promoting protein and polyunsaturated fatty acid synthesis, activating the cell cycle to increase cell proliferation and thus promoting their biomass increase. These results uncovered molecular mechanisms underlying growth regulation, nutrition quality, and pigmentation and provided candidate biomarkers for molecular breeding in oysters. SIGNIFICANCE: Rapid growth has always been the primary breeding objective to increase the production profits of Pacific oyster (Crassostrea gigas), while favorable nutritional quality and beautiful color add commercial value. In recent years, proteomic and metabolomic techniques have been widely used in marine organisms, although these techniques are seldom utilized to study oyster growth and development. In this study, two C. gigas strains with contrasted phenotypes in growth and shell color provided an ideal model for unraveling the molecular basis of growth and nutrient composition through a comparison of the proteome and metabolome. Since proteins and metabolites are the critical undertakers and the end products of cellular regulatory processes, identifying the differentially expressed proteins and metabolites would allow for discovering biomarkers and pathways that were implicated in cell growth, proliferation, and other critical functions. This work provides valuable resources in assistance with molecular breeding of oyster strains with superior production traits of fast-growth and high-quality nutrient value.
Collapse
Affiliation(s)
- Ying Tan
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Yongjing Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Liting Ren
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Huiru Fu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Qi Li
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China
| | - Shikai Liu
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, and College of Fisheries, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
3
|
Pedernera E, Morales-Vásquez F, Gómora MJ, Almaraz MA, Mena E, Pérez-Montiel D, Rendon E, López-Basave H, Maldonado-Cubas J, Méndez C. 17β-hydroxysteroid dehydrogenase type 1 improves survival in serous epithelial ovarian tumors. Endocr Connect 2023; 12:e230315. [PMID: 37924640 PMCID: PMC10762561 DOI: 10.1530/ec-23-0315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023]
Abstract
The incidence of ovarian cancer has been epidemiologically related to female reproductive events and hormone replacement therapy after menopause. This highlights the importance of evaluating the role of sexual steroid hormones in ovarian cancer by the expression of enzymes related to steroid hormone biosynthesis in the tumor cells. This study was aimed to evaluate the presence of 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1), aromatase and estrogen receptor alpha (ERα) in the tumor cells and their association with the overall survival in 111 patients diagnosed with primary ovarian tumors. Positive immunoreactivity for 17β-HSD1 was observed in 74% of the tumors. In the same samples, aromatase and ERα revealed 66% and 47% positivity, respectively. No association was observed of 17β-HSD1 expression with the histological subtypes and clinical stages of the tumor. The overall survival of patients was improved in 17β-HSD1-positive group in Kaplan-Meier analysis (P = 0.028), and 17β-HSD1 expression had a protective effect from multivariate proportional regression evaluation (HR = 0.44; 95% CI 0.24-0.9; P = 0.040). The improved survival was observed in serous epithelial tumors but not in nonserous ovarian tumors. The expression of 17β-HSD1 in the cells of the serous epithelial ovarian tumors was associated with an improved overall survival, whereas aromatase and ERα were not related to a better survival. The evaluation of hazard risk factors demonstrated that age and clinical stage showed worse prognosis, and 17β-HSD1 expression displayed a protective effect with a better survival outcome in patients of epithelial ovarian tumors.
Collapse
Affiliation(s)
- Enrique Pedernera
- Universidad Nacional Autónoma de México, Facultad de Medicina, Departamento de Embriología y Genética, Ciudad de México, México
| | | | - María J Gómora
- Universidad Nacional Autónoma de México, Facultad de Medicina, Departamento de Embriología y Genética, Ciudad de México, México
| | - Miguel A Almaraz
- Universidad Nacional Autónoma de México, Facultad de Medicina, Departamento de Embriología y Genética, Ciudad de México, México
| | - Esteban Mena
- Universidad Nacional Autónoma de México, Facultad de Medicina, Secretaría General, Ciudad de México, México
- Universidad La Salle, Posgrado de la Facultad de Ciencias Químicas, Ciudad de México, México
| | | | - Elizabeth Rendon
- Hospital Militar de Especialidades de la Mujer y Neonatología. Ciudad de México, México
| | | | - Juan Maldonado-Cubas
- Universidad La Salle, Posgrado de la Facultad de Ciencias Químicas, Ciudad de México, México
| | - Carmen Méndez
- Universidad Nacional Autónoma de México, Facultad de Medicina, Departamento de Embriología y Genética, Ciudad de México, México
| |
Collapse
|
4
|
Clavijo-Buriticá DC, Sosa CC, Heredia RC, Mosquera AJ, Álvarez A, Medina J, Quimbaya M. Use of Arabidopsis thaliana as a model to understand specific carcinogenic events: Comparison of the molecular machinery associated with cancer-hallmarks in plants and humans. Heliyon 2023; 9:e15367. [PMID: 37101642 PMCID: PMC10123165 DOI: 10.1016/j.heliyon.2023.e15367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/28/2023] Open
Abstract
Model organisms are fundamental in cancer research given that they rise the possibility to characterize in a quantitative-objective fashion the organisms as a whole in ways that are infeasible in humans. From this perspective, model organisms with short generation times and established protocols for genetic manipulation allow the understanding of basic biology principles that might guide carcinogenic onset. The cancer-hallmarks (CHs) approach, a modular perspective for cancer understanding, stands that underlying the variability among different cancer types, critical events support the carcinogenic origin and progression. Thus, CHs as interconnected genetic circuitry, have a causal effect over cancer biogenesis and might represent a comparison scaffold among model organisms to identify and characterize evolutionarily conserved modules to understand cancer. Nevertheless, the identification of novel cancer regulators by comparative genomics approaches relies on selecting specific biological processes or related signaling cascades that limit the type of detected regulators, even more, holistic analysis from a systemic perspective is absent. Similarly, although the plant Arabidopsis thaliana has been used as a model organism to dissect specific disease-associated mechanisms, given the evolutionary distance between plants and humans, a general concern about the utility of using A. thaliana as a cancer model persists. In the present research, we take advantage of the CHs paradigm as a framework to establish a functional systemic comparison between plants and humans, that allowed the identification not only of specific novel key genetic regulators, but also, biological processes, metabolic systems, and genetic modules that might contribute to the neoplastic transformation. We propose five cancer-hallmarks that overlapped in conserved mechanisms and processes between Arabidopsis and human and thus, represent mechanisms which study can be prioritized in A. thaliana as an alternative model for cancer research. Additionally, derived from network analyses and machine learning strategies, a new set of potential candidate genes that might contribute to neoplastic transformation is described. These findings postulate A. thaliana as a suitable model to dissect, not all, but specific cancer properties, highlighting the importance of using alternative complementary models to understand carcinogenesis.
Collapse
Affiliation(s)
| | - Chrystian C. Sosa
- Pontificia Universidad Javeriana Cali, Department of Natural Sciences and Mathematics, Cali, Colombia
- Grupo de Investigación en Evolución, Ecología y Conservación EECO, Programa de Biología, Facultad de Ciencias Básicas y Tecnologías, Universidad del Quindío, Armenia, Colombia
| | - Rafael Cárdenas Heredia
- Pontificia Universidad Javeriana Cali, Department of Natural Sciences and Mathematics, Cali, Colombia
| | - Arlen James Mosquera
- Pontificia Universidad Javeriana Cali, Department of Natural Sciences and Mathematics, Cali, Colombia
| | - Andrés Álvarez
- Pontificia Universidad Javeriana Cali, Department of Natural Sciences and Mathematics, Cali, Colombia
| | - Jan Medina
- Pontificia Universidad Javeriana Cali, Department of Natural Sciences and Mathematics, Cali, Colombia
| | - Mauricio Quimbaya
- Pontificia Universidad Javeriana Cali, Department of Natural Sciences and Mathematics, Cali, Colombia
- Corresponding author.
| |
Collapse
|
5
|
Estradiol and Estrone Have Different Biological Functions to Induce NF-κB-Driven Inflammation, EMT and Stemness in ER+ Cancer Cells. Int J Mol Sci 2023; 24:ijms24021221. [PMID: 36674737 PMCID: PMC9865376 DOI: 10.3390/ijms24021221] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
In general, the risk of being diagnosed with cancer increases with age; however, the development of estrogen-receptor-positive (ER+) cancer types in women are more closely related to menopausal status than age. In fact, the general risk factors for cancer development, such as obesity-induced inflammation, show differences in their association with ER+ cancer risk in pre- and postmenopausal women. Here, we tested the role of the principal estrogens in the bloodstream before and after menopause, estradiol (E2) and estrone (E1), respectively, on inflammation, epithelial-to-mesenchymal transition (EMT) and cancer stem cell enrichment in the human ER+ cervical cancer cell line HeLa. Our results demonstrate that E1, contrary to E2, is pro-inflammatory, increases embryonic stem-transcription factors (ES-TFs) expression and induces EMT in ER+ HeLa cells. Moreover, we observed that high intratumoural expression levels of 17β-Hydroxysteroid dehydrogenase (HSD17B) isoforms involved in E1 synthesis is a poor prognosis factor, while overexpression of E2-synthetizing HSD17B isoforms is associated with a better outcome, for patients diagnosed with ER+ ovarian and uterine corpus carcinomas. This work demonstrates that E1 and E2 have different biological functions in ER+ gynaecologic cancers. These results open a new line of research in the study of ER+ cancer subtypes, highlighting the potential key oncogenic role of E1 and HSD17B E1-synthesizing enzymes in the development and progression of these diseases.
Collapse
|
6
|
Moon YA. Emerging roles of polyunsaturated fatty acid synthesis pathway in colorectal cancer. Anim Cells Syst (Seoul) 2023; 27:61-71. [PMID: 36970499 PMCID: PMC10035963 DOI: 10.1080/19768354.2023.2189933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
The development of colorectal cancer typically involves the accumulated influences of genetic alterations, medical issues, lifestyle, and diet. Dietary fatty acids appear to affect the tumorigenesis and progression of colorectal cancer. Despite conflicting results, the current consensus on the effects of very long-chain polyunsaturated fatty acids on colorectal cancer is that low levels of eicosapentaenoic acid and docosahexaenoic acid, and high levels of arachidonic acid are associated with an increased risk of colorectal cancer. Altered levels of arachidonic acid in membrane phospholipids can change the levels of prostaglandin E2, which affect the biological activities of cancer cells in multiple stages. Arachidonic acid and other very long-chain polyunsaturated fatty acids can affect tumorigenesis in prostaglandin E2-independent manners as well, including stabilization of β-catenine, ferroptosis, ROS generation, regulation of transcription factors, and de novo lipogenesis. Recent studies have revealed an association between the activities of enzymes synthesizing very long-chain polyunsaturated fatty acids and tumorigenesis and cancer progression, although the mechanisms are still unknown. In this study, PUFA effects on tumorigenesis, the endogenous very long-chain polyunsaturated fatty acid synthesis pathway, metabolites of arachidonic acid and their effects on tumorigenesis and progression of CRC, and current knowledge that supports the association of the enzymes involved in the polyunsaturated fatty acid synthesis pathway with colorectal cancer tumorigenesis and progression are reviewed.
Collapse
Affiliation(s)
- Young-Ah Moon
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea
- Young-Ah Moon Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea
| |
Collapse
|
7
|
Giordo R, Gulsha R, Kalla S, Calin GA, Lipovich L. LncRNA-Associated Genetic Etiologies Are Shared between Type 2 Diabetes and Cancers in the UAE Population. Cancers (Basel) 2022; 14:3313. [PMID: 35884374 PMCID: PMC9313416 DOI: 10.3390/cancers14143313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/22/2022] [Accepted: 05/06/2022] [Indexed: 12/13/2022] Open
Abstract
Numerous epidemiological studies place patients with T2D at a higher risk for cancer. Many risk factors, such as obesity, ageing, poor diet and low physical activity, are shared between T2D and cancer; however, the biological mechanisms linking the two diseases remain largely unknown. The advent of genome wide association studies (GWAS) revealed large numbers of genetic variants associated with both T2D and cancer. Most significant disease-associated variants reside in non-coding regions of the genome. Several studies show that single nucleotide polymorphisms (SNPs) at or near long non-coding RNA (lncRNA) genes may impact the susceptibility to T2D and cancer. Therefore, the identification of genetic variants predisposing individuals to both T2D and cancer may help explain the increased risk of cancer in T2D patients. We aim to investigate whether lncRNA genetic variants with significant diabetes and cancer associations overlap in the UAE population. We first performed an annotation-based analysis of UAE T2D GWAS, confirming the high prevalence of variants at or near non-coding RNA genes. We then explored whether these T2D SNPs in lncRNAs were relevant to cancer. We highlighted six non-coding genetic variants, jointly reaching statistical significance in T2D and cancer, implicating a shared genetic architecture between the two diseases in the UAE population.
Collapse
Affiliation(s)
- Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (R.G.); (R.G.); (S.K.)
| | - Rida Gulsha
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (R.G.); (R.G.); (S.K.)
| | - Sarah Kalla
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (R.G.); (R.G.); (S.K.)
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Leonard Lipovich
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (R.G.); (R.G.); (S.K.)
| |
Collapse
|
8
|
Pavlič R, Gjorgoska M, Rižner TL. Model Cell Lines and Tissues of Different HGSOC Subtypes Differ in Local Estrogen Biosynthesis. Cancers (Basel) 2022; 14:cancers14112583. [PMID: 35681563 PMCID: PMC9179372 DOI: 10.3390/cancers14112583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Ovarian cancer (OC) comprises a heterogeneous group of hormone-dependent diseases with very high mortality. Estrogens have been shown to promote the progression of OC; however, their exact role in OC subtypes remains unknown. Here, we investigated the local estrogen biosynthesis in OC. We performed targeted transcriptomics and estrogen metabolism analyses in high-grade serous OC (HGSOC) cell lines that differed in chemoresistance status and compared these data with publicly available transcriptome and proteome data for HGSOC tissues. In HGSOC cells, estrogen metabolism decreased with increasing chemoresistance. In highly chemoresistant cells and platinum-resistant HGSOC tissues, HSD17B14 expression was increased. Proteome data showed differential levels of HSD17B10, SULT1E1, CYP1B1, and NQO1 between the four HGSOC subtypes. Our results confirm that estrogen biosynthesis differs between different HGSOC cell models and possibly between different HGSOC subtypes. Such differentially expressed enzymes have potential as targets in the search of new treatment options. Abstract Ovarian cancer (OC) is highly lethal and heterogeneous. Several hormones are involved in OC etiology including estrogens; however, their role in OC is not completely understood. Here, we performed targeted transcriptomics and estrogen metabolism analyses in high-grade serous OC (HGSOC), OVSAHO, Kuramochi, COV632, and immortalized normal ovarian epithelial HIO-80 cells. We compared these data with public transcriptome and proteome data for the HGSOC tissues. In all model systems, high steroid sulfatase expression and weak/undetected aromatase (CYP19A1) expression indicated the formation of estrogens from the precursor estrone-sulfate (E1-S). In OC cells, the metabolism of E1-S to estradiol was the highest in OVSAHO, followed by Kuramochi and COV362 cells, and decreased with increasing chemoresistance. In addition, higher HSD17B14 and CYP1A2 expressions were observed in highly chemoresistant COV362 cells and platinum-resistant tissues compared to those in HIO-80 cells and platinum-sensitive tissues. The HGSOC cell models differed in HSD17B10, CYP1B1, and NQO1 expression. Proteomic data also showed different levels of HSD17B10, CYP1B1, NQO1, and SULT1E1 between the four HGSOC subtypes. These results suggest that different HGSOC subtypes form different levels of estrogens and their metabolites and that the estrogen-biosynthesis-associated targets should be further studied for the development of personalized treatment.
Collapse
|
9
|
Lu M, Feng R, Qin Y, Deng H, Lian B, Yin C, Xiao Y. Identifying Environmental Endocrine Disruptors Associated With the Age at Menarche by Integrating a Transcriptome-Wide Association Study With Chemical-Gene-Interaction Analysis. Front Endocrinol (Lausanne) 2022; 13:836527. [PMID: 35282430 PMCID: PMC8907571 DOI: 10.3389/fendo.2022.836527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022] Open
Abstract
Menarche is the first occurrence of menstrual bleeding and one of the most important events of female puberty. Alarmingly, over the last several decades, the mean age at menarche (AAM) has decreased. Environmental endocrine disruptors (EEDs) are chemicals that may interfere with the endocrine system, resulting in adverse developmental, immunological, neurological, and reproductive effects in humans. Thus, the effects of EEDs on fertility and reproduction are growing concerns in modern societies. In this study, we aimed to determine the influence of genetic and environmental factors on AAM. We used data from an AAM genome-wide association study of 329,345 women to conduct a transcriptome-wide association study (TWAS) with FUSION software. As references, we determined the gene-expression levels in the hypothalamus, pituitary gland, ovaries, uterus, and whole blood. We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses using the significantly dysregulated genes identified by the TWAS. Using the STRING database, we also generated a protein-protein-interaction network to analyze common AAM-specific genes identified by the TWAS with different tissues. We performed chemical-related gene set enrichment analysis (CGSEA) and identified significant TWAS genes to uncover relationships between different chemicals and AAM. The TWAS identified 9,848 genes; among these, 1580 genes were significant (P < 0.05), and 11 genes were significant among the hypothalamus, pituitary, ovary, uterus, and whole blood. CGSEA identified 1,634 chemicals, including 120 chemicals significantly correlated with AAM. In summary, we performed a TWAS (for genetic factors) and CGSEA (for environmental factors) focusing on AAM and identified several AAM-associated genes and EEDs. The results of this study expand our understanding of genetic and environmental factors related to the onset of female puberty.
Collapse
Affiliation(s)
- Mengnan Lu
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Ruoyang Feng
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiao Tong University, Xi'an, China
| | - Yujie Qin
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Hongyang Deng
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Biyao Lian
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| |
Collapse
|
10
|
Joshi H, Vastrad B, Joshi N, Vastrad C, Tengli A, Kotturshetti I. Identification of Key Pathways and Genes in Obesity Using Bioinformatics Analysis and Molecular Docking Studies. Front Endocrinol (Lausanne) 2021; 12:628907. [PMID: 34248836 PMCID: PMC8264660 DOI: 10.3389/fendo.2021.628907] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Obesity is an excess accumulation of body fat. Its progression rate has remained high in recent years. Therefore, the aim of this study was to diagnose important differentially expressed genes (DEGs) associated in its development, which may be used as novel biomarkers or potential therapeutic targets for obesity. The gene expression profile of E-MTAB-6728 was downloaded from the database. After screening DEGs in each ArrayExpress dataset, we further used the robust rank aggregation method to diagnose 876 significant DEGs including 438 up regulated and 438 down regulated genes. Functional enrichment analysis was performed. These DEGs were shown to be significantly enriched in different obesity related pathways and GO functions. Then protein-protein interaction network, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. The module analysis was performed based on the whole PPI network. We finally filtered out STAT3, CORO1C, SERPINH1, MVP, ITGB5, PCM1, SIRT1, EEF1G, PTEN and RPS2 hub genes. Hub genes were validated by ICH analysis, receiver operating curve (ROC) analysis and RT-PCR. Finally a molecular docking study was performed to find small drug molecules. The robust DEGs linked with the development of obesity were screened through the expression profile, and integrated bioinformatics analysis was conducted. Our study provides reliable molecular biomarkers for screening and diagnosis, prognosis as well as novel therapeutic targets for obesity.
Collapse
Affiliation(s)
- Harish Joshi
- Department of Endocrinology, Endocrine and Diabetes Care Center, Hubbali, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, India
| | - Nidhi Joshi
- Department of Medicine, Dr. D. Y. Patil Medical College, Kolhapur, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, India
- *Correspondence: Chanabasayya Vastrad,
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, India
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, India
| |
Collapse
|
11
|
Lin Y, Meng Y, Zhang J, Ma L, Jiang L, Zhang Y, Yuan M, Ren A, Zhu W, Li S, Shu Y, Du M, Zhu L. Functional genetic variant of HSD17B12 in the fatty acid biosynthesis pathway predicts the outcome of colorectal cancer. J Cell Mol Med 2020; 24:14160-14170. [PMID: 33118286 PMCID: PMC7754038 DOI: 10.1111/jcmm.16026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 12/24/2022] Open
Abstract
Fatty acids are involved in the development and progression of colorectal cancer (CRC). However, genetic effects of fatty acid biosynthesis pathway on CRC outcome are unclear. Cox regression model was used to evaluate genetic effects on CRC overall survival (OS) and progression‐free survival (PFS), accompanied by calculating hazard ratios (HRs) and confidence intervals (CIs). Differential expression analysis, expression quantitative trait loci analysis, dual‐luciferase reporter assay and chromatin immunoprecipitation assay were performed to explore the genetically biological mechanism. The rs10838164 C>T in HSD17B12 was significantly associated with an increased risk of death and progression of CRC (OS, HR = 2.12, 95% CI = 1.40‐3.22, P = 4.03 × 10−4; PFS, HR = 1.64, 95% CI = 1.11‐2.44, P = 1.35 × 10−2), of which T allele could increase HSD17B12 expression (P = 1.78 × 10−11). Subsequently, the functional experiments indicated that rs10838164 T allele could not only enhance the binding affinity of transcription factor YY1 to HSD17B12 region harbouring rs10838164 but also promote the transcriptional activity of HSD17B12, which was significantly up‐regulated in colorectal tumour tissues. Our findings suggest that genetic variants in fatty acid biosynthesis pathway play an important role in CRC outcome.
Collapse
Affiliation(s)
- Yu Lin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yixuan Meng
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jinying Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lu Jiang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming Yuan
- Department of Oncology, The Jiangyin People's Hospital, Wuxi, China
| | - Anjing Ren
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weiyou Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuwei Li
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mulong Du
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Oncology, The Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Heikelä H, Ruohonen ST, Adam M, Viitanen R, Liljenbäck H, Eskola O, Gabriel M, Mairinoja L, Pessia A, Velagapudi V, Roivainen A, Zhang FP, Strauss L, Poutanen M. Hydroxysteroid (17β) dehydrogenase 12 is essential for metabolic homeostasis in adult mice. Am J Physiol Endocrinol Metab 2020; 319:E494-E508. [PMID: 32691632 DOI: 10.1152/ajpendo.00042.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hydroxysteroid 17β dehydrogenase 12 (HSD17B12) is suggested to be involved in the elongation of very long chain fatty acids. Previously, we have shown a pivotal role for the enzyme during mouse development. In the present study we generated a conditional Hsd17b12 knockout (HSD17B12cKO) mouse model by breeding mice homozygous for a floxed Hsd17b12 allele with mice expressing the tamoxifen-inducible Cre recombinase at the ROSA26 locus. Gene inactivation was induced by administering tamoxifen to adult mice. The gene inactivation led to a 20% loss of body weight within 6 days, associated with drastic reduction in both white (83% males, 75% females) and brown (65% males, 60% females) fat, likely due to markedly reduced food and water intake. Furthermore, the knockout mice showed sickness behavior and signs of liver toxicity, specifically microvesicular hepatic steatosis and increased serum alanine aminotransferase (4.6-fold in males, 7.7-fold in females). The hepatic changes were more pronounced in females than males. Proinflammatory cytokines, such as interleukin-6 (IL-6), IL-17, and granulocyte colony-stimulating factor, were increased in the HSD17B12cKO mice indicating an inflammatory response. Serum lipidomics study showed an increase in the amount of dihydroceramides, despite the dramatic overall loss of lipids. In line with the proposed role for HSD17B12 in fatty acid elongation, we observed accumulation of ceramides, dihydroceramides, hexosylceramides, and lactosylceramides with shorter than 18-carbon fatty acid side chains in the serum. The results indicate that HSD17B12 is essential for proper lipid homeostasis and HSD17B12 deficiency rapidly results in fatal systemic inflammation and lipolysis in adult mice.
Collapse
Affiliation(s)
- Hanna Heikelä
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Marion Adam
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Heidi Liljenbäck
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku PET Centre, University of Turku, Turku, Finland
| | - Olli Eskola
- Turku PET Centre, University of Turku, Turku, Finland
| | - Michael Gabriel
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura Mairinoja
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Alberto Pessia
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Vidya Velagapudi
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Anne Roivainen
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Fu-Ping Zhang
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Leena Strauss
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Poutanen
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Internal Medicine, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
13
|
Tsachaki M, Strauss P, Dunkel A, Navrátilová H, Mladenovic N, Odermatt A. Impact of 17β-HSD12, the 3-ketoacyl-CoA reductase of long-chain fatty acid synthesis, on breast cancer cell proliferation and migration. Cell Mol Life Sci 2020; 77:1153-1175. [PMID: 31302749 PMCID: PMC7109200 DOI: 10.1007/s00018-019-03227-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 07/02/2019] [Accepted: 07/09/2019] [Indexed: 12/21/2022]
Abstract
Metabolic reprogramming of tumor cells involves upregulation of fatty acid (FA) synthesis to support high bioenergetic demands and membrane synthesis. This has been shown for cytosolic synthesis of FAs with up to 16 carbon atoms. Synthesis of long-chain fatty acids (LCFAs), including ω-6 and ω-3 polyunsaturated FAs, takes place at the endoplasmic reticulum. Despite increasing evidence for an important role of LCFAs in cancer, the impact of their synthesis in cancer cell growth has scarcely been studied. Here, we demonstrated that silencing of 17β-hydroxysteroid dehydrogenase type 12 (17β-HSD12), essentially catalyzing the 3-ketoacyl-CoA reduction step in LCFA production, modulates proliferation and migration of breast cancer cells in a cell line-dependent manner. Increased proliferation and migration after 17β-HSD12 knockdown were partly mediated by metabolism of arachidonic acid towards COX2 and CYP1B1-derived eicosanoids. Decreased proliferation was rescued by increased glucose concentration and was preceded by reduced ATP production through oxidative phosphorylation and spare respiratory capacity. In addition, 17β-HSD12 silencing was accompanied by alterations in unfolded protein response, including a decrease in CHOP expression and increase in eIF2α activation and the folding chaperone ERp44. Our study highlights the significance of LCFA biosynthesis for tumor cell physiology and unveils unknown aspects of breast cancer cell heterogeneity.
Collapse
Affiliation(s)
- Maria Tsachaki
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Pirmin Strauss
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Anja Dunkel
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Hana Navrátilová
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05, Hradec Kralove, Czech Republic
| | - Natasa Mladenovic
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
14
|
Nadaraja S, Schledermann D, Herrstedt J, Østrup O, Ditzel HJ. ARAP1 is an independent prognostic biomarker in older women with ovarian high-grade serous adenocarcinoma receiving first-line platinum-based antineoplastic therapy. Acta Oncol 2020; 59:40-47. [PMID: 31478407 DOI: 10.1080/0284186x.2019.1657941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Little is known about the biological factors influencing ovarian cancer (OC) patient outcome, especially in older patients who are often underrepresented in clinical trials. We examined alterations in the transcriptomic profile of primary high-grade serous carcinoma (HGSC) samples from older OC patients (>70 years) receiving first-line platinum-based treatment to identify potential biomarkers for prediction of response to this therapy.Material and methods: Tumor samples from 50 HGSC patients were identified from a retrospective cohort, analyzed by gene expression array. The protein expression of selected biomarkers was examined using immunohistochemistry (IHC).Results: Gene expression profiling revealed 81 genes with significantly altered expression in patients experiencing progression after first-line platinum-based treatment within 6 months versus those who progressed later than 12 months. Expression of ankyrin repeat and PH domain 1 (ARAP1) was significantly lower in the group with early versus late progression (p ≤ .01). Correlation between ARAP1 expression and outcome was further confirmed by IHC staining in the discovery cohort (χ2-test, p = .004) and in independent validation cohorts. The sensitivity of ARAP1 allowed identification of 64.7% of patients with early progression in the discovery population, with a specificity of 78.6% and a negative predictive value of 78.6%. Multivariate regression analysis identified ARAP1 as an independent prognostic factor.Conclusions: This hypothesis generating study suggests that low expression of ARAP1 is an independent prognostic biomarker of shorter RFS in older patients with HGSC receiving first-line platinum-based antineoplastic therapy, which could be used to identify patients who should receive more intensive treatment and closer surveillance.
Collapse
Affiliation(s)
- Sambavy Nadaraja
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | - Doris Schledermann
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Jørn Herrstedt
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
- Department of Clinical Oncology, Zealand University Hospital, Roskilde, Denmark
| | - Olga Østrup
- Center for Genomic Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - Henrik J. Ditzel
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
15
|
Kouba S, Ouldamer L, Garcia C, Fontaine D, Chantome A, Vandier C, Goupille C, Potier-Cartereau M. Lipid metabolism and Calcium signaling in epithelial ovarian cancer. Cell Calcium 2019; 81:38-50. [PMID: 31200184 DOI: 10.1016/j.ceca.2019.06.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 02/06/2023]
Abstract
Epithelial Ovarian cancer (EOC) is the deadliest gynecologic malignancy and represents the fifth leading cause of all cancer-related deaths in women. The majority of patients are diagnosed at an advanced stage of the disease that has spread beyond the ovaries to the peritoneum or to distant organs (stage FIGO III-IV) with a 5-year overall survival of about 29%. Consequently, it is necessary to understand the pathogenesis of this disease. Among the factors that contribute to cancer development, lipids and ion channels have been described to be associated to cancerous diseases particularly in breast, colorectal and prostate cancers. Here, we reviewed the literature data to determine how lipids or lipid metabolites may influence EOC risk or progression. We also highlighted the role and the expression of the calcium (Ca2+) and calcium-activated potassium (KCa) channels in EOC and how lipids might regulate them. Although lipids and some subclasses of nutritional lipids may be associated to EOC risk, lipid metabolism of LPA (lysophosphatidic acid) and AA (arachidonic acid) emerges as an important signaling network in EOC. Clinical data showed that they are found at high concentrations in EOC patients and in vitro and in vivo studies referred to them as triggers of the Ca2+entry in the cancer cells inducing their proliferation, migration or drug resistance. The cross-talk between lipid mediators and Ca2+ and/or KCa channels needs to be elucidated in EOC in order to facilitate the understanding of its outcomes and potentially suggest novel therapeutic strategies including treatment and prevention.
Collapse
Affiliation(s)
- Sana Kouba
- Université de Tours, INSERM, N2C UMR 1069, Faculté de Médecine, Tours, France; Réseau Molécules Marines, Métabolisme et Cancer du Cancéropôle Grand Ouest, France
| | - Lobna Ouldamer
- Université de Tours, INSERM, N2C UMR 1069, Faculté de Médecine, Tours, France; Université de Tours, INSERM, N2C UMR 1069, CHRU de Tours, Service de gynécologie et d'obstétrique, Tours, France
| | - Céline Garcia
- Université de Tours, INSERM, N2C UMR 1069, Faculté de Médecine, Tours, France; Réseau Molécules Marines, Métabolisme et Cancer du Cancéropôle Grand Ouest, France
| | - Delphine Fontaine
- Université de Tours, INSERM, N2C UMR 1069, Faculté de Médecine, Tours, France; Réseau Molécules Marines, Métabolisme et Cancer du Cancéropôle Grand Ouest, France
| | - Aurélie Chantome
- Réseau Molécules Marines, Métabolisme et Cancer du Cancéropôle Grand Ouest, France; Université de Tours, INSERM, N2C UMR 1069, Faculté de Pharmacie, Tours, France
| | - Christophe Vandier
- Université de Tours, INSERM, N2C UMR 1069, Faculté de Médecine, Tours, France; Réseau Molécules Marines, Métabolisme et Cancer du Cancéropôle Grand Ouest, France
| | - Caroline Goupille
- Réseau CASTOR du Cancéropôle Grand Ouest, France; Université de Tours, INSERM, N2C UMR 1069, CHRU de Tours, Faculté de Médecine, Tours, France
| | - Marie Potier-Cartereau
- Université de Tours, INSERM, N2C UMR 1069, Faculté de Médecine, Tours, France; Réseau Molécules Marines, Métabolisme et Cancer du Cancéropôle Grand Ouest, France.
| |
Collapse
|
16
|
Tsachaki M, Odermatt A. Subcellular localization and membrane topology of 17β-hydroxysteroid dehydrogenases. Mol Cell Endocrinol 2019; 489:98-106. [PMID: 30864548 DOI: 10.1016/j.mce.2018.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 06/18/2018] [Accepted: 07/03/2018] [Indexed: 01/09/2023]
Abstract
The 17β-hydroxysteroid dehydrogenases (17β-HSDs) comprise enzymes initially identified by their ability to interconvert active and inactive forms of sex steroids, a vital process for the tissue-specific control of estrogen and androgen balance. However, most 17β-HSDs have now been shown to accept substrates other than sex steroids, including bile acids, retinoids and fatty acids, thereby playing unanticipated roles in cell physiology. This functional divergence is often reflected by their different subcellular localization, with 17β-HSDs found in the cytosol, peroxisome, mitochondria, endoplasmic reticulum and in lipid droplets. Moreover, a subset of 17β-HSDs are integral membrane proteins, with their specific topology dictating the cellular compartment in which they exert their enzymatic activity. Here, we summarize the present knowledge on the subcellular localization and membrane topology of the 17β-HSD enzymes and discuss the correlation with their biological functions.
Collapse
Affiliation(s)
- Maria Tsachaki
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
17
|
Zhang Y, Wu J, Jing H, Huang G, Dong J, Cui Z. Increased DHRS12 expression independently predicts poor survival in patients with high-grade serous ovarian cancer. Future Oncol 2018; 14:2579-2588. [PMID: 29783891 DOI: 10.2217/fon-2018-0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To explore the expression profile of some DHRS genes in high-grade serous ovarian cancer (SOVC) and to study their prognostic values. PATIENTS & METHODS A retrospective bioinformatic analysis was performed using data in the Gene Expression Omnibus, the Human Protein Atlas and the Cancer Genome Atlas-Ovarian Cancer. RESULTS Increased DHRS12 expression was an independent indicator of poor overall survival (hazard ratio [HR]: 1.265, 95% CI: 1.075-1.488; p = 0.005) and recurrence-free survival (RFS; HR: 2.242, 95%CI: 1.464-3.432; p < 0.001) in patients with high-grade SOVC. DNA deletion was associated with decreased DHRS12 expression, as well as the best overall survival and RFS among the three copy number alteration groups. CONCLUSION DHRS12 might serve as a valuable prognostic biomarker in high-grade SOVC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng, 475000, PR China
| | - Jiang Wu
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng, 475000, PR China
| | - Hong Jing
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng, 475000, PR China
| | - Gui Huang
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng, 475000, PR China
| | - Jinlong Dong
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng, 475000, PR China
| | - Zhanjun Cui
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, PR China
| |
Collapse
|
18
|
Yang Y, Fang X, Yang R, Yu H, Jiang P, Sun B, Zhao Z. MiR-152 Regulates Apoptosis and Triglyceride Production in MECs via Targeting ACAA2 and HSD17B12 Genes. Sci Rep 2018; 8:417. [PMID: 29323178 PMCID: PMC5765104 DOI: 10.1038/s41598-017-18804-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/18/2017] [Indexed: 01/11/2023] Open
Abstract
Mammary epithelial cells (MECs) affect milk production capacity during lactation and are critical for the maintenance of tissue homeostasis. Our previous studies have revealed that the expression of miR-152 was increased significantly in MECs of cows with high milk production. In the present study, bioinformatics analysis identified ACAA2 and HSD17B12 as the potential targets of miR-152, which were further validated by dual-luciferase repoter assay. In addition, the expressions of miR-152 was shown to be negatively correlated with levels of mRNA and protein of ACAA2, HSD17B12 genes by qPCR and western bot analysis. Furthermore, transfection with miR-152 significantly up-regulated triglyceride production, promoted proliferation and inhibited apoptosis in MECs. Furthermore, overexpression of ACAA2 and HSD17B12 could inhibit triglyceride production, cells proliferation and induce apoptosis; but sh234-ACAA2-181/sh234-HSD17B12-474 could reverse the trend. These findings suggested that miR-152 could significantly influence triglyceride production and suppress apoptosis, possibly via the expression of target genes ACAA2 and HSD17B12.
Collapse
Affiliation(s)
- Yuwei Yang
- College of Animal Science, Jilin University, Xi An Road 5333, Changchun, Jilin, 130062, P.R. China
| | - Xibi Fang
- College of Animal Science, Jilin University, Xi An Road 5333, Changchun, Jilin, 130062, P.R. China
| | - Runjun Yang
- College of Animal Science, Jilin University, Xi An Road 5333, Changchun, Jilin, 130062, P.R. China
| | - Haibin Yu
- College of Animal Science, Jilin University, Xi An Road 5333, Changchun, Jilin, 130062, P.R. China
| | - Ping Jiang
- College of Animal Science, Jilin University, Xi An Road 5333, Changchun, Jilin, 130062, P.R. China
| | - Boxing Sun
- College of Animal Science, Jilin University, Xi An Road 5333, Changchun, Jilin, 130062, P.R. China.
| | - Zhihui Zhao
- Agricultural College, Guangdong Ocean University, Zhanjiang, 524088, China.
| |
Collapse
|
19
|
Szajnik M, Czystowska-Kuźmicz M, Elishaev E, Whiteside TL. Biological markers of prognosis, response to therapy and outcome in ovarian carcinoma. Expert Rev Mol Diagn 2016; 16:811-26. [PMID: 27268121 DOI: 10.1080/14737159.2016.1194758] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Ovarian cancer (OvCa) is among the most common types of cancer and is the leading cause of death from gynecological malignancies in western countries. Cancer biomarkers have a potential for improving the management of OvCa patients at every point from screening and detection, diagnosis, prognosis, follow up, response to therapy and outcome. AREAS COVERED The literature search has indicated a number of candidate biomarkers have recently emerged that could facilitate the molecular definition of OvCa, providing information about prognosis and predicting response to therapy. These potentially promising biomarkers include immune cells and their products, tumor-derived exosomes, nucleic acids and epigenetic biomarkers. Expert commentary: Although most of the biomarkers available today require prospective validation, the development of noninvasive liquid biopsy-based monitoring promises to improve their utility for evaluations of prognosis, response to therapy and outcome in OvCa.
Collapse
Affiliation(s)
- Marta Szajnik
- a Department of Gynecology and Gynecologic Oncology , Military Institute of Medicine , Warsaw , Poland.,b Department of Immunology, Centre of Biostructure Research , Medical University of Warsaw , Warsaw , Poland
| | | | - Esther Elishaev
- c Department of Pathology , University of Pittsburgh, School of Medicine , Pittsburgh , PA , USA
| | - Theresa L Whiteside
- c Department of Pathology , University of Pittsburgh, School of Medicine , Pittsburgh , PA , USA.,d University of Pittsburgh Cancer Institute , Pittsburgh , PA , USA
| |
Collapse
|
20
|
Lu J, Chu P, Wang H, Jin Y, Han S, Han W, Tai J, Guo Y, Ni X. Candidate Gene Association Analysis of Neuroblastoma in Chinese Children Strengthens the Role of LMO1. PLoS One 2015; 10:e0127856. [PMID: 26030754 PMCID: PMC4452511 DOI: 10.1371/journal.pone.0127856] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/21/2015] [Indexed: 11/25/2022] Open
Abstract
Neuroblastoma (NB) is the most common extra-cranial solid tumor in children and the most frequently diagnosed cancer in the first year of life. Previous genome-wide association studies (GWAS) of Caucasian and African populations have shown that common single nucleotide polymorphisms (SNPs) in several genes are associated with the risk of developing NB, while few studies have been performed on Chinese children. Herein, we examined the association between the genetic polymorphisms in candidate genes and the risk of NB in Chinese children. In total, 127 SNPs in nine target genes, revealed by GWAS studies of other ethnic groups and four related lincRNAs, were genotyped in 549 samples (244 NB patients and 305 healthy controls). After adjustment for gender and age, there were 21 SNPs associated with NB risk at the two-sided P < 0.05 level, 11 of which were located in LMO1. After correction for multiple comparisons, only rs204926 in LMO1 remained significantly different between cases and controls (OR = 0.45, 95% CI: 0.31–0.65, adjusted P = 0.003). In addition, 16 haplotypes in four separate genes were significantly different between case and control groups at an unadjusted P value < 0.05, 11 of which were located in LMO1. A major haplotype, ATC, containing rs204926, rs110420, and rs110419, conferred a significant increase in risk for NB (OR = 1.82, 95% CI: 1.41–2.36, adjusted P < 0.001). The major finding of our study was obtained for risk alleles within the LMO1 gene. Our data suggest that genetic variants in LMO1 are associated with increased NB risk in Chinese children.
Collapse
Affiliation(s)
- Jie Lu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Ping Chu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Huanmin Wang
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Shujing Han
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Wei Han
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Jun Tai
- Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
- * E-mail: (XN); (YG)
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
- Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, Beijing, China
- * E-mail: (XN); (YG)
| |
Collapse
|
21
|
Tsolis KC, Bei ES, Papathanasiou I, Kostopoulou F, Gkretsi V, Kalantzaki K, Malizos K, Zervakis M, Tsezou A, Economou A. Comparative proteomic analysis of hypertrophic chondrocytes in osteoarthritis. Clin Proteomics 2015; 12:12. [PMID: 25945082 PMCID: PMC4415313 DOI: 10.1186/s12014-015-9085-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/15/2015] [Indexed: 02/07/2023] Open
Abstract
Background Osteoarthritis (OA) is a multi-factorial disease leading progressively to loss of articular cartilage and subsequently to loss of joint function. While hypertrophy of chondrocytes is a physiological process implicated in the longitudinal growth of long bones, hypertrophy-like alterations in chondrocytes play a major role in OA. We performed a quantitative proteomic analysis in osteoarthritic and normal chondrocytes followed by functional analyses to investigate proteome changes and molecular pathways involved in OA pathogenesis. Methods Chondrocytes were isolated from articular cartilage of ten patients with primary OA undergoing knee replacement surgery and six normal donors undergoing fracture repair surgery without history of joint disease and no OA clinical manifestations. We analyzed the proteome of chondrocytes using high resolution mass spectrometry and quantified it by label-free quantification and western blot analysis. We also used WebGestalt, a web-based enrichment tool for the functional annotation and pathway analysis of the differentially synthesized proteins, using the Wikipathways database. ClueGO, a Cytoscape plug-in, is also used to compare groups of proteins and to visualize the functionally organized Gene Ontology (GO) terms and pathways in the form of dynamical network structures. Results The proteomic analysis led to the identification of a total of ~2400 proteins. 269 of them showed differential synthesis levels between the two groups. Using functional annotation, we found that proteins belonging to pathways associated with regulation of the actin cytoskeleton, EGF/EGFR, TGF-β, MAPK signaling, integrin-mediated cell adhesion, and lipid metabolism were significantly enriched in the OA samples (p ≤10−5). We also observed that the proteins GSTP1, PLS3, MYOF, HSD17B12, PRDX2, APCS, PLA2G2A SERPINH1/HSP47 and MVP, show distinct synthesis levels, characteristic for OA or control chondrocytes. Conclusion In this study we compared the quantitative changes in proteins synthesized in osteoarthritic compared to normal chondrocytes. We identified several pathways and proteins to be associated with OA chondrocytes. This study provides evidence for further testing on the molecular mechanism of the disease and also propose proteins as candidate markers of OA chondrocyte phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s12014-015-9085-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Konstantinos C Tsolis
- Institute of Molecular Biology and Biotechnology - FoRTH, Iraklio, Greece ; Department of Microbiology and Immunology, Rega Institute for Medical Research, KULeuven, Leuven, Belgium
| | - Ekaterini S Bei
- School of Electronic and Computer Engineering, Technical Univ. of Crete, Chania, Greece
| | - Ioanna Papathanasiou
- Department of Biology, University of Thessaly, Faculty of Medicine, Larissa, Greece ; Institute for Research & Technology-Thessaly/Centre for Research & Technology-Hellas (CE.R.T.H), Larissa, Greece
| | - Fotini Kostopoulou
- Department of Biology, University of Thessaly, Faculty of Medicine, Larissa, Greece ; Institute for Research & Technology-Thessaly/Centre for Research & Technology-Hellas (CE.R.T.H), Larissa, Greece
| | - Vassiliki Gkretsi
- Institute for Research & Technology-Thessaly/Centre for Research & Technology-Hellas (CE.R.T.H), Larissa, Greece
| | - Kalliopi Kalantzaki
- School of Electronic and Computer Engineering, Technical Univ. of Crete, Chania, Greece
| | - Konstantinos Malizos
- Department of Orthopedics, University of Thessaly, Faculty of Medicine, Larissa, Greece
| | - Michalis Zervakis
- School of Electronic and Computer Engineering, Technical Univ. of Crete, Chania, Greece
| | - Aspasia Tsezou
- Department of Biology, University of Thessaly, Faculty of Medicine, Larissa, Greece ; Institute for Research & Technology-Thessaly/Centre for Research & Technology-Hellas (CE.R.T.H), Larissa, Greece
| | - Anastassios Economou
- Institute of Molecular Biology and Biotechnology - FoRTH, Iraklio, Greece ; Department of Microbiology and Immunology, Rega Institute for Medical Research, KULeuven, Leuven, Belgium
| |
Collapse
|
22
|
Determination of the topology of endoplasmic reticulum membrane proteins using redox-sensitive green-fluorescence protein fusions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1672-82. [PMID: 25889538 DOI: 10.1016/j.bbamcr.2015.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/30/2015] [Accepted: 04/02/2015] [Indexed: 02/01/2023]
Abstract
Membrane proteins of the endoplasmic reticulum (ER) are involved in a wide array of essential cellular functions. Identification of the topology of membrane proteins can provide significant insight into their mechanisms of action and biological roles. This is particularly important for membrane enzymes, since their topology determines the subcellular site where a biochemical reaction takes place and the dependence on luminal or cytosolic co-factor pools and substrates. The methods currently available for the determination of topology of proteins are rather laborious and require post-lysis or post-fixation manipulation of cells. In this work, we have developed a simple method for defining intracellular localization and topology of ER membrane proteins in living cells, based on the fusion of the respective protein with redox-sensitive green-fluorescent protein (roGFP). We validated the method and demonstrated that roGFP fusion proteins constitute a reliable tool for the study of ER membrane protein topology, using as control microsomal 11β-hydroxysteroid dehydrogenase (11β-HSD) proteins whose topology has been resolved, and comparing with an independent approach. We then implemented this method to determine the membrane topology of six microsomal members of the 17β-hydroxysteroid dehydrogenase (17β-HSD) family. The results revealed a luminal orientation of the catalytic site for three enzymes, i.e. 17β-HSD6, 7 and 12. Knowledge of the intracellular location of the catalytic site of these enzymes will enable future studies on their biological functions and on the role of the luminal co-factor pool.
Collapse
|
23
|
Yang Y, Xiong J, Zhou Z, Huo F, Miao W, Ran C, Liu Y, Zhang J, Feng J, Wang M, Wang M, Wang L, Yao B. The genome of the myxosporean Thelohanellus kitauei shows adaptations to nutrient acquisition within its fish host. Genome Biol Evol 2014; 6:3182-98. [PMID: 25381665 PMCID: PMC4986447 DOI: 10.1093/gbe/evu247] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Members of Myxozoa, a parasitic metazoan taxon, have considerable detrimental effects on fish hosts and also have been associated with human food-borne illness. Little is known about their biology and metabolism. Analysis of the genome of Thelohanellus kitauei and comparative analysis with genomes of its two free-living cnidarian relatives revealed that T. kitauei has adapted to parasitism, as indicated by the streamlined metabolic repertoire and the tendency toward anabolism rather than catabolism. Thelohanellus kitauei mainly secretes proteases and protease inhibitors for nutrient digestion (parasite invasion), and depends on endocytosis (mainly low-density lipoprotein receptors-mediated type) and secondary carriers for nutrient absorption. Absence of both classic and complementary anaerobic pathways and gluconeogenesis, the lack of de novo synthesis and reduced activity in hydrolysis of fatty acids, amino acids, and nucleotides indicated that T. kitauei in this vertebrate host-parasite system has adapted to inhabit a physiological environment extremely rich in both oxygen and nutrients (especially glucose), which is consistent with its preferred parasitic site, that is, the host gut submucosa. Taking advantage of the genomic and transcriptomic information, 23 potential nutrition-related T. kitauei-specific chemotherapeutic targets were identified. This first genome sequence of a myxozoan will facilitate development of potential therapeutics for efficient control of myxozoan parasites and ultimately prevent myxozoan-induced fish-borne illnesses in humans.
Collapse
Affiliation(s)
- Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Jie Xiong
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Zhigang Zhou
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Fengmin Huo
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Wei Miao
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Yuchun Liu
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Jinyong Zhang
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Jinmei Feng
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Meng Wang
- Tianjin Biochip Corporation, Tianjin, People's Republic of China
| | - Min Wang
- TEDA School of Biological Sciences and Biotechnology, Nankai University, Tianjin, People's Republic of China
| | - Lei Wang
- TEDA School of Biological Sciences and Biotechnology, Nankai University, Tianjin, People's Republic of China
| | - Bin Yao
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| |
Collapse
|
24
|
Trottier A, Maltais R, Poirier D. Identification of a first enzymatic activator of a 17β-hydroxysteroid dehydrogenase. ACS Chem Biol 2014; 9:1668-73. [PMID: 24910887 DOI: 10.1021/cb500109e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Small molecule activators that directly modulate the activity of an enzyme are uncommon entities, and such activators had never yet been identified for any 17β-hydroxysteroid dehydrogenase (17β-HSD). We hereby report the fortuitous discovery of a steroid derivative that caused an up to 3-fold increase in the activity of 17β-HSD12. The stimulation of estrone to estradiol conversion has been characterized in intact and homogenized stably transfected HEK-293 cells and has also been observed in T47D breast cancer cells. Structure-activity relationships closely linked to the nature of the substituent on the [1,3]oxazinan-2-one ring of an estradiol derivative emerged from this study and may help in the identification of a previously unsuspected endogenous activation of 17β-HSD12. This activator will therefore be a useful tool to study this relatively unknown enzyme as well as the possible activation of other 17β-HSD family members.
Collapse
Affiliation(s)
- Alexandre Trottier
- Laboratory
of Medicinal Chemistry,
Endocrinology and Nephrology Unit, CHU de Québec (CHUL, T4),
and Faculty of Medicine, Laval University, Québec, Québec G1V 4G2, Canada
| | - René Maltais
- Laboratory
of Medicinal Chemistry,
Endocrinology and Nephrology Unit, CHU de Québec (CHUL, T4),
and Faculty of Medicine, Laval University, Québec, Québec G1V 4G2, Canada
| | - Donald Poirier
- Laboratory
of Medicinal Chemistry,
Endocrinology and Nephrology Unit, CHU de Québec (CHUL, T4),
and Faculty of Medicine, Laval University, Québec, Québec G1V 4G2, Canada
| |
Collapse
|