1
|
Brascher TC, de Bortoli L, Toledo-Silva G, Zacchi FL, Razzera G. In silico structural features of the CgNR5A: CgDAX complex and its role in regulating gene expression of CYP target genes in Crassostrea gigas. CHEMOSPHERE 2024; 361:142443. [PMID: 38815811 DOI: 10.1016/j.chemosphere.2024.142443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Contamination of aquatic environments has been steadily increasing due to human activities. The Pacific oyster Crassostrea gigas has been used as a key species in studies assessing the impacts of contaminants on human health and the aquatic biome. In this context, cytochrome P450 (CYPs) play a crucial role in xenobiotic metabolism. In vertebrates many of these CYPs are regulated by nuclear receptors (NRs) and little is known about the NRs role in C. gigas. Particularly, the CgNR5A represents a homologue of SF1 and LRH-1 found in vertebrates. Members of this group can regulate genes of CYPs involved in lipid/steroid metabolism, with their activity regulated by other NR, called as DAX-1, generating a NR complex on DNA response elements (REs). As C. gigas does not exhibit steroid biosynthesis pathways, CgNR5A may play other physiological roles. To clarify this issue, we conducted an in silico investigation of the interaction between CgNR5A and DNA to identify potential C. gigas CYP target genes. Using molecular docking and dynamics simulations of the CgNR5A on DNA molecules, we identified a monomeric interaction with extended REs. This RE was found in the promoter region of 30 CYP genes and also the NR CgDAX. When the upstream regulatory region was analyzed, CYP2C39, CYP3A11, CYP4C21, CYP7A1, CYP17A1, and CYP27C1 were mapped as the main genes regulated by CgNR5A. These identified CYPs belong to families known for their involvement in xenobiotic and lipid/steroid metabolism. Furthermore, we reconstructed a trimeric complex, previously proposed for vertebrates, with CgNR5A:CgDAX and subjected it to molecular dynamics simulations analysis. Heterotrimeric complex remained stable during the simulations, suggesting that CgDAX may modulate CgNR5A transcriptional activity. This study provides insights into the potential physiological processes involving these NRs in the regulation of CYPs associated with xenobiotic and steroid/lipid metabolism.
Collapse
Affiliation(s)
- Theo Cardozo Brascher
- Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil; Laboratório de Biomarcadores de Contaminação Aquática e Imunoquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Leonardo de Bortoli
- Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil; Laboratório de Biomarcadores de Contaminação Aquática e Imunoquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil; Laboratório de Genômica, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Guilherme Toledo-Silva
- Laboratório de Genômica, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Flávia Lucena Zacchi
- Laboratório de Moluscos Marinhos, Universidade Federal de Santa Catarina, Florianópolis, SC, 88061-600, Brazil
| | - Guilherme Razzera
- Programa de Pós-Graduação em Bioquímica, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil; Laboratório de Biomarcadores de Contaminação Aquática e Imunoquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
| |
Collapse
|
2
|
Takahashi F, Baba T, Christianto A, Yanai S, Lee-Okada HC, Ishiwata K, Nakabayashi K, Hata K, Ishii T, Hasegawa T, Yokomizo T, Choi MH, Morohashi KI. Development of sexual dimorphism of skeletal muscles through the adrenal cortex, caused by androgen-induced global gene suppression. Cell Rep 2024; 43:113715. [PMID: 38306273 DOI: 10.1016/j.celrep.2024.113715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/08/2023] [Accepted: 01/11/2024] [Indexed: 02/04/2024] Open
Abstract
The zona fasciculata (zF) in the adrenal cortex contributes to multiple physiological actions through glucocorticoid synthesis. The size, proliferation, and glucocorticoid synthesis characteristics are all female biased, and sexual dimorphism is established by androgen. In this study, transcriptomes were obtained to unveil the sex differentiation mechanism. Interestingly, both the amount of mRNA and the expressions of nearly all genes were higher in females. The expression of Nr5a1, which is essential for steroidogenic cell differentiation, was also female biased. Whole-genome studies demonstrated that NR5A1 regulates nearly all gene expression directly or indirectly. This suggests that androgen-induced global gene suppression is potentially mediated by NR5A1. Using Nr5a1 heterozygous mice, whose adrenal cortex is smaller than the wild type, we demonstrated that the size of skeletal muscles is possibly regulated by glucocorticoid synthesized by zF. Taken together, considering the ubiquitous presence of glucocorticoid receptors, our findings provide a pathway for sex differentiation through glucocorticoid synthesis.
Collapse
Affiliation(s)
- Fumiya Takahashi
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takashi Baba
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Antonius Christianto
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shogo Yanai
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hyeon-Cheol Lee-Okada
- Department of Biochemistry, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Keisuke Ishiwata
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-0074, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-0074, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-0074, Japan; Department of Human Molecular Genetics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tomohiro Ishii
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Man Ho Choi
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Ken-Ichirou Morohashi
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Internal Medicine, Kurume University School of Medicine, 67 Asahimachi, Kurume 830-0011, Japan.
| |
Collapse
|
3
|
Campbell AN, Choi WJ, Chi ES, Orun AR, Poland JC, Stivison EA, Kubina JN, Hudson KL, Loi MNC, Bhatia JN, Gilligan JW, Quintanà AA, Blind RD. Steroidogenic Factor-1 form and function: From phospholipids to physiology. Adv Biol Regul 2024; 91:100991. [PMID: 37802761 PMCID: PMC10922105 DOI: 10.1016/j.jbior.2023.100991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023]
Abstract
Steroidogenic Factor-1 (SF-1, NR5A1) is a member of the nuclear receptor superfamily of ligand-regulated transcription factors, consisting of a DNA-binding domain (DBD) connected to a transcriptional regulatory ligand binding domain (LBD) via an unstructured hinge domain. SF-1 is a master regulator of development and adult function along the hypothalamic pituitary adrenal and gonadal axes, with strong pathophysiological association with endometriosis and adrenocortical carcinoma. SF-1 was shown to bind and be regulated by phospholipids, one of the most interesting aspects of SF-1 regulation is the manner in which SF-1 interacts with phospholipids: SF-1 buries the phospholipid acyl chains deep in the hydrophobic core of the SF-1 protein, while the lipid headgroups remain solvent-exposed on the exterior of the SF-1 protein surface. Here, we have reviewed several aspects of SF-1 structure, function and physiology, touching on other transcription factors that help regulate SF-1 target genes, non-canonical functions of SF-1, the DNA-binding properties of SF-1, the use of mass spectrometry to identify lipids that associate with SF-1, how protein phosphorylation regulates SF-1 and the structural biology of the phospholipid-ligand binding domain. Together this review summarizes the form and function of Steroidogenic Factor-1 in physiology and in human disease, with particular emphasis on adrenal cancer.
Collapse
Affiliation(s)
- Alexis N Campbell
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Woong Jae Choi
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ethan S Chi
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Abigail R Orun
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - James C Poland
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Elizabeth A Stivison
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jakub N Kubina
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Kimora L Hudson
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Mong Na Claire Loi
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jay N Bhatia
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Joseph W Gilligan
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Adrian A Quintanà
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Raymond D Blind
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| |
Collapse
|
4
|
Lalli E. A reappraisal of transcriptional regulation by NR5A1 and beta-catenin in adrenocortical carcinoma. Front Endocrinol (Lausanne) 2023; 14:1303332. [PMID: 38155952 PMCID: PMC10753177 DOI: 10.3389/fendo.2023.1303332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/17/2023] [Indexed: 12/30/2023] Open
Abstract
Background Overexpression of the transcription factor NR5A1 and constitutive activation of canonical Wnt signalling leading to nuclear translocation of beta-catenin are hallmarks of malignancy in adrenocortical carcinoma (ACC). Based on the analysis of genomic profiles in H295R ACC cells, Mohan et al. (Cancer Res. 2023; 83: 2123-2141) recently suggested that a major determinant driving proliferation and differentiation in malignant ACC is the interaction of NR5A1 and beta-catenin on chromatin to regulate gene expression. Methods I reanalyzed the same set of data generated by Mohan et al. and other published data of knockdown-validated NR5A1 and beta-catenin target genes. Results Beta-catenin is mainly found in association to canonical T cell factor/lymphoid enhancer factor (TCF/LEF) motifs in genomic DNA. NR5A1 and beta-catenin regulate distinct target gene sets in ACC cells. Conclusion Overall, my analysis suggests a model where NR5A1 overexpression and beta-catenin activation principally act independently, rather than functionally interacting, to drive ACC malignancy.
Collapse
Affiliation(s)
- Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, Valbonne, France
- Université Côte d’Azur, Valbonne, France
- Inserm, Valbonne, France
| |
Collapse
|
5
|
Hur SK, Somerville TD, Wu XS, Maia-Silva D, Demerdash OE, Tuveson DA, Notta F, Vakoc CR. p73 activates transcriptional signatures of basal lineage identity and ciliogenesis in pancreatic ductal adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.537667. [PMID: 37131797 PMCID: PMC10153254 DOI: 10.1101/2023.04.20.537667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
During the progression of pancreatic ductal adenocarcinoma (PDAC), tumor cells are known to acquire transcriptional and morphological properties of the basal (also known as squamous) epithelial lineage, which leads to more aggressive disease characteristics. Here, we show that a subset of basal-like PDAC tumors aberrantly express p73 (TA isoform), which is a known transcriptional activator of basal lineage identity, ciliogenesis, and tumor suppression in normal tissue development. Using gain- and loss- of function experiments, we show that p73 is necessary and sufficient to activate genes related to basal identity (e.g. KRT5), ciliogenesis (e.g. FOXJ1), and p53-like tumor suppression (e.g. CDKN1A) in human PDAC models. Owing to the paradoxical combination of oncogenic and tumor suppressive outputs of this transcription factor, we propose that PDAC cells express a low level of p73 that is optimal for promoting lineage plasticity without severe impairment of cell proliferation. Collectively, our study reinforces how PDAC cells exploit master regulators of the basal epithelial lineage during disease progression.
Collapse
Affiliation(s)
- Stella K. Hur
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, U.S.A
| | | | - Xiaoli S. Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, U.S.A
| | - Diogo Maia-Silva
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, U.S.A
| | | | - David A. Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, U.S.A
| | - Faiyaz Notta
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
6
|
Steroidogenic Factor 1, a Goldilocks Transcription Factor from Adrenocortical Organogenesis to Malignancy. Int J Mol Sci 2023; 24:ijms24043585. [PMID: 36835002 PMCID: PMC9959402 DOI: 10.3390/ijms24043585] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Steroidogenic factor-1 (SF-1, also termed Ad4BP; NR5A1 in the official nomenclature) is a nuclear receptor transcription factor that plays a crucial role in the regulation of adrenal and gonadal development, function and maintenance. In addition to its classical role in regulating the expression of P450 steroid hydroxylases and other steroidogenic genes, involvement in other key processes such as cell survival/proliferation and cytoskeleton dynamics have also been highlighted for SF-1. SF-1 has a restricted pattern of expression, being expressed along the hypothalamic-pituitary axis and in steroidogenic organs since the time of their establishment. Reduced SF-1 expression affects proper gonadal and adrenal organogenesis and function. On the other hand, SF-1 overexpression is found in adrenocortical carcinoma and represents a prognostic marker for patients' survival. This review is focused on the current knowledge about SF-1 and the crucial importance of its dosage for adrenal gland development and function, from its involvement in adrenal cortex formation to tumorigenesis. Overall, data converge towards SF-1 being a key player in the complex network of transcriptional regulation within the adrenal gland in a dosage-dependent manner.
Collapse
|
7
|
Moore EC, Ioannou L, Ruseckaite R, Serpell J, Ahern S. Hereditary Endocrine Tumor Registries. J Endocr Soc 2022; 7:bvac194. [PMID: 36632485 PMCID: PMC9825730 DOI: 10.1210/jendso/bvac194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Indexed: 12/24/2022] Open
Abstract
Context Endocrine neoplasia syndromes are phenotypically complex, and there is a misconception that they are universally rare. Genetic alterations are increasingly recognized; however, true prevalence is unknown. The purpose of a clinical registry is to monitor the quality of health care delivered to a specified group of patients through the collection, analysis, and reporting of relevant health-related information. This leads to improved clinical practice, decision-making, patient satisfaction, and outcome. Objective This review aims to identify, compare, and contrast active registries worldwide that capture data relevant to hereditary endocrine tumors (HETs). Methods Clinical registries were identified using a systematic approach from publications (Ovid MEDLINE, EMBASE) peer consultation, clinical trials, and web searches. Inclusion criteria were hereditary endocrine tumors, clinical registries, and English language. Exclusion criteria were institutional audits, absence of clinical data, or inactivity. Details surrounding general characteristics, funding, data fields, collection periods, and entry methods were collated. Results Fifteen registries specific for HET were shortlisted with 136 affiliated peer-reviewed manuscripts. Conclusion There are few clinical registries specific to HET. Most of these are European, and the data collected are highly variable. Further research into their effectiveness is warranted. We note the absence of an Australian registry for all HET, which would provide potential health and economic gains. This review presents a unique opportunity to harmonize registry data for HET locally and further afield.
Collapse
Affiliation(s)
- Edwina C Moore
- Correspondence: Edwina C. Moore, MBBS (HONS), BMedSci, Peninsula Private Hospital, 525 McClelland Dr, Ste 16, Langwarrin, VIC, 3199, Australia.
| | - Liane Ioannou
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3800, Australia
| | - Rasa Ruseckaite
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3800, Australia
| | - Jonathan Serpell
- Department of Breast, Endocrine and General Surgery, Alfred Health, Monash University, Melbourne, Victoria 3800, Australia
| | - Susannah Ahern
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3800, Australia
| |
Collapse
|
8
|
Lin YF, Schang G, Buddle ERS, Schultz H, Willis TL, Ruf-Zamojski F, Zamojski M, Mendelev N, Boehm U, Sealfon SC, Andoniadou CL, Bernard DJ. Steroidogenic Factor 1 Regulates Transcription of the Inhibin B Coreceptor in Pituitary Gonadotrope Cells. Endocrinology 2022; 163:6661776. [PMID: 35957608 PMCID: PMC9761571 DOI: 10.1210/endocr/bqac131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Indexed: 11/19/2022]
Abstract
The inhibins control reproduction by suppressing follicle-stimulating hormone synthesis in pituitary gonadotrope cells. The newly discovered inhibin B coreceptor, TGFBR3L, is selectively and highly expressed in gonadotropes in both mice and humans. Here, we describe our initial characterization of mechanisms controlling cell-specific Tgfbr3l/TGFBR3L transcription. We identified two steroidogenic factor 1 (SF-1 or NR5A1) cis-elements in the proximal Tgfbr3l promoter in mice. SF-1 induction of murine Tgfbr3l promoter-reporter activity was inhibited by mutations in one or both sites in heterologous cells. In homologous cells, mutation of these cis-elements or depletion of endogenous SF-1 similarly decreased reporter activity. We observed nearly identical results when using a human TGFBR3L promoter-reporter. The Tgfbr3l gene was tightly compacted and Tgfbr3l mRNA expression was essentially absent in gonadotropes of SF-1 (Nr5a1) conditional knockout mice. During murine embryonic development, Tgfbr3l precedes Nr5a1 expression, though the two transcripts are fully colocalized by embryonic day 18.5 and thereafter. Collectively, these data indicate that SF-1 directly regulates Tgfbr3l/TGFBR3L transcription and is required for postnatal expression of the gene in gonadotropes.
Collapse
Affiliation(s)
- Yeu-Farn Lin
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Gauthier Schang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Evan R S Buddle
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Hailey Schultz
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
| | - Thea L Willis
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 1UL, UK
| | - Frederique Ruf-Zamojski
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michel Zamojski
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Natalia Mendelev
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg 66421, Germany
| | - Stuart C Sealfon
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 1UL, UK
| | - Daniel J Bernard
- Correspondence: Daniel J. Bernard, PhD, Department of Pharmacology and Therapeutics, 3655 Promenade Sir William Osler, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Sigala S, Rossini E, Abate A, Tamburello M, Bornstein SR, Hantel C. An update on adrenocortical cell lines of human origin. Endocrine 2022; 77:432-437. [PMID: 35764904 PMCID: PMC9385758 DOI: 10.1007/s12020-022-03112-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/10/2022] [Indexed: 12/23/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare, heterogenous and highly malignant disease. Management of ACC is dependent on disease stage with complete surgical resection as the only potentially curative option. However, advanced, un-resectable, metastatic stages and also recurrences often require systemic treatments, which are unfortunately nowadays still unsatisfactory. The scarcity of preclinical models reflecting patient heterogeneities and furthermore drug-resistant phenotypes, has hampered the progress and development of new therapies in recent years. In this review, we provide an overview on the classical models and substantial progress which has been made over the last years in context of this aggressive disease.
Collapse
Affiliation(s)
- Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Elisa Rossini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Stefan R Bornstein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091, Zürich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Dresden, Germany
- Diabetes and Nutritional Sciences, King's College London, London, WC2R 2LS, UK
- Center for Regenerative Therapies, Technische Universität Dresden, 01307, Dresden, Germany
- Paul-Langerhans-Institute Dresden, Helmholtz Center Munich, University Hospital Carl Gustav Carus, Faculty of Medicine, Technische Universität Dresden, 01307, Dresden, Germany
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091, Zürich, Switzerland.
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Dresden, Germany.
| |
Collapse
|
10
|
Lalli E, Figueiredo BC. Prolactin as an adrenocorticotropic hormone: Prolactin signalling is a conserved key regulator of sexually dimorphic adrenal gland function in health and disease. Bioessays 2022; 44:e2200109. [PMID: 36000778 DOI: 10.1002/bies.202200109] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
A large number of previous reports described an effect of the pituitary hormone prolactin (PRL) on steroid hormone production by the adrenal cortex. However, those studies remained anecdotal and were never converted into a conceptual and mechanistic framework, let alone being translated into clinical care. In the light of our recently published landmark study where we described PRL signalling as a pivotal regulator of the sexually dimorphic adrenal phenotype in mouse and of adrenal androgen production in humans, we present here the overarching hypothesis that PRL signalling increases the activity of Steroidogenic Factor-1 (SF-1/NR5A1), a transcription factor that has an essential role in adrenal gland development and function, to regulate adrenal cortex growth and hormonal production in physiological and pathological conditions. PRL can then be considered as a bona fide adrenocorticotropic hormone synergizing with ACTH in the endocrine control of adrenal cortex function.
Collapse
Affiliation(s)
- Enzo Lalli
- EXPOGEN-CANCER CNRS International Research Project, 660 route des Lucioles, Sophia Antipolis, Valbonne, 06560, France.,Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.,Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.,Pelé Pequeno Principe Research Institute, Curitiba, PR, Brazil
| | - Bonald C Figueiredo
- EXPOGEN-CANCER CNRS International Research Project, 660 route des Lucioles, Sophia Antipolis, Valbonne, 06560, France.,Pelé Pequeno Principe Research Institute, Curitiba, PR, Brazil
| |
Collapse
|
11
|
Ko MC, Frankl-Vilches C, Bakker A, Gahr M. The Gene Expression Profile of the Song Control Nucleus HVC Shows Sex Specificity, Hormone Responsiveness, and Species Specificity Among Songbirds. Front Neurosci 2021; 15:680530. [PMID: 34135731 PMCID: PMC8200640 DOI: 10.3389/fnins.2021.680530] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/28/2021] [Indexed: 11/17/2022] Open
Abstract
Singing occurs in songbirds of both sexes, but some species show typical degrees of sex-specific performance. We studied the transcriptional sex differences in the HVC, a brain nucleus critical for song pattern generation, of the forest weaver (Ploceus bicolor), the blue-capped cordon-bleu (Uraeginthus cyanocephalus), and the canary (Serinus canaria), which are species that show low, medium, and high levels of sex-specific singing, respectively. We observed persistent sex differences in gene expression levels regardless of the species-specific sexual singing phenotypes. We further studied the HVC transcriptomes of defined phenotypes of canary, known for its testosterone-sensitive seasonal singing. By studying both sexes of canaries during both breeding and non-breeding seasons, non-breeding canaries treated with testosterone, and spontaneously singing females, we found that the circulating androgen levels and sex were the predominant variables associated with the variations in the HVC transcriptomes. The comparison of natural singing with testosterone-induced singing in canaries of the same sex revealed considerable differences in the HVC transcriptomes. Strong transcriptional changes in the HVC were detected during the transition from non-singing to singing in canaries of both sexes. Although the sex-specific genes of singing females shared little resemblance with those of males, our analysis showed potential functional convergences. Thus, male and female songbirds achieve comparable singing behaviours with sex-specific transcriptomes.
Collapse
Affiliation(s)
- Meng-Ching Ko
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | - Carolina Frankl-Vilches
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | - Antje Bakker
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | - Manfred Gahr
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| |
Collapse
|
12
|
Šemanjski K, Majdič G, Kozina V, Ježek D. Sexual dimorphism of the extraorbital lacrimal glands in SF-1 knockout mice. Acta Histochem 2021; 123:151669. [PMID: 33360489 DOI: 10.1016/j.acthis.2020.151669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 01/15/2023]
Abstract
Sexual dimorphism (SD) represents all the differences between males and females of the same species. SD of the murine lacrimal gland and the major effect of testosterone on its formation are well documented. Steroidogenic factor-1 (SF-1, NR5a1) is a nuclear receptor essential for the fetal development of steroid hormones producing organs and SF-1 knockout mice (Sf-1 KO) are therefore born without gonads and adrenal glands. The aim of this study was to investigate whether SD in lacrimal glands is present in the absence of exposure to sex hormones during development. Lacrimal glands from adult Sf-1 KO male and female mice without hormonal exposure, and from males that were treated with testosterone propionate (TP) prior to sacrifice, were examined. After sacrifice, glandular tissue was processed using standard histological procedures. Paraffin sections were analysed by stereology and immunostained against the androgen receptor (AR). Our results showed that there were no statistically significant differences in the mean volumes of acini, connective tissue or ductal system between males, females, and males on TP. The same pertains to the mean length of the ducts in all three groups. In the absence of sex hormones, sex chromosomes proved to be insufficient in inducing sexual dimorphism in LG. However, nuclei of the acinar cells in males on TP were positive for AR, whereas in males without TP no expression of AR was detected. Administration of TP induced the expression of AR in the nuclei of acinar cells of males but did not affect the morphology of LG. We conclude that SD in the lacrimal gland is not present in Sf-1 KO mice and this suggests that sex hormones have a major role in the development of SD in the lacrimal gland.
Collapse
Affiliation(s)
- Kristina Šemanjski
- Clinical Hospital "Sveti Duh", Zagreb, Department of Surgery, Sveti Duh 64, 10000, Zagreb, Croatia.
| | - Gregor Majdič
- University of Ljubljana, Veterinary Faculty, Centre for Animal Genomics, Croatia
| | - Viviana Kozina
- University of Zagreb, School of Medicine, Department of Histology and Embryology, Croatia
| | - Davor Ježek
- University of Zagreb, School of Medicine, Department of Histology and Embryology, Croatia
| |
Collapse
|
13
|
Morohashi KI, Inoue M, Baba T. Coordination of Multiple Cellular Processes by NR5A1/Nr5a1. Endocrinol Metab (Seoul) 2020; 35:756-764. [PMID: 33397036 PMCID: PMC7803590 DOI: 10.3803/enm.2020.402] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/27/2020] [Indexed: 11/11/2022] Open
Abstract
The agenesis of the gonads and adrenal gland in revealed by knockout mouse studies strongly suggested a crucial role for Nr5a1 (SF-1 or Ad4BP) in organ development. In relation to these striking phenotypes, NR5A1/Nr5a1 has the potential to reprogram cells to steroidogenic cells, endow pluripotency, and regulate cell proliferation. However, due to limited knowledge regarding NR5A1 target genes, the mechanism by which NR5A1/Nr5a1 regulates these fundamental processes has remained unknown. Recently, newlyestablished technologies have enabled the identification of NR5A1 target genes related to multiple metabolic processes, as well as the aforementioned biological processes. Considering that active cellular processes are expected to be accompanied by active metabolism, NR5A1 may act as a key factor for processes such as cell differentiation, proliferation, and survival by coordinating these processes with cellular metabolism. A complete and definite picture of the cellular processes coordinated by NR5A1/Nr5a1 could be depicted by accumulating evidence of the potential target genes through whole genome studies.
Collapse
Affiliation(s)
- Ken-Ichirou Morohashi
- Division of Biology of Sex Differences, Graduate School of Medical Sciences, and Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Miki Inoue
- Division of Biology of Sex Differences, Graduate School of Medical Sciences, and Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Baba
- Division of Biology of Sex Differences, Graduate School of Medical Sciences, and Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
14
|
Yang D, Wu X, Zhou Y, Wang W, Wang Z. The microRNA/TET3/REST axis is required for olfactory globose basal cell proliferation and male behavior. EMBO Rep 2020; 21:e49431. [PMID: 32677323 DOI: 10.15252/embr.201949431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 06/14/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
In the main olfactory epithelium (MOE), new olfactory sensory neurons (OSNs) are persistently generated to replace lost neurons throughout an organism's lifespan. This process predominantly depends on the proliferation of globose basal cells (GBCs), the actively dividing stem cells in the MOE. Here, by using CRISPR/Cas9 and RNAi coupled with adeno-associated virus (AAV) nose delivery approaches, we demonstrated that knockdown of miR-200b/a in the MOE resulted in supernumerary Mash1-marked GBCs and decreased numbers of differentiated OSNs, accompanied by abrogation of male behaviors. We further showed that in the MOE, miR-200b/a targets the ten-eleven translocation methylcytosine dioxygenase TET3, which cooperates with RE1-silencing transcription factor (REST) to exert their functions. Deficiencies including proliferation, differentiation, and behaviors illustrated in miR-200b/a knockdown mice were rescued by suppressing either TET3 or REST. Our work describes a mechanism of coordination of GBC proliferation and differentiation in the MOE and olfactory male behaviors through miR-200/TET3/REST signaling.
Collapse
Affiliation(s)
- Dong Yang
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Xiangbo Wu
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Yanfen Zhou
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Weina Wang
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Zhenshan Wang
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| |
Collapse
|
15
|
Yamauchi K, Ikeda T, Hosokawa M, Nakatsuji N, Kawase E, Chuma S, Hasegawa K, Suemori H. Overexpression of Nuclear Receptor 5A1 Induces and Maintains an Intermediate State of Conversion between Primed and Naive Pluripotency. Stem Cell Reports 2020; 14:506-519. [PMID: 32084386 PMCID: PMC7066342 DOI: 10.1016/j.stemcr.2020.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 10/31/2022] Open
Abstract
Naive and primed human pluripotent stem cells (hPSCs) have provided useful insights into the regulation of pluripotency. However, the molecular mechanisms regulating naive conversion remain elusive. Here, we report intermediate naive conversion induced by overexpressing nuclear receptor 5A1 (NR5A1) in hPSCs. The cells displayed some naive features, such as clonogenicity, glycogen synthase kinase 3β, and mitogen-activated protein kinase (MAPK) independence, expression of naive-associated genes, and two activated X chromosomes, but lacked others, such as KLF17 expression, transforming growth factor β independence, and imprinted gene demethylation. Notably, NR5A1 negated MAPK activation by fibroblast growth factor 2, leading to cell-autonomous self-renewal independent of MAPK inhibition. These phenotypes may be associated with naive conversion, and were regulated by a DPPA2/4-dependent pathway that activates the selective expression of naive-associated genes. This study increases our understanding of the mechanisms regulating the conversion from primed to naive pluripotency.
Collapse
Affiliation(s)
- Kaori Yamauchi
- Laboratory of Embryonic Stem Cell Research, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Tatsuhiko Ikeda
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8351, Japan
| | - Mihoko Hosokawa
- Laboratory of Developmental Epigenome, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Norio Nakatsuji
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8351, Japan; Laboratory of Developmental Epigenome, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Eihachiro Kawase
- Laboratory of Embryonic Stem Cell Research, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Shinichiro Chuma
- Laboratory of Developmental Epigenome, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Kouichi Hasegawa
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8351, Japan; Institute for Stem Cell Biology and Regenerative Medicine, NCBS Campus, GKVK, Bangalore 560065, India
| | - Hirofumi Suemori
- Laboratory of Embryonic Stem Cell Research, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
16
|
Hiranuma N, Lundberg SM, Lee SI. AIControl: replacing matched control experiments with machine learning improves ChIP-seq peak identification. Nucleic Acids Res 2019; 47:e58. [PMID: 30869146 PMCID: PMC6547432 DOI: 10.1093/nar/gkz156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/15/2019] [Accepted: 02/28/2019] [Indexed: 01/24/2023] Open
Abstract
ChIP-seq is a technique to determine binding locations of transcription factors, which remains a central challenge in molecular biology. Current practice is to use a 'control' dataset to remove background signals from a immunoprecipitation (IP) 'target' dataset. We introduce the AIControl framework, which eliminates the need to obtain a control dataset and instead identifies binding peaks by estimating the distributions of background signals from many publicly available control ChIP-seq datasets. We thereby avoid the cost of running control experiments while simultaneously increasing the accuracy of binding location identification. Specifically, AIControl can (i) estimate background signals at fine resolution, (ii) systematically weigh the most appropriate control datasets in a data-driven way, (iii) capture sources of potential biases that may be missed by one control dataset and (iv) remove the need for costly and time-consuming control experiments. We applied AIControl to 410 IP datasets in the ENCODE ChIP-seq database, using 440 control datasets from 107 cell types to impute background signal. Without using matched control datasets, AIControl identified peaks that were more enriched for putative binding sites than those identified by other popular peak callers that used a matched control dataset. We also demonstrated that our framework identifies binding sites that recover documented protein interactions more accurately.
Collapse
Affiliation(s)
- Naozumi Hiranuma
- Paul G. Allen School of Computer Science and Engineering, University of Washington, WA, USA, 98195-2350
| | - Scott M Lundberg
- Paul G. Allen School of Computer Science and Engineering, University of Washington, WA, USA, 98195-2350
| | - Su-In Lee
- Paul G. Allen School of Computer Science and Engineering, University of Washington, WA, USA, 98195-2350
| |
Collapse
|
17
|
Jiang DN, Mustapha UF, Shi HJ, Huang YQ, Si-Tu JX, Wang M, Deng SP, Chen HP, Tian CX, Zhu CH, Li MH, Li GL. Expression and transcriptional regulation of gsdf in spotted scat (Scatophagus argus). Comp Biochem Physiol B Biochem Mol Biol 2019; 233:35-45. [PMID: 30980893 DOI: 10.1016/j.cbpb.2019.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/31/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022]
Abstract
Gonadal soma-derived factor (Gsdf) is critical for testicular differentiation and early germ cell development in teleosts. The spotted scat (Scatophagus argus), with a stable XX-XY sex-determination system and the candidate sex determination gene dmrt1, provides a good model for understanding the mechanism of sex determination and differentiation in teleosts. In this study, we analyzed spotted scat gsdf tissue distribution and gene expression patterns in gonads, as well as further analysis of transcriptional regulation. Tissue distribution analysis showed that gsdf was only expressed in testis and ovary. Real-time PCR showed that both gsdf and dmrt1 were expressed significantly higher in testes at different phases (phase III, IV and V) compared to ovaries at phase II, III and IV, while gsdf was expressed significantly higher in phase II ovaries than those of phase III and IV. Western blot analysis also showed that Gsdf was more highly expressed in the testis than ovary. Immunohistochemistry analysis showed that Gsdf was expressed in Sertoli cells surrounding spermatogonia in the testis, while it was expressed in the somatic cells surrounding the oogonia of the ovary. Approximately 2.7 kb of the 5' upstream region of gsdf was cloned from the spotted scat genomic DNA and in silico promoter analysis revealed the putative transcription factor binding sites of Dmrt1 and Sf1. The luciferase reporter assay, using the human embryonic kidney cells, demonstrated that Dmrt1 activated gsdf expression in a dose-dependent manner in the presence of Sf1 in spotted scat. These results suggest that Gsdf could play a role in regulating the development of spermatogonia and oogonia, and also participate in male sex differentiation by acting as a downstream gene of Dmrt1 in spotted scat.
Collapse
Affiliation(s)
- Dong-Neng Jiang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Umar Farouk Mustapha
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Hong-Juan Shi
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yuan-Qing Huang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Jia-Xin Si-Tu
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Mei Wang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Si-Ping Deng
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Hua-Pu Chen
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Chang-Xu Tian
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Chun-Hua Zhu
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ming-Hui Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Guang-Li Li
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
18
|
Doghman-Bouguerra M, Lalli E. ER-mitochondria interactions: Both strength and weakness within cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:650-662. [PMID: 30668969 DOI: 10.1016/j.bbamcr.2019.01.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/10/2019] [Accepted: 01/17/2019] [Indexed: 12/22/2022]
Abstract
ER-mitochondria contact sites represent hubs for signaling that control mitochondrial biology related to several aspects of cellular survival, metabolism, cell death sensitivity and metastasis, which all contribute to tumorigenesis. Altered ER-mitochondria contacts can deregulate Ca2+ homeostasis, phospholipid metabolism, mitochondrial morphology and dynamics. MAM represent both a hot spot in cancer onset and progression and an Achilles' heel of cancer cells that can be exploited for therapeutic perspectives. Over the past years, an increasing number of cancer-related proteins, including oncogenes and tumor suppressors, have been localized in MAM and exert their pro- or antiapoptotic functions through the regulation of Ca2+ transfer and signaling between the two organelles. In this review, we highlight the central role of ER-mitochondria contact sites in tumorigenesis and focus on chemotherapeutic drugs or potential targets that act on MAM properties for new therapeutic approaches in cancer.
Collapse
Affiliation(s)
- Mabrouka Doghman-Bouguerra
- Université Côte d'Azur, Valbonne 06560, France; CNRS UMR 7275, Sophia Antipolis, Valbonne 06560, France; EXPOGEN-CANCER CNRS International Associated Laboratory, Valbonne 06560, France; Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne 06560, France.
| | - Enzo Lalli
- Université Côte d'Azur, Valbonne 06560, France; CNRS UMR 7275, Sophia Antipolis, Valbonne 06560, France; EXPOGEN-CANCER CNRS International Associated Laboratory, Valbonne 06560, France; Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne 06560, France.
| |
Collapse
|
19
|
Knarston IM, Robevska G, van den Bergen JA, Eggers S, Croft B, Yates J, Hersmus R, Looijenga LHJ, Cameron FJ, Monhike K, Ayers KL, Sinclair AH. NR5A1 gene variants repress the ovarian-specific WNT signaling pathway in 46,XX disorders of sex development patients. Hum Mutat 2018; 40:207-216. [PMID: 30350900 PMCID: PMC6492147 DOI: 10.1002/humu.23672] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/08/2018] [Accepted: 10/20/2018] [Indexed: 12/12/2022]
Abstract
Several recent reports have described a missense variant in the gene NR5A1 (c.274C>T; p.Arg92Trp) in a significant number of 46,XX ovotesticular or testicular disorders of sex development (DSDs) cases. The affected residue falls within the DNA‐binding domain of the NR5A1 protein, however the exact mechanism by which it causes testicular development in 46,XX individuals remains unclear. We have screened a cohort of 26 patients with 46,XX (ovo)testicular DSD and identified three unrelated individuals with this NR5A1 variant (p.Arg92Trp), as well as one patient with a novel NR5A1 variant (c.779C>T; p.Ala260Val). We examined the functional effect of these changes, finding that while protein levels and localization were unaffected, variant NR5A1 proteins repress the WNT signaling pathway and have less ability to upregulate the anti‐testis gene NR0B1. These findings highlight how NR5A1 variants impact ovarian differentiation across multiple pathways, resulting in a switch from ovarian to testis development in genetic females.
Collapse
Affiliation(s)
- Ingrid M Knarston
- Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | | | | | | | - Brittany Croft
- Murdoch Children's Research Institute, Melbourne, Australia.,The Hudson Institute of Medical Research, Monash University, Melbourne, Australia
| | - Jason Yates
- The Townsville Hospital, Department of Health, Queensland, Australia
| | - Remko Hersmus
- Department of Pathology, Josephine Nefkens Building, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Leendert H J Looijenga
- Department of Pathology, Josephine Nefkens Building, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Fergus J Cameron
- Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia.,Department of Endocrinology and Diabetes, Royal Children's Hospital, Melbourne, Australia
| | - Klaus Monhike
- Otto-von-Guericke Universität, Universitätskinderklinik, Magdeburg, Germany
| | - Katie L Ayers
- Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Andrew H Sinclair
- Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
20
|
Abstract
INTRODUCTION Adrenocortical carcinoma (ACC) is a rare cancer, with an incidence less than 0.7-1.5 per 1 million people per year, with a poor prognosis. The overall survival (OS) depends on the ENSAT stage: in particular in metastatic ACC the OS varies from 10 to 20 months, with a 5-year survival around 10%. ACC has a different behavior, probably due to a different biology. For this reason, a careful prognostic classification is mandatory, in order to stratify the patients and propose a specific management. EVIDENCE ACQUISITION Prognostic factors can be divides in three groups: clinical factors (tumor stage, age, hormone-related symptoms), pathological factors (Weiss Score, mitotic count, Ki-67, SF-1 and AVA2, P53, beta-catenin immunohistochemistry, resection status), molecular factors (chromosomal aberrations, methylation profile, altered gene expression and miRNA expression, gene mutations). EVIDENCE SYNTHESIS The best way to stratify ACC patients and propose the best therapeutic option is to combine clinical, pathological and molecular factors. CONCLUSIONS Individualizing patients' prognosis and tumor biology appears as a necessary step for personalized medicine. In addition to tumor stage and tumor grade, the genomic classification may precise the risk stratification and thus help defining therapeutic strategy.
Collapse
Affiliation(s)
- Rossella Libé
- French Network for Adrenal Cancer, Department of Endocrinology, Cochin Hospital, Paris, France -
| |
Collapse
|
21
|
Broad phenotypes in heterozygous NR5A1 46,XY patients with a disorder of sex development: an oligogenic origin? Eur J Hum Genet 2018; 26:1329-1338. [PMID: 29891883 DOI: 10.1038/s41431-018-0202-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 05/23/2018] [Accepted: 05/26/2018] [Indexed: 11/08/2022] Open
Abstract
SF-1/NR5A1 is a transcriptional regulator of adrenal and gonadal development. NR5A1 disease-causing variants cause disorders of sex development (DSD) and adrenal failure, but most affected individuals show a broad DSD/reproductive phenotype only. Most NR5A1 variants show in vitro pathogenic effects, but not when tested in heterozygote state together with wild-type NR5A1 as usually seen in patients. Thus, the genotype-phenotype correlation for NR5A1 variants remains an unsolved question. We analyzed heterozygous 46,XY SF-1/NR5A1 patients by whole exome sequencing and used an algorithm for data analysis based on selected project-specific DSD- and SF-1-related genes. The variants detected were evaluated for their significance in literature, databases and checked in silico using webtools. We identified 19 potentially deleterious variants (one to seven per patient) in 18 genes in four 46,XY DSD subjects carrying heterozygous NR5A1 disease-causing variants. We constructed a scheme of all these hits within the landscape of currently known genes involved in male sex determination and differentiation. Our results suggest that the broad phenotype in these heterozygous NR5A1 46,XY DSD subjects may well be explained by an oligogenic mode of inheritance, in which multiple hits, individually non-deleterious, may contribute to a DSD phenotype unique to each heterozygous SF-1/NR5A1 individual.
Collapse
|
22
|
Lalli E, Luconi M. The next step: mechanisms driving adrenocortical carcinoma metastasis. Endocr Relat Cancer 2018; 25:R31-R48. [PMID: 29142005 DOI: 10.1530/erc-17-0440] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/20/2022]
Abstract
Endocrine tumors have the peculiarity to become clinically evident not only due to symptoms related to space occupation by the growing lesion, similarly to most other tumors, but also, and most often, because of their specific hormonal secretion, which significantly contributes to their pathological burden. Malignant endocrine tumors, in addition, have the ability to produce distant metastases. Here, we critically review the current knowledge about mechanisms and biomarkers characterizing the metastatic process in adrenocortical carcinoma (ACC), a rare endocrine malignancy with a high risk of relapse and metastatization even when the primary tumor is diagnosed and surgically removed at an early stage. We highlight perspectives of future research in the domain and possible new therapeutic avenues based on targeting factors having an important role in the metastatic process of ACC.
Collapse
Affiliation(s)
- Enzo Lalli
- Université Côte d'AzurValbonne, France
- CNRS UMR7275Valbonne, France
- NEOGENEX CNRS International Associated LaboratoryValbonne, France
- Institut de Pharmacologie Moléculaire et CellulaireValbonne, France
| | - Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio'University of Florence, Florence, Italy
| |
Collapse
|
23
|
Molatore S, Kügler A, Irmler M, Wiedemann T, Neff F, Feuchtinger A, Beckers J, Robledo M, Roncaroli F, Pellegata NS. Characterization of neuroendocrine tumors in heterozygous mutant MENX rats: a novel model of invasive medullary thyroid carcinoma. Endocr Relat Cancer 2018; 25:145-162. [PMID: 29142006 DOI: 10.1530/erc-17-0456] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 11/15/2017] [Indexed: 12/18/2022]
Abstract
Rats affected by the MENX syndrome spontaneously develop multiple neuroendocrine tumors (NETs) including adrenal, pituitary and thyroid gland neoplasms. MENX was initially reported to be inherited as a recessive trait and affected rats were found to be homozygous for the predisposing Cdkn1b mutation encoding p27. We here report that heterozygous MENX-mutant rats (p27+/mut) develop the same spectrum of NETs seen in the homozygous (p27mut/mut) animals but with slower progression. Consequently, p27+/mut rats have a significantly shorter lifespan compared with their wild-type (p27+/+) littermates. In the tumors of p27+/mut rats, the wild-type Cdkn1b allele is neither lost nor silenced, implying that p27 is haploinsufficient for tumor suppression in this model. Transcriptome profiling of rat adrenal (pheochromocytoma) and pituitary tumors having different p27 dosages revealed a tissue-specific, dose-dependent effect of p27 on gene expression. In p27+/mut rats, thyroid neoplasms progress to invasive and metastatic medullary thyroid carcinomas (MTCs) accompanied by increased calcitonin levels, as in humans. Comparison of expression signatures of late-stage vs early-stage MTCs from p27+/mut rats identified genes potentially involved in tumor aggressiveness. The expression of a subset of these genes was evaluated in human MTCs and found to be associated with aggressive RET-M918T-positive tumors. Altogether, p27 haploinsufficiency in MENX rats uncovered a novel, representative model of invasive and metastatic MTC exploitable for translational studies of this often aggressive and incurable cancer.
Collapse
Affiliation(s)
- Sara Molatore
- Institute for Diabetes and CancerHelmholtz Zentrum München, Neuherberg, Germany
| | - Andrea Kügler
- Institute for Diabetes and CancerHelmholtz Zentrum München, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental GeneticsHelmholtz Zentrum München, Neuherberg, Germany
| | - Tobias Wiedemann
- Institute for Diabetes and CancerHelmholtz Zentrum München, Neuherberg, Germany
| | - Frauke Neff
- Institute of Experimental GeneticsHelmholtz Zentrum München, Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical PathologyHelmholtz Zentrum München, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental GeneticsHelmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD)Neuherberg, Germany
- Technische Universität MünchenChair of Experimental Genetics, Freising, Germany
| | - Mercedes Robledo
- Hereditary Endocrine Cancer GroupSpanish National Cancer Research Centre (CNIO) and ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Federico Roncaroli
- Division of Neuroscience and Experimental PsychologyFaculty of Medicine, University of Manchester, Manchester, UK
| | - Natalia S Pellegata
- Institute for Diabetes and CancerHelmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
24
|
Frigola J, Iturbide A, Lopez-Bigas N, Peiro S, Gonzalez-Perez A. Altered oncomodules underlie chromatin regulatory factors driver mutations. Oncotarget 2017; 7:30748-59. [PMID: 27095575 PMCID: PMC5058714 DOI: 10.18632/oncotarget.8752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/31/2016] [Indexed: 11/25/2022] Open
Abstract
Chromatin regulatory factors (CRFs), are known to be involved in tumorigenesis in several cancer types. Nevertheless, the molecular mechanisms through which driver alterations of CRFs cause tumorigenesis remain unknown. Here, we developed a CRFs Oncomodules Discovery approach, which mines several sources of cancer genomics and perturbaomics data. The approach prioritizes sets of genes significantly miss-regulated in primary tumors (oncomodules) bearing mutations of driver CRFs. We applied the approach to eleven TCGA tumor cohorts and uncovered oncomodules potentially associated to mutations of five driver CRFs in three cancer types. Our results revealed, for example, the potential involvement of the mTOR pathway in the development of tumors with loss-of-function mutations of MLL2 in head and neck squamous cell carcinomas. The experimental validation that MLL2 loss-of-function increases the sensitivity of cancer cell lines to mTOR inhibition lends further support to the validity of our approach. The potential oncogenic modules detected by our approach may guide experiments proposing ways to indirectly target driver mutations of CRFs.
Collapse
Affiliation(s)
- Joan Frigola
- Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| | - Ane Iturbide
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain
| | - Nuria Lopez-Bigas
- Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Sandra Peiro
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain
| | - Abel Gonzalez-Perez
- Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| |
Collapse
|
25
|
Wang L, Peng Z, Wang K, Qi Y, Yang Y, Zhang Y, An X, Luo S, Zheng J. NDUFA4L2 is associated with clear cell renal cell carcinoma malignancy and is regulated by ELK1. PeerJ 2017; 5:e4065. [PMID: 29158991 PMCID: PMC5695248 DOI: 10.7717/peerj.4065] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 10/29/2017] [Indexed: 01/04/2023] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is the most common and lethal cancer of the adult kidney. However, its pathogenesis has not been fully understood till now, which hinders the therapeutic development of ccRCC. NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4-like 2 (NDUFA4L2) was found to be upregulated and play an important role in ccRCC. We aimed to further investigate the underlying mechanisms by which NDUFA4L2 exerted function and its expression level was upregulated. Methods The Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) data were mined to verify the change of NDUFA4L2 expression level in ccRCC tissues. The correlation between expression level of NDUFA4L2 and cell proliferation/apoptosis was explored by Gene Set Enrichment Analysis (GSEA). Protein-protein interaction (PPI) network of NDUFA4L2 was constructed. Biological process and involved pathways of NDUFA4L2 were analyzed by gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway. The transcription factors (TFs) which can induce the expression of NDUFA4L2 were explored in clinical samples by correlation analysis and its regulation on the expression of NDUFA4L2 was verified by knockdown experiment. Results NDUFA4L2 was verified to be overexpressed in ccRCC tissues and its expression level was increased accordingly as the American Joint Committee on Cancer (AJCC) stage progressed. A high NDUFA4L2 level predicted the poor prognosis of ccRCC patients and correlated with enhanced cell proliferation and anti-apoptosis. NDUFA4L2 may interact with 14 tumor-related proteins, participate in growth and death processes and be involved in ccRCC-related pathways, such as insulin-like growth factor 1 (IGF-1), mammalian target of Rapamycin (mTOR) and phosphoinositide 3 kinase serine/threonine protein kinase (PI3K/AKT). ETS domain-containing protein ELK1 level positively correlated with the level of NDUFA4L2 in ccRCC tissues and ELK1 could regulate the expression of NDUFA4L2 in ccRCC cells. Discussion NDUFA4L2 upregulation was associated with ccRCC malignancy. NDUFA4L2 expression was regulated by ELK1 in ccRCC cells. Our study provided potential mechanisms by which NDUFA4L2 affected ccRCC occurrence and progression.
Collapse
Affiliation(s)
- Lei Wang
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhiqiang Peng
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Kaizhen Wang
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Yijun Qi
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Ying Yang
- Core Facilities Center, Capital Medical University, Beijing, China
| | - Yue Zhang
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Xinyuan An
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Shudong Luo
- Key Laboratory of Biology of Insect-Pollinator, Ministry of Agriculture, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Junfang Zheng
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Tumor Invasion and Metastasis, Cancer Institute of Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Sbiera S, Sbiera I, Ruggiero C, Doghman-Bouguerra M, Korpershoek E, de Krijger RR, Ettaieb H, Haak H, Volante M, Papotti M, Reimondo G, Terzolo M, Luconi M, Nesi G, Mannelli M, Libé R, Ragazzon B, Assié G, Bertherat J, Altieri B, Fadda G, Rogowski-Lehmann N, Reincke M, Beuschlein F, Fassnacht M, Lalli E. Assessment of VAV2 Expression Refines Prognostic Prediction in Adrenocortical Carcinoma. J Clin Endocrinol Metab 2017; 102:3491-3498. [PMID: 28911143 DOI: 10.1210/jc.2017-00984] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/28/2017] [Indexed: 11/19/2022]
Abstract
CONTEXT Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with overall poor prognosis. The Ki67 labeling index (LI) has a major prognostic role in localized ACC after complete resection, but its estimates may suffer from considerable intra- and interobserver variability. VAV2 overexpression induced by increased Steroidogenic Factor-1 dosage is an essential factor driving ACC tumor cell invasion. OBJECTIVE To assess the prognostic role of VAV2 expression in ACC by investigation of a large cohort of patients. DESIGN, SETTING, AND PARTICIPANTS A total of 171 ACC cases (157 primary tumors, six local recurrences, eight metastases) from seven European Network for the Study of Adrenal Tumors centers were studied. OUTCOME MEASUREMENTS H-scores were generated to quantify VAV2 expression. VAV2 expression was divided into two categories: low (H-score, <2) and high (H-score, ≥2). The Ki67 LI retrieved from patients' pathology records was also categorized into low (<20%) and high (≥20%). Clinical and immunohistochemical markers were correlated with progression-free survival (PFS) and overall survival (OS). RESULTS VAV2 expression and Ki67 LI were significantly correlated with each other and with PFS and OS. Heterogeneity of VAV2 expression inside the same tumor was very low. Combined assessment of VAV2 expression and Ki67 LI improved patient stratification to low-risk and high-risk groups. CONCLUSION Combined assessment of Ki67 LI and VAV2 expression improves prognostic prediction in ACC.
Collapse
Affiliation(s)
- Silviu Sbiera
- Department of Internal Medicine I - Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Wurzburg, Germany
| | - Iuliu Sbiera
- Department of Internal Medicine I - Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Wurzburg, Germany
| | - Carmen Ruggiero
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France
- CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France
- NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| | - Mabrouka Doghman-Bouguerra
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France
- CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France
- NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| | - Esther Korpershoek
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Ronald R de Krijger
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Pathology, Reinier de Graaf Hospital, 2625 AD Delft, The Netherlands
| | - Hester Ettaieb
- Department of Internal Medicine, Máxima Medical Centre, 5631 BM Eindhoven/Veldhoven, The Netherlands
| | - Harm Haak
- Department of Internal Medicine, Máxima Medical Centre, 5631 BM Eindhoven/Veldhoven, The Netherlands
- Department of Internal Medicine, Division of General Internal Medicine, Maastricht University Medical Centre+, 6202 AZ Maastricht, The Netherlands
- Maastricht University, CAPHRI School for Public Health and Primary Care, Ageing and Long-Term Care, 6200 MD Maastricht, The Netherlands
| | - Marco Volante
- Department of Oncology, University of Turin at San Luigi Hospital, 10043 Orbassano, Italy
| | - Mauro Papotti
- Department of Oncology, University of Turin at San Luigi Hospital, 10043 Orbassano, Italy
| | - Giuseppe Reimondo
- Department of Clinical and Biological Sciences, University of Turin at San Luigi Hospital, 10043 Orbassano, Italy
| | - Massimo Terzolo
- Department of Clinical and Biological Sciences, University of Turin at San Luigi Hospital, 10043 Orbassano, Italy
| | - Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, 50139 Florence, Italy
| | - Gabriella Nesi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, 50139 Florence, Italy
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, 50139 Florence, Italy
| | - Rossella Libé
- Inserm U1016, Institut Cochin, 75014 Paris, France
- CNRS UMR8104, 75014 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Bruno Ragazzon
- Inserm U1016, Institut Cochin, 75014 Paris, France
- CNRS UMR8104, 75014 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Guillaume Assié
- Inserm U1016, Institut Cochin, 75014 Paris, France
- CNRS UMR8104, 75014 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Jérôme Bertherat
- Inserm U1016, Institut Cochin, 75014 Paris, France
- CNRS UMR8104, 75014 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Barbara Altieri
- Department of Internal Medicine I - Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Wurzburg, Germany
- Division of Endocrinology and Metabolic Diseases, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Guido Fadda
- Division of Anatomic Pathology and Histology, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | | | - Martin Reincke
- Medizinische Klinik and Poliklinik IV, Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Felix Beuschlein
- Medizinische Klinik and Poliklinik IV, Ludwig-Maximilians-Universität, 80336 Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, 8091 Zurich, Switzerland
| | - Martin Fassnacht
- Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Wurzburg, Germany
| | - Enzo Lalli
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France
- CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France
- NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France
- Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| |
Collapse
|
27
|
Ruggiero C, Doghman-Bouguerra M, Sbiera S, Sbiera I, Parsons M, Ragazzon B, Morin A, Robidel E, Favier J, Bertherat J, Fassnacht M, Lalli E. Dosage-dependent regulation of VAV2 expression by steroidogenic factor-1 drives adrenocortical carcinoma cell invasion. Sci Signal 2017; 10:10/469/eaal2464. [PMID: 28270555 DOI: 10.1126/scisignal.aal2464] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with a dismal prognosis. Genomic studies have enabled progress in our understanding of the molecular bases of ACC, but factors that influence its prognosis are lacking. Amplification of the gene encoding the transcription factor steroidogenic factor-1 (SF-1; also known as NR5A1) is one of the genetic alterations common in ACC. We identified a transcriptional regulatory mechanism involving increased abundance of VAV2, a guanine nucleotide exchange factor for small GTPases that control the cytoskeleton, driven by increased expression of the gene encoding SF-1 in ACC. Manipulating SF-1 and VAV2 abundance in cultured ACC cells revealed that VAV2 was a critical factor for SF-1-induced cytoskeletal remodeling and invasion in culture (Matrigel) and in vivo (chicken chorioallantoic membrane) models. Analysis of ACC patient cohorts indicated that greater VAV2 abundance robustly correlated with poor prognosis in ACC patients. Because VAV2 is a druggable target, our findings suggest that blocking VAV2 may be a new therapeutic approach to inhibit metastatic progression in ACC patients.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France.,CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France.,NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France.,Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| | - Mabrouka Doghman-Bouguerra
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France.,CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France.,NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France.,Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Iuliu Sbiera
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, U.K
| | - Bruno Ragazzon
- Inserm, U1016, Institut Cochin, 75014 Paris, France.,CNRS UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France
| | - Aurélie Morin
- Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France.,Inserm, UMR970, Paris Cardiovascular Research Centre, 75015 Paris, France
| | - Estelle Robidel
- Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France.,Inserm, UMR970, Paris Cardiovascular Research Centre, 75015 Paris, France
| | - Judith Favier
- Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France.,Inserm, UMR970, Paris Cardiovascular Research Centre, 75015 Paris, France
| | - Jérôme Bertherat
- Inserm, U1016, Institut Cochin, 75014 Paris, France.,CNRS UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 74014 Paris, France
| | - Martin Fassnacht
- Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Würzburg, Germany
| | - Enzo Lalli
- Université Côte d'Azur, Sophia Antipolis, 06560 Valbonne, France. .,CNRS UMR7275, Sophia Antipolis, 06560 Valbonne, France.,NEOGENEX CNRS International Associated Laboratory, Sophia Antipolis, 06560 Valbonne, France.,Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, 06560 Valbonne, France
| |
Collapse
|
28
|
Doghman-Bouguerra M, Lalli E. The ER-mitochondria couple: In life and death from steroidogenesis to tumorigenesis. Mol Cell Endocrinol 2017; 441:176-184. [PMID: 27594532 DOI: 10.1016/j.mce.2016.08.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 02/07/2023]
Abstract
Steroidogenesis is a multistep process where interorganelle communications between the endoplasmic reticulum and mitochondria are critical. These intimate interactions physically occur through the Mitochondria-Associated ER membranes called MAMs. MAMs play important roles in mitochondrial morphology and in many cellular functions ranging from lipid metabolism, to calcium signaling and apoptosis together with a critical effect on steroidogenesis. Moreover, our recent characterization of new MAM resident proteins in adrenocortical cells extends the function of MAM in the mechanism of resistance of cancer cells to apoptotic stimuli and offers new perspectives in targeted therapeutic approaches for adrenocortical tumorigenesis.
Collapse
Affiliation(s)
- Mabrouka Doghman-Bouguerra
- Université Côte d'Azur, France; CNRS UMR 7275, France; NEOGENEX CNRS International Associated Laboratory, France; Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), France.
| | - Enzo Lalli
- Université Côte d'Azur, France; CNRS UMR 7275, France; NEOGENEX CNRS International Associated Laboratory, France; Inserm, France; Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), France
| |
Collapse
|
29
|
Affiliation(s)
- Mabrouka Doghman-Bouguerra
- a IPMC, Université Côte d'Azur , Valbonne , France.,b CNRS UMR7275 , Valbonne , France.,c NEOGENEX CNRS International Associated Laboratory , Valbonne , France.,d Institut de Pharmacologie Moléculaire et Cellulaire , Sophia Antipolis, Valbonne , France
| | - Enzo Lalli
- a IPMC, Université Côte d'Azur , Valbonne , France.,b CNRS UMR7275 , Valbonne , France.,c NEOGENEX CNRS International Associated Laboratory , Valbonne , France.,d Institut de Pharmacologie Moléculaire et Cellulaire , Sophia Antipolis, Valbonne , France
| |
Collapse
|
30
|
Doghman-Bouguerra M, Granatiero V, Sbiera S, Sbiera I, Lacas-Gervais S, Brau F, Fassnacht M, Rizzuto R, Lalli E. FATE1 antagonizes calcium- and drug-induced apoptosis by uncoupling ER and mitochondria. EMBO Rep 2016; 17:1264-80. [PMID: 27402544 PMCID: PMC5007562 DOI: 10.15252/embr.201541504] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 06/10/2016] [Accepted: 06/16/2016] [Indexed: 11/09/2022] Open
Abstract
Several stimuli induce programmed cell death by increasing Ca(2+) transfer from the endoplasmic reticulum (ER) to mitochondria. Perturbation of this process has a special relevance in pathologies as cancer and neurodegenerative disorders. Mitochondrial Ca(2+) uptake mainly takes place in correspondence of mitochondria-associated ER membranes (MAM), specialized contact sites between the two organelles. Here, we show the important role of FATE1, a cancer-testis antigen, in the regulation of ER-mitochondria distance and Ca(2+) uptake by mitochondria. FATE1 is localized at the interface between ER and mitochondria, fractionating into MAM FATE1 expression in adrenocortical carcinoma (ACC) cells under the control of the transcription factor SF-1 decreases ER-mitochondria contact and mitochondrial Ca(2+) uptake, while its knockdown has an opposite effect. FATE1 also decreases sensitivity to mitochondrial Ca(2+)-dependent pro-apoptotic stimuli and to the chemotherapeutic drug mitotane. In patients with ACC, FATE1 expression in their tumor is inversely correlated with their overall survival. These results show that the ER-mitochondria uncoupling activity of FATE1 is harnessed by cancer cells to escape apoptotic death and resist the action of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Mabrouka Doghman-Bouguerra
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275 Sophia Antipolis, Valbonne, France NEOGENEX CNRS International Associated Laboratory, Valbonne, France University of Nice - Sophia Antipolis, Valbonne, France
| | - Veronica Granatiero
- Department of Biomedical Sciences, University of Padova, Padova, Italy CNR Neuroscience Institute, Padova, Italy
| | - Silviu Sbiera
- Department of Internal Medicine I - Endocrine Unit, University Hospital University of Würzburg, Würzburg, Germany
| | - Iuliu Sbiera
- Department of Internal Medicine I - Endocrine Unit, University Hospital University of Würzburg, Würzburg, Germany
| | | | - Frédéric Brau
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275 Sophia Antipolis, Valbonne, France University of Nice - Sophia Antipolis, Valbonne, France
| | - Martin Fassnacht
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padova, Italy CNR Neuroscience Institute, Padova, Italy
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275 Sophia Antipolis, Valbonne, France NEOGENEX CNRS International Associated Laboratory, Valbonne, France University of Nice - Sophia Antipolis, Valbonne, France
| |
Collapse
|
31
|
Jiang DN, Yang HH, Li MH, Shi HJ, Zhang XB, Wang DS. gsdf
is a downstream gene of dmrt1
that functions in the male sex determination pathway of the Nile tilapia. Mol Reprod Dev 2016; 83:497-508. [DOI: 10.1002/mrd.22642] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/24/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Dong-Neng Jiang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education); Key Laboratory of Aquatic Science of Chongqing; School of Life Sciences; Southwest University; Beibei Chongqing China
| | - Hui-Hui Yang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education); Key Laboratory of Aquatic Science of Chongqing; School of Life Sciences; Southwest University; Beibei Chongqing China
| | - Ming-Hui Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education); Key Laboratory of Aquatic Science of Chongqing; School of Life Sciences; Southwest University; Beibei Chongqing China
| | - Hong-Juan Shi
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education); Key Laboratory of Aquatic Science of Chongqing; School of Life Sciences; Southwest University; Beibei Chongqing China
| | - Xian-Bo Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education); Key Laboratory of Aquatic Science of Chongqing; School of Life Sciences; Southwest University; Beibei Chongqing China
| | - De-Shou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education); Key Laboratory of Aquatic Science of Chongqing; School of Life Sciences; Southwest University; Beibei Chongqing China
| |
Collapse
|
32
|
Vasquez YM, Wu SP, Anderson ML, Hawkins SM, Creighton CJ, Ray M, Tsai SY, Tsai MJ, Lydon JP, DeMayo FJ. Endometrial Expression of Steroidogenic Factor 1 Promotes Cystic Glandular Morphogenesis. Mol Endocrinol 2016; 30:518-32. [PMID: 27018534 DOI: 10.1210/me.2015-1215] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Epigenetic silencing of steroidogenic factor 1 (SF1) is lost in endometriosis, potentially contributing to de novo local steroidogenesis favoring inflammation and growth of ectopic endometrial tissue. In this study, we examine the impact of SF1 expression in the eutopic uterus by a novel mouse model that conditionally expresses SF1 in endometrium. In vivo SF1 expression promoted the development of enlarged endometrial glands and attenuated estrogen and progesterone responsiveness. Endometriosis induction by autotransplantation of uterine tissue to the mesenteric membrane resulted in the increase in size of ectopic lesions from SF1-expressing mice. By integrating the SF1-dependent transcriptome with the whole genome binding profile of SF1, we identified uterine-specific SF1-regulated genes involved in Wingless and Progesterone receptor-Hedgehog-Chicken ovalbumin upstream promoter transcription factor II signaling for gland development and epithelium-stroma interaction, respectively. The present results indicate that SF1 directly contributes to the abnormal uterine gland morphogenesis, an inhibition of steroid hormone signaling and activation of an immune response, in addition to previously postulated estrogen production.
Collapse
Affiliation(s)
- Yasmin M Vasquez
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - San-Pin Wu
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Matthew L Anderson
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Shannon M Hawkins
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Chad J Creighton
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Madhumita Ray
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Sophia Y Tsai
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - John P Lydon
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Francesco J DeMayo
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| |
Collapse
|
33
|
Ruggiero C, Doghman M, Lalli E. How genomic studies have improved our understanding of the mechanisms of transcriptional regulation by NR5A nuclear receptors. Mol Cell Endocrinol 2015; 408:138-44. [PMID: 25449416 DOI: 10.1016/j.mce.2014.10.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/27/2014] [Indexed: 02/03/2023]
Abstract
SF-1 and LRH-1 are transcription factors that belong to the NR5A family of nuclear receptors that both have an essential role during development. Recent studies at the genome-wide scale have enabled the characterization of the cistrome and transcriptome regulated by SF-1 and LRH-1 in different cell lines and tissues. Those studies have allowed us to make a significant leap forward in our understanding of the mechanisms of transcriptional regulation of NR5A nuclear receptors in stem cells and cancer.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, Sophia Antipolis, Valbonne, France; Laboratoire International Associé (LIA) CNRS NEOGENEX, Sophia Antipolis, Valbonne, France; Université de Nice, Sophia Antipolis, Valbonne, France
| | - Mabrouka Doghman
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, Sophia Antipolis, Valbonne, France; Laboratoire International Associé (LIA) CNRS NEOGENEX, Sophia Antipolis, Valbonne, France; Université de Nice, Sophia Antipolis, Valbonne, France
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, Sophia Antipolis, Valbonne, France; Laboratoire International Associé (LIA) CNRS NEOGENEX, Sophia Antipolis, Valbonne, France; Université de Nice, Sophia Antipolis, Valbonne, France.
| |
Collapse
|
34
|
Lalli E, Figueiredo BC. Pediatric adrenocortical tumors: what they can tell us on adrenal development and comparison with adult adrenal tumors. Front Endocrinol (Lausanne) 2015; 6:23. [PMID: 25741319 PMCID: PMC4332354 DOI: 10.3389/fendo.2015.00023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 02/08/2015] [Indexed: 12/05/2022] Open
Abstract
Adrenocortical tumors (ACT) in children are very rare and are most frequently diagnosed in the context of the Li-Fraumeni syndrome, a multiple cancer syndrome linked to germline mutations of the tumor suppressor gene TP53 with loss of heterozygosity in the tumors. A peak of children ACT incidence is present in the states of southern Brazil, where they are linked to the high prevalence in the population of a specific TP53 mutation (R337H). Children ACT have specific features distinguishing them from adult tumors in their pathogenetic mechanisms, genomic profiles, and prognosis. Epidemiological and molecular evidence suggests that in most cases they are derived from the fetal adrenal.
Collapse
Affiliation(s)
- Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Valbonne, France
- University of Nice-Sophia-Antipolis, Valbonne, France
- Associated International Laboratory (LIA) NEOGENEX, CNRS, Valbonne, France
- *Correspondence: Enzo Lalli, Institut de Pharmacologie Moléculaire et Cellulaire CNRS, 660 route des Lucioles – Sophia Antipolis, Valbonne 06560, France e-mail: ; Bonald C. Figueiredo, Instituto de Pesquisa Pelé Pequeno Principe, Av. Silva Jardim, 1632, Curitiba, Paraná CEP 80250-060, Brazil e-mail:
| | - Bonald C. Figueiredo
- Associated International Laboratory (LIA) NEOGENEX, CNRS, Valbonne, France
- Federal University of Paraná, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Principe, Curitiba, Brazil
- *Correspondence: Enzo Lalli, Institut de Pharmacologie Moléculaire et Cellulaire CNRS, 660 route des Lucioles – Sophia Antipolis, Valbonne 06560, France e-mail: ; Bonald C. Figueiredo, Instituto de Pesquisa Pelé Pequeno Principe, Av. Silva Jardim, 1632, Curitiba, Paraná CEP 80250-060, Brazil e-mail:
| |
Collapse
|
35
|
Latre de Late P, El Wakil A, Jarjat M, de Krijger RR, Heckert LL, Naquet P, Lalli E. Vanin-1 inactivation antagonizes the development of adrenocortical neoplasia in Sf-1 transgenic mice. Endocrinology 2014; 155:2349-54. [PMID: 24712878 DOI: 10.1210/en.2014-1088] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
SF-1 (NR5A1) overexpression can induce adrenocortical tumor formation in transgenic mice and is associated with more severe prognosis in patients with adrenocortical cancer. In this study we have identified Vanin-1 (Vnn1), a SF-1 target gene, as a novel modulator of the tumorigenic effect of Sf-1 overexpression in the adrenal cortex. Vanin-1 is endowed with pantetheinase activity, releasing cysteamine in tissues and regulating cell response to oxidative stress by modulating the production of glutathione. Sf-1 transgenic mice developed adrenocortical neoplastic lesions (both dysplastic and nodular) with a frequency increasing with age. Genetic ablation of the Vnn1 gene in Sf-1 transgenic mice significantly reduced the severity of neoplastic lesions in the adrenal cortex. This effect could be reversed by treatment of Sf-1 transgenic/Vnn1 null mice with cysteamine. These data show that alteration of the mechanisms controlling intracellular redox and detoxification mechanisms is relevant to the pathogenesis of adrenocortical neoplasia induced by SF-1 overexpression.
Collapse
Affiliation(s)
- Perle Latre de Late
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS (P.L.D.L., A.E.W., M.J., E.L.) Valbonne 06560, France; Associated International Laboratory for the Study of the ALterations of Gene Expression in Childhood Cancers CNRS (P.L.D.L., A.E.W., M.J., E.L.), Valbonne 06560, France; Université de Nice-Sophia Antipolis (P.L.D.L., A.E.W., M.J., E.L.), Valbonne 06560, France; Department of Pathology (R.R.K.), Erasmus MC-University Medical Center, Rotterdam 3015, The Netherlands; Department of Molecular and Integrative Physiology (L.L.H.), University of Kansas Medical Center, Kansas City, Kansas 66160; Centre d'Immunologie de Marseille-Luminy (P.N.), Aix-Marseille University, UM2, Marseille, France; Institut National de la Santé et de la Recherche Médicale (P.N.), U1104, Marseille, France; and Centre National de la Recherche Scientifique (P.N.), UMR7280, Marseille, France
| | | | | | | | | | | | | |
Collapse
|
36
|
Role of Orphan Nuclear Receptor DAX-1/NR0B1 in Development, Physiology, and Disease. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/582749] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
DAX-1/NR0B1 is an unusual orphan receptor that has a pivotal role in the development and function of steroidogenic tissues and of the reproductive axis. Recent studies have also indicated that this transcription factor has an important function in stem cell biology and in several types of cancer. Here I critically review the most important findings on the role of DAX-1 in development, physiology, and disease of endocrine tissues since the cloning of its gene twenty years ago.
Collapse
|
37
|
Imamichi Y, Mizutani T, Ju Y, Matsumura T, Kawabe S, Kanno M, Yazawa T, Miyamoto K. Transcriptional regulation of human ferredoxin reductase through an intronic enhancer in steroidogenic cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1839:33-42. [PMID: 24321386 DOI: 10.1016/j.bbagrm.2013.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 11/15/2013] [Accepted: 11/25/2013] [Indexed: 02/08/2023]
Abstract
Ferredoxin reductase (FDXR, also known as adrenodoxin reductase) is a mitochondrial flavoprotein that transfers electrons from NADPH to mitochondrial cytochrome P450 enzymes, mediating the function of an iron-sulfur cluster protein, ferredoxin. FDXR functions in various metabolic processes including steroidogenesis. It is well known that multiple steroidogenic enzymes are regulated by a transcription factor steroidogenic factor-1 (SF-1, also known as Ad4BP). Previously, we have shown that SF-1 transduction causes human mesenchymal stem cell differentiation into steroidogenic cells. Genome-wide analysis of differentiated cells, using a combination of DNA microarray and promoter tiling array analyses, showed that FDXR is a novel SF-1 target gene. In this study, the transcriptional regulatory mechanism of FDXR was examined in steroidogenic cells. A chromatin immunoprecipitation assay revealed that a novel SF-1 binding region was located within intron 2 of the human FDXR gene. Luciferase reporter assays showed that FDXR transcription was activated through the novel SF-1 binding site within intron 2. Endogenous SF-1 knockdown in human adrenocortical H295R and KGN cells decreased FDXR expression. In H295R cells, strong binding of two histone markers of active enhancers, histones H3K27ac and H3K4me2, were detected near the SF-1 binding site within intron 2. Furthermore, the binding of these histone markers was decreased concurrent with SF-1 knockdown in H295R cells. These results indicated that abundant FDXR expression in these steroidogenic cells was maintained through SF-1 binding to the intronic enhancer of the FDXR gene.
Collapse
Affiliation(s)
- Yoshitaka Imamichi
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan; Translational Research Center, Organization for Life Science Advancement Programs, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan
| | - Tetsuya Mizutani
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan; Translational Research Center, Organization for Life Science Advancement Programs, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan.
| | - Yunfeng Ju
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan
| | - Takehiro Matsumura
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan
| | - Shinya Kawabe
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan; Translational Research Center, Organization for Life Science Advancement Programs, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan
| | - Masafumi Kanno
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan; Translational Research Center, Organization for Life Science Advancement Programs, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan
| | - Kaoru Miyamoto
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan; Translational Research Center, Organization for Life Science Advancement Programs, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji, Fukui 910-1193, Japan
| |
Collapse
|
38
|
Abstract
The nuclear receptor superfamily includes many receptors, identified based on their similarity to steroid hormone receptors but without a known ligand. The study of how these receptors are diversely regulated to interact with genomic regions to control a plethora of biological processes has provided critical insight into development, physiology, and the molecular pathology of disease. Here we provide a compendium of these so-called orphan receptors and focus on what has been learned about their modes of action, physiological functions, and therapeutic promise.
Collapse
Affiliation(s)
- Shannon E Mullican
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|