1
|
Sanders K, Kooistra HS, van den Heuvel M, Mokry M, Grinwis GCM, van den Dungen NAM, van Steenbeek FG, Galac S. Transcriptome sequencing reveals two subtypes of cortisol-secreting adrenocortical tumours in dogs and identifies CYP26B1 as a potential new therapeutic target. Vet Comp Oncol 2023; 21:100-110. [PMID: 36582114 DOI: 10.1111/vco.12871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022]
Abstract
Cushing's syndrome (CS) is a serious endocrine disorder that is relatively common in dogs, but rare in humans. In ~15%-20% of cases, CS is caused by a cortisol-secreting adrenocortical tumour (csACT). To identify differentially expressed genes that can improve prognostic predictions after surgery and represent novel treatment targets, we performed RNA sequencing on csACTs (n = 48) and normal adrenal cortices (NACs; n = 10) of dogs. A gene was declared differentially expressed when the adjusted p-value was <.05 and the log2 fold change was >2 or < -2. Between NACs and csACTs, 98 genes were differentially expressed. Based on the principal component analysis (PCA) the csACTs were separated in two groups, of which Group 1 had significantly better survival after adrenalectomy (p = .002) than Group 2. Between csACT Group G1 and Group 2, 77 genes were differentially expressed. One of these, cytochrome P450 26B1 (CYP26B1), was significantly associated with survival in both our canine csACTs and in a publicly available data set of 33 human cortisol-secreting adrenocortical carcinomas. In the validation cohort, CYP26B1 was also expressed significantly higher (p = .012) in canine csACTs compared with NACs. In future studies it would be interesting to determine whether CYP26B1 inhibitors could inhibit csACT growth in both dogs and humans.
Collapse
Affiliation(s)
- Karin Sanders
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Hans S Kooistra
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marieke van den Heuvel
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Michal Mokry
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Guy C M Grinwis
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Noortje A M van den Dungen
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Frank G van Steenbeek
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sara Galac
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
2
|
Bueno AC, More CB, Marrero-Gutiérrez J, de Almeida E Silva DC, Leal LF, Montaldi AP, Ramalho FS, Vêncio RZN, de Castro M, Antonini SRR. Vitamin D receptor activation is a feasible therapeutic target to impair adrenocortical tumorigenesis. Mol Cell Endocrinol 2022; 558:111757. [PMID: 36049598 DOI: 10.1016/j.mce.2022.111757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To evaluate the therapeutic potential of vitamin D receptor (VDR) signaling in adrenocortical carcinoma (ACC) cells. METHODS We evaluated VDR's methylation pattern in H295R ACC cells, and investigated the effects of calcitriol and seocalcitol treatments on adrenocortical tumorigenesis. RESULTS VDR was hypermethylated and underexpressed in basal H295R cells. Treatments with calcitriol and seocalcitol restored VDR signaling, resulted in antiproliferative effects, and impaired Wnt/B-catenin signaling. RNAseq of treated cells demonstrated VDR activation on steroid hormones biosynthesis and Rap1 signaling, among others. In vivo, seocalcitol constrained the growth of H295R xenografts and reduced autonomous tumor steroid secretion without hypercalcemia-associated side effects. CONCLUSIONS H295R cells present VDR hypermethylation, which can be responsible for its underexpression and signaling inactivation under basal conditions. VDR signaling promoted antiproliferative effects in vitro and in vivo, suggesting that it may be a potential therapeutic target for ACC and a valuable tool for patient's clinical management.
Collapse
Affiliation(s)
- Ana Carolina Bueno
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Candy Bellido More
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Junier Marrero-Gutiérrez
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Danillo C de Almeida E Silva
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Leticia Ferro Leal
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Ana Paula Montaldi
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Fernando Silva Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Ricardo Zorzetto Nicoliello Vêncio
- Department of Computation and Mathematics, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Margaret de Castro
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Sonir Roberto R Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
3
|
Bueno AC, Stecchini MF, Marrero-Gutiérrez J, More CB, Leal LF, Gomes DC, de Lima Neto DF, Brandalise SR, Cardinalli IA, Yunes JA, Junqueira T, Scrideli CA, Molina CAF, Ramalho FS, Tucci S, Coeli-Lacchini FB, Moreira AC, Ramalho L, Vêncio RZN, De Castro M, Antonini SRR. Vitamin D receptor hypermethylation as a biomarker for pediatric adrenocortical tumors. Eur J Endocrinol 2022; 186:573-585. [PMID: 35290212 DOI: 10.1530/eje-21-0879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/14/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Pediatric adrenocortical tumors (pACT) display complex genomic backgrounds, lacking robust prognostic markers and targeted therapeutic options. Vitamin D3 receptor (VDR) promoter hypermethylation and underexpression were reported in adrenocortical carcinomas from adult patients. In this study, we aimed to investigate VDR expression levels and methylation status in pACT and their clinical and prognostic significance. DESIGN Retrospective cross-sectional study enrolling pediatric patients with ACT from two tertiary referral institutions. METHODS We evaluated clinicopathological features, VDR mRNA (qPCR) and protein (immunohistochemistry) expression, and VDR-wide methylation of ACT samples from 108 pediatric patients. Fourteen pediatric and 32 fetal and postnatal normal adrenals were used as controls. RESULTS Unlike in pre- and post-natal normal adrenals, most pACT lacked nuclear VDR expression and had reduced mRNA levels, especially the carcinomas. Unsupervised analysis of VDR methylation data revealed two groups of pACT with distinct disease features and outcomes. Tumors with high VDR methylation presented lower mRNA levels, and the respective patients presented advanced disease and reduced disease-free and overall survival. CONCLUSIONS VDR has a role in normal adrenocortical development and homeostasis, which is impaired during tumorigenesis. VDR hypermethylation and underexpression may be both predictive and prognostic biomarkers for pACT.
Collapse
Affiliation(s)
- Ana Carolina Bueno
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Mônica F Stecchini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Junier Marrero-Gutiérrez
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Candy Bellido More
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Leticia Ferro Leal
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Daniel Ferreira de Lima Neto
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | | | - José Andres Yunes
- Boldrini Children's Center, State University of Campinas, Campinas, Sao Paulo, Brazil
| | - Thais Junqueira
- Boldrini Children's Center, State University of Campinas, Campinas, Sao Paulo, Brazil
| | - Carlos Alberto Scrideli
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Carlos Augusto Fernandes Molina
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Fernando Silva Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Silvio Tucci
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Ayrton Custodio Moreira
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Leandra Ramalho
- Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Ricardo Zorzetto Nicoliello Vêncio
- Department of Computation and Mathematics, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of São Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Margaret De Castro
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Sonir Roberto R Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
4
|
Viëtor CL, Creemers SG, van Kemenade FJ, van Ginhoven TM, Hofland LJ, Feelders RA. How to Differentiate Benign from Malignant Adrenocortical Tumors? Cancers (Basel) 2021; 13:cancers13174383. [PMID: 34503194 PMCID: PMC8431066 DOI: 10.3390/cancers13174383] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Adrenocortical carcinoma is a rare cancer with a poor prognosis. Adrenal tumors are, however, commonly identified in clinical practice. Discrimination between benign and malignant adrenal tumors is of great importance to determine the appropriate treatment and follow-up strategy. This review summarizes the current diagnostic strategies and challenges to distinguish benign from malignant adrenal lesions. We will focus both on radiological and biochemical assessments, enabling diagnosis of the adrenal lesion preoperatively, and on histopathological and a wide variety of molecular assessments that can be done after surgical removal of the adrenal lesion. Furthermore, new non-invasive strategies such as liquid biopsies, in which blood samples are used to study circulating tumor cells, tumor DNA and microRNA, will be addressed in this review. Abstract Adrenocortical carcinoma (ACC) is a rare cancer with a poor prognosis. Adrenal incidentalomas are, however, commonly identified in clinical practice. Discrimination between benign and malignant adrenal tumors is of great importance considering the large differences in clinical behavior requiring different strategies. Diagnosis of ACC starts with a thorough physical examination, biochemical evaluation, and imaging. Computed tomography is the first-level imaging modality in adrenal tumors, with tumor size and Hounsfield units being important features for determining malignancy. New developments include the use of urine metabolomics, also enabling discrimination of ACC from adenomas preoperatively. Postoperatively, the Weiss score is used for diagnosis of ACC, consisting of nine histopathological criteria. Due to known limitations as interobserver variability and lack of accuracy in borderline cases, much effort has been put into new tools to diagnose ACC. Novel developments vary from immunohistochemical markers and pathological scores, to markers at the level of DNA, methylome, chromosome, or microRNA. Molecular studies have provided insights into the most promising and most frequent alterations in ACC. The use of liquid biopsies for diagnosis of ACC is studied, although in a small number of patients, requiring further investigation. In this review, current diagnostic modalities and challenges in ACC will be addressed.
Collapse
Affiliation(s)
- Charlotte L. Viëtor
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015GD Rotterdam, The Netherlands; (C.L.V.); (T.M.v.G.)
| | - Sara G. Creemers
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC University Medical Center, 3015GD Rotterdam, The Netherlands; (S.G.C.); (L.J.H.)
| | - Folkert J. van Kemenade
- Department of Pathology, Erasmus MC University Medical Center, 3015GD Rotterdam, The Netherlands;
| | - Tessa M. van Ginhoven
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015GD Rotterdam, The Netherlands; (C.L.V.); (T.M.v.G.)
| | - Leo J. Hofland
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC University Medical Center, 3015GD Rotterdam, The Netherlands; (S.G.C.); (L.J.H.)
| | - Richard A. Feelders
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC University Medical Center, 3015GD Rotterdam, The Netherlands; (S.G.C.); (L.J.H.)
- Correspondence:
| |
Collapse
|
5
|
Manso J, Sharifi-Rad J, Zam W, Tsouh Fokou PV, Martorell M, Pezzani R. Plant Natural Compounds in the Treatment of Adrenocortical Tumors. Int J Endocrinol 2021; 2021:5516285. [PMID: 34567112 PMCID: PMC8463247 DOI: 10.1155/2021/5516285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/14/2021] [Accepted: 08/31/2021] [Indexed: 01/08/2023] Open
Abstract
Plant natural products are a plethora of diverse and complex molecules produced by the plant secondary metabolism. Among these, many can reserve beneficial or curative properties when employed to treat human diseases. Even in cancer, they can be successfully used and indeed numerous phytochemicals exert antineoplastic activity. The most common molecules derived from plants and used in the fight against cancer are polyphenols, i.e., quercetin, genistein, resveratrol, curcumin, etc. Despite valuable data especially in preclinical models on such compounds, few of them are currently used in the medical practice. Also, in adrenocortical tumors (ACT), phytochemicals are scarcely or not at all used. This work summarizes the available research on phytochemicals used against ACT and adrenocortical cancer, a very rare disease with poor prognosis and high metastatic potential, and wants to contribute to stimulate preclinical and clinical research to find new therapeutic strategies among the overabundance of biomolecules produced by the plant kingdom.
Collapse
Affiliation(s)
- Jacopo Manso
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Via Ospedale 105, Padova 35128, Italy
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Wissam Zam
- Analytical and Food Chemistry Department, Faculty of Pharmacy, Tartous University, Tartous, Syria
| | | | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, Centre for Healthy Living, University of Concepción, Concepción 4070386, Chile
- Universidad de Concepción, Unidad de Desarrollo Tecnológico, UDT, Concepción 4070386, Chile
| | - Raffaele Pezzani
- Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Via Ospedale 105, Padova 35128, Italy
- Phytotherapy Lab, Endocrinology Unit, Department of Medicine (DIMED), University of Padova, via Ospedale 105, 35128 Padova, Italy
| |
Collapse
|
6
|
Surakhy M, Wallace M, Bond E, Grochola LF, Perez H, Di Giovannantonio M, Zhang P, Malkin D, Carter H, Parise IZS, Zambetti G, Komechen H, Paraizo MM, Pagadala MS, Pinto EM, Lalli E, Figueiredo BC, Bond GL. A common polymorphism in the retinoic acid pathway modifies adrenocortical carcinoma age-dependent incidence. Br J Cancer 2020; 122:1231-1241. [PMID: 32147670 PMCID: PMC7156685 DOI: 10.1038/s41416-020-0764-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 01/09/2020] [Accepted: 02/04/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have enriched the fields of genomics and drug development. Adrenocortical carcinoma (ACC) is a rare cancer with a bimodal age distribution and inadequate treatment options. Paediatric ACC is frequently associated with TP53 mutations, with particularly high incidence in Southern Brazil due to the TP53 p.R337H (R337H) germline mutation. The heterogeneous risk among carriers suggests other genetic modifiers could exist. METHODS We analysed clinical, genotype and gene expression data derived from paediatric ACC, R337H carriers, and adult ACC patients. We restricted our analyses to single nucleotide polymorphisms (SNPs) previously identified in GWASs to associate with disease or human traits. RESULTS A SNP, rs971074, in the alcohol dehydrogenase 7 gene significantly and reproducibly associated with allelic differences in ACC age-of-onset in both cohorts. Patients homozygous for the minor allele were diagnosed up to 16 years earlier. This SNP resides in a gene involved in the retinoic acid (RA) pathway and patients with differing levels of RA pathway gene expression in their tumours associate with differential ACC progression. CONCLUSIONS These results identify a novel genetic component to ACC development that resides in the retinoic acid pathway, thereby informing strategies to develop management, preventive and therapeutic treatments for ACC.
Collapse
Affiliation(s)
- Mirvat Surakhy
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Marsha Wallace
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Elisabeth Bond
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Lukasz Filip Grochola
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Department of Surgery, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Husein Perez
- Faculty of Technology, Design and Environment, Oxford Brookes University, Oxford, UK
| | - Matteo Di Giovannantonio
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ping Zhang
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - David Malkin
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, USA
| | - Ivy Zortea S Parise
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Gerard Zambetti
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Heloisa Komechen
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Mariana M Paraizo
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Meghana S Pagadala
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, USA
| | - Emilia M Pinto
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Université Côte D'Azur, Inserm, Valbonne, France
| | - Bonald C Figueiredo
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil.
- Departamento de Saúde Coletiva, Universidade Federal do Paraná, Curitiba, PR, Brazil.
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Curitiba, PR, Brazil.
| | - Gareth L Bond
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
Xu WH, Wu J, Wang J, Wan FN, Wang HK, Cao DL, Qu YY, Zhang HL, Ye DW. Screening and Identification of Potential Prognostic Biomarkers in Adrenocortical Carcinoma. Front Genet 2019; 10:821. [PMID: 31572440 PMCID: PMC6749084 DOI: 10.3389/fgene.2019.00821] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 08/08/2019] [Indexed: 12/27/2022] Open
Abstract
Objective: Adrenocortical carcinoma (ACC) is a rare but aggressive malignant cancer that has been attracting growing attention over recent decades. This study aims to integrate protein interaction networks with gene expression profiles to identify potential biomarkers with prognostic value in silico. Methods: Three microarray data sets were downloaded from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) according to the normalization annotation information. Enrichment analyses were utilized to describe biological functions. A protein-protein interaction network (PPI) of the DEGs was developed, and the modules were analyzed using STRING and Cytoscape. LASSO Cox regression was used to identify independent prognostic factors. The Kaplan-Meier method for the integrated expression score was applied to analyze survival outcomes. A receiver operating characteristic (ROC) curve was constructed with area under curve (AUC) analysis to determine the diagnostic ability of the candidate biomarkers. Results: A total of 150 DEGs and 24 significant hub genes with functional enrichment were identified as candidate prognostic biomarkers. LASSO Cox regression suggested that ZWINT, PRC1, CDKN3, CDK1 and CCNA2 were independent prognostic factors in ACC. In multivariate Cox analysis, the integrated expression scores of the modules showed statistical significance in predicting disease-free survival (DFS, P = 0.019) and overall survival (OS, P < 0.001). Meanwhile, ROC curves were generated to validate the ability of the Cox model to predict prognosis. The AUC index for the integrated genes scores was 0.861 (P < 0.0001). Conclusion: In conclusion, the present study identifies DEGs and hub genes that may be involved in poor prognosis and early recurrence of ACC. The expression levels of ZWINT, PRC1, CDKN3, CDK1 and CCNA2 are of high prognostic value, and may help us understand better the underlying carcinogenesis or progression of ACC. Further studies are required to elucidate molecular pathogenesis and alteration in signaling pathways for these genes in ACC.
Collapse
Affiliation(s)
- Wen-Hao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang-Ning Wan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Kai Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Da-Long Cao
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan-Yuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hai-Liang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding-Wei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Pereira SS, Monteiro MP, Antonini SR, Pignatelli D. Apoptosis regulation in adrenocortical carcinoma. Endocr Connect 2019; 8:R91-R104. [PMID: 30978697 PMCID: PMC6510712 DOI: 10.1530/ec-19-0114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Apoptosis evading is a hallmark of cancer. Tumor cells are characterized by having an impaired apoptosis signaling, a fact that deregulates the balance between cell death and survival, leading to tumor development, invasion and resistance to treatment. In general, patients with adrenocortical carcinomas (ACC) have an extremely bad prognosis, which is related to disease progression and significant resistance to treatments. In this report, we performed an integrative review about the disruption of apoptosis in ACC that may underlie the characteristic poor prognosis in these patients. Although the apoptosis has been scarcely studied in ACC, the majority of the deregulation phenomena already described are anti-apoptotic. Most importantly, in a near future, targeting apoptosis modulation in ACC patients may become a promising therapeutic.
Collapse
Affiliation(s)
- Sofia S Pereira
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Endocrine, Cardiovascular & Metabolic Research, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal
| | - Mariana P Monteiro
- Endocrine, Cardiovascular & Metabolic Research, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal
| | - Sonir R Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Duarte Pignatelli
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Department of Endocrinology, Hospital S. João, Porto, Portugal
- Correspondence should be addressed to D Pignatelli:
| |
Collapse
|
9
|
Xiao H, Xu D, Chen P, Zeng G, Wang X, Zhang X. Identification of Five Genes as a Potential Biomarker for Predicting Progress and Prognosis in Adrenocortical Carcinoma. J Cancer 2018; 9:4484-4495. [PMID: 30519354 PMCID: PMC6277665 DOI: 10.7150/jca.26698] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022] Open
Abstract
Background: Adrenocortical carcinoma (ACC) is a limited endocrine fatality with a minor diagnosis and rare remedial options. The progressive and predictive meaning of message RNA (mRNA) expression oddity in ACC has been studied extensively in recent years. However, differences in measurement platforms and lab protocols as well as small sample sizes can render gene expression levels incomparable. Methods: An extensive study of GEO datasets was conducted to define potential mRNA biomarkers for ACC. The study compared the mRNA expression profiles of ACC tissues and neighboring noncancerous adrenal tissues in the pair. The study covered a sum of 165 tumors and 36 benign control samples. Hub genes were identified through a protein-protein interaction (PPI) network and Robust Rank Aggregation method. Then the Cancer Genome Atlas (TCGA) and Oncomine database were used to perform the validation of hub genes. 4 ACC tissues and 4 normal tissues were collected and then Polymerase Chain Reaction (PCR), Western-blot and immunofluorescence were conducted to validate the expression of five hub genes. Results: We identified five statistically significant genes (TOP2A, NDC80, CEP55, CDKN3, CDK1) corrected with clinical features. The expression of five hub genes in TCGA and Oncomine database were significantly overexpressed in ACC compared with normal ones. Among all the TCGA ACC cases, the strong expression of TOP2A (logrank p=1.4e-04, HR=4.7), NDC80 (logrank p=8.8e-05, HR=4.9), CEP55 (logrank p=5.2e-07, HR=8.6), CDKN3 (log rank p=2.3e-06, HR=7.6) and CDK1 (logrank p=7e-08, HR=11) were correlated with low comprehensive survival, disease free survival (logrank p < 0.001), pathology stage and pathology T stage (FDR < 0.001). PCR results showed that the transcriptional levels of these five genes were significantly higher in ACC tissues than in normal tissues. The western blotting results also showed that the translational level of TOP2A was significantly higher in tumor tissues than in normal tissues. The results of immunofluorescence showed that TOP2A was abundantly observed in the adrenal cortical cell membrane and nucleus and its expression in ACC tissues was significantly higher than that in normal tissues. Conclusions: The distinguished five genes may be utilized to form a board of progressive and predictive biomarkers for ACC for clinical purpose.
Collapse
Affiliation(s)
- He Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Deqiang Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Guang Zeng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China.,Biomedical Engineering, Stony Brook University, New York 11790
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Xinhua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| |
Collapse
|
10
|
Pereira SS, Monteiro MP, Bourdeau I, Lacroix A, Pignatelli D. MECHANISMS OF ENDOCRINOLOGY: Cell cycle regulation in adrenocortical carcinoma. Eur J Endocrinol 2018; 179:R95-R110. [PMID: 29773584 DOI: 10.1530/eje-17-0976] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/15/2018] [Indexed: 12/27/2022]
Abstract
Adrenocortical carcinomas (ACCs) are rather rare endocrine tumors that often have a poor prognosis. The reduced survival rate associated with these tumors is due to their aggressive biological behavior, combined with the scarcity of effective treatment options that are currently available. The recent identification of the genomic alterations present in ACC have provided further molecular mechanisms to develop consistent strategies for the diagnosis, prevention of progression and treatment of advanced ACCs. Taken together, molecular and genomic advances could be leading the way to develop personalized medicine in ACCs similarly to similar developments in lung or breast cancers. In this review, we focused our attention to systematically compile and summarize the alterations in the cell cycle regulation that were described so far in ACC as they are known to play a crucial role in cell differentiation and growth. We have divided the analysis according to the major transition phases of the cell cycle, G1 to S and G2 to M. We have analyzed the most extensively studied checkpoints: the p53/Rb1 pathway, CDC2/cyclin B and topoisomerases (TOPs). We reached the conclusion that the most important alterations having a potential application in clinical practice are the ones related to p53/Rb1 and TOP 2. We also present a brief description of on-going clinical trials based on molecular alterations in ACC. The drugs have targeted the insulin-like growth factor receptor 1, TOP 2, polo-like kinase1, cyclin-dependent kinase inhibitors, p53 reactivation and CDC25.
Collapse
Affiliation(s)
- Sofia S Pereira
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Clinical and Experimental Endocrinology, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal
| | - Mariana P Monteiro
- Clinical and Experimental Endocrinology, Department of Anatomy, Multidisciplinary Unit for Biomedical Research (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal
| | - Isabelle Bourdeau
- Endocrinology Division, Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Canada
| | - André Lacroix
- Endocrinology Division, Department of Medicine, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Canada
| | - Duarte Pignatelli
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Department of Endocrinology, Hospital S. João, Porto, Portugal
| |
Collapse
|
11
|
Bonnet-Serrano F, Bertherat J. Genetics of tumors of the adrenal cortex. Endocr Relat Cancer 2018; 25:R131-R152. [PMID: 29233839 DOI: 10.1530/erc-17-0361] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/12/2017] [Indexed: 01/23/2023]
Abstract
This review describes the molecular alterations observed in the various types of tumors of the adrenal cortex, excluding Conn adenomas, especially the alterations identified by genomic approaches these last five years. Two main forms of bilateral adrenocortical tumors can be distinguished according to size and aspect of the nodules: primary pigmented nodular adrenal disease (PPNAD), which can be sporadic or part of Carney complex and primary bilateral macro nodular adrenal hyperplasia (PBMAH). The bilateral nature of the tumors suggests the existence of an underlying genetic predisposition. PPNAD and Carney complex are mainly due to germline-inactivating mutations of PRKAR1A, coding for a regulatory subunit of PKA, whereas PBMAH genetic seems more complex. However, genome-wide approaches allowed the identification of a new tumor suppressor gene, ARMC5, whose germline alteration could be responsible for at least 25% of PBMAH cases. Unilateral adrenocortical tumors are more frequent, mostly adenomas. The Wnt/beta-catenin pathway can be activated in both benign and malignant tumors by CTNNB1 mutations and by ZNRF3 inactivation in adrenal cancer (ACC). Some other signaling pathways are more specific of the tumor dignity. Thus, somatic mutations of cAMP/PKA pathway genes, mainly PRKACA, coding for the catalytic alpha-subunit of PKA, are found in cortisol-secreting adenomas, whereas IGF-II overexpression and alterations of p53 signaling pathway are observed in ACC. Genome-wide approaches including transcriptome, SNP, methylome and miRome analysis have identified new genetic and epigenetic alterations and the further clustering of ACC in subgroups associated with different prognosis, allowing the development of new prognosis markers.
Collapse
Affiliation(s)
- Fidéline Bonnet-Serrano
- Institut CochinINSERM U1016, CNRS UMR8104, Paris Descartes University, Paris, France
- Hormonal Biology LaboratoryAssistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Jérôme Bertherat
- Institut CochinINSERM U1016, CNRS UMR8104, Paris Descartes University, Paris, France
- Department of EndocrinologyAssistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| |
Collapse
|
12
|
Brossaud J, Pallet V, Corcuff JB. Vitamin A, endocrine tissues and hormones: interplay and interactions. Endocr Connect 2017; 6:R121-R130. [PMID: 28720593 PMCID: PMC5551430 DOI: 10.1530/ec-17-0101] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/03/2017] [Accepted: 07/18/2017] [Indexed: 12/12/2022]
Abstract
Vitamin A (retinol) is a micronutrient critical for cell proliferation and differentiation. In adults, vitamin A and metabolites such as retinoic acid (RA) play major roles in vision, immune and brain functions, and tissue remodelling and metabolism. This review presents the physiological interactions of retinoids and endocrine tissues and hormonal systems. Two endocrine systems have been particularly studied. In the pituitary, retinoids targets the corticotrophs with a possible therapeutic use in corticotropinomas. In the thyroid, retinoids interfere with iodine metabolism and vitamin A deficiency aggravates thyroid dysfunction caused by iodine-deficient diets. Retinoids use in thyroid cancer appears less promising than expected. Recent and still controversial studies investigated the relations between retinoids and metabolic syndrome. Indeed, retinoids contribute to pancreatic development and modify fat and glucose metabolism. However, more detailed studies are needed before planning any therapeutic use. Finally, retinoids probably play more minor roles in adrenal and gonads development and function apart from their major effects on spermatogenesis.
Collapse
Affiliation(s)
- Julie Brossaud
- J Brossaud, Nuclear Medicine, University hospital of Bordeaux, Pessac, France
| | - Veronique Pallet
- V Pallet, NutriNeurO-INRA 1286 - Université Bdx 2, University of Bordeaux, Bordeaux, 33076 BORDEAUX , France
| | - Jean-Benoit Corcuff
- J Corcuff, Nuclear Medicine, University hospital of Bordeaux, Pessac, 33604, France
| |
Collapse
|
13
|
Pennanen M, Hagström J, Heiskanen I, Sane T, Mustonen H, Arola J, Haglund C. C-myc expression in adrenocortical tumours. J Clin Pathol 2017; 71:129-134. [PMID: 28801349 DOI: 10.1136/jclinpath-2017-204503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/29/2017] [Accepted: 06/08/2017] [Indexed: 11/04/2022]
Abstract
AIMS Widespread use of high-resolution imaging techniques and thus increased prevalence of adrenal lesions has made diagnostics of adrenocortical tumours an increasingly important clinical issue. In non-metastatic tumours, diagnosis is based on histology. New or enhanced information for clinicopathological diagnosis, revealing the malignant potential of the tumour, could emerge by means of biomarkers. The connection of proto-oncogene c-myc to adrenocortical neoplasias is poorly known, although the Wnt/beta-catenin pathway, one of the signalling pathways leading to induction of c-myc expression, has been connected to development of adrenocortical neoplasias. We studied c-myc expression in adrenocortical tumours and investigated molecules associated with the signalling pathway of c-myc, including cell cycle-related proteins p27, cyclin E and cyclin D1. METHODS We studied 195 consecutive adult patients with 197 primary adrenocortical tumours. Histopathological diagnosis was determined by Weiss score and the novel Helsinki score. C-myc, cyclin D1, cyclin E and p27 expressions were determined by immunohistochemistry. RESULTS Benign adenomas showed prominent nuclear c-myc expression comparable to that of normal adrenocortical cells, whereas carcinomas showed increased cytoplasmic expression. Strong cytoplasmic and weak nuclear c-myc expressions associated with malignancy and adverse outcome. C-myc staining did not correlate with cyclin E. Cyclin D1 correlated with cytoplasmic c-myc expression and to a lesser extent with nuclear c-myc. P27 correlated with cytoplasmic c-myc, but not with nuclear c-myc. P27 correlated with cyclin E. CONCLUSIONS Strong cytoplasmic c-myc expression and weak nuclear expression in adrenocortical tumours associated with malignancy and shorter survival.
Collapse
Affiliation(s)
- Mirkka Pennanen
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki, Finland
| | - Jaana Hagström
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki, Finland
| | - Ilkka Heiskanen
- Department of Surgery, Helsinki University Hospital, and University of Helsinki, Helsinki, Finland
| | - Timo Sane
- Division of Endocrinology, Department of Medicine, Helsinki University Hospital, and University of Helsinki, Helsinki, Finland
| | - Harri Mustonen
- Department of Surgery, Helsinki University Hospital, and University of Helsinki, Helsinki, Finland
| | - Johanna Arola
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, Helsinki University Hospital, and University of Helsinki, Helsinki, Finland.,Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
14
|
Kulshrestha A, Suman S, Ranjan R. Network analysis reveals potential markers for pediatric adrenocortical carcinoma. Onco Targets Ther 2016; 9:4569-81. [PMID: 27555782 PMCID: PMC4968868 DOI: 10.2147/ott.s108485] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pediatric adrenocortical carcinoma (ACC) is a rare malignancy with a poor outcome. Molecular mechanisms of pediatric ACC oncogenesis and advancement are not well understood. Accurate and timely diagnosis of the disease requires identification of new markers for pediatric ACC. Differentially expressed genes (DEGs) were identified from the gene expression profile of pediatric ACC and obtained from Gene Expression Omnibus. Gene Ontology functional and pathway enrichment analysis was implemented to recognize the functions of DEGs. A protein–protein interaction (PPI) and gene–gene functional interaction (GGI) network of DEGs was constructed. Hub gene detection and enrichment analysis of functional modules were performed. Furthermore, a gene regulatory network incorporating DEGs–microRNAs–transcription factors was constructed and analyzed. A total of 431 DEGs including 228 upregulated and 203 downregulated DEGs were screened. These genes were largely involved in cell cycle, steroid biosynthesis, and p53 signaling pathways. Upregulated genes, CDK1, CCNB1, CDC20, and BUB1B, were identified as the common hubs of PPI and GGI networks. All the four common hub genes were also part of modules of the PPI network. Moreover, all the four genes were also present in the largest module of GGI network. A gene regulatory network consisting of 82 microRNAs and 100 transcription factors was also constructed. CDK1, CCNB1, CDC20, and BUB1B may serve as potential biomarker of pediatric ACC and as potential targets for therapeutic approach, although experimental studies are required to authenticate our findings.
Collapse
Affiliation(s)
- Anurag Kulshrestha
- Bioinformatics Division, National Bureau of Animal Genetic Resources, Karnal
| | - Shikha Suman
- Division of Applied Sciences, Indian Institute of Information Technology, Allahabad, India
| | - Rakesh Ranjan
- Bioinformatics Division, National Bureau of Animal Genetic Resources, Karnal
| |
Collapse
|
15
|
Faillot S, Assie G. ENDOCRINE TUMOURS: The genomics of adrenocortical tumors. Eur J Endocrinol 2016; 174:R249-65. [PMID: 26739091 DOI: 10.1530/eje-15-1118] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/06/2016] [Indexed: 01/01/2023]
Abstract
The last decade witnessed the emergence of genomics, a set of high-throughput molecular measurements in biological samples. These pan-genomic and agnostic approaches have revolutionized the molecular biology and genetics of malignant and benign tumors. These techniques have been applied successfully to adrenocortical tumors. Exome sequencing identified new major drivers in all tumor types, including KCNJ5, ATP1A1, ATP2B3 and CACNA1D mutations in aldosterone-producing adenomas (APA), PRKACA mutations in cortisol-producing adenomas (CPA), ARMC5 mutations in primary bilateral macronodular adrenocortical hyperplasia (PBMAH) and ZNRF3 mutations in adrenocortical carcinomas (ACC). Moreover, the various genomic approaches - including exome sequencing, transcriptome, miRNome, genome and methylome - converge into a single molecular classification of adrenocortical tumors. Especially for ACC, two main molecular groups have emerged, showing major differences in outcomes. These ACC groups differ by their gene expression profiles, but also by recurrent mutations and specific DNA hypermethylation patterns in the subgroup of poor outcome. The clinical impact of these findings is just starting. The main altered signaling pathways now become therapeutic targets. The molecular groups of diseases individualize robust subtypes within diseases such as APA, CPA, PBMAH and ACC. A revised nosology of adrenocortical tumors should impact the clinical research. Obvious consequences also include genetic counseling for the new genetic diseases such as ARMC5 mutations in PBMAH, and a better prognostication of ACC based on targeted measurements of a few discriminant molecular alterations. Identifying the main molecular groups of adrenocortical tumors by extensively gathering the molecular variations is a significant step forward towards precision medicine.
Collapse
Affiliation(s)
- Simon Faillot
- Institut CochinINSERM U1016, CNRS 8104, Paris Descartes University, Paris, FranceSIRIC (Site de Recherche Intégré sur le Cancer) CARPEM (CAncer Research for PErsonalized Medicine)Assistance Publique Hôpitaux de Paris, Paris, FranceDepartment of EndocrinologyReference Center for Rare Adrenal Diseases, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, 27 rue du Faubourg Saint Jacques, 75014 Paris, France Institut CochinINSERM U1016, CNRS 8104, Paris Descartes University, Paris, FranceSIRIC (Site de Recherche Intégré sur le Cancer) CARPEM (CAncer Research for PErsonalized Medicine)Assistance Publique Hôpitaux de Paris, Paris, FranceDepartment of EndocrinologyReference Center for Rare Adrenal Diseases, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, 27 rue du Faubourg Saint Jacques, 75014 Paris, France
| | - Guillaume Assie
- Institut CochinINSERM U1016, CNRS 8104, Paris Descartes University, Paris, FranceSIRIC (Site de Recherche Intégré sur le Cancer) CARPEM (CAncer Research for PErsonalized Medicine)Assistance Publique Hôpitaux de Paris, Paris, FranceDepartment of EndocrinologyReference Center for Rare Adrenal Diseases, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, 27 rue du Faubourg Saint Jacques, 75014 Paris, France Institut CochinINSERM U1016, CNRS 8104, Paris Descartes University, Paris, FranceSIRIC (Site de Recherche Intégré sur le Cancer) CARPEM (CAncer Research for PErsonalized Medicine)Assistance Publique Hôpitaux de Paris, Paris, FranceDepartment of EndocrinologyReference Center for Rare Adrenal Diseases, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, 27 rue du Faubourg Saint Jacques, 75014 Paris, France
| |
Collapse
|
16
|
Effect of retinoic acid on human adrenal corticosteroid synthesis. Life Sci 2016; 151:277-280. [PMID: 26979774 DOI: 10.1016/j.lfs.2016.03.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/08/2016] [Accepted: 03/11/2016] [Indexed: 11/23/2022]
Abstract
AIMS Retinoic acid has recently yielded promising results in the treatment of Cushing's disease, i.e., excess cortisol secretion due to a pituitary corticotropin (ACTH)-secreting adenoma. In addition to its effect on the tumoral corticotrope cell, clinical results suggest an additional adrenal site of action. Aim of this study was to evaluate whether retinoic acid modulates cortisol synthesis and secretion by human adrenals in vitro. MAIN METHODS Primary cultures from 10 human adrenals specimens were incubated with 10nM, 100nM and 1μM retinoic acid with and without 10nM ACTH for 24h. Cortisol levels were measured by radioimmunoassay and CYP11A1, STAR and MC2R gene expression analyzed by real-time PCR. KEY FINDINGS Retinoic acid increased cortisol secretion (149.5±33.01%, 151.3±49.45% and 129.3±8.32% control secretion for 10nM, 100nM and 1μM respectively, p<0.05) and potentiated STAR expression (1.51±0.22, 1.56±0.15 and 1.59±0.14 fold change over baseline, for 10nM, 100nM and 1μM respectively, p<0.05). Concurrently, retinoic acid markedly blunted constitutional and ACTH-induced MC2R expression (0.66±0.11, 0.62±0.08 and 0.53±0.07 fold change over baseline, for 10nM, 100nM and 1μM respectively, p<0.05; 0.71±0.10, 0.51±0.07 and 0.51±0.08 fold change over ACTH alone, for 10nM, 100nM and 1μM respectively, p<0.05). No effect on CYP11A1 was observed. SIGNIFICANCE Retinoic acid stimulates cortisol synthesis and secretion in human adrenals and at the same time markedly blunts ACTH receptor transcription. These results reveal a novel, adrenal effect of retinoic acid which may contribute to its efficacy in patients with Cushing's disease.
Collapse
|
17
|
5th International ACC Symposium: Classification of Adrenocortical Cancers from Pathology to Integrated Genomics: Real Advances or Lost in Translation? Discov Oncol 2015; 7:3-8. [DOI: 10.1007/s12672-015-0242-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 10/22/2022] Open
|
18
|
Brown TC, Juhlin CC, Healy JM, Stenman A, Rubinstein JC, Korah R, Carling T. DNA copy amplification and overexpression of SLC12A7 in adrenocortical carcinoma. Surgery 2015; 159:250-7. [PMID: 26454676 DOI: 10.1016/j.surg.2015.08.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 07/19/2015] [Accepted: 08/11/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Overexpression of Solute carrier family 12 member 7 (SLC12A7) promotes tumor aggressiveness in various cancers. Previous studies have identified the 5p15.33 region, containing the SLC12A7 locus, as being amplified frequently in adrenocortical carcinoma (ACC). Copy number amplifications (CNAs) may alter gene expression levels and occur frequently in ACC; however, SLC12A7 gene amplifications or expression levels have not been studied in ACC. METHODS Fifty-five cases of clinically well-characterized ACCs were recruited for this study. Whole-exome sequencing was used to predict CNAs in 19 samples. CNA analysis was performed on an expanded cohort of 26 samples with the use of TaqMan Copy Number Assays. SLC12A7 mRNA expression was analyzed in 32 samples with real-time quantitative polymerase chain reaction and protein expression was assessed by immunohistochemistry. SLC12A7 CNAs and expression patterns were evaluated for correlation with patient and tumor characteristics. RESULTS Whole-exome sequencing and TaqMan Copy Number Assays demonstrated SLC12A7 amplifications in 68.4% and 65.4% of ACCs tested, respectively. Furthermore, SLC12A7 copy gains were associated with increased gene expression (P < .05) and non-functional tumors (P < .05). SLC12A7 gene expression levels were increased in ACCs compared with normal adrenal tissue (P < .05). CONCLUSION SLC12A7 gene amplification and overexpression occurs frequently in ACCs and may represent a novel molecular event associated with ACC.
Collapse
Affiliation(s)
- Taylor C Brown
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - C Christofer Juhlin
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT; Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - James M Healy
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - Adam Stenman
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jill C Rubinstein
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - Reju Korah
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT
| | - Tobias Carling
- Department of Surgery & Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
19
|
Abstract
Advances in genomics accelerated greatly progress in the study of the genetics adrenocortical tumors. Bilateral nodular hyperplasias causing Cushing's syndrome are frequently caused by germline alterations leading to cAMP/PKA pathway activation (micronodular) and ARMC5 inactivation (macronodular). Somatic mutations of β-catenin and PRKACA are observed in non secreting or cortisol producing adenomas, respectively. Alterations of the β-catenin (CTNN1B, ZNFR3) or TP53 pathways are found in carcinomas. Mutations in cancers are more common in aggressive tumors and correlate with transcriptome or methylation profiles. Identification of these alterations helps to refine the molecular classification of these tumors and to develop molecular diagnostic tools.
Collapse
Affiliation(s)
- Stéphanie Espiard
- Cochin Institut, INSERM U1016, 24 rue du Faubourg Saint Jacques, Paris 75014, France; Cochin Institut, CNRS UMR8104, 24 rue du Faubourg Saint-Jacques, Paris 75014, France; Paris Descartes University, 12 rue de l'Ecole de Médecine, Paris 75006, France
| | - Jérôme Bertherat
- Cochin Institut, INSERM U1016, 24 rue du Faubourg Saint Jacques, Paris 75014, France; Cochin Institut, CNRS UMR8104, 24 rue du Faubourg Saint-Jacques, Paris 75014, France; Paris Descartes University, 12 rue de l'Ecole de Médecine, Paris 75006, France; Endocrinology Department, Center for Rare Adrenal Diseases, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, 27 Rue du Fg-St-Jacques, Paris F-75014, France.
| |
Collapse
|
20
|
Cerquetti L, Sampaoli C, De Salvo M, Bucci B, Argese N, Chimento A, Vottari S, Marchese R, Pezzi V, Toscano V, Stigliano A. C-MYC modulation induces responsiveness to paclitaxel in adrenocortical cancer cell lines. Int J Oncol 2015; 46:2231-40. [PMID: 25708932 DOI: 10.3892/ijo.2015.2902] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/23/2014] [Indexed: 11/06/2022] Open
Abstract
C-MYC is overexpressed in many types of cancer linked to poor prognosis. We examined the c-Myc protein expression in adrenocortical cancer (ACC) cells to investigate the role of this protein in the neoplasm, its involvement in chemotherapy and finally to determine whether c-Myc could be considered a prognostic factor in patients with ACC. H295R and SW13 cell lines were treated with paclitaxel. c-Myc overexpressing cell clones were achieved by transfecting the H295R cell line with the pcDNA3-hMYC plasmid expressing the full-lengh C-MYC coding sequence. The SW13 cell line was transfected with siRNA oligonucleotides for C-MYC. Cell cycle analysis was evaluated by flow cytometry. c-Myc, cyclin B1 and pro caspase expression levels were evaluated by western blot analysis. We found that expression of c-Myc was highly expressed in the SW13 cells, whereas the protein was undetectable in the H295R cells. Different doses of paclitaxel were required in the two ACC cell line to induce a block in the G2 phase, characterized by increased cyclin B1 levels and to induce apoptosis by pro-caspase-3 activation. Interestingly, the silencing of C-MYC mRNA prevented paclitaxel induced apoptosis in SW13 cells, whereas in the H295R cells the overexpression of C-MYC rendered the cells more prone to growth inhibition after paclitaxel exposure. The present study directly demonstrates that C-MYC plays a central role in controlling proliferation in ACC cells after paclitaxel treatment and that c-Myc could be considered as a marker for predicting response to chemotherapeutic agents in ACC cell lines.
Collapse
Affiliation(s)
- Lidia Cerquetti
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, 'Sapienza' University of Rome, Rome, Italy
| | - Camilla Sampaoli
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, 'Sapienza' University of Rome, Rome, Italy
| | | | | | - Nicola Argese
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, 'Sapienza' University of Rome, Rome, Italy
| | - Adele Chimento
- Department of Pharmacy, University of Calabria, Cosenza, Italy
| | - Sebastiano Vottari
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, 'Sapienza' University of Rome, Rome, Italy
| | | | - Vincenzo Pezzi
- Department of Pharmacy, University of Calabria, Cosenza, Italy
| | - Vincenzo Toscano
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, 'Sapienza' University of Rome, Rome, Italy
| | - Antonio Stigliano
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, 'Sapienza' University of Rome, Rome, Italy
| |
Collapse
|
21
|
Igaz P, Igaz I, Nagy Z, Nyírő G, Szabó PM, Falus A, Patócs A, Rácz K. MicroRNAs in adrenal tumors: relevance for pathogenesis, diagnosis, and therapy. Cell Mol Life Sci 2015; 72:417-428. [PMID: 25297921 PMCID: PMC11114066 DOI: 10.1007/s00018-014-1752-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 09/16/2014] [Accepted: 09/29/2014] [Indexed: 12/11/2022]
Abstract
Several lines of evidence support the relevance of microRNAs in both adrenocortical and adrenomedullary (pheochromocytomas) tumors. Significantly differentially expressed microRNAs have been described among benign and malignant adrenocortical tumors and different forms of pheochromocytomas that might affect different pathogenic pathways. MicroRNAs can be exploited as markers of malignancy or disease recurrence. Besides tissue microRNAs, novel data show that microRNAs are released in body fluids, and blood-borne microRNAs can be envisaged as minimally invasive markers of malignancy or prognosis. MicroRNAs might even serve as treatment targets that could expand the rather-limited therapeutic repertoire in the field of adrenal tumors. In this review, we present a critical synopsis of the recent observations made in the field of adrenal tumor-associated microRNAs regarding their pathogenic, diagnostic, and potential therapeutic relevance.
Collapse
Affiliation(s)
- Peter Igaz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, 1088, Budapest, Hungary.
| | - Ivan Igaz
- Department of Gastroenterology, Szent Imre Teaching Hospital, Tétényi str. 12-16, 1115, Budapest, Hungary
| | - Zoltán Nagy
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, 1088, Budapest, Hungary
| | - Gábor Nyírő
- Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Szentkirályi str. 46, 1088, Budapest, Hungary
| | - Peter M Szabó
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, 1088, Budapest, Hungary
| | - András Falus
- Department of Genetics Cell- and Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad sq. 4, 1089, Budapest, Hungary
| | - Attila Patócs
- Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Szentkirályi str. 46, 1088, Budapest, Hungary
- "Lendület-2013" Research Group, Hungarian Academy of Sciences and Semmelweis University, Szentkirályi str. 46, 1088, Budapest, Hungary
| | - Károly Rácz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, 1088, Budapest, Hungary
- Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Szentkirályi str. 46, 1088, Budapest, Hungary
| |
Collapse
|
22
|
Abstract
The adrenal gland consists of two distinct parts, the cortex and the medulla. Molecular mechanisms controlling differentiation and growth of the adrenal gland have been studied in detail using mouse models. Knowledge also came from investigations of genetic disorders altering adrenal development and/or function. During embryonic development, the adrenal cortex acquires a structural and functional zonation in which the adrenal cortex is divided into three different steroidogenic zones. Significant progress has been made in understanding adrenal zonation. Recent lineage tracing experiments have accumulated evidence for a centripetal differentiation of adrenocortical cells from the subcapsular area to the inner part of the adrenal cortex. Understanding of the mechanism of adrenocortical cancer (ACC) development was stimulated by knowledge of adrenal gland development. ACC is a rare cancer with a very poor overall prognosis. Abnormal activation of the Wnt/β-catenin as well as the IGF2 signaling plays an important role in ACC development. Studies examining rare genetic syndromes responsible for familial ACT have played an important role in identifying genetic alterations in these tumors (like TP53 or CTNNB1 mutations as well as IGF2 overexpression). Recently, genomic analyses of ACT have shown gene expression profiles associated with malignancy as well as chromosomal and methylation alterations in ACT and exome sequencing allowed to describe the mutational landscape of these tumors. This progress leads to a new classification of these tumors, opening new perspectives for the diagnosis and prognostication of ACT. This review summarizes current knowledge of adrenocortical development, growth, and tumorigenesis.
Collapse
Affiliation(s)
- Lucile Lefèvre
- Inserm, U1016, Institut Cochin, Paris, France Cnrs, UMR8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, France Department of Endocrinology, Referral Center for Rare Adrenal Diseases, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | | | | |
Collapse
|
23
|
Gordon BL, Finnerty BM, Aronova A, Fahey TJ. Genomic medicine for cancer diagnosis. J Surg Oncol 2015; 111:24-30. [DOI: 10.1002/jso.23778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Benjamin L. Gordon
- Research Fellow, Department of Surgery; Weill Cornell Medical College/New York Presbyterian Hospital; NY New York
| | - Brendan M. Finnerty
- Research Fellow, Department of Surgery; Weill Cornell Medical College/New York Presbyterian Hospital; NY New York
| | - Anna Aronova
- Research Fellow, Department of Surgery; Weill Cornell Medical College/New York Presbyterian Hospital; NY New York
| | - Thomas J. Fahey
- Chief of Endocrine Surgery and Professor of Surgery, Department of Surgery; Weill Cornell Medical College/New York Presbyterian Hospital; NY New York
| |
Collapse
|
24
|
Antonini SR, Leal LF, Cavalcanti MM. Pediatric adrenocortical tumors: diagnosis, management and advancements in the understanding of the genetic basis and therapeutic implications. Expert Rev Endocrinol Metab 2014; 9:445-464. [PMID: 30736208 DOI: 10.1586/17446651.2014.941813] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adrenocortical tumors (ACTs) may be sporadic or related to inherited genetic syndromes. Uncovering the molecular defects underlying these genetic syndromes has revealed key signaling pathways involved in adrenocortical tumorigenesis. Although the understanding of ACT biology has improved, to date, very few potential prognostic molecular markers of childhood ACTs have been identified. In this review, we summarize the current knowledge of the epidemiology, clinical presentation, diagnosis, prognosis and treatment options for pediatric patients with ACTs. A review of the genetic basis of adrenocortical tumorigenesis is presented, focusing on the main molecular abnormalities involved in the tumorigenic process and potential novel therapy targets that have been generated, or are being generated, with the discovery of these molecular defects.
Collapse
Affiliation(s)
| | - Letícia F Leal
- a Department of Pediatrics, Ribeirao Preto Medical-School - University of Sao Paulo, Sao Paulo, Brazil
| | - Marcelo M Cavalcanti
- a Department of Pediatrics, Ribeirao Preto Medical-School - University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
25
|
|
26
|
Abstract
Pan-genomic analyses of genetic and epigenetic alterations and gene expression profiles are providing important new insights into the pathogenesis and molecular classification of cancers. The technologies and methods used for these studies are rapidly diversifying and improving. The use of such methodologies for the analysis of adrenocortical tumours has revealed clear transcriptomic (mRNA and microRNA expression profiles), epigenomic (DNA methylation profiles) and genomic (DNA mutations and chromosomal alterations) differences between benign and malignant tumours. Interestingly, genomic studies of adrenal cancers have also identified subtypes of malignant tumours, which demonstrate distinct patterns of molecular alterations and are associated with different clinical outcomes. These discoveries have created the opportunity for classifying adrenocortical tumours on the basis of molecular analyses. Following these genomic studies, efforts to develop new molecular tools that improve diagnosis and prognostication of patients with adrenocortical tumours have also been made. This Review describes the progress that has been made towards classification of adrenocortical tumours to date based on key genomic approaches. In addition, the potential for the development and use of various molecular tools to personalize the management of patients with adrenocortical tumours is discussed.
Collapse
Affiliation(s)
- Guillaume Assié
- 1] Department of Endocrinology, Referral Centre for Rare Adrenal Diseases, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, 27 rue du Fg-St-Jacques, 75014 Paris, France. [2] INSERM U1016, CNRS UMR 8104, Paris Descartes University, Institut Cochin, 75014 Paris, France
| | - Anne Jouinot
- INSERM U1016, CNRS UMR 8104, Paris Descartes University, Institut Cochin, 75014 Paris, France
| | - Jérôme Bertherat
- 1] Department of Endocrinology, Referral Centre for Rare Adrenal Diseases, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, 27 rue du Fg-St-Jacques, 75014 Paris, France. [2] INSERM U1016, CNRS UMR 8104, Paris Descartes University, Institut Cochin, 75014 Paris, France
| |
Collapse
|
27
|
Szabó DR, Baghy K, Szabó PM, Zsippai A, Marczell I, Nagy Z, Varga V, Éder K, Tóth S, Buzás EI, Falus A, Kovalszky I, Patócs A, Rácz K, Igaz P. Antitumoral effects of 9-cis retinoic acid in adrenocortical cancer. Cell Mol Life Sci 2014; 71:917-32. [PMID: 23807211 PMCID: PMC11113805 DOI: 10.1007/s00018-013-1408-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/10/2013] [Accepted: 06/11/2013] [Indexed: 01/08/2023]
Abstract
The currently available medical treatment options of adrenocortical cancer (ACC) are limited. In our previous meta-analysis of adrenocortical tumor genomics data, ACC was associated with reduced retinoic acid production and retinoid X receptor-mediated signaling. Our objective has been to study the potential antitumoral effects of 9-cis retinoic acid (9-cisRA) on the ACC cell line NCI-H295R and in a xenograft model. Cell proliferation, hormone secretion, and gene expression have been studied in the NCI-H295R cell line. A complex bioinformatics approach involving pathway and network analysis has been performed. Selected genes have been validated by real-time qRT-PCR. Athymic nude mice xenografted with NCI-H295R have been used in a pilot in vivo xenograft model. 9-cisRA significantly decreased cell viability and steroid hormone secretion in a concentration- and time-dependent manner in the NCI-H295R cell line. Four major molecular pathways have been identified by the analysis of gene expression data. Ten genes have been successfully validated involved in: (1) steroid hormone secretion (HSD3B1, HSD3B2), (2) retinoic acid signaling (ABCA1, ABCG1, HMGCR), (3) cell-cycle damage (GADD45A, CCNE2, UHRF1), and the (4) immune response (MAP2K6, IL1R2). 9-cisRA appears to directly regulate the cell cycle by network analysis. 9-cisRA also reduced tumor growth in the in vivo xenograft model. In conclusion, 9-cisRA might represent a promising new candidate in the treatment of hormone-secreting adrenal tumors and adrenocortical cancer.
Collapse
Affiliation(s)
- Diana Rita Szabó
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| | - Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Üllői Str. 26, Budapest, 1088 Hungary
| | - Peter M. Szabó
- Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| | - Adrienn Zsippai
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| | - István Marczell
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| | - Zoltán Nagy
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| | - Vivien Varga
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| | - Katalin Éder
- Department of Genetics, Cell- and Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad Sq. 4, Budapest, 1089 Hungary
| | - Sára Tóth
- Department of Genetics, Cell- and Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad Sq. 4, Budapest, 1089 Hungary
| | - Edit I. Buzás
- Department of Genetics, Cell- and Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad Sq. 4, Budapest, 1089 Hungary
| | - András Falus
- Department of Genetics, Cell- and Immunobiology, Faculty of Medicine, Semmelweis University, Nagyvárad Sq. 4, Budapest, 1089 Hungary
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Üllői Str. 26, Budapest, 1088 Hungary
| | - Attila Patócs
- Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| | - Károly Rácz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| | - Peter Igaz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi Str. 46, Budapest, 1088 Hungary
| |
Collapse
|
28
|
Bi C, Li B, Du L, Wang L, Zhang Y, Cheng Z, Zhai A. Vitamin D receptor, an important transcription factor associated with aldosterone-producing adenoma. PLoS One 2013; 8:e82309. [PMID: 24376526 PMCID: PMC3869669 DOI: 10.1371/journal.pone.0082309] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 10/22/2013] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To explore the endocrine mechanisms of aldosterone-producing adenoma (APA) by using the microarray expression profiles of normal and APA samples. METHODS The gene expression profile GSE8514 was downloaded from Gene Expression Omnibus database, including samples from normal adrenals (n = 5) and APAs (n = 10). The differentially expressed genes (DEGs) were identified by samr package and endocrine DEGs were obtained according to Clinical Genome Database. Then, functional enrichment analysis of screened DEGs was performed by DAVID (Database for Annotation, Visualization and Integrated Discovery). Finally, a regulatory network was constructed to screen endocrine genes related with adrenal dysfunction and pathway enrichment analysis for the constructed network was performed. RESULTS A total of 2149 DEGs were identified including 379 up- and 1770 down-regulated genes. And 26 endocrine genes were filtered from the DEGs. Furthermore, the down-regulated DEGs are mainly related to protein kinase cascade, response to molecule of bacterial origin, response to lipopolysaccharide, cellular macromolecule catabolic process and macromolecule catabolic process, while the up-regulated DEGs are related with regulation of ion transport. The target genes of VDR (vitamin D receptor), one of the three endocrine genes differentially expressed in the regulatory network, were endocrine genes including CYP24A1 (25-hydroxyvitamin D-24-hydroxylase) and PTH (parathyroid hormone). Three pathways may be associated with APA pathogenesis including cytokine-cytokine receptor interaction, pathways in cancer and autoimmune thyroid disease. CONCLUSION The VDR is the most significant transcription factor and related endocrine genes might play important roles in the endocrine mechanisms of APA.
Collapse
Affiliation(s)
- Changlong Bi
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Li
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lili Du
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lishan Wang
- FengHe (ShangHai) Information Technology Co., Ltd., Shanghai, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai ,China
| | - Yingqi Zhang
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhifeng Cheng
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Aixia Zhai
- Department of Microbiology, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Science and Technology Innovation Team in Higher Education Institutes for Infection and Immunity, Harbin Medical University, Harbin , China
- Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin Medical University, Harbin, China
- *
| |
Collapse
|
29
|
Zsippai A, Szabó PM, Szabó DR, Nagy Z, Patócs A, Rácz K, Igaz P. In silico analysis of pathways affected by differentially expressed microRNA in adrenocortical tumors. J Endocrinol Invest 2013; 36:1011-9. [PMID: 23812403 DOI: 10.3275/9024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND MicroRNA are involved in the pathogenesis of several tumors, and several studies have been performed on the microRNA profile of adrenocortical tumors to date. The pathways affected by these microRNA, however, have not been analyzed yet by a systematic approach. AIM To perform an in silico bioinformatics analysis of microRNA commonly altered in at least two studies and to decipher the pathways affected by microRNA in adrenocortical tumors. METHODS Datasets on microRNA and mRNA expression have been retrieved from 5 and 3 studies, respectively. MicroRNA mRNA targets have been identified by our tissue specific target prediction pipeline, and mRNA have been subjected to Ingenuity Pathway Analysis. RESULTS Thirty- nine microRNA were identified as commonly altered in two studies. Altogether 49,817 mRNA targets have been found for these microRNA. One-hundred and seventy-eight significant pathways associating with these have been identified and were found in all studies. We have selected 12 pathways involving retinoic acid signaling (lipopolysaccharide/ interleukin-1 mediated inhibition of retinoic X receptor (RXR) function, peroxisome proliferator-activated receptor (PPAR)α/RXRα activation, retinoic A receptor activation and PPAR signaling pathways) and cell cycle alterations (aryl hydrocarbon receptor signaling, growth arrest and DNA damage-inducible 45 signaling, integrin signaling, G2/M DNA damage checkpoint regulation, cyclins and cell cycle regulation and cell cycle control of chromosomal replication pathways) as these have been also established in our previous study on the functional genomics meta-analysis of adrenocortical tumors. Several microRNA have been identified that could affect these pathways. CONCLUSIONS MicroRNA might affect several pathogenic pathways in adrenocortical tumors. Validation studies are required to confirm the biological relevance of these findings.
Collapse
Affiliation(s)
- A Zsippai
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, H-1088 Budapest, Szentkirályi str. 46, Hungary
| | | | | | | | | | | | | |
Collapse
|
30
|
De Martino MC, Al Ghuzlan A, Aubert S, Assié G, Scoazec JY, Leboulleux S, Do Cao C, Libè R, Nozières C, Lombès M, Pattou F, Borson-Chazot F, Hescot S, Mazoyer C, Young J, Borget I, Colao A, Pivonello R, Soria JC, Bertherat J, Schlumberger M, Lacroix L, Baudin E. Molecular screening for a personalized treatment approach in advanced adrenocortical cancer. J Clin Endocrinol Metab 2013; 98:4080-8. [PMID: 23979958 DOI: 10.1210/jc.2013-2165] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Adrenocortical cancer (ACC) is a rare cancer with poor prognosis and scant treatment options. In ACC, no personalized approach has emerged but no extensive molecular screening has been performed to date. OBJECTIVE The objective of the study was to evaluate the presence of a large number of potentially targetable molecular events in a large cohort of advanced ACC. DESIGN, SETTING, AND PARTICIPANTS We used hot spot gene sequencing (Ion Torrent, 40 patients) and comparative genomic hybridization (CGH; 28 patients; a subset of the entire cohort) in adult stage III-IV ACC samples to screen for mutations and copy number abnormalities of potential interest for therapeutic use in 46 and 130 genes, respectively. RESULTS At least one copy number alteration or mutation was found in 19 patients (47.5%). The most frequent mutations were detected on TP53, ATM, and CTNNB1 [6 of 40 (15%), 5 of 40 (12.5%), and 4 of 40 (10%), respectively]. The most frequent copy number alterations identified were: amplification of the CDK4 oncogene (5 of 28; 17.9%) and deletion of the CDKN2A (4 of 28; 14.3%) and CDKN2B (3 of 28; 10.7%) tumor suppressor genes. Amplifications of FGFR1, FGF9, or FRS2 were discovered in three subjects (10.7%). Associated alterations were: deletions of CDKN2A, CDKN2B with ATM mutations, and TP53 mutations with CTNNB1 mutations. CONCLUSIONS No simple targetable molecular event emerged. Drugs targeting the cell cycle could be the most relevant new therapeutic approach for patients with advanced ACC. Inhibitors of the fibroblast growth factor receptor pathway could also be a therapeutic option in a subset of patients, whereas other targeted therapies should be considered on a case-by-case basis.
Collapse
Affiliation(s)
- Maria Cristina De Martino
- PhD, Translational Research Laboratory and Biobank, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ronchi CL, Sbiera S, Leich E, Henzel K, Rosenwald A, Allolio B, Fassnacht M. Single nucleotide polymorphism array profiling of adrenocortical tumors--evidence for an adenoma carcinoma sequence? PLoS One 2013; 8:e73959. [PMID: 24066089 PMCID: PMC3774745 DOI: 10.1371/journal.pone.0073959] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 07/25/2013] [Indexed: 12/20/2022] Open
Abstract
Adrenocortical tumors consist of benign adenomas and highly malignant carcinomas with a still incompletely understood pathogenesis. A total of 46 adrenocortical tumors (24 adenomas and 22 carcinomas) were investigated aiming to identify novel genes involved in adrenocortical tumorigenesis. High-resolution single nucleotide polymorphism arrays (Affymetrix) were used to detect copy number alterations (CNAs) and copy neutral losses of heterozygosity (cnLOH). Genomic clustering showed good separation between adenomas and carcinomas, with best partition including only chromosome 5, which was highly amplified in 17/22 malignant tumors. The malignant tumors had more relevant genomic aberrations than benign tumors, such as a higher median number of recurrent CNA (2631 vs 94), CNAs >100 Kb (62.5 vs 7) and CN losses (72.5 vs 5.5), and a higher percentage of samples with cnLOH (91% vs 29%). Within the carcinoma cohort, a precise genetic pattern (i.e. large gains at chr 5, 7, 12, and 19, and losses at chr 1, 2, 13, 17, and 22) was associated with a better prognosis (overall survival: 72.2 vs 35.4 months, P=0.063). Interestingly, >70% of gains frequent in beningn were also present in malignant tumors. Notch signaling was the most frequently involved pathway in both tumor entities. Finally, a CN gain at imprinted “IGF2” locus chr 11p15.5 appeared to be an early alteration in a multi-step tumor progression, followed by the loss of one or two alleles, associated with increased IGF2 expression, only in carcinomas. Our study serves as database for the identification of genes and pathways, such as Notch signaling, which could be involved in the pathogenesis of adrenocortical tumors. Using these data, we postulate an adenoma-carcinoma sequence for these tumors.
Collapse
Affiliation(s)
- Cristina L. Ronchi
- Endocrine and Diabetes Unit, Department of Internal Medicine I, University Hospital, University of Würzburg, Wuerzburg, Germany
- * E-mail:
| | - Silviu Sbiera
- Endocrine and Diabetes Unit, Department of Internal Medicine I, University Hospital, University of Würzburg, Wuerzburg, Germany
- Department of Internal Medicine IV, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Ellen Leich
- Institute of Pathology, University of Würzburg, Wuerzburg, Germany
| | - Katharina Henzel
- Endocrine and Diabetes Unit, Department of Internal Medicine I, University Hospital, University of Würzburg, Wuerzburg, Germany
| | | | - Bruno Allolio
- Endocrine and Diabetes Unit, Department of Internal Medicine I, University Hospital, University of Würzburg, Wuerzburg, Germany
| | - Martin Fassnacht
- Endocrine and Diabetes Unit, Department of Internal Medicine I, University Hospital, University of Würzburg, Wuerzburg, Germany
- Department of Internal Medicine IV, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| |
Collapse
|
32
|
Szabó PM, Butz H, Igaz P, Rácz K, Hunyady L, Patócs A. Minireview: miRomics in endocrinology: a novel approach for modeling endocrine diseases. Mol Endocrinol 2013; 27:573-85. [PMID: 23349525 PMCID: PMC5416806 DOI: 10.1210/me.2012-1220] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 12/31/2012] [Indexed: 01/26/2023] Open
Abstract
MicroRNAs (miRNAs) are small (16-24 nucleotides) noncoding RNAs that negatively regulate gene expression. Growing evidence demonstrates that miRNAs participate in the regulation of numerous physiological and pathological processes. The clinical utility of the cell-type-specific miRNA expression profile (miRomics) has been directly demonstrated in molecular classification of tumor samples and in prediction of prognosis or therapeutic responsiveness. Identification of the relevant miRNAs and their targets requires both in silico and molecular biological methods. In this review, we summarize the methodological arsenal used in miRNA-related research, and through our own data on adrenal tumors, we present how miRNA could be integrated into omics-based networks. The expanding knowledge obtained from miRNA research may lead to the development of novel diagnostic and treatment modalities in future.
Collapse
Affiliation(s)
- Péter M Szabó
- Molecular Medicine Research Group, Hungarian Academy of Sciences and Second Department of Medicine, Semmelweis University, 46 Szentkirályi Street, Budapest, Hungary H-1088
| | | | | | | | | | | |
Collapse
|
33
|
A data similarity-based strategy for meta-analysis of transcriptional profiles in cancer. PLoS One 2013; 8:e54979. [PMID: 23383020 PMCID: PMC3558433 DOI: 10.1371/journal.pone.0054979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 12/22/2012] [Indexed: 11/22/2022] Open
Abstract
Background Robust transcriptional signatures in cancer can be identified by data similarity-driven meta-analysis of gene expression profiles. An unbiased data integration and interrogation strategy has not previously been available. Methods and Findings We implemented and performed a large meta-analysis of breast cancer gene expression profiles from 223 datasets containing 10,581 human breast cancer samples using a novel data similarity-based approach (iterative EXALT). Cancer gene expression signatures extracted from individual datasets were clustered by data similarity and consolidated into a meta-signature with a recurrent and concordant gene expression pattern. A retrospective survival analysis was performed to evaluate the predictive power of a novel meta-signature deduced from transcriptional profiling studies of human breast cancer. Validation cohorts consisting of 6,011 breast cancer patients from 21 different breast cancer datasets and 1,110 patients with other malignancies (lung and prostate cancer) were used to test the robustness of our findings. During the iterative EXALT analysis, 633 signatures were grouped by their data similarity and formed 121 signature clusters. From the 121 signature clusters, we identified a unique meta-signature (BRmet50) based on a cluster of 11 signatures sharing a phenotype related to highly aggressive breast cancer. In patients with breast cancer, there was a significant association between BRmet50 and disease outcome, and the prognostic power of BRmet50 was independent of common clinical and pathologic covariates. Furthermore, the prognostic value of BRmet50 was not specific to breast cancer, as it also predicted survival in prostate and lung cancers. Conclusions We have established and implemented a novel data similarity-driven meta-analysis strategy. Using this approach, we identified a transcriptional meta-signature (BRmet50) in breast cancer, and the prognostic performance of BRmet50 was robust and applicable across a wide range of cancer-patient populations.
Collapse
|
34
|
Fonseca AL, Healy J, Kunstman JW, Korah R, Carling T. Gene expression and regulation in adrenocortical tumorigenesis. BIOLOGY 2012; 2:26-39. [PMID: 24832650 PMCID: PMC4009874 DOI: 10.3390/biology2010026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/01/2012] [Accepted: 12/14/2012] [Indexed: 11/21/2022]
Abstract
Adrenocortical tumors are frequently found in the general population, and may be benign adrenocortical adenomas or malignant adrenocortical carcinomas. Unfortunately the clinical, biochemical and histopathological distinction between benign and malignant adrenocortical tumors may be difficult in the absence of widely invasive or metastatic disease, and hence attention has turned towards a search for molecular markers. The study of rare genetic diseases that are associated with the development of adrenocortical carcinomas has contributed to our understanding of adrenocortical tumorigenesis. In addition, comprehensive genomic hybridization, methylation profiling, and genome wide mRNA and miRNA profiling have led to improvements in our understanding, as well as demonstrated several genes and pathways that may serve as diagnostic or prognostic markers.
Collapse
Affiliation(s)
- Annabelle L Fonseca
- Department of Surgery, Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, 333 Cedar Street, TMP202 Box 208062, New Haven, CT 06520, USA.
| | - James Healy
- Department of Surgery, Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, 333 Cedar Street, TMP202 Box 208062, New Haven, CT 06520, USA.
| | - John W Kunstman
- Department of Surgery, Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, 333 Cedar Street, TMP202 Box 208062, New Haven, CT 06520, USA.
| | - Reju Korah
- Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Tobias Carling
- Department of Surgery, Yale Endocrine Neoplasia Laboratory, Yale University School of Medicine, 333 Cedar Street, TMP202 Box 208062, New Haven, CT 06520, USA.
| |
Collapse
|
35
|
Szabó PM, Pintér M, Szabó DR, Zsippai A, Patócs A, Falus A, Rácz K, Igaz P. Integrative analysis of neuroblastoma and pheochromocytoma genomics data. BMC Med Genomics 2012; 5:48. [PMID: 23106811 PMCID: PMC3495658 DOI: 10.1186/1755-8794-5-48] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 10/26/2012] [Indexed: 12/26/2022] Open
Abstract
Background Pheochromocytoma and neuroblastoma are the most common neural crest-derived tumors in adults and children, respectively. We have performed a large-scale in silico analysis of altogether 1784 neuroblastoma and 531 pheochromocytoma samples to establish similarities and differences using analysis of mRNA and microRNA expression, chromosome aberrations and a novel bioinformatics analysis based on cooperative game theory. Methods Datasets obtained from Gene Expression Omnibus and ArrayExpress have been subjected to a complex bioinformatics analysis using GeneSpring, Gene Set Enrichment Analysis, Ingenuity Pathway Analysis and own software. Results Comparison of neuroblastoma and pheochromocytoma with other tumors revealed the overexpression of genes involved in development of noradrenergic cells. Among these, the significance of paired-like homeobox 2b in pheochromocytoma has not been reported previously. The analysis of similar expression patterns in neuroblastoma and pheochromocytoma revealed the same anti-apoptotic strategies in these tumors. Cancer regulation by stathmin turned out to be the major difference between pheochromocytoma and neuroblastoma. Underexpression of genes involved in neuronal cell-cell interactions was observed in unfavorable neuroblastoma. By the comparison of hypoxia- and Ras-associated pheochromocytoma, we have found that enhanced insulin like growth factor 1 signaling may be responsible for the activation of Src homology 2 domain containing transforming protein 1, the main co-factor of RET. Hypoxia induced factor 1α and vascular endothelial growth factor signaling included the most prominent gene expression changes between von Hippel-Lindau- and multiple endocrine neoplasia type 2A-associated pheochromocytoma. Conclusions These pathways include previously undescribed pathomechanisms of neuroblastoma and pheochromocytoma and associated gene products may serve as diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Peter M Szabó
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str, 46, Budapest, H-1088, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Lebastchi AH, Kunstman JW, Carling T. Adrenocortical Carcinoma: Current Therapeutic State-of-the-Art. JOURNAL OF ONCOLOGY 2012; 2012:234726. [PMID: 23125857 PMCID: PMC3483813 DOI: 10.1155/2012/234726] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Revised: 08/13/2012] [Accepted: 08/27/2012] [Indexed: 12/24/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare, aggressive malignancy that generally conveys a poor prognosis. Currently, surgical resection is considered the lone curative treatment modality. In addition, the low prevalence of ACC has limited effective clinical trial design to develop evidence-based approaches to ACC therapy. The proper role of radio- and chemotherapy treatment for ACC is still being defined. Similarly, the molecular pathogenesis of ACC remains to be fully characterized. Despite these challenges, progress has been made in several areas. After years of refinement, an internationally accepted staging system has been defined. International collaborations have facilitated increasingly robust clinical trials, especially regarding agent choice and patient selection for chemotherapeutics. Genetic array data and molecular profiling have identified new potential targets for rational drug design as well as potential tumor markers and predictors of therapeutic response. However, these advances have not yet been translated into a large outcomes benefit for ACC patients. In this paper, we summarize established therapy for ACC and highlight recent findings in the field that are impacting clinical practice.
Collapse
Affiliation(s)
- Amir H. Lebastchi
- Department of Surgery, Yale Endocrine Neoplasia Laboratory, Yale School of Medicine, 333 Cedar Street, TMP202, Box 208062, New Haven, CT 06520, USA
| | - John W. Kunstman
- Department of Surgery, Yale Endocrine Neoplasia Laboratory, Yale School of Medicine, 333 Cedar Street, TMP202, Box 208062, New Haven, CT 06520, USA
| | - Tobias Carling
- Department of Surgery, Yale Endocrine Neoplasia Laboratory, Yale School of Medicine, 333 Cedar Street, TMP202, Box 208062, New Haven, CT 06520, USA
| |
Collapse
|
37
|
Single nucleotide polymorphism microarray analysis in cortisol-secreting adrenocortical adenomas identifies new candidate genes and pathways. Neoplasia 2012; 14:206-18. [PMID: 22496620 DOI: 10.1593/neo.111758] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 02/13/2012] [Accepted: 02/13/2012] [Indexed: 02/04/2023] Open
Abstract
The genetic mechanisms underlying adrenocortical tumor development are still largely unknown. We used high-resolution single nucleotide polymorphism microarrays (Affymetrix SNP 6.0) to detect copy number alterations (CNAs) and copy-neutral losses of heterozygosity (cnLOH) in 15 cortisol-secreting adrenocortical adenomas with matched blood samples. We focused on microalterations aiming to discover new candidate genes involved in early tumorigenesis and/or autonomous cortisol secretion. We identified 962 CNAs with a median of 18 CNAs per sample. Half of them involved noncoding regions, 89% were less than 100 kb, and 28% were found in at least two samples. The most frequently gained regions were 5p15.33, 6q16.1, 7p22.3-22.2, 8q24.3, 9q34.2-34.3, 11p15.5, 11q11, 12q12, 16q24.3, 20p11.1-20q21.11, and Xq28 (≥20% of cases), most of them being identified in the same three adenomas. These regions contained among others genes like NOTCH1, CYP11B2, HRAS, and IGF2. Recurrent losses were less common and smaller than gains, being mostly localized at 1p, 6q, and 11q. Pathway analysis revealed that Notch signaling was the most frequently altered. We identified 46 recurrent CNAs that each affected a single gene (31 gains and 15 losses), including genes involved in steroidogenesis (CYP11B1) or tumorigenesis (CTNNB1, EPHA7, SGK1, STIL, FHIT). Finally, 20 small cnLOH in four cases affecting 15 known genes were found. Our findings provide the first high-resolution genome-wide view of chromosomal changes in cortisol-secreting adenomas and identify novel candidate genes, such as HRAS, EPHA7, and SGK1. Furthermore, they implicate that the Notch1 signaling pathway might be involved in the molecular pathogenesis of adrenocortical tumors.
Collapse
|
38
|
Lehmann T, Wrzesinski T. The molecular basis of adrenocortical cancer. Cancer Genet 2012; 205:131-7. [PMID: 22559973 DOI: 10.1016/j.cancergen.2012.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 02/04/2012] [Accepted: 02/21/2012] [Indexed: 12/12/2022]
Abstract
Adrenocortical tumors (ACTs) are common, and most are benign adrenocortical adenomas (ACAs). Malignant adrenocortical carcinoma (ACC) is a rare tumor type and is observed at the rate of one or two cases per million annually. ACTs are classified as either ACAs or ACCs by histopathologic methods that are based on nine Weiss scoring criteria, including the nuclear grade, mitotic rate, presence of necrosis, and others. In this review, we describe the findings of studies that have examined the molecular basis of ACTs, and we compare transcriptome analysis with other diagnostic approaches. ACTs are occasionally difficult to classify. Therefore, molecular techniques, such as microarray analysis, have recently been applied to overcome some of these diagnostic problems. We also discuss the likelihood of the diagnosis and discernment between ACAs and ACCs based on the molecular tests. To show the recent progress in understanding the etiology of ACTs, we highlight the relationship between genetic analysis and transcriptome analysis. We attempt to understand the role of abnormal cell growth and steroid hormone secretion. Genetic and transcriptome analyses have improved our understanding of ACTs considerably, yet many unanswered questions remain.
Collapse
Affiliation(s)
- Tomasz Lehmann
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, Poznan, Poland.
| | | |
Collapse
|
39
|
Jain M, Rechache N, Kebebew E. Molecular markers of adrenocortical tumors. J Surg Oncol 2012; 106:549-56. [PMID: 22504887 DOI: 10.1002/jso.23119] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 03/23/2012] [Indexed: 12/18/2022]
Abstract
Adrenocortical tumors are common and incidentally discovered in up to 14% of axial imaging studies performed for other indications. Most of these tumors are nonfunctioning but may require removal because of the risk of adrenocortical carcinoma. Unfortunately, most clinical and imaging features are still not accurate enough to allow definitive diagnosis and an increasing number of patients undergo adrenalectomy to exclude a cancer diagnosis. Adrenocortical carcinoma is an aggressive malignancy with no effective therapy for patients with locally advanced and metastatic disease. Studies using new genomic approaches including mRNA, miRNA, methylation, and CGH profiling have identified dysregulated genes and pathways that may have clinical implications in improved molecular diagnosis and prognostication of adrenocortical cancer (ACC). In this review, we highlight recent advances in the molecular diagnosis of adrenocortical tumors.
Collapse
Affiliation(s)
- Meenu Jain
- Endocrine Oncology Section, Surgery Branch, National Cancer Institute, Maryland 20892, USA
| | | | | |
Collapse
|
40
|
Assie G, Giordano TJ, Bertherat J. Gene expression profiling in adrenocortical neoplasia. Mol Cell Endocrinol 2012; 351:111-7. [PMID: 22056416 DOI: 10.1016/j.mce.2011.09.044] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 09/14/2011] [Accepted: 09/14/2011] [Indexed: 12/29/2022]
Abstract
Transcriptome studies of adrenocortical tumors have shown clear differences between adenomas and carcinomas and identified two subgroups of carcinomas with different prognoses. This review focuses on how transcriptomes have enriched our knowledge about genes previously identified by classical candidate gene approaches, uncovered novel genes relevant to adrenocortical tumor biology, helped to identify and understand specific pathway alterations, and advanced the overall translational relevance of this field of research.
Collapse
Affiliation(s)
- G Assie
- INSERM U1016, Institut Cochin, Paris, France.
| | | | | |
Collapse
|
41
|
Zhao Y, Li Y, Lu H, Chen J, Zhang Z, Zhu ZZ. Association of copy number loss of CDKN2B and PTCH1 with poor overall survival in patients with pulmonary squamous cell carcinoma. Clin Lung Cancer 2012; 12:328-34. [PMID: 21889114 DOI: 10.1016/j.cllc.2011.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 02/13/2011] [Accepted: 02/22/2011] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND PURPOSE Although lung cancer is the leading cause of cancer deaths worldwide, reliable markers allowing prediction of patient survival at the time of initial diagnosis are still lacking. Copy number alterations (CNAs) in tumor tissue DNA have been associated with tumorigenesis and malignant progression. We aimed at identification of gene-level CNAs with prognostic value for survival in pulmonary squamous cell carcinoma (SCC). METHODS The CNA status of a panel of 44 genes was analyzed by high-resolution array comparative genomic hybridization (CGH) in 49 SCC samples. Overall survival information (median follow-up, 40 months) for the patients was collected and used to assess outcome correlations with gene CNAs. RESULTS Survival analysis showed that both CDKN2B loss and PTCH1 loss were associated with poor survival (both P < .001, log-rank test). Multivariate Cox analysis, including CDKN2B loss and PTCH1 loss as well as age, sex, cigarette smoking status, tumor size, tumor differentiation, and TNM stage showed that CDKN2B loss (hazard ratio [HR], 17.88; 95% confidence interval [CI], 4.40-72.67; P < .001) and PTCH1 loss (HR, 10.81; 95% CI, 1.92-60.98; P = .007) were independent prognostic factors for poor survival. In addition the PTCH1 loss was more frequently found in moderately or poorly differentiated tumors than in well-differentiated tumors (P = .007). CONCLUSION These findings suggest that 2 genes of loss, CDKN2B and PTCH1, are associated with poor overall survival in patients with SCC of the lung and may be useful as prognostic markers.
Collapse
Affiliation(s)
- Yushi Zhao
- Department of Cardiovascular Surgery, the Fourth Affiliated Hospital, Harbin Medical University, Heilongjiang, P.R. China
| | | | | | | | | | | |
Collapse
|
42
|
O’Connell S, Tuite N, Slattery C, Ryan MP, McMorrow T. Cyclosporine A–Induced Oxidative Stress in Human Renal Mesangial Cells: A Role for ERK 1/2 MAPK Signaling. Toxicol Sci 2011; 126:101-13. [DOI: 10.1093/toxsci/kfr330] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
43
|
Antonini SRR, Colli LM, Ferro L, Mermejo L, Castro MD. Tumores adrenocorticais na criança: da abordagem clínica à avaliação molecular. ACTA ACUST UNITED AC 2011; 55:599-606. [DOI: 10.1590/s0004-27302011000800014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 10/16/2011] [Indexed: 11/21/2022]
Abstract
Tumores do córtex adrenal (TCA) são mais frequentes em crianças, mas podem ocorrer em qualquer faixa etária. São classificados como funcionantes, não funcionantes (predominam no adulto), e mistos. O diagnóstico é baseado na avaliação clínica, hormonal e exames de imagem. Em crianças, o método de escolha para diferenciar entre benigno ou maligno é a classificação baseada no estadiamento do tumor. Alguns marcadores moleculares merecem destaque: além de mutações inativadoras no gene supressor tumoral TP53, há evidências de envolvimento do IGF2 em 90% de TAC malignos, e mutações no éxon 3 do gene CTNNB1 foram encontradas em 6% dos TAC pediátricos. Além disso, microRNAs podem atuar como reguladores negativos da expressão gênica e participar da tumorigênese adrenocortical. Métodos para análise da expressão gênica permitem identificar TCA com prognóstico bom ou ruim, e espera-se que esses estudos possam facilitar o desenvolvimento de drogas para tratar pacientes de acordo com as vias de sinalização específicas que estiverem alteradas.
Collapse
|
44
|
Abstract
Adrenocortical carcinoma is a rare heterogeneous neoplasm with an incompletely understood pathogenesis and a poor prognosis. Previous studies have identified overexpression of insulin-like growth factor 2 (IGF-2) and constitutive activation of β-catenin as key factors involved in the development of adrenocortical carcinoma. Most patients present with steroid hormone excess, for example Cushing syndrome or virilization, or abdominal mass effects, but a growing proportion of patients with adrenocortical carcinoma (currently >15%) is initially diagnosed incidentally. No general consensus on the diagnostic and therapeutic measures for adrenocortical carcinoma exists, but collaborative efforts, such as international conferences and networks, including the European Network for the Study of Adrenal Tumors (ENSAT), have substantially advanced the field. In patients with suspected adrenocortical carcinoma, a thorough endocrine and imaging work-up is recommended to guide the surgical approach aimed at complete resection of the tumor. To establish an adequate basis for treatment decisions, pathology reports include the Weiss score to assess malignancy, the resection status and the Ki67 index. As recurrence is frequent, close follow-up initially every 3 months is mandatory. Most patients benefit from adjuvant mitotane treatment. In metastatic disease, mitotane is the cornerstone of initial treatment, and cytotoxic drugs should be added in case of progression. Results of a large phase III trial in advanced adrenocortical carcinoma are anticipated for 2011 and will hopefully establish a benchmark therapy. New targeted therapies, for example, IGF-1 receptor inhibitors, are under investigation and may soon improve current treatment options.
Collapse
Affiliation(s)
- Martin Fassnacht
- Department of Internal Medicine I, Endocrine Unit, University Hospital, University of Würzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany.
| | | | | | | |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Adrenocortical carcinoma is an aggressive, lethal malignancy of the adrenal cortex. The rarity of the disease has stymied therapeutic development. Recent work toward understanding the molecular pathogenesis of the disease has identified several potential new diagnostic and therapeutic targets. RECENT FINDINGS The molecular characterization of adrenocortical carcinoma has identified dysregulation of the Gap 2/mitosis transition and the insulin-like growth factor 1 receptor signaling cascade as two major pathways for therapeutic development. These studies have also highlighted an unappreciated heterogeneity of the disease at the gene level that nevertheless seems to converge onto common cellular pathways. Additionally, the characterization of Wnt signaling through β-catenin in adrenal development, the demonstration of the involvement of BMP signaling in adrenocortical carcinoma growth regulation, and the discovery that ERCC1 expression levels can predict therapeutic response to platinum are just a few of the recent advances that promise to shed light on adrenocortical carcinoma biology. SUMMARY Short-term, therapeutic development should target the Gap 2/mitosis transition and the downstream signaling of the insulin-like growth factor 1 receptor receptor. Long-term, additional characterization of patient samples, particularly at the sequence level, is required to fully understand adrenocortical carcinoma biology and apply that knowledge to clinical practice.
Collapse
|
46
|
Martini P, Risso D, Sales G, Romualdi C, Lanfranchi G, Cagnin S. Statistical Test of Expression Pattern (STEPath): a new strategy to integrate gene expression data with genomic information in individual and meta-analysis studies. BMC Bioinformatics 2011; 12:92. [PMID: 21481242 PMCID: PMC3094239 DOI: 10.1186/1471-2105-12-92] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 04/11/2011] [Indexed: 01/21/2023] Open
Abstract
Background In the last decades, microarray technology has spread, leading to a dramatic increase of publicly available datasets. The first statistical tools developed were focused on the identification of significant differentially expressed genes. Later, researchers moved toward the systematic integration of gene expression profiles with additional biological information, such as chromosomal location, ontological annotations or sequence features. The analysis of gene expression linked to physical location of genes on chromosomes allows the identification of transcriptionally imbalanced regions, while, Gene Set Analysis focuses on the detection of coordinated changes in transcriptional levels among sets of biologically related genes. In this field, meta-analysis offers the possibility to compare different studies, addressing the same biological question to fully exploit public gene expression datasets. Results We describe STEPath, a method that starts from gene expression profiles and integrates the analysis of imbalanced region as an a priori step before performing gene set analysis. The application of STEPath in individual studies produced gene set scores weighted by chromosomal activation. As a final step, we propose a way to compare these scores across different studies (meta-analysis) on related biological issues. One complication with meta-analysis is batch effects, which occur because molecular measurements are affected by laboratory conditions, reagent lots and personnel differences. Major problems occur when batch effects are correlated with an outcome of interest and lead to incorrect conclusions. We evaluated the power of combining chromosome mapping and gene set enrichment analysis, performing the analysis on a dataset of leukaemia (example of individual study) and on a dataset of skeletal muscle diseases (meta-analysis approach). In leukaemia, we identified the Hox gene set, a gene set closely related to the pathology that other algorithms of gene set analysis do not identify, while the meta-analysis approach on muscular disease discriminates between related pathologies and correlates similar ones from different studies. Conclusions STEPath is a new method that integrates gene expression profiles, genomic co-expressed regions and the information about the biological function of genes. The usage of the STEPath-computed gene set scores overcomes batch effects in the meta-analysis approaches allowing the direct comparison of different pathologies and different studies on a gene set activation level.
Collapse
Affiliation(s)
- Paolo Martini
- CRIBI Biotechnology Centre, Department of Biology, University of Padova, via U, Bassi 58/B, 35121 Padova, Italy
| | | | | | | | | | | |
Collapse
|
47
|
Almeida MQ, Harran M, Bimpaki EI, Hsiao HP, Horvath A, Cheadle C, Watkins T, Nesterova M, Stratakis CA. Integrated genomic analysis of nodular tissue in macronodular adrenocortical hyperplasia: progression of tumorigenesis in a disorder associated with multiple benign lesions. J Clin Endocrinol Metab 2011; 96:E728-38. [PMID: 21252250 PMCID: PMC3070257 DOI: 10.1210/jc.2010-2420] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Massive macronodular adrenocortical disease or ACTH-independent macronodular adrenal hyperplasia (AIMAH) is a clinically and genetically heterogeneous disorder. OBJECTIVE AND DESIGN Whole-genome expression profiling and oligonucleotide array comparative genomic hybridization changes were analyzed in samples of different nodules from the same patients with AIMAH. Quantitative RT-PCR and staining were employed to validate the mRNA array data. RESULTS Chromosomal gains were more frequent in larger nodules when compared with smaller nodules from the same patients. Among the 50 most overexpressed genes, 50% had a chromosomal locus that was amplified in the comparative genomic hybridization data. Although the list of most over- and underexpressed genes was similar between the nodules of different size, the gene set enrichment analysis identified different pathways associated with AIMAH that corresponded to the size; the smaller nodules were mainly enriched for metabolic pathways, whereas p53 signaling and cancer genes were enriched in larger nodules. Confirmatory studies demonstrated that BCL2, E2F1, EGF, c-KIT, MYB, PRKCA, and CTNNB1 were overexpressed in the larger nodules at messenger and/or protein levels. Chromosomal enrichment analysis showed that chromosomes 20q13 and 14q23 might be involved in progression of AIMAH from smaller to larger tumors. CONCLUSION Integrated transcriptomic and genomic data for AIMAH provides supporting evidence to the hypothesis that larger adrenal lesions, in the context of this chronic, polyclonal hyperplasia, accumulate an increased number of genomic and, subsequently, transcript abnormalities. The latter shows that the disease appears to start with mainly tissue metabolic derangements, as suggested by the study of the smaller nodules, but larger lesions showed aberrant expression of oncogenic pathways.
Collapse
Affiliation(s)
- Madson Q Almeida
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
LIU YU, PATEL SANJAY, NIBBE ROD, MAXWELL SEAN, CHOWDHURY SALIMA, KOYUTURK MEHMET, ZHU XIAOFENG, LARKIN EMMAK, BUXBAUM SARAHG, PUNJABI NARESHM, GHARIB SINAA, REDLINE SUSAN, CHANCE MARKR. Systems biology analyses of gene expression and genome wide association study data in obstructive sleep apnea. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2011:14-25. [PMID: 21121029 PMCID: PMC4465214 DOI: 10.1142/9789814335058_0003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The precise molecular etiology of obstructive sleep apnea (OSA) is unknown; however recent research indicates that several interconnected aberrant pathways and molecular abnormalities are contributors to OSA. Identifying the genes and pathways associated with OSA can help to expand our understanding of the risk factors for the disease as well as provide new avenues for potential treatment. Towards these goals, we have integrated relevant high dimensional data from various sources, such as genome-wide expression data (microarray), protein-protein interaction (PPI) data and results from genome-wide association studies (GWAS) in order to define sub-network elements that connect some of the known pathways related to the disease as well as define novel regulatory modules related to OSA. Two distinct approaches are applied to identify sub-networks significantly associated with OSA. In the first case we used a biased approach based on sixty genes/proteins with known associations with sleep disorders and/or metabolic disease to seed a search using commercial software to discover networks associated with disease followed by information theoretic (mutual information) scoring of the sub-networks. In the second case we used an unbiased approach and generated an interactome constructed from publicly available gene expression profiles and PPI databases, followed by scoring of the network with p-values from GWAS data derived from OSA patients to uncover sub-networks significant for the disease phenotype. A comparison of the approaches reveals a number of proteins that have been previously known to be associated with OSA or sleep. In addition, our results indicate a novel association of Phosphoinositide 3-kinase, the STAT family of proteins and its related pathways with OSA.
Collapse
Affiliation(s)
- YU LIU
- Center for Proteomics & Bioinformatics, Case Western Reserve University (CWRU), Cleveland, Ohio, 44106, USA
| | - SANJAY PATEL
- Division of Pulmonary, Critical Care and Sleep Medicine, CWRU, Cleveland, Ohio, 44106, USA
| | - ROD NIBBE
- Center for Proteomics & Bioinformatics, CWRU, Cleveland, Ohio, 44106, USA
| | - SEAN MAXWELL
- Center for Proteomics & Bioinformatics, CWRU, Cleveland, Ohio, 44106, USA
| | - SALIM A. CHOWDHURY
- Department of Electrical Engineering & Computer Science, CWRU, Cleveland, Ohio, 44106, USA
| | - MEHMET KOYUTURK
- Department of Electrical Engineering & Computer Science, CWRU, Cleveland, Ohio, 44106, USA
| | - XIAOFENG ZHU
- Department of Epidemiology and Biostatistics, CWRU, Cleveland, Ohio, 44106, USA
| | - EMMA K. LARKIN
- Division of Allergy, Pulmonary and Critical Care, Vanderbilt University Medical Center, 1215 21st Ave S., Nashville, Tennessee, 37232, USA
| | - SARAH G BUXBAUM
- Jackson Heart Study, Jackson State University, Jackson, MS 39213, USA
| | - NARESH M. PUNJABI
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - SINA A. GHARIB
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98109, USA
| | - SUSAN REDLINE
- Department of Medicine, CWRU, Cleveland, Ohio, 44106, and Depart of Medicine, Brigham & Women’s Hospital and Beth Israel Deaconess Medical School, Harvard Medical School, Boston, MA, 02115
| | - MARK R. CHANCE
- Center for Proteomics & Bioinformatics, Department of Genetics, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
49
|
Ragazzon B, Libé R, Gaujoux S, Assié G, Fratticci A, Launay P, Clauser E, Bertagna X, Tissier F, de Reyniès A, Bertherat J. Transcriptome analysis reveals that p53 and {beta}-catenin alterations occur in a group of aggressive adrenocortical cancers. Cancer Res 2010; 70:8276-81. [PMID: 20959480 DOI: 10.1158/0008-5472.can-10-2014] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adrenocortical carcinoma (ACC) is a rare disease with an overall poor but heterogeneous prognosis. This heterogeneity could reflect different mechanisms of tumor development. Gene expression profiling by transcriptome analysis led to ACC being divided into two groups of tumors with very different outcomes. Somatic inactivating mutations of the tumor suppressor gene TP53 and activating mutations of the proto-oncogene β-catenin (CTNNB1) are the most frequent mutations identified in ACC. This study investigates the correlation between p53 and β-catenin alterations and the molecular classification of ACC by transcriptome analysis of 51 adult sporadic ACCs. All TP53 and CTNNB1 mutations seemed to be mutually exclusive and were observed only in the poor-outcome ACC group. Most of the abnormal p53 and β-catenin immunostaining was also found in this group. Fifty-two percent of the poor-outcome ACC group had TP53 or CTNNB1 mutations and 60% had abnormal p53 or β-catenin immunostaining. Unsupervised clustering transcriptome analysis of this poor-outcome group revealed three different subgroups, two of them being associated with p53 or β-catenin alterations, respectively. Analysis of p53 and β-catenin target gene expressions in each cluster confirmed a profound and anticipated effect on tumor biology, with distinct profiles logically associated with the respective pathway alterations. The third group had no p53 or β-catenin alteration, suggesting other unidentified molecular defects. This study shows the important respective roles of p53 and β-catenin in ACC development, delineating subgroups of ACC with different tumorigenesis and outcomes.
Collapse
Affiliation(s)
- Bruno Ragazzon
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, Inserm, U1016, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Szabó D, Zsippai A, Bendes M, Tömböl Z, Szabó PM, Rácz K, Igaz P. Pathogenesis of adrenocortical cancer. Orv Hetil 2010; 151:1163-70. [DOI: 10.1556/oh.2010.28931] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A mellékvesekéreg-carcinoma ritka, rossz prognózisú daganat. Döntően sporadikus előfordulású, de ismertek nagyon ritka öröklődő formái is, amelyek a patogenezis megértésében nagy segítséget nyújtanak. A mellékvesekéreg-daganatokra hajlamosító öröklődő szindrómák közé tartozik a Li–Fraumeni-szindróma, a Beckwith–Wiedemann-szindróma, a familiáris adenomatosus polyposis, illetve a döntően benignus daganatokkal társuló multiplex endokrin neoplasia 1-es típusa (MEN1), Carney-komplex és McCune–Albright-szindróma. A mellékvesekéreg-daganatok patogenezisében szereplő főbb mechanizmusok közé tartozik az inzulinszerű növekedési faktor-2 fokozott expressziója, a Wnt/β-katenin és a cAMP-proteinkináz-A jelátviteli utak aktivációja, valamint a p53 és MEN1 gének mutációi. A mellékvesekéreg-carcinoma kezelésében a gyógyszeres lehetőségek meglehetősen korlátozottak. Az utóbbi évek molekuláris-bioinformatikai kutatásai számos eddig ismeretlen patogenetikai út szerepét vetették fel, amelyek új gyógyszeres támadáspontok lehetőségét is jelenthetik. E tanulmányban a szerzők az öröklődő daganatszindrómák patogenezisét, a sporadikus daganatokban észlelt eltéréseket és a legújabb molekuláris-bioinformatikai eredményeket ismertetik.
Collapse
Affiliation(s)
- Diána Szabó
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Adrienn Zsippai
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Melinda Bendes
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Zsófia Tömböl
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Péter M. Szabó
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Károly Rácz
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| | - Péter Igaz
- 1 Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest Szentkirályi u. 46. 1088
| |
Collapse
|