1
|
Araujo-Castro M, Álvarez-Escola C, Casteràs A, Carmona-Bayonas A, Chiara MD, Hanzu FA, Hernando J, Vercher-Conejero JL, Rodríguez-Fraile M, Gómez Dos Santos V, Jimenez-Fonseca P, Giraldo A, Valdés N, Vidal O, Del Olmo-García M, Capdevila J. Spanish consensus on the diagnosis and management of adrenocortical carcinoma. Endocr Relat Cancer 2025; 32:e250034. [PMID: 40215284 PMCID: PMC12053981 DOI: 10.1530/erc-25-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with an estimated incidence of 0.7-2 cases per million/year. The rarity of this disease, coupled with limited preclinical models and clinical trials, has hindered progress, resulting in poor outcomes, with a 5-year survival rate of approximately 35%. Currently, the only available curative treatment is complete surgical resection of the adrenal tumor. For unresectable or metastatic ACC, the current standard therapeutic modalities are mitotane, chemotherapy, radiotherapy and locoregional treatments; however, these are noncurative. Mitotane has an adrenolytic and anti-steroidogenic effect, and it is used in the adjuvant setting for high-risk patients, as systemic therapy for metastatic disease, and/or to control hormonal secretion. While key pathways in ACC pathogenesis have been identified as potential therapeutic targets, results with targeted therapies remain modest, showing that there is a clinical unmet need for novel treatments or new combinations of exiting drugs. Effective management requires a multidisciplinary team of experts to optimize outcomes for patients. This article presents a multidisciplinary consensus on the diagnosis, management, prognosis and follow-up of patients with ACC, and the approach to two special contexts, ACC in pregnant women and hormone-producing ACC. The consensus was coordinated by the Spanish Society of Endocrinology and Nutrition (SEEN) and the Spanish Group of Neuroendocrine and Endocrine Tumors (GETNE), with contribution from experts from related societies including the Spanish Association of Surgeons (AEC), Spanish Society of Urology (AEU), Anatomic-Pathology (SEAP), Nuclear Medicine (SEMNIM), Medical Oncology (SEOM) and Radiotherapeutic Oncology (SEOR).
Collapse
Affiliation(s)
- Marta Araujo-Castro
- Endocrinology & Nutrition Department. Hospital Universitario Ramón y Cajal Madrid, Madrid, Spain
- Instituto de Investigación Biomédica Ramón y Cajal (IRYCIS), Madrid, Spain
| | | | - Ana Casteràs
- Endocrinology & Nutrition Department. Hospital Universitario Vall d´Hebron, Barcelona, Spain
| | - Alberto Carmona-Bayonas
- Medical Oncology Department, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB, Murcia, Spain
| | - María-Dolores Chiara
- Health Research Institute of the Principality of Asturias, Oviedo, Spain
- Institute of Oncology of the Principality of Asturias, University of Oviedo, Oviedo, Spain
| | - Felicia A Hanzu
- Endocrinology & Nutrition Department, Hospital Clinic, University of Barcelona, IDIBPAS, CIBERDEM, Barcelona, Spain
| | - Jorge Hernando
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Hospital Universitario Vall D'Hebron, Vall Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | | - Victoria Gómez Dos Santos
- Instituto de Investigación Biomédica Ramón y Cajal (IRYCIS), Madrid, Spain
- Urology Department, Hospital Universitario Ramón y Cajal Madrid, Madrid, Spain
| | - Paula Jimenez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Alexandra Giraldo
- Radiotherapy Oncology Department, Hospital Universitario Vall D' Hebron, VHIO, Barcelona, Spain
| | - Nuria Valdés
- Endocrinology & Nutrition Department, Hospital Universitario Cruces, Barakaldo, Spain
- UPV/EHU, Biobizkaia, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Oscar Vidal
- General Surgery Department. Hospital Clinic de Barcelona, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Maribel Del Olmo-García
- Endocrinology & Nutrition Department. Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Hospital Universitario Vall D'Hebron, Vall Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
2
|
Sun J, Huai J, Zhang W, Zhao T, Shi R, Wang X, Li M, Jiao X, Zhou X. Therapeutic strategies for adrenocortical carcinoma: integrating genomic insights, molecular targeting, and immunotherapy. Front Immunol 2025; 16:1545012. [PMID: 40145087 PMCID: PMC11937102 DOI: 10.3389/fimmu.2025.1545012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Adrenocortical carcinoma (ACC) is an uncommon and highly aggressive cancer originating in the adrenal cortex, characterized by a high likelihood of recurrence and unfavorable survival rates, particularly in the advanced disease stages. This review discusses the complex molecular pathogenesis of ACC, focusing on critical pathways implicated in the tumorigenesis and providing potential targets for therapy: the Wnt/β-catenin signaling pathway, the IGF2/IGF1R axis, and the apoptosis pathway regulated by p53. Current treatment strategies include surgical resection and mitotane, the sole adrenolytic agent approved by the FDA; however, its effects in advanced disease are suboptimal. Cytotoxic chemotherapy combined with mitotane may be applied, but survival benefits are limited so far. In the following review, we outline emerging targeted therapies, such as mTOR inhibitors and tyrosine kinase inhibitors (TKIs), which show favorable preclinical and clinical data, especially in treatment-resistant ACC. We also emphasize the possible role of immune checkpoint inhibitors (ICIs) in the management of ACC, although their effectiveness is still under study. Upcoming trends in treatment involve forms of personalized medicine, where molecular profiling is integrated to identify actionable biomarkers for administered therapies. This review will attempt to provide a comprehensive framework on how recent breakthroughs in the genomics of ACC, coupled with advances in targeted therapies and immunotherapy, can improve management.
Collapse
Affiliation(s)
- Jing Sun
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiaxuan Huai
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenhui Zhang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianyu Zhao
- Institute and Clinic for Occupational, Social and Environmental Medicine, Ludwig Maximilians University (LMU) University Hospital Munich, Munich, Germany
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacology, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Minglun Li
- Department of Radiation Oncology, Lueneburg Hospital, Lueneburg, Germany
| | - Xuehua Jiao
- Department of Endocrinology, Suzhou Ninth People’s Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Xiqiao Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
3
|
Chu Y, Setayesh J, Dumontet T, Krumeich L, Werner J, Moretti IF, De Sousa K, Kennedy C, La Pensee C, Lerario AM, Hammer GD. Adrenocortical stem cells in health and disease. Nat Rev Endocrinol 2025:10.1038/s41574-025-01091-2. [PMID: 40065108 DOI: 10.1038/s41574-025-01091-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 04/13/2025]
Abstract
The adrenal cortex is the major site of production of steroid hormones, which are essential for life. The normal development and homeostatic renewal of the adrenal cortex depend on capsular stem cells and cortical progenitor cells. These cell populations are highly plastic and support adaptation to physiological demands, injury and disease, linking steroid production and adrenal (organ) homeostasis with systemic endocrine cues and organismal homeostasis. This Review integrates findings from the past decade, outlining the mechanisms that govern the establishment and maintenance of the adrenal stem cell niche under different physiological and pathological conditions. The sophisticated regulation of the stem cell niche by gene regulatory networks, coordinated through paracrine and endocrine signalling, is highlighted in a context-dependent and sex-specific manner. We discuss how dysregulation of this intricate regulatory network is implicated in a wide range of adrenal diseases, and how emerging knowledge from adrenal stem cell research is inspiring the future development of gene-based and cell-based therapeutic strategies.
Collapse
Affiliation(s)
- Yulan Chu
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jordan Setayesh
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Lauren Krumeich
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Johanna Werner
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Isabele F Moretti
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Kelly De Sousa
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Christopher Kennedy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Christopher La Pensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Antonio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Gary D Hammer
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Endocrine Oncology Program, Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Mariniello K, Pittaway JFH, Altieri B, Borges KS, Hadjidemetriou I, Ribeiro C, Ruiz-Babot G, Tourigny DS, Lim JA, Foster J, Cleaver J, Sosabowski J, Rahman N, Doroszko M, Hantel C, Sigala S, Abate A, Tamburello M, Kiseljak-Vassiliades K, Wierman M, Hall C, Parvanta L, Abdel-Aziz TE, Chung TT, Marco AD, Palazzo F, Gomez-Sanchez CE, Taylor DR, Rayner O, Ronchi CL, Gaston-Massuet C, Sbiera S, Drake WM, Rognoni E, Kroiss M, Breault DT, Fassnacht M, Guasti L. Dlk1 is a novel adrenocortical stem/progenitor cell marker that predicts malignancy in adrenocortical carcinoma. Cancer Commun (Lond) 2025. [PMID: 40035383 DOI: 10.1002/cac2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/13/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025] Open
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James F H Pittaway
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Kleiton Silva Borges
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Irene Hadjidemetriou
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technical, University Dresden, Dresden, Germany
| | | | - Jiang A Lim
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Foster
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Julie Cleaver
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Jane Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Nafis Rahman
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Milena Doroszko
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes at Rocky Mountain Regional Veterans Affair Medical Center, Washington, District of Columbia, USA
| | - Margaret Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes at Rocky Mountain Regional Veterans Affair Medical Center, Washington, District of Columbia, USA
| | - Charlotte Hall
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Laila Parvanta
- Department of Surgery, St Bartholomew's Hospital, West Smithfield, London, UK
| | - Tarek E Abdel-Aziz
- Department of Surgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Teng-Teng Chung
- Department of Endocrinology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Aimee Di Marco
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London, UK
| | - Fausto Palazzo
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London, UK
| | - Celso E Gomez-Sanchez
- Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center and the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - David R Taylor
- Department of Clinical Biochemistry (Synnovis Analytics), King's College Hospital, London, UK
| | - Oliver Rayner
- Department of Clinical Biochemistry (Synnovis Analytics), King's College Hospital, London, UK
| | - Cristina L Ronchi
- Institute of Metabolism and System Research College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - William M Drake
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Emanuel Rognoni
- Centre for Cell Biology & Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthias Kroiss
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, Würzburg, Germany
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, München, Germany
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
You M, Lu Z, Wang B, Liu M, Zhou Q, Li L, Tong D, Zhao Y, Zhang H, Bai Z, Wang L, Cao T, Gao P, Yan Z, Zhu Z. Meriones unguiculatus serves as a spontaneous primary aldosteronism rodent model. PLoS One 2025; 20:e0314943. [PMID: 39946402 PMCID: PMC11824956 DOI: 10.1371/journal.pone.0314943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/19/2024] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Primary aldosteronism (PA) is the most common form of endocrine hypertension. The available animal models of PA rely on gene manipulation, thus fail to duplicate the general pathological process of PA in humans. Meriones unguiculatus (MU) has been reported to possess a large size of adrenal gland and an elevated ability to save water. In this study, we aimed to confirm whether MU can serve as an ideal animal model of PA. METHODS Sprague Dawley rats of the same body weight (SD1) or age (SD2) as MU were used as control groups. Blood pressure and serum aldosterone, renin and electrolyte levels were measured, and the oral salt loading test was used as confirmatory test to compare the inhibition level of the renin angiotensin aldosterone system (RAAS) among the three groups. The expression and distribution of CYP11B2 (aldosterone synthase) were evaluated in the adrenal gland of each group. RESULTS MU exhibited typical clinical manifestations of PA, including hypertension, hyperaldosteronism, low renin levels and strong sodium retention and potassium excretion abilities. Compared with control groups, the inhibitory effect of a high-sodium diet on the RAAS was milder in MU, accompanied by significant cardiac dysfunction. The protein expression level and distribution area of CYP11B2 were significantly increased in the adrenal gland of MU. CONCLUSION The current study reveals that MU could serve as an ideal spontaneous PA model. The increased expression and distribution of CYP11B2 stimulate the excessive aldosterone production in a renin-independent manner, leading to a significant increase in blood pressure in MU.
Collapse
Affiliation(s)
- Mei You
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Bowen Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Min Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Qing Zhou
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Li Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Dan Tong
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Yu Zhao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Hexuan Zhang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Zhongping Bai
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Lijuan Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Tingbing Cao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, China
- Chongqing Institute for Brain and Intelligence, Chongqing, China
| |
Collapse
|
6
|
Nair V, Demitri C, Thankam FG. Competitive signaling and cellular communications in myocardial infarction response. Mol Biol Rep 2025; 52:129. [PMID: 39820809 PMCID: PMC11739196 DOI: 10.1007/s11033-025-10236-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Cell communication and competition pathways are malleable to Myocardial Infarction (MI). Key signals, transcriptive regulators, and metabolites associated with apoptotic responses such as Myc, mTOR, and p53 are important players in the myocardium. The individual state of cardiomyocytes, fibroblasts, and macrophages in the heart tissue are adaptable in times of stress. The overlapping communication pathways of Wnt/β-catenin, Notch, and c-Kit exhibit the involvement of important factors in cell competition in the myocardium. Depending on the effects of these pathways on genetic expression and signal amplification, the proliferative capacities of the previously stated cells that make up the myocardium, amplify or diminish. This creates a distinct classification of "fit" and "unfit" cells. Beyond straightforward traits, the intricate metabolite interactions between neighboring cells unveil a complex battle. Strategic manipulation of these pathways holds translational promise for rapid cardiac recovery post-trauma.
Collapse
Affiliation(s)
- Vishnu Nair
- Department of Molecular, Cell, & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Christian Demitri
- Department of Experimental Medicine, University of Salento, Lecce, 73100, Italy
| | - Finosh G Thankam
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
7
|
Zou C, Zhang Y, Liu C, Li Y, Lin C, Chen H, Hou J, Gao G, Liu Z, Yan Q, Su W. Identification of CENPM as a key gene driving adrenocortical carcinoma metastasis via physical interaction with immune checkpoint ligand FGL1. Clin Transl Med 2025; 15:e70182. [PMID: 39778025 PMCID: PMC11707433 DOI: 10.1002/ctm2.70182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Distant metastasis occurs in the majority of adrenocortical carcinoma (ACC), leading to an extremely poor prognosis. However, the key genes driving ACC metastasis remain unclear. METHODS Weighted gene co-expression network analysis (WGCNA) and functional enrichment analysis were conducted to identify ACC metastasis-related genes. Data from RNA-seq and microarray were analyzed to reveal correlations of the CENPM gene with cancer, metastasis, and survival in ACC. Immunohistochemistry was used to assess CENPM protein expression. The impact of CENPM on metastasis behaviour was verified in ACC (H295R and SW-13) cells and xenograft NPG mice. DIA quantitative proteomics analysis, western blot, immunofluorescence, and co-immunoprecipitation assay were performed to identify the downstream target of CENPM. RESULTS Among the 12 035 analyzed genes, 363 genes were related to ACC metastasis and CENPM was identified as the hub gene. CENPM was upregulated in ACC samples and associated with metastasis and poor prognosis. Knockdown of CENPM inhibited proliferation, invasion, and migration of ACC cells and suppressed liver metastasis in xenograft NPG mice. Collagen-containing extracellular matrix signalling was primarily downregulated when CENPM was knocked down. FGL1, important components of ECM signalling and immune checkpoint ligand of LAG3, were downregulated following CENPM silence, overexpressed in human advanced ACC samples, and colocalized with CENPM. Physical interaction between CENPM and FGL1 was identified. Overexpression of FGL1 rescued migration and invasion of CENPM knockdown ACC cells. CONCLUSIONS CENPM is a key gene in driving ACC metastasis. CENPM promotes ACC metastasis through physical interaction with the immune checkpoint ligand FGL1. CENPM can be used as a new prognostic biomarker and therapeutic target for metastatic ACC. HIGHLIGHTS CENPM is the key gene that drives ACC metastasis, and a robust biomarker for ACC prognosis. Silencing CENPM impedes ACC metastasis in vitro and in vivo by physical interaction with immune checkpoint ligand FGL1. FGL1 is overexpressed in ACC and promotes ACC metastasis.
Collapse
Affiliation(s)
- Cunru Zou
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Yu Zhang
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Chengyue Liu
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Yaxin Li
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Congjie Lin
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Hao Chen
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Jiangping Hou
- Department of OphthalmologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Guojun Gao
- Department of Urology SurgeryAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Zheng Liu
- Department of Urology SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Qiupeng Yan
- Department of Teaching and Research Section of Introduction to Basic MedicineSchool of Basic MedicineShandong Second Medical UniversityWeifangChina
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher EducationShandong Second Medical UniversityWeifangChina
| | - Wenxia Su
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| |
Collapse
|
8
|
Borges KS, Little DW, Magalhães TDA, Ribeiro C, Dumontet T, Lapensee C, Basham KJ, Seth A, Azova S, Guagliardo NA, Barrett PQ, Berber M, O'Connell AE, Turcu AF, Lerario AM, Mohan DR, Rainey W, Carlone DL, Hirschhorn JN, Salic A, Breault DT, Hammer GD. Non-canonical Wnt signaling triggered by WNT2B drives adrenal aldosterone production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609423. [PMID: 39229119 PMCID: PMC11370552 DOI: 10.1101/2024.08.23.609423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The steroid hormone aldosterone, produced by the zona glomerulosa (zG) of the adrenal gland, is a master regulator of plasma electrolytes and blood pressure. While aldosterone control by the renin-angiotensin system is well understood, other key regulatory factors have remained elusive. Here, we replicated a prior association between a non-coding variant in WNT2B and an increased risk of primary aldosteronism, a prevalent and debilitating disease caused by excessive aldosterone production. We further show that in both mice and humans, WNT2B is expressed in the mesenchymal capsule surrounding the adrenal cortex, in close proximity to the zG. Global loss of Wnt2b in the mouse results in a dysmorphic and hypocellular zG, with impaired aldosterone production. Similarly, humans harboring WNT2B loss-of-function mutations develop a novel form of Familial Hyperreninemic Hypoaldosteronism, designated here as Type 4. Additionally, we demonstrate that WNT2B signals by activating the non-canonical Wnt/planar cell polarity pathway. Our findings identify WNT2B as a key regulator of zG function and aldosterone production with important clinical implications.
Collapse
Affiliation(s)
- Kleiton S Borges
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Donald W Little
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chris Lapensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Aishwarya Seth
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Svetlana Azova
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Nick A Guagliardo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Paula Q Barrett
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0735, USA
| | - Mesut Berber
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Amy E O'Connell
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Adina F Turcu
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dipika R Mohan
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - William Rainey
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Joel N Hirschhorn
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge MA, 02142
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
9
|
Mariniello K, Pittaway JFH, Altieri B, Borges KS, Hadjidemetriou I, Ribeiro C, Ruiz-Babot G, Lim JA, Foster J, Cleaver J, Sosabowski J, Rahman N, Doroszko M, Hantel C, Sigala S, Abate A, Tamburello M, Kiseljak-Vassiliades K, Wierman M, Parvanta L, Abdel-Aziz TE, Chung TT, Di Marco A, Palazzo F, Gomez-Sanchez CE, Taylor DR, Rayner O, Ronchi CL, Gaston-Massuet C, Sbiera S, Drake WM, Rognoni E, Kroiss M, Breault DT, Fassnacht M, Guasti L. Dlk1 is a novel adrenocortical stem/progenitor cell marker that predicts malignancy in adrenocortical carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609117. [PMID: 39229217 PMCID: PMC11370565 DOI: 10.1101/2024.08.22.609117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Disruption of processes involved in tissue development and homeostatic self-renewal is increasingly implicated in cancer initiation, progression, and recurrence. The adrenal cortex is a dynamic tissue that undergoes life-long turnover. Here, using genetic fate mapping and murine adrenocortical carcinoma (ACC) models, we have identified a population of adrenocortical stem cells that express delta-like non-canonical Notch ligand 1 (DLK1). These cells are active during development, near dormant postnatally but are re-expressed in ACC. In a study of over 200 human ACC samples, we have shown DLK1 expression is ubiquitous and is an independent prognostic marker of recurrence-free survival. Paradoxically, despite its progenitor role, spatial transcriptomic analysis has identified DLK1 expressing cell populations to have increased steroidogenic potential in human ACC, a finding also observed in four human and one murine ACC cell lines. Finally, the cleavable DLK1 ectodomain is measurable in patients' serum and can discriminate between ACC and other adrenal pathologies with high sensitivity and specificity to aid in diagnosis and follow-up of ACC patients. These data demonstrate a prognostic role for DLK1 in ACC, detail its hierarchical expression in homeostasis and oncogenic transformation and propose a role for its use as a biomarker in this malignancy.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James F H Pittaway
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Kleiton Silva Borges
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Irene Hadjidemetriou
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claudio Ribeiro
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technical, University Dresden, Dresden, Germany
| | - Jiang A Lim
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Foster
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Julie Cleaver
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Jane Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Nafis Rahman
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Milena Doroszko
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091 Zurich, Switzerland
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124 Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124 Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124 Brescia, Italy
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes at Rocky Mountain Regional Veterans Affair Medical Center, Washington, DC, USA
| | - Margaret Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Endocrinology, Metabolism and Diabetes at Rocky Mountain Regional Veterans Affair Medical Center, Washington, DC, USA
| | - Laila Parvanta
- Department of Surgery, St Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, United Kingdom
| | - Tarek E Abdel-Aziz
- Department of Surgery, University College London Hospitals NHS Foundation Trust, London NW1 2PG, United Kingdom
| | - Teng-Teng Chung
- Department of Endocrinology, University College London Hospitals NHS Foundation Trust, London NW1 2PG, United Kingdom
| | - Aimee Di Marco
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London W12 0HS, United Kingdom
| | - Fausto Palazzo
- Department of Endocrine and Thyroid Surgery, Hammersmith Hospital, Imperial College London, London W12 0HS, United Kingdom
| | - Celso E Gomez-Sanchez
- Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center and the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - David R Taylor
- Department of Clinical Biochemistry (Synnovis Analytics), King's College Hospital, London SE5 9RS, United Kingdom
| | - Oliver Rayner
- Department of Clinical Biochemistry (Synnovis Analytics), King's College Hospital, London SE5 9RS, United Kingdom
| | - Cristina L Ronchi
- Institute of Metabolism and System Research College of Medical and Dental Sciences, University of Birmingham, B15 2TT, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - William M Drake
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Emanuel Rognoni
- Centre for Cell Biology & Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthias Kroiss
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Ziemssenstraße 5, 80336 München, Germany
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Dept. of Medicine, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
10
|
Cioppi F, Cantini G, Ercolino T, Chetta M, Zanatta L, Nesi G, Mannelli M, Maggi M, Canu L, Luconi M. Targeted Next Generation Sequencing molecular profiling and its clinical application in adrenocortical cancer. Eur J Endocrinol 2024; 191:17-30. [PMID: 38917236 DOI: 10.1093/ejendo/lvae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
OBJECTIVE Adrenal cortical carcinoma (ACC) is a rare malignancy with a generally poor but heterogeneous prognosis, especially depending on the tumour stage at diagnosis. Identification of somatic gene alterations combined with clinical/histopathological evaluation of the tumour can help improve prognostication. We applied a simplified targeted-Next-Generation Sequencing (NGS) panel to characterise the mutational profiles of ACCs, providing potentially relevant information for better patient management. DESIGN AND METHODS Thirty frozen tumour specimens from a local ACC series were retrospectively analysed by a custom-NGS panel (CDKN2A, CTNNB1, DAXX, MED12, NF1, PRKAR1A, RB1, TERT, TP53, ZNRF3) to detect somatic prioritised single-nucleotide variants. This cohort was integrated with 86 patients from the ACC-TCGA series bearing point-mutations in the same genes and their combinations identified by our panel. Primary endpoints of the analysis on the total cohort (113 patients) were overall survival (OS) and progression-free survival (PFS), and hazard ratio (HR) for the different alterations grouped by the signalling pathways/combinations affected. RESULTS Different PFS, OS, and HR were associated to the different pathways/combinations, being NF1 + TP53 and Wnt/β-catenin + Rb/p53 combined mutations the most deleterious, with a statistical significance for progression HR which is retained only in low-(I/II) stages-NF1 + TP53 combination: HR = 2.96[1.01-8.69] and HR = 13.23[3.15-55.61], all and low stages, respectively; Wnt/β-catenin + Rb/p53 combined pathways: HR = 6.47[2.54-16.49] and HR = 16.24[3.87-68.00], all and low-stages, respectively. CONCLUSIONS A simplified targeted-NGS approach seems the best routinely applicable first step towards somatic genetic characterisation of ACC for prognostic assessment. This approach proved to be particularly promising in low-stage cases, suggesting the need for more stringent surveillance and personalised treatment.
Collapse
Affiliation(s)
- Francesca Cioppi
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
| | - Giulia Cantini
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
| | - Tonino Ercolino
- Azienda Ospedaliero-Universitaria Careggi, (AOUC), 50139 Florence, Italy
| | - Massimiliano Chetta
- Medical Genetics, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Cardarelli, Padiglione, 80131 Naples, Italy
| | - Lorenzo Zanatta
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Azienda Ospedaliero-Universitaria Careggi, (AOUC), 50139 Florence, Italy
| | - Gabriella Nesi
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Massimo Mannelli
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
| | - Mario Maggi
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- Azienda Ospedaliero-Universitaria Careggi, (AOUC), 50139 Florence, Italy
| | - Letizia Canu
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- Azienda Ospedaliero-Universitaria Careggi, (AOUC), 50139 Florence, Italy
| | - Michaela Luconi
- European Network for the Study of Adrenal Tumours (ENSAT) Centre of Excellence, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, Endocrinology Section, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
| |
Collapse
|
11
|
Zhang J, Wu L, Su T, Liu H, Jiang L, Jiang Y, Wu Z, Chen L, Li H, Zheng J, Sun Y, Peng H, Han R, Ning G, Ye L, Wang W. Pharmacogenomic analysis in adrenocortical carcinoma reveals genetic features associated with mitotane sensitivity and potential therapeutics. Front Endocrinol (Lausanne) 2024; 15:1365321. [PMID: 38779454 PMCID: PMC11109426 DOI: 10.3389/fendo.2024.1365321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Background Adrenocortical carcinoma (ACC) is an aggressive endocrine malignancy with limited therapeutic options. Treating advanced ACC with mitotane, the cornerstone therapy, remains challenging, thus underscoring the significance to predict mitotane response prior to treatment and seek other effective therapeutic strategies. Objective We aimed to determine the efficacy of mitotane via an in vitro assay using patient-derived ACC cells (PDCs), identify molecular biomarkers associated with mitotane response and preliminarily explore potential agents for ACC. Methods In vitro mitotane sensitivity testing was performed in 17 PDCs and high-throughput screening against 40 compounds was conducted in 8 PDCs. Genetic features were evaluated in 9 samples using exomic and transcriptomic sequencing. Results PDCs exhibited variable sensitivity to mitotane treatment. The median cell viability inhibition rate was 48.4% (IQR: 39.3-59.3%) and -1.2% (IQR: -26.4-22.1%) in responders (n=8) and non-responders (n=9), respectively. Median IC50 and AUC were remarkably lower in responders (IC50: 53.4 µM vs 74.7 µM, P<0.0001; AUC: 158.0 vs 213.5, P<0.0001). Genomic analysis revealed CTNNB1 somatic alterations were only found in responders (3/5) while ZNRF3 alterations only in non-responders (3/4). Transcriptomic profiling found pathways associated with lipid metabolism were upregulated in responder tumors whilst CYP27A1 and ABCA1 expression were positively correlated to in vitro mitotane sensitivity. Furthermore, pharmacologic analysis identified that compounds including disulfiram, niclosamide and bortezomib exhibited efficacy against PDCs. Conclusion ACC PDCs could be useful for testing drug response, drug repurposing and guiding personalized therapies. Our results suggested response to mitotane might be associated with the dependency on lipid metabolism. CYP27A1 and ABCA1 expression could be predictive markers for mitotane response, and disulfiram, niclosamide and bortezomib could be potential therapeutics, both warranting further investigation.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luming Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingwei Su
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Jiang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiran Jiang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyuan Wu
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Chen
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haorong Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingkai Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hangya Peng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rulai Han
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Tai Y, Shang J. Wnt/β-catenin signaling pathway in the tumor progression of adrenocortical carcinoma. Front Endocrinol (Lausanne) 2024; 14:1260701. [PMID: 38269250 PMCID: PMC10806569 DOI: 10.3389/fendo.2023.1260701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024] Open
Abstract
Adrenocortical carcinoma (ACC) is an uncommon, aggressive endocrine malignancy with a high rate of recurrence, a poor prognosis, and a propensity for metastasis. Currently, only mitotane has received certification from both the US Food and Drug Administration (FDA) and the European Medicines Agency for the therapy of advanced ACC. However, treatment in the advanced periods of the disorders is ineffective and has serious adverse consequences. Completely surgical excision is the only cure but has failed to effectively improve the survival of advanced patients. The aberrantly activated Wnt/β-catenin pathway is one of the catalysts for adrenocortical carcinogenesis. Research has concentrated on identifying methods that can prevent the stimulation of the Wnt/β-catenin pathway and are safe and advantageous for patients in view of the absence of effective treatments and the frequent alteration of the Wnt/β-catenin pathway in ACC. Comprehending the complex connection between the development of ACC and Wnt/β-catenin signaling is essential for accurate pharmacological targets. In this review, we summarize the potential targets between adrenocortical carcinoma and the Wnt/β-catenin signaling pathway. We analyze the relevant targets of drugs or inhibitors that act on the Wnt pathway. Finally, we provide new insights into how drugs or inhibitors may improve the treatment of ACC.
Collapse
Affiliation(s)
- Yanghao Tai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Jiwen Shang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
- Department of Ambulatory Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
13
|
Li H, Li J, Ma Y, Wang Z, Peng Z, Xu H, Bi H, Althaf Hussain S, Li Z. The active phthalate metabolite, DHEP, induces proliferation and metastasis of prostate cancer cells via upregulation of β-catenin and downregulation of KLF7. Bioorg Chem 2023; 141:106864. [PMID: 37734194 DOI: 10.1016/j.bioorg.2023.106864] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
Phthalates such as DHEP are among the widely used compounds in industry. It has been shown that DHEP can convey various biological consequences in mammalian cells, among them, the carcinogenic effects of DHEP are emphasized. The present study aimed to assess the impact of DHEP exposure on the proliferation and invasiveness of DU145 prostate cancer cells through in vitro and in vivo models. The DU145 cells were treated with increasing concentrations of DHEP and the tumorigenic parameters were analyzed. KLF7 as a probable mediator of the effect of DHEP was either overexpressed or knocked down in DU145 to evaluate the probable impact of KLF7 on the biological effects of DHEP. The effect of DHEP was also studied in a DU145 xenograft tumor model. The moderate doses of DHEP increased the proliferation and migration of DU145 cells. In the case of gene expression patterns, DHEP reduced the levels of p53 and KLF7 while elevated the expression of β-catenin. The knock-down of KLF7 conveyed comparable effects to that of DHEP to some degree and increased the proliferation of DU145 cells, while the transduction of KLF7 increased the expressions of p53 and p21 along with controlling the tumor size. The present study demonstrated the potential of DHEP in increasing the tumorigenic properties of DU145 cells along with a focus on the underlying mechanisms. Sustained exposure to DHEP can cause a dysregulation in balance between oncogenes and tumor suppressor genes which is the hallmark of malignant transformation. Thus, special considerations seem necessary for the safe exploitation of phthalates.
Collapse
Affiliation(s)
- Hecheng Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jianping Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yubo Ma
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Ziming Wang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Zihe Peng
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Hang Xu
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Hang Bi
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Zhaolun Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
14
|
Ghosh C, Hu J, Kebebew E. Advances in translational research of the rare cancer type adrenocortical carcinoma. Nat Rev Cancer 2023; 23:805-824. [PMID: 37857840 DOI: 10.1038/s41568-023-00623-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2023] [Indexed: 10/21/2023]
Abstract
Adrenocortical carcinoma is a rare malignancy with an annual worldwide incidence of 1-2 cases per 1 million and a 5-year survival rate of <60%. Although adrenocortical carcinoma is rare, such rare cancers account for approximately one third of patients diagnosed with cancer annually. In the past decade, there have been considerable advances in understanding the molecular basis of adrenocortical carcinoma. The genetic events associated with adrenocortical carcinoma in adults are distinct from those of paediatric cases, which are often associated with germline or somatic TP53 mutations and have a better prognosis. In adult primary adrenocortical carcinoma, the main somatic genetic alterations occur in genes that encode proteins involved in the WNT-β-catenin pathway, cell cycle and p53 apoptosis pathway, chromatin remodelling and telomere maintenance pathway, cAMP-protein kinase A (PKA) pathway or DNA transcription and RNA translation pathways. Recently, integrated molecular studies of adrenocortical carcinomas, which have characterized somatic mutations and the methylome as well as gene and microRNA expression profiles, have led to a molecular classification of these tumours that can predict prognosis and have helped to identify new therapeutic targets. In this Review, we summarize these recent translational research advances in adrenocortical carcinoma, which it is hoped could lead to improved patient diagnosis, treatment and outcome.
Collapse
Affiliation(s)
| | - Jiangnan Hu
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Electron Kebebew
- Department of Surgery, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
15
|
Dev A, Vachher M, Prasad CP. β-catenin inhibitors in cancer therapeutics: intricacies and way forward. Bioengineered 2023; 14:2251696. [PMID: 37655825 PMCID: PMC10478749 DOI: 10.1080/21655979.2023.2251696] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023] Open
Abstract
β-catenin is an evolutionary conserved, quintessential, multifaceted protein that plays vital roles in cellular homeostasis, embryonic development, organogenesis, stem cell maintenance, cell proliferation, migration, differentiation, apoptosis, and pathogenesis of various human diseases including cancer. β-catenin manifests both signaling and adhesive features. It acts as a pivotal player in intracellular signaling as a component of versatile WNT signaling cascade involved in embryonic development, homeostasis as well as in carcinogenesis. It is also involved in Ca2+ dependent cell adhesion via interaction with E-cadherin at the adherens junctions. Aberrant β-catenin expression and its nuclear accumulation promote the transcription of various oncogenes including c-Myc and cyclinD1, thereby contributing to tumor initiation, development, and progression. β-catenin's expression is closely regulated at various levels including its stability, sub-cellular localization, as well as transcriptional activity. Understanding the molecular mechanisms of regulation of β-catenin and its atypical expression will provide researchers not only the novel insights into the pathogenesis and progression of cancer but also will help in deciphering new therapeutic avenues. In the present review, we have summarized the dual functions of β-catenin, its role in signaling, associated mutations as well as its role in carcinogenesis and tumor progression of various cancers. Additionally, we have discussed the challenges associated with targeting β-catenin molecule with the presently available drugs and suggested the possible way forward in designing new therapeutic alternatives against this oncogene.
Collapse
Affiliation(s)
- Arundhathi Dev
- Department of Medical Oncology (Laboratory), DR BRAIRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Meenakshi Vachher
- Department of Biochemistry, Institute of Home Economics, University of Delhi, New Delhi, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology (Laboratory), DR BRAIRCH, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
Lippert J, Dischinger U, Appenzeller S, Prete A, Kircher S, Skordilis K, Elhassan YS, Altieri B, Fassnacht M, Ronchi CL. Performance of DNA-based biomarkers for classification of adrenocortical carcinoma: a prognostic study. Eur J Endocrinol 2023; 189:262-270. [PMID: 37590967 DOI: 10.1093/ejendo/lvad112] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/16/2023] [Accepted: 06/14/2023] [Indexed: 08/19/2023]
Abstract
OBJECTIVE Adrenocortical carcinoma (ACC) is a rare aggressive malignancy with heterogeneous clinical outcomes. Recent studies proposed a combination of clinical/histopathological parameters (S-GRAS score) or molecular biomarkers (BMs) to improve prognostication. We performed a comparative analysis of DNA-based BMs by evaluating their added prognostic value to the S-GRAS score. DESIGN AND METHODS A total of 194 formalin-fixed, paraffin-embedded (FFPE) ACC samples were analysed, including a retrospective training cohort (n = 107) and a prospective validation cohort (n = 87). Targeted DNA sequencing and pyrosequencing were used to detect somatic single-nucleotide variations in ACC-specific genes and methylation in the promoter region of paired box 5 (PAX5). The European Network for the Study of Adrenocortical Tumors (ENSAT) tumour stage, age, symptoms at presentation, resection status, and Ki-67 were combined to calculate S-GRAS. Endpoints were overall (OS), progression-free (PFS), and disease-free survival (DFS). Prognostic role was evaluated by multivariable survival analysis and their performance compared by Harrell's concordance index (C index). RESULTS In training cohort, an independent prognostic role was confirmed at multivariate analysis for two DNA-based BMs: alterations in Wnt/β-catenin and Rb/p53 pathways and hypermethylated PAX5 (both P< .05 for PFS and DFS, hazard ratio [HR] 1.47-2.33). These were combined to S-GRAS to obtain a combined (COMBI) score. At comparative analysis, the best discriminative prognostic model was COMBI score in both cohorts for all endpoints, followed by S-GRAS score (C index for OS 0.724 and 0.765, PFS 0.717 and 0.670, and DFS 0.699 and 0.644, respectively). CONCLUSIONS Targeted DNA-based BM evaluated on routinely available FFPE samples improves prognostication of ACC beyond routinely available clinical and histopathological parameters. This approach may help to better individualise patient's management.
Collapse
Affiliation(s)
- Juliane Lippert
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Ulrich Dischinger
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Silke Appenzeller
- Core Unit Bioinformatics, Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97078 Würzburg, Germany
| | - Alessandro Prete
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B152TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham B152TT, United Kingdom
- NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham B152TT, United Kingdom
| | - Stefan Kircher
- Department of Pathology, University of Würzburg, 97078 Würzburg, Germany
| | - Kassiani Skordilis
- Department of Histopathology, University Hospitals Birmingham NHS Foundation Trust, Birmingham B152GW, United Kingdom
| | - Yasir S Elhassan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B152TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham B152TT, United Kingdom
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, 97080 Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken (CCCM), University of Würzburg, 97078 Würzburg, Germany
- Central Labor, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Cristina L Ronchi
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, 97080 Würzburg, Germany
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B152TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham B152TT, United Kingdom
| |
Collapse
|
17
|
Penny MK, Lerario AM, Basham KJ, Chukkapalli S, Mohan DR, LaPensee C, Converso-Baran K, Hoenerhoff MJ, Suárez-Fernández L, del Rey CG, Giordano TJ, Han R, Newman EA, Hammer GD. Targeting Oncogenic Wnt/β-Catenin Signaling in Adrenocortical Carcinoma Disrupts ECM Expression and Impairs Tumor Growth. Cancers (Basel) 2023; 15:3559. [PMID: 37509222 PMCID: PMC10377252 DOI: 10.3390/cancers15143559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/12/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare but highly aggressive cancer with limited treatment options and poor survival for patients with advanced disease. An improved understanding of the transcriptional programs engaged in ACC will help direct rational, targeted therapies. Whereas activating mutations in Wnt/β-catenin signaling are frequently observed, the β-catenin-dependent transcriptional targets that promote tumor progression are poorly understood. To address this question, we analyzed ACC transcriptome data and identified a novel Wnt/β-catenin-associated signature in ACC enriched for the extracellular matrix (ECM) and predictive of poor survival. This suggested an oncogenic role for Wnt/β-catenin in regulating the ACC microenvironment. We further investigated the minor fibrillar collagen, collagen XI alpha 1 (COL11A1), and found that COL11A1 expression originates specifically from cancer cells and is strongly correlated with both Wnt/β-catenin activation and poor patient survival. Inhibition of constitutively active Wnt/β-catenin signaling in the human ACC cell line, NCI-H295R, significantly reduced the expression of COL11A1 and other ECM components and decreased cancer cell viability. To investigate the preclinical potential of Wnt/β-catenin inhibition in the adrenal microenvironment, we developed a minimally invasive orthotopic xenograft model of ACC and demonstrated that treatment with the newly developed Wnt/β-catenin:TBL1 inhibitor Tegavivint significantly reduced tumor growth. Together, our data support that the inhibition of aberrantly active Wnt/β-catenin disrupts transcriptional reprogramming of the microenvironment and reduces ACC growth and survival. Furthermore, this β-catenin-dependent oncogenic program can be therapeutically targeted with a newly developed Wnt/β-catenin inhibitor. These results show promise for the further clinical development of Wnt/β-catenin inhibitors in ACC and unveil a novel Wnt/β-catenin-regulated transcriptome.
Collapse
Affiliation(s)
- Morgan K. Penny
- Doctoral Program in Cancer Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Antonio M. Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kaitlin J. Basham
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sahiti Chukkapalli
- Mott Solid Tumor Oncology Program, C.S. Mott Children’s and Women’s Hospital, Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dipika R. Mohan
- Doctoral Program in Cancer Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Chris LaPensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kimber Converso-Baran
- UMH Frankel Cardiovascular Center Physiology and Phenotyping Core, Ann Arbor, MI 48109, USA
| | - Mark J. Hoenerhoff
- In Vivo Animal Core, Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Laura Suárez-Fernández
- Department Head and Neck Oncology, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain
| | - Carmen González del Rey
- Department of Pathology, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain
| | - Thomas J. Giordano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Ruolan Han
- Iterion Therapeutics, Inc., Houston, TX 77021, USA
| | - Erika A. Newman
- Mott Solid Tumor Oncology Program, C.S. Mott Children’s and Women’s Hospital, Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gary D. Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan Health System, Ann Arbor, MI 48109, USA
| |
Collapse
|
18
|
Mohan DR, Borges KS, Finco I, LaPensee CR, Rege J, Solon AL, Little DW, Else T, Almeida MQ, Dang D, Haggerty-Skeans J, Apfelbaum AA, Vinco M, Wakamatsu A, Mariani BMP, Amorim LC, Latronico AC, Mendonca BB, Zerbini MCN, Lawlor ER, Ohi R, Auchus RJ, Rainey WE, Marie SKN, Giordano TJ, Venneti S, Fragoso MCBV, Breault DT, Lerario AM, Hammer GD. β-Catenin-Driven Differentiation Is a Tissue-Specific Epigenetic Vulnerability in Adrenal Cancer. Cancer Res 2023; 83:2123-2141. [PMID: 37129912 PMCID: PMC10330305 DOI: 10.1158/0008-5472.can-22-2712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/19/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
Adrenocortical carcinoma (ACC) is a rare cancer in which tissue-specific differentiation is paradoxically associated with dismal outcomes. The differentiated ACC subtype CIMP-high is prevalent, incurable, and routinely fatal. CIMP-high ACC possess abnormal DNA methylation and frequent β-catenin-activating mutations. Here, we demonstrated that ACC differentiation is maintained by a balance between nuclear, tissue-specific β-catenin-containing complexes, and the epigenome. On chromatin, β-catenin bound master adrenal transcription factor SF1 and hijacked the adrenocortical super-enhancer landscape to maintain differentiation in CIMP-high ACC; off chromatin, β-catenin bound histone methyltransferase EZH2. SF1/β-catenin and EZH2/β-catenin complexes present in normal adrenals persisted through all phases of ACC evolution. Pharmacologic EZH2 inhibition in CIMP-high ACC expelled SF1/β-catenin from chromatin and favored EZH2/β-catenin assembly, erasing differentiation and restraining cancer growth in vitro and in vivo. These studies illustrate how tissue-specific programs shape oncogene selection, surreptitiously encoding targetable therapeutic vulnerabilities. SIGNIFICANCE Oncogenic β-catenin can use tissue-specific partners to regulate cellular differentiation programs that can be reversed by epigenetic therapies, identifying epigenetic control of differentiation as a viable target for β-catenin-driven cancers.
Collapse
Affiliation(s)
- Dipika R. Mohan
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
| | - Kleiton S. Borges
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Isabella Finco
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Christopher R. LaPensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - April L. Solon
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Donald W. Little
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Tobias Else
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Madson Q. Almeida
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Departamento de Clínica Médica, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
- Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | - Derek Dang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, University of Michigan, Ann Arbor, MI, USA
| | - James Haggerty-Skeans
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, University of Michigan, Ann Arbor, MI, USA
| | - April A. Apfelbaum
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA
- Seattle Children’s Research Institute, University of Washington, Seattle, WA, USA
| | - Michelle Vinco
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Alda Wakamatsu
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | - Beatriz M. P. Mariani
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Departamento de Clínica Médica, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | - Larissa Costa Amorim
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Departamento de Clínica Médica, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
- Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | - Ana Claudia Latronico
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Departamento de Clínica Médica, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | - Berenice B. Mendonca
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Departamento de Clínica Médica, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | | | - Elizabeth R. Lawlor
- Seattle Children’s Research Institute, University of Washington, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Ryoma Ohi
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Richard J. Auchus
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - William E. Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Suely K. N. Marie
- Laboratório de Biologia Molecular e Celular/LIM15, Departamento de Neurologia, Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | - Thomas J. Giordano
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center Endocrine Oncology Program, University of Michigan, Ann Arbor, MI, USA
| | - Sriram Venneti
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - Maria Candida Barisson Villares Fragoso
- Unidade de Suprarrenal, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Departamento de Clínica Médica, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
- Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, SP, Brazil
| | - David T. Breault
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Co-senior authors
| | - Gary D. Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center Endocrine Oncology Program, University of Michigan, Ann Arbor, MI, USA
- Co-senior authors
| |
Collapse
|
19
|
Warde KM, Smith LJ, Liu L, Stubben CJ, Lohman BK, Willett PW, Ammer JL, Castaneda-Hernandez G, Imodoye SO, Zhang C, Jones KD, Converso-Baran K, Ekiz HA, Barry M, Clay MR, Kiseljak-Vassiliades K, Giordano TJ, Hammer GD, Basham KJ. Senescence-induced immune remodeling facilitates metastatic adrenal cancer in a sex-dimorphic manner. NATURE AGING 2023; 3:846-865. [PMID: 37231196 PMCID: PMC11534150 DOI: 10.1038/s43587-023-00420-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 04/12/2023] [Indexed: 05/27/2023]
Abstract
Aging markedly increases cancer risk, yet our mechanistic understanding of how aging influences cancer initiation is limited. Here we demonstrate that the loss of ZNRF3, an inhibitor of Wnt signaling that is frequently mutated in adrenocortical carcinoma, leads to the induction of cellular senescence that remodels the tissue microenvironment and ultimately permits metastatic adrenal cancer in old animals. The effects are sexually dimorphic, with males exhibiting earlier senescence activation and a greater innate immune response, driven in part by androgens, resulting in high myeloid cell accumulation and lower incidence of malignancy. Conversely, females present a dampened immune response and increased susceptibility to metastatic cancer. Senescence-recruited myeloid cells become depleted as tumors progress, which is recapitulated in patients in whom a low myeloid signature is associated with worse outcomes. Our study uncovers a role for myeloid cells in restraining adrenal cancer with substantial prognostic value and provides a model for interrogating pleiotropic effects of cellular senescence in cancer.
Collapse
Affiliation(s)
- Kate M Warde
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Lorenzo J Smith
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Lihua Liu
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chris J Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Brian K Lohman
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Parker W Willett
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Julia L Ammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | | | - Sikiru O Imodoye
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chenge Zhang
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kara D Jones
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kimber Converso-Baran
- Frankel Cardiovascular Center Physiology and Phenotyping Core, University of Michigan, Ann Arbor, MI, USA
| | - H Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla Izmir, Turkey
| | - Marc Barry
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Michael R Clay
- Department of Pathology, University of Colorado School of Medicine at Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine at Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas J Giordano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
20
|
Lyraki R, Grabek A, Tison A, Weerasinghe Arachchige LC, Peitzsch M, Bechmann N, Youssef SA, de Bruin A, Bakker ERM, Claessens F, Chaboissier MC, Schedl A. Crosstalk between androgen receptor and WNT/β-catenin signaling causes sex-specific adrenocortical hyperplasia in mice. Dis Model Mech 2023; 16:dmm050053. [PMID: 37102205 PMCID: PMC10184674 DOI: 10.1242/dmm.050053] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/29/2023] [Indexed: 04/28/2023] Open
Abstract
Female bias is highly prevalent in conditions such as adrenal cortex hyperplasia and neoplasia, but the reasons behind this phenomenon are poorly understood. In this study, we show that overexpression of the secreted WNT agonist R-spondin 1 (RSPO1) leads to ectopic activation of WNT/β-catenin signaling and causes sex-specific adrenocortical hyperplasia in mice. Although female adrenals show ectopic proliferation, male adrenals display excessive immune system activation and cortical thinning. Using a combination of genetic manipulations and hormonal treatment, we show that gonadal androgens suppress ectopic proliferation in the adrenal cortex and determine the selective regulation of the WNT-related genes Axin2 and Wnt4. Notably, genetic removal of androgen receptor (AR) from adrenocortical cells restores the mitogenic effect of WNT/β-catenin signaling. This is the first demonstration that AR activity in the adrenal cortex determines susceptibility to canonical WNT signaling-induced hyperplasia.
Collapse
Affiliation(s)
- Rodanthi Lyraki
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| | - Anaëlle Grabek
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| | - Amélie Tison
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| | | | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Sameh A. Youssef
- Dutch Molecular Pathology Center, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL, Utrecht, the Netherlands
- Janssen Research and Development, 2340 Beerse, Belgium
| | - Alain de Bruin
- Dutch Molecular Pathology Center, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL, Utrecht, the Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands
| | - Elvira R. M. Bakker
- Department of Pathology, University Medical Center Utrecht, 3508 AB, Utrecht, the Netherlands
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | | | - Andreas Schedl
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| |
Collapse
|
21
|
Sedlack AJH, Hatfield SJ, Kumar S, Arakawa Y, Roper N, Sun NY, Nilubol N, Kiseljak-Vassiliades K, Hoang CD, Bergsland EK, Hernandez JM, Pommier Y, del Rivero J. Preclinical Models of Adrenocortical Cancer. Cancers (Basel) 2023; 15:2873. [PMID: 37296836 PMCID: PMC10251941 DOI: 10.3390/cancers15112873] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023] Open
Abstract
Adrenocortical cancer is an aggressive endocrine malignancy with an incidence of 0.72 to 1.02 per million people/year, and a very poor prognosis with a five-year survival rate of 22%. As an orphan disease, clinical data are scarce, meaning that drug development and mechanistic research depend especially on preclinical models. While a single human ACC cell line was available for the last three decades, over the last five years, many new in vitro and in vivo preclinical models have been generated. Herein, we review both in vitro (cell lines, spheroids, and organoids) and in vivo (xenograft and genetically engineered mouse) models. Striking leaps have been made in terms of the preclinical models of ACC, and there are now several modern models available publicly and in repositories for research in this area.
Collapse
Affiliation(s)
- Andrew J. H. Sedlack
- Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Samual J. Hatfield
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Suresh Kumar
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yasuhiro Arakawa
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Nitin Roper
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Nai-Yun Sun
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Naris Nilubol
- Surgical Oncology Program National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO 80016, USA
| | - Chuong D. Hoang
- Thoracic Surgery Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Emily K. Bergsland
- University of California, San Francisco (UCSF) Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | | | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jaydira del Rivero
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Ectopic localization of CYP11B1 and CYP11B2-expressing cells in the normal human adrenal gland. PLoS One 2022; 17:e0279682. [PMID: 36584094 PMCID: PMC9803228 DOI: 10.1371/journal.pone.0279682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022] Open
Abstract
The sharp line of demarcation between zona glomerulosa (ZG) and zona fasciculata (ZF) has been recently challenged suggesting that this interface is no longer a compartment boundary. We have used immunohistochemical analyses to study the steroid 11β-hydroxylase (CYP11B1) and aldosterone synthase (CYP11B2) pattern of expression and investigate the remodeling of the adrenal cortex in relation to aging. We analyzed human adrenal glands prepared from 47 kidney donors. No aldosterone-producing micronodules (APMs) were detectable in the younger donors aged between 22-39 but the functional ZG depicted by positive CYP11B2 staining demonstrated a lack of continuity. In contrast, the development of APMs was found in samples from individuals aged 40-70. Importantly, the progressive replacement of CYP11B2-expressing cells in the histological ZG by CYP11B1-expressing cells highlights the remodeling capacity of the adrenal cortex. In 70% of our samples, immunofluorescence studies revealed the presence of isolated or clusters of CYP11B2 positive cells in the ZF and zona reticularis. Our data emphasize that mineralocorticoid- and glucocorticoid-producing cells are distributed throughout the cortex and the medulla making the determination of the functional status of a cell or group of cells a unique tool in deciphering the changes occurring in adrenal gland particularly during aging. They also suggest that, in humans, steroidogenic cell phenotype defined by function is a stable feature and thus, the functional zonation might be not solely maintained by cell lineage conversion/migration.
Collapse
|
23
|
Lerario AM, Mohan DR, Hammer GD. Update on Biology and Genomics of Adrenocortical Carcinomas: Rationale for Emerging Therapies. Endocr Rev 2022; 43:1051-1073. [PMID: 35551369 PMCID: PMC9695111 DOI: 10.1210/endrev/bnac012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Indexed: 11/19/2022]
Abstract
The adrenal glands are paired endocrine organs that produce steroid hormones and catecholamines required for life. Adrenocortical carcinoma (ACC) is a rare and often fatal cancer of the peripheral domain of the gland, the adrenal cortex. Recent research in adrenal development, homeostasis, and disease have refined our understanding of the cellular and molecular programs controlling cortical growth and renewal, uncovering crucial clues into how physiologic programs are hijacked in early and late stages of malignant neoplasia. Alongside these studies, genome-wide approaches to examine adrenocortical tumors have transformed our understanding of ACC biology, and revealed that ACC is composed of distinct molecular subtypes associated with favorable, intermediate, and dismal clinical outcomes. The homogeneous transcriptional and epigenetic programs prevailing in each ACC subtype suggest likely susceptibility to any of a plethora of existing and novel targeted agents, with the caveat that therapeutic response may ultimately be limited by cancer cell plasticity. Despite enormous biomedical research advances in the last decade, the only potentially curative therapy for ACC to date is primary surgical resection, and up to 75% of patients will develop metastatic disease refractory to standard-of-care adjuvant mitotane and cytotoxic chemotherapy. A comprehensive, integrated, and current bench-to-bedside understanding of our field's investigations into adrenocortical physiology and neoplasia is crucial to developing novel clinical tools and approaches to equip the one-in-a-million patient fighting this devastating disease.
Collapse
Affiliation(s)
- Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | - Dipika R Mohan
- Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| |
Collapse
|
24
|
Bridging the Scientific Gaps to Identify Effective Treatments in Adrenocortical Cancer. Cancers (Basel) 2022; 14:cancers14215245. [DOI: 10.3390/cancers14215245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
Adrenocortical cancer (ACC) typically presents in advanced stages of disease and has a dismal prognosis. One of the foremost reasons for this is the lack of available systemic therapies, with mitotane remaining the backbone of treatment since its discovery in the 1960s, despite underwhelming efficacy. Surgery remains the only potentially curative option, but about half of patients will recur post-operatively, often with metastatic disease. Other local treatment options have been attempted but are only used practically on a case-by-case basis. Over the past few decades there have been significant advances in understanding the molecular background of ACC, but this has not yet translated to better treatment options. Attempts at novel treatment strategies have not provided significant clinical benefit. This paper reviews our current treatment options and molecular understanding of ACC and the reasons why a successful treatment has remained elusive. Additionally, we discuss the knowledge gaps that need to be overcome to bring us closer to successful treatment and ways to bridge them.
Collapse
|
25
|
Context dependent role of p53 during the interaction of hepatocellular carcinoma and endothelial cells. Microvasc Res 2022; 142:104374. [PMID: 35523268 DOI: 10.1016/j.mvr.2022.104374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND During the progression of hepatocellular carcinoma (HCC), several angiogenic factors are overexpressed in the hepatic microenvironment, which play a critical role in governing the phenotype of the endothelial cells. Mutation in the p53 gene (TP53) is a common event in HCC that may dysregulate the angiogenic signals. However, their functional messages remain largely unexplored at the onset of metastasis. METHODS Role of p53 was studied by siRNA mediated silencing of p53 in HepG2 cells (WTp53), collecting and analyzing their conditioned medium, followed by indirect co-culture with endothelial cells (HUVECs). Gene and protein expression in HCC cells and endothelial cells was studied by RT-qPCR and western blotting respectively. β-catenin protein expression and localization were analyzed by immunocytochemistry. RESULTS We have studied a cell-to-cell interaction model to investigate the crosstalk of endothelial and hepatoma cells by either knocking down p53 or by using p53 null low metastatic HCC cell line. In the absence of p53, the HCC cells influence the migration and vascular network formation of endothelial cells through paracrine signaling of VEGF. Secretory VEGF activated the VEGF receptor-2 along with the survival signaling in endothelial cells. However, the β-catenin signal is upregulated in endothelial cells only during interaction with metastatic set up irrespective of absence and presence of p53, indicating context-dependent participation of p53 during communication between hepatoma cells and endothelial cells. CONCLUSION This study highlights that the role of p53 on cellular responses during interaction of hepatocellular carcinoma and endothelial cells is distinct to cell types and context.
Collapse
|
26
|
Feng Y, Wang Y, Guo K, Feng J, Shao C, Pan M, Ding P, Liu H, Duan H, Lu D, Wang Z, Zhang Y, Zhang Y, Han J, Li X, Yan X. The value of WNT5A as prognostic and immunological biomarker in pan-cancer. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:466. [PMID: 35571400 PMCID: PMC9096401 DOI: 10.21037/atm-22-1317] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/13/2022] [Indexed: 11/25/2022]
Abstract
Background Finding new immune-related biomarkers is one of the promising research directions for tumor immunotherapy. The WNT5A gene could stimulate the WNT pathway and regulate the progression of various tumors. Recent studies have partially revealed the relationship between WNT5A and tumor immunity, but the correlation and underlying mechanisms in pan-cancer remain obscure. Thus, we conducted this study aiming to characterize the prognostic value and immunological portrait of WNT5A in cancer. Methods The data obtained from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Cancer Cell Line Encyclopedia (CCLE) databases was utilized to analyze WNT5A expression levels by Kruskal-Wallis test and correlation to prognosis by Cox regression test and Kaplan-Meier test, while the data was also used to study the association between WNT5A expression and immune microenvironment, immune neoantigens, immune checkpoints, tumor mutational burden (TMB), and microsatellite instability (MSI) in pan-cancer. Gene set enrichment analysis (GSEA) was used to clarify the relevant signaling pathways. The R package was used for data analysis and to create the plots. Results The pan-cancer analysis revealed that the expression level of WNT5A is generally elevated in most tumors (19/34, 55.88%), and high WNT5A expression was correlated with poor prognosis in esophageal carcinoma (ESCA, P<0.05), low-grade glioma (LGG, P<0.01), adrenocortical carcinoma (ACC, P<0.01), pancreatic adenocarcinoma (PAAD, P<0.01), and head and neck squamous cell carcinoma (HNSC, P<0.05). In addition, WNT5A expression was positively associated with immune infiltration, stromal score, and immune checkpoints in most cancers, and correlated to immune neoantigens, TMB, and MSI. Finally, GSEA indicated that WNT5A is implicated in the transforming growth factor β (TGFβ), Notch, and Hedgehog signaling pathways, which may be related to tumor immunity. Conclusions The expression of WNT5A is elevated in most tumors and associated with tumor prognosis. Furthermore, WNT5A is associated with tumor immunity and may be an immunological biomarker in cancer.
Collapse
Affiliation(s)
- Yingtong Feng
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China.,Department of Cardiothoracic Surgery, The 71st Group Army Hospital of PLA/The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuanyong Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Kai Guo
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Junjun Feng
- Department of Human Resource Management, The 71st Group Army Hospital of PLA/The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, China
| | - Changjian Shao
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Minghong Pan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Peng Ding
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Honggang Liu
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Hongtao Duan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Di Lu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Zhaoyang Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Yimeng Zhang
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Yujing Zhang
- Department of Cardiothoracic Surgery, The 71st Group Army Hospital of PLA/The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| |
Collapse
|
27
|
Xiao Q, Werner J, Venkatachalam N, Boonekamp KE, Ebert MP, Zhan T. Cross-Talk between p53 and Wnt Signaling in Cancer. Biomolecules 2022; 12:453. [PMID: 35327645 PMCID: PMC8946298 DOI: 10.3390/biom12030453] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 11/16/2022] Open
Abstract
Targeting cancer hallmarks is a cardinal strategy to improve antineoplastic treatment. However, cross-talk between signaling pathways and key oncogenic processes frequently convey resistance to targeted therapies. The p53 and Wnt pathway play vital roles for the biology of many tumors, as they are critically involved in cancer onset and progression. Over recent decades, a high level of interaction between the two pathways has been revealed. Here, we provide a comprehensive overview of molecular interactions between the p53 and Wnt pathway discovered in cancer, including complex feedback loops and reciprocal transactivation. The mutational landscape of genes associated with p53 and Wnt signaling is described, including mutual exclusive and co-occurring genetic alterations. Finally, we summarize the functional consequences of this cross-talk for cancer phenotypes, such as invasiveness, metastasis or drug resistance, and discuss potential strategies to pharmacologically target the p53-Wnt interaction.
Collapse
Affiliation(s)
- Qiyun Xiao
- Department of Medicine II, Mannheim University Hospital, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (Q.X.); (N.V.); (M.P.E.)
| | - Johannes Werner
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), and Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg University, D-69120 Heidelberg, Germany; (J.W.); (K.E.B.)
| | - Nachiyappan Venkatachalam
- Department of Medicine II, Mannheim University Hospital, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (Q.X.); (N.V.); (M.P.E.)
| | - Kim E. Boonekamp
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), and Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg University, D-69120 Heidelberg, Germany; (J.W.); (K.E.B.)
| | - Matthias P. Ebert
- Department of Medicine II, Mannheim University Hospital, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (Q.X.); (N.V.); (M.P.E.)
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| | - Tianzuo Zhan
- Department of Medicine II, Mannheim University Hospital, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany; (Q.X.); (N.V.); (M.P.E.)
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| |
Collapse
|
28
|
Nukala SB, Jousma J, Cho Y, Lee WH, Ong SG. Long non-coding RNAs and microRNAs as crucial regulators in cardio-oncology. Cell Biosci 2022; 12:24. [PMID: 35246252 PMCID: PMC8895873 DOI: 10.1186/s13578-022-00757-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/10/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality worldwide. Significant improvements in the modern era of anticancer therapeutic strategies have increased the survival rate of cancer patients. Unfortunately, cancer survivors have an increased risk of cardiovascular diseases, which is believed to result from anticancer therapies. The emergence of cardiovascular diseases among cancer survivors has served as the basis for establishing a novel field termed cardio-oncology. Cardio-oncology primarily focuses on investigating the underlying molecular mechanisms by which anticancer treatments lead to cardiovascular dysfunction and the development of novel cardioprotective strategies to counteract cardiotoxic effects of cancer therapies. Advances in genome biology have revealed that most of the genome is transcribed into non-coding RNAs (ncRNAs), which are recognized as being instrumental in cancer, cardiovascular health, and disease. Emerging studies have demonstrated that alterations of these ncRNAs have pathophysiological roles in multiple diseases in humans. As it relates to cardio-oncology, though, there is limited knowledge of the role of ncRNAs. In the present review, we summarize the up-to-date knowledge regarding the roles of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) in cancer therapy-induced cardiotoxicities. Moreover, we also discuss prospective therapeutic strategies and the translational relevance of these ncRNAs.
Collapse
Affiliation(s)
- Sarath Babu Nukala
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, COMRB 4100, Chicago, IL, 60612, USA
| | - Jordan Jousma
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, COMRB 4100, Chicago, IL, 60612, USA
| | - Yoonje Cho
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, COMRB 4100, Chicago, IL, 60612, USA
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, ABC-1 Building, 425 North 5th Street, Phoenix, AZ, 85004, USA.
| | - Sang-Ging Ong
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, COMRB 4100, Chicago, IL, 60612, USA.
- Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, COMRB 4100, Chicago, IL, 60612, USA.
| |
Collapse
|
29
|
Ruggiero C, Doghman-Bouguerra M, Lalli E. How good are the current models of adrenocortical carcinoma for novel drug discovery? Expert Opin Drug Discov 2022; 17:211-213. [PMID: 34666583 DOI: 10.1080/17460441.2022.1993817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/12/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR, Valbonne, France
- Université Côte d'Azur, Valbonne, France
| | - Mabrouka Doghman-Bouguerra
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR, Valbonne, France
- Université Côte d'Azur, Valbonne, France
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR, Valbonne, France
- Université Côte d'Azur, Valbonne, France
- Inserm, Valbonne, France
| |
Collapse
|
30
|
Sandru F, Petca RC, Carsote M, Petca A, Dumitrascu M, Ghemigian A. Adrenocortical carcinoma: Pediatric aspects (Review). Exp Ther Med 2022; 23:287. [DOI: 10.3892/etm.2022.11216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 11/05/2022] Open
Affiliation(s)
- Florica Sandru
- Department of Dermatology, ‘Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Răzvan-Cosmin Petca
- Department of Urology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mara Carsote
- Department of Endocrinology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Aida Petca
- Department of Obstetrics and Gynecology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mihai Dumitrascu
- Department of Obstetrics and Gynecology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Adina Ghemigian
- Department of Endocrinology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
31
|
Li Y, Lv M, Lu M, Guan H. miR-124a Involves in the Regulation of Wnt/ β-Catenin and P53 Pathways to Inhibit Abdominal Aortic Aneurysm via Targeting BRD4. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9241959. [PMID: 35096137 PMCID: PMC8799344 DOI: 10.1155/2022/9241959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) belongs to a progressive, gradual aortic rupture, which can lead to death without surgical intervention. The key factors regulating the occurrence and progress of AAA are not clear. Increasing studies have indicated that microRNA (miRNA) plays an important role in cancer development. miR-124a serves as a tumor suppressor in several neoplasms, and its upregulation can greatly inhibit the life activities such as malignant growth and migration of tumor cells. AIM The objective of this study is to explore the association of miR-124a with AAA and to uncover the regulated mechanism of miR-124a on AAA progression. METHODS The specimens from the AAA patients were used for observing the miR-124a expression, and human aortic endothelial cells (hAoECs) were treated with AngII to establish the AAA cell models. The quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), CCK-8, transwell assay, flow cytometry assay, and western blot were conducted to unearth the regulation mechanism of miR-124a on AAA, and the dual-luciferase reporter assay was employed to investigate the downstream target of miR-124a. RESULTS miR-124a was significantly downregulated in the whole blood of the patients, and the decreased miR-124a was also observed in AAA cell models. Overexpressing miR-124a could effectively inhibit the proliferation and migration and promote the apoptosis of the AAA cells. The dual-luciferase reporter assay confirmed that BRD4 was a downstream target of miR-124a, and BRD4 upregulation could obviously reverse the effects of miR-124a on the phenotype of AAA cells. Moreover, it was found that miR-124a could regulate the activities of Wnt/β-catenin and P53 pathways via targeting the BRD4. CONCLUSION Our data suggested that miR-124a could regulate the activities of Wnt/β-catenin and P53 to suppress the AAA progression via targeting the BRD4.
Collapse
Affiliation(s)
- Yunhui Li
- Department of Vascular Surgery, Jinan People's Hospital Affiliated to Shandong First Medical University, China
| | - Meifeng Lv
- Pharmacy Department of Jinan Second Maternal and Child Health Hospital, China
| | - Mingshu Lu
- Department of Vascular Surgery, Jinan People's Hospital Affiliated to Shandong First Medical University, China
| | - Hongliang Guan
- Department of Vascular Surgery, Shandong Shanxian Central Hospital, China
| |
Collapse
|
32
|
Ma Z, Cai S, Xiong Q, Liu W, Xia H, Zhu Z, Huang Z, Yan X, Wang Q. WNT signaling modulates chemoresistance to temozolomide in p53-mutant glioblastoma multiforme. Apoptosis 2022; 27:80-89. [PMID: 35037107 DOI: 10.1007/s10495-021-01704-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2021] [Indexed: 11/02/2022]
Abstract
Glioblastoma multiforme (GBM) has been characterized by the high incidence, therapy tolerance and relapse. The molecular events controlling GBM resistant to chemotherapy temozolomide (TMZ) remain to be elusive. Here, we identified WNT signaling was amplified by TMZ and mediated drug response in GBM. We found O6-methylguanine DNA methyltransferase (MGMT) was redundant to WNT-mediated chemoresistance, which was highly associated with p53 mutation status. In GBM with p53 mutation, loss of function of p53 downregulated miR-34a expression, which represses transcription of WNT ligand 6 (WNT6) by directly binding to 3' UTR of WNT6 mRNA, leading to activation of WNT signaling, and the eventual WNT-mediated chemoresistance to TMZ. Combined treatment of TMZ with WNT inhibitor or miR34a mimic induced drug sensitivity of p53-mutant GBM cells and extended survival in xenograft mice in vivo. Our findings provide insight into understanding the molecular mechanism of GBM chemoresistance to TMZ and facilitating to develop novel treatment strategy to combat p53-mutant GBM by targeting miR-34a/WNT6 axis.
Collapse
Affiliation(s)
- Zhourui Ma
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Shizhong Cai
- Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Qianwei Xiong
- Department of Urology, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Wei Liu
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Hongliang Xia
- Department of Urology, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Zhenhong Zhu
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Zhijian Huang
- Department of Burns and Plastic Surgery, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Xiangming Yan
- Department of Urology, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Qian Wang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China.
| |
Collapse
|
33
|
VALD-3 inhibits proliferation and induces apoptosis of colorectal cancer cells via upregulating tumor suppressor activity of p53 to inhibit Wnt/β-catenin signal pathway. Anticancer Drugs 2021; 32:1046-1057. [PMID: 34419958 DOI: 10.1097/cad.0000000000001116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Colorectal cancer is the third most common malignant tumor and a leading cause of cancer death. Currently lacks effective therapies available to improve the prognosis. In the present study, VALD-3, an important Schiff base ligand from o-vanillin derivatives was evaluated for its anti-cancer activity in vitro and in vivo against colorectal cancer. The effect of VALD-3 on colorectal cancer cells proliferation was assessed using MTT assay and the cell migration was evaluated using wound healing scratch assay. The appearance of apoptotic colorectal cancer cells was detected by flowcytometry analysis. Morphological changes caused by VALD-3 induced apoptosis were also observed by Hoechst 33258 staining. The flow cytometry assay was also used to measure cell cycle arrest. The expression levels of TP53 and Bad were analyzed using quantitative real-time PCR. Protein expression of P53, Wnt/β-catenin signaling pathway proteins, apoptosis proteins and cell cycle-related protein were viewed by Western blotting. In addition, HT-29 cells xenograft tumor model was used for the study in vivo. Immunohistochemistry (IHC) staining was employed to detect the P53 protein expression. The results showed that VALD-3 obviously inhibited the proliferation and migration for colorectal cancer cells. In addition, flow cytometry analysis demonstrated that VALD-3 markedly increased early and late apoptosis on colorectal cancer cells, respectively. VALD-3 induced cell cycle arrest at the G0/G1 phase. Most importantly, tumor growth in HT-29 xenograft mice was suppressed by VALD-3, but no significant change in body weight. As confirmed by IHC staining from tumor tissue, the P53 proteins expression increased. These results suggested that VALD-3 represses cell proliferation and induces apoptosis associated with upregulating tumor suppressor activity of p53 to inhibit Wnt/β-catenin signal pathway, and it is a potential anticancer agent for colorectal cancer.
Collapse
|
34
|
Abstract
Resident progenitor and/or stem cell populations in the adult adrenal cortex enable cortical cells to undergo homeostatic renewal and regeneration after injury. Renewal occurs predominantly in the outer layers of the adrenal gland but newly formed cells undergo centripetal migration, differentiation and lineage conversion in the process of forming the different functional steroidogenic zones. Over the past 10 years, advances in the genetic characterization of adrenal diseases and studies of mouse models with altered adrenal phenotypes have helped to elucidate the molecular pathways that regulate adrenal tissue renewal, several of which are fine-tuned via complex paracrine and endocrine influences. Moreover, the adrenal gland is a sexually dimorphic organ, and testicular androgens have inhibitory effects on cell proliferation and progenitor cell recruitment in the adrenal cortex. This Review integrates these advances, including the emerging role of sex hormones, into existing knowledge on adrenocortical cell renewal. An in-depth understanding of these mechanisms is expected to contribute to the development of novel therapies for severe endocrine diseases, for which current treatments are unsatisfactory.
Collapse
Affiliation(s)
- Rodanthi Lyraki
- Université Côte d'Azur, INSERM, CNRS, Institut de Biologie Valrose, Nice, France
| | - Andreas Schedl
- Université Côte d'Azur, INSERM, CNRS, Institut de Biologie Valrose, Nice, France.
| |
Collapse
|
35
|
Maria AG, Silva Borges K, Lira RCP, Hassib Thomé C, Berthon A, Drougat L, Kiseljak-Vassiliades K, Wierman ME, Faucz FR, Faça VM, Tone LG, Stratakis CA. Inhibition of Aurora kinase A activity enhances the antitumor response of beta-catenin blockade in human adrenocortical cancer cells. Mol Cell Endocrinol 2021; 528:111243. [PMID: 33716050 PMCID: PMC8297658 DOI: 10.1016/j.mce.2021.111243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022]
Abstract
Adrenocortical cancer (ACC) is a rare and aggressive type of endocrine tumor with high risk of recurrence and metastasis. The overall survival of patients diagnosed with ACC is low and treatment for metastatic stages remain limited to mitotane, which has low efficiency in advanced stages of the disease and is associated with high toxicity. Therefore, identification of new biological targets to improve ACC treatment is crucial. Blockade of the Wnt/beta-catenin pathway decreased adrenal steroidogenesis and increased apoptosis of NCI-H295 human ACC cells, in vitro and in a xenograft mouse model. Aurora kinases play important roles in cell division during the G1-M phase and their aberrant expression is correlated with a poor prognosis in different types of tumors. Hence, we hypothesized that inhibition of aurora kinases activity combined with the beta-catenin pathway blockade would improve the impairment of ACC cell growth in vitro. We studied the combinatorial effects of AMG 900, an aurora kinase inhibitor and PNU-74654, a beta-catenin pathway blocker, on proliferation, survival and tumor progression in multiple ACC cell lines: NCI-H295, CU-ACC1 and CU-ACC2. Exposure of ACC cells to the combination of AMG 900 with PNU-74654 decreased cell proliferation and viability compared to either treatment alone. In addition, AMG 900 inhibited cell invasion and clonogenesis compared to PNU-74654, and the combination showed no greater effects. In contrast, PNU-74654 was more effective in decreasing cortisol secretion. These data suggest that inhibition of aurora kinases activity combined with blockade of the beta-catenin pathway may provide a combinatorial approach for targeting ACC tumors.
Collapse
Affiliation(s)
- Andrea Gutierrez Maria
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA; Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.
| | - Kleiton Silva Borges
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - R C P Lira
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Carolina Hassib Thomé
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Annabel Berthon
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Ludivine Drougat
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO, 80045, USA; Research Service Veterans Affairs Medical Center, Denver, CO, 80045, USA
| | - Margaret E Wierman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO, 80045, USA; Research Service Veterans Affairs Medical Center, Denver, CO, 80045, USA
| | - Fabio R Faucz
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Luiz Gonzaga Tone
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA; Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD20892, USA
| |
Collapse
|
36
|
The Sexually Dimorphic Adrenal Cortex: Implications for Adrenal Disease. Int J Mol Sci 2021; 22:ijms22094889. [PMID: 34063067 PMCID: PMC8124132 DOI: 10.3390/ijms22094889] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Many adrenocortical diseases are more prevalent in women than in men, but the reasons underlying this sex bias are still unknown. Recent studies involving gonadectomy and sex hormone replacement experiments in mice have shed some light onto the molecular basis of sexual dimorphism in the adrenal cortex. Indeed, it has been shown that gonadal hormones influence many aspects of adrenal physiology, ranging from stem cell-dependent tissue turnover to steroidogenesis and X-zone dynamics. This article reviews current knowledge on adrenal cortex sexual dimorphism and the potential mechanisms underlying sex hormone influence of adrenal homeostasis. Both topics are expected to contribute to personalized and novel therapeutic approaches in the future.
Collapse
|
37
|
Amodru V, Garcia ME, Libe R, Brue T, Reznik Y, Castinetti F. Medical management of adrenocortical carcinoma: Current recommendations, new therapeutic options and future perspectives. ANNALES D'ENDOCRINOLOGIE 2020; 82:52-58. [PMID: 33279475 DOI: 10.1016/j.ando.2020.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022]
Abstract
Adrenocortical carcinoma is a rare malignant tumor of poor prognosis, frequently requiring additional treatments after initial surgery. Due to its adrenolytic action, mitotane has become the first-line medical treatment in patients with aggressive adrenocortical carcinoma. Over the last 2years, apart from the classical chemotherapy based on etoposide and platinum salts, several studies reported the use of drugs such as temozolomide, tyrosine kinase inhibitors or immunotherapy, with more or less convincing results. The aim of this review is to give further insights in the use of these drugs, and to describe potential therapeutic perspectives based on recent pangenomic studies, for the future management of these still difficult to treat tumors.
Collapse
Affiliation(s)
- Vincent Amodru
- Aix-Marseille University, Marseille Medical Genetics, INSERM, Department of endocrinology, La Conception Hospital, Marseille, France
| | - Marie-Eve Garcia
- Aix-Marseille University, Assistance publique-Hôpitaux de Marseille, Multidisciplinary Oncology & Therapeutic Innovations department, Marseille, France
| | - Rossella Libe
- Réseau National "ENDOCAN-COMETE-Cancers de la surrénale", Service d'Endocrinologie, Hôpital Cochin, 27, rue du Faubourg-Saint-Jacques, 75014 Paris, France
| | - Thierry Brue
- Aix-Marseille University, Marseille Medical Genetics, INSERM, Department of endocrinology, La Conception Hospital, Marseille, France
| | - Yves Reznik
- Department of Endocrinology-Diabetology, Caen University Hospital, Caen, France
| | - Frederic Castinetti
- Aix-Marseille University, Marseille Medical Genetics, INSERM, Department of endocrinology, La Conception Hospital, Marseille, France.
| |
Collapse
|