1
|
Monteagudo M, Calsina B, Salazar-Hidalgo ME, Martínez-Montes ÁM, Piñeiro-Yáñez E, Caleiras E, Martín MC, Rodríguez-Perales S, Letón R, Gil E, Buffet A, Burnichon N, Fernández-Sanromán Á, Díaz-Talavera A, Mellid S, Arroba E, Reglero C, Martínez-Puente N, Roncador G, Del Olmo MI, Corrales PJP, Oliveira CL, Álvarez-Escolá C, Gutiérrez MC, López-Fernández A, García NP, Regojo RM, Díaz LR, Laorden NR, Guadarrama OS, Bechmann N, Beuschlein F, Canu L, Eisenhofer G, Fassnacht M, Nölting S, Quinkler M, Rapizzi E, Remde H, Timmers HJ, Favier J, Gimenez-Roqueplo AP, Rodriguez-Antona C, Currás-Freixes M, Al-Shahrour F, Cascón A, Leandro-García LJ, Montero-Conde C, Robledo M. MAML3-fusions modulate vascular and immune tumour microenvironment and confer high metastatic risk in pheochromocytoma and paraganglioma. Best Pract Res Clin Endocrinol Metab 2024:101931. [PMID: 39218714 DOI: 10.1016/j.beem.2024.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pheochromocytomas and paragangliomas are rare neuroendocrine tumours. Around 20-25 % of patients develop metastases, for which there is an urgent need of prognostic markers and therapeutic stratification strategies. The presence of a MAML3-fusion is associated with increased metastatic risk, but neither the processes underlying disease progression, nor targetable vulnerabilities have been addressed. We have compiled a cohort of 850 patients, which has shown a 3.65 % fusion prevalence and represents the largest MAML3-positive series reported to date. While MAML3-fusions mainly cause single pheochromocytomas, we also observed somatic post-zygotic events, resulting in multiple tumours in the same patient. MAML3-tumours show increased expression of neuroendocrine-to-mesenchymal transition markers, MYC-targets, and angiogenesis-related genes, leading to a distinct tumour microenvironment with unique vascular and immune profiles. Importantly, our findings have identified MAML3-tumours specific vulnerabilities beyond Wnt-pathway dysregulation, such as a rich vascular network, and overexpression of PD-L1 and CD40, suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- María Monteagudo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, Madrid, Spain
| | - Bruna Calsina
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Milton E Salazar-Hidalgo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ángel M Martínez-Montes
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elena Piñeiro-Yáñez
- Bioinformatics Unit, Structural Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Core Unit Biotechnology Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Carmen Martín
- Molecular Citogenetic Unit Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sandra Rodríguez-Perales
- Molecular Citogenetic Unit Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rocío Letón
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Gil
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alexandre Buffet
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Nelly Burnichon
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Ángel Fernández-Sanromán
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alberto Díaz-Talavera
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Sara Mellid
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ester Arroba
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Clara Reglero
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Natalia Martínez-Puente
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, Madrid, Spain
| | - Giovanna Roncador
- Monoclonal Antibodies Core Unit Biotechnology Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Isabel Del Olmo
- Department of Endocrinology and Nutrition, University Hospital La Fe, Valencia, Spain
| | | | - Cristina Lamas Oliveira
- Department of Endocrinology and Nutrition Albacete University Hospital, SESCAM, Albacete, Spain
| | | | | | | | | | | | - Luis Robles Díaz
- Department of Oncology, 12 de Octubre University Hospital, Madrid, Spain
| | | | | | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden Germany, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV Klinikum der Universität München, Munich, Germany; Klinik für Endokrinologie Diabetologie und Klinische Ernährung UniversitätsSpital Zürich, Zürich, Switzerland; LOOP Zurich - Medical Research Center, Zurich, Switzerland
| | - Letizia Canu
- Department of Experimental and Clinical Medicine University of Florence, Florence, Italy
| | - Graeme Eisenhofer
- Department of Medicine III University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | - Martin Fassnacht
- Department of Internal Medicine I Division of Endocrinology and Diabetes University Hospital Würzburg University of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken University of Würzburg, Würzburg, Germany
| | - Svenja Nölting
- Klinik für Endokrinologie Diabetologie und Klinische Ernährung UniversitätsSpital Zürich, Zürich, Switzerland
| | - Marcus Quinkler
- Endocrinology in Charlottenburg Stuttgarter Platz 1, Berlin, Germany
| | - Elena Rapizzi
- Department of Experimental and Clinical Medicine University of Florence, Florence, Italy
| | - Hanna Remde
- Comprehensive Cancer Center Mainfranken University of Würzburg, Würzburg, Germany
| | - Henri J Timmers
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Judith Favier
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Cristina Rodriguez-Antona
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Currás-Freixes
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Fatima Al-Shahrour
- Bioinformatics Unit, Structural Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alberto Cascón
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Luis J Leandro-García
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cristina Montero-Conde
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
| |
Collapse
|
2
|
Wang X, Sun J, Feng G, Tian X, Zhao Y, Gao Z, Sun W. Proteomic characterization of head and neck paraganglioma and its molecular classification. Front Mol Neurosci 2024; 17:1391568. [PMID: 39234408 PMCID: PMC11371750 DOI: 10.3389/fnmol.2024.1391568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Background Head and neck paragangliomas (HNPGLs) are rare neuroendocrine tumors that pose significant challenges in both diagnosis and treatment. The pathogenic mechanism remains unclear, and there is no proteomic analysis-based molecular classification. Therefore, gaining a deeper understanding of this disease from the protein level is crucial because proteins play a fundamental role in the occurrence and development of tumors. Methods We collected 44 tumor samples from patients diagnosed with HNPGL. The adrenal paraganglioma tissue (N = 46) was used as the disease control group and the chorda tympani nerves (N = 18) were used as the control group. High-pH reversed-phase liquid chromatography and liquid chromatography with tandem mass spectrometry analyses were used to build an integrated protein database of tumor samples. We then obtained two sets of differentially expressed proteins between the tumor group and the control group to identify the unique proteomic signatures of HNPGLs. Ingenuity pathway analysis annotations were used to perform the functional analysis. Subsequently, we developed a clinically relevant molecular classification for HNPGLs that connected the clinical characteristics with meaningful proteins and pathways to explain the varied clinical manifestations. Results We identified 6,640 proteins in the HNPGL group, and 314 differentially expressed proteins unique to HNPGL were discovered via inter-group comparison. We identified two HNPGL subgroups that significantly differed in clinical manifestation and proteomic characteristics. On the basis of the proteomic results, we proposed a pathogenic mechanism underlying HNPGL. Conclusion We conducted a comprehensive analysis of the molecular mechanisms of HNPGL to build, for the first time, a clinically relevant molecular classification. By focusing on differential proteomic analyses between different types of paragangliomas, we were able to obtain a comprehensive description of the proteomic characteristics of HNPGL, which will be valuable for the search for significant biomarkers as a new treatment method for HNPGL.
Collapse
Affiliation(s)
- Xi Wang
- Department of Otorhinolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiameng Sun
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guodong Feng
- Department of Otorhinolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Tian
- Department of Otorhinolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Zhao
- Department of Otorhinolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiqiang Gao
- Department of Otorhinolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Sun
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Courcelles L, Stoenoiu M, Haufroid V, Lopez-Sublet M, Boland L, Wauthier L, Beauloye C, Maiter D, Januszewicz A, Kreutz R, Persu A, Gruson D. Laboratory Testing for Endocrine Hypertension: Current and Future Perspectives. Clin Chem 2024; 70:709-726. [PMID: 38484135 DOI: 10.1093/clinchem/hvae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/22/2023] [Indexed: 05/03/2024]
Abstract
BACKGROUND Secondary hypertension (SH) is a form of high blood pressure caused by an identifiable underlying condition. Although, it accounts for a small fraction of the overall hypertensive population, detection and management of SH is of utmost importance, because SH phenotypes carry a high cardiovascular risk and can possibly be cured by timely treatment. CONTENT This review focuses on the endocrine causes of SH, such as primary aldosteronism, Cushing syndrome, thyroid disease, pheochromocytoma and paraganglioma, acromegaly, and rare monogenic forms. It discusses current biomarkers, analytical methods, and diagnostic strategies, highlighting advantages and limitations of each approach. It also explores the emerging -omics technologies that can provide a comprehensive and multidimensional assessment of SH and its underlying mechanisms. SUMMARY Endocrine SH is a heterogeneous and complex condition that requires proper screening and confirmatory tests to avoid diagnostic delays and improve patient outcomes. Careful biomarker interpretation is essential due to potential interferences, variability, and method-dependent differences. Liquid chromatography-tandem mass spectrometry is a superior method for measuring low-concentration hormones and metabolites involved in SH, but it requires expertise. Omics approaches have great potential to identify novel biomarkers, pathways, and targets for SH diagnosis and treatment, especially considering its multifactorial nature.
Collapse
Affiliation(s)
- Louisiane Courcelles
- Department of Laboratory Medicine, Cliniques Universitaires St-Luc and Université Catholique de Louvain, Brussels, Belgium
| | - Maria Stoenoiu
- Department of Internal Medicine, Rheumatology, Cliniques Universitaires Saint-Luc, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Vincent Haufroid
- Department of Laboratory Medicine, Cliniques Universitaires St-Luc and Université Catholique de Louvain, Brussels, Belgium
- Louvain centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Marilucy Lopez-Sublet
- AP-HP, Hôpital Avicenne, Centre d'Excellence Européen en Hypertension Artérielle, Service de Médecine Interne, Paris, France
- INSERM UMR 942 MASCOT, Paris 13-Université Paris Nord, Bobigny, France
- FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), CHRU de Nancy - Hôpitaux de Brabois, Vandoeuvre-lès-Nancy, France
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Lidvine Boland
- Department of Laboratory Medicine, Cliniques Universitaires St-Luc and Université Catholique de Louvain, Brussels, Belgium
- Louvain centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Loris Wauthier
- Department of Laboratory Medicine, Cliniques Universitaires St-Luc and Université Catholique de Louvain, Brussels, Belgium
| | - Christophe Beauloye
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Dominique Maiter
- Department of Endocrinology and Nutrition, Cliniques Universitaires St-Luc and Université Catholique de Louvain, Brussels, Belgium
| | - Andrzej Januszewicz
- Department of Hypertension, National Institute of Cardiology, Warsaw, Poland
| | - Reinhold Kreutz
- Charité-Universitätsmedizin Berlin, Institute of Clinical Pharmacology and Toxicology, Charitéplatz 1, 10117 Berlin, Germany
| | - Alexandre Persu
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Damien Gruson
- Department of Laboratory Medicine, Cliniques Universitaires St-Luc and Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
4
|
Gallinson DG, Kozakiewicz CP, Rautsaw RM, Beer MA, Ruiz-Aravena M, Comte S, Hamilton DG, Kerlin DH, McCallum HI, Hamede R, Jones ME, Storfer A, McMinds R, Margres MJ. Intergenomic signatures of coevolution between Tasmanian devils and an infectious cancer. Proc Natl Acad Sci U S A 2024; 121:e2307780121. [PMID: 38466855 PMCID: PMC10962979 DOI: 10.1073/pnas.2307780121] [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: 06/07/2023] [Accepted: 01/17/2024] [Indexed: 03/13/2024] Open
Abstract
Coevolution is common and frequently governs host-pathogen interaction outcomes. Phenotypes underlying these interactions often manifest as the combined products of the genomes of interacting species, yet traditional quantitative trait mapping approaches ignore these intergenomic interactions. Devil facial tumor disease (DFTD), an infectious cancer afflicting Tasmanian devils (Sarcophilus harrisii), has decimated devil populations due to universal host susceptibility and a fatality rate approaching 100%. Here, we used a recently developed joint genome-wide association study (i.e., co-GWAS) approach, 15 y of mark-recapture data, and 960 genomes to identify intergenomic signatures of coevolution between devils and DFTD. Using a traditional GWA approach, we found that both devil and DFTD genomes explained a substantial proportion of variance in how quickly susceptible devils became infected, although genomic architectures differed across devils and DFTD; the devil genome had fewer loci of large effect whereas the DFTD genome had a more polygenic architecture. Using a co-GWA approach, devil-DFTD intergenomic interactions explained ~3× more variation in how quickly susceptible devils became infected than either genome alone, and the top genotype-by-genotype interactions were significantly enriched for cancer genes and signatures of selection. A devil regulatory mutation was associated with differential expression of a candidate cancer gene and showed putative allele matching effects with two DFTD coding sequence variants. Our results highlight the need to account for intergenomic interactions when investigating host-pathogen (co)evolution and emphasize the importance of such interactions when considering devil management strategies.
Collapse
Affiliation(s)
- Dylan G. Gallinson
- Department of Integrative Biology, University of South Florida, Tampa, FL33620
- College of Public Health, University of South Florida, Tampa, FL33620
| | - Christopher P. Kozakiewicz
- School of Biological Sciences, Washington State University, Pullman, WA99163
- W.K. Kellogg Biological Station, Department of Integrative Biology, Michigan State University, Hickory Corners, MI49060
| | - Rhett M. Rautsaw
- Department of Integrative Biology, University of South Florida, Tampa, FL33620
- School of Biological Sciences, Washington State University, Pullman, WA99163
| | - Marc A. Beer
- School of Biological Sciences, Washington State University, Pullman, WA99163
| | - Manuel Ruiz-Aravena
- School of Natural Sciences, University of Tasmania, Hobart, TAS7001, Australia
- Department of Public and Ecosystem Health, Cornell University, Ithaca, NY14853
| | - Sebastien Comte
- School of Natural Sciences, University of Tasmania, Hobart, TAS7001, Australia
- New South Wales Department of Primary Industries, Vertebrate Pest Research Unit, Orange, NSW2800, Australia
| | - David G. Hamilton
- School of Natural Sciences, University of Tasmania, Hobart, TAS7001, Australia
| | - Douglas H. Kerlin
- Centre for Planetary Health and Food Security, Griffith University, Nathan, QLD4111, Australia
| | - Hamish I. McCallum
- Centre for Planetary Health and Food Security, Griffith University, Nathan, QLD4111, Australia
| | - Rodrigo Hamede
- School of Natural Sciences, University of Tasmania, Hobart, TAS7001, Australia
- CANECEV Centre de Recherches Ecologiques et Evolutives sur le Cancer, Montpellier34394, France
| | - Menna E. Jones
- School of Natural Sciences, University of Tasmania, Hobart, TAS7001, Australia
| | - Andrew Storfer
- School of Biological Sciences, Washington State University, Pullman, WA99163
| | - Ryan McMinds
- Department of Integrative Biology, University of South Florida, Tampa, FL33620
- College of Public Health, University of South Florida, Tampa, FL33620
| | - Mark J. Margres
- Department of Integrative Biology, University of South Florida, Tampa, FL33620
| |
Collapse
|
5
|
Lei J, Qu T, Cha L, Tian L, Qiu F, Guo W, Cao J, Sun C, Zhou B. Clinicopathological characteristics of pheochromocytoma/paraganglioma and screening of prognostic markers. J Surg Oncol 2023; 128:510-518. [PMID: 37272486 DOI: 10.1002/jso.27358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/31/2023] [Accepted: 05/03/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Malignant pheochromocytoma/paraganglioma (PCPG) is lethal and difficult to diagnose before metastasis. This study is aiming to characterize the PCPG and explore novel prognostic markers. METHODS Clinical data of patients with pathologically confirmed invasive and noninvasive PCPG were collected and analyzed. Then, the differentially expressed genes (DEGs) and HUB genes were identified by R package "limma" in GSE67066-GPL570. Afterward, the prognostic markers were screened out using R packages of "survival" and "survminer" based on the TCGA data. RESULTS The 34 invasive PCPGs were characterized by irregular contour and unclear boundary on CT and capsule/extracapsule tissue invasion on pathology compared with the 42 noninvasive PCPGs. Then, 29 upregulated and 30 downregulated DEGs were identified in malignant PCPG compared with benign, which were mainly enriched in the terms of calcium ion binding, neuron cell-cell adhesion, axon, regulation of hormone levels, and regulation of secretion by cell. Of which, nine DEGs were furtherly selected as the HUB genes. Finally, CNTN4 and SH3GL2 were found to be highly expressed in malignant PCPGs and negatively correlated with progression-free interval. CONCLUSIONS Malignant PCPGs tend to be aggressive in imaging and pathology. The high expression of CNTN4 and SH3GL2 in PCPGs may indicate a poor prognosis.
Collapse
Affiliation(s)
- Jinghao Lei
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tengfei Qu
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lichao Cha
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lantian Tian
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fabo Qiu
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Weidong Guo
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jingyu Cao
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chuandong Sun
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
6
|
Karakaya S, Gunnesson L, Elias E, Martos-Salvo P, Robledo M, Nilsson O, Wängberg B, Abel F, Påhlman S, Muth A, Mohlin S. Cytoplasmic HIF-2α as tissue biomarker to identify metastatic sympathetic paraganglioma. Sci Rep 2023; 13:11588. [PMID: 37463949 PMCID: PMC10354100 DOI: 10.1038/s41598-023-38606-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/11/2023] [Indexed: 07/20/2023] Open
Abstract
Pheochromocytomas (PCCs) and paragangliomas (PGLs) are rare neuroendocrine tumors. PGLs can further be divided into sympathetic (sPGLs) and head-and-neck (HN-PGLs). There are virtually no treatment options, and no cure, for metastatic PCCs and PGLs (PPGLs). Here, we composed a tissue microarray (TMA) consisting of 149 PPGLs, reflecting clinical features, presenting as a useful resource. Mutations in the pseudohypoxic marker HIF-2α correlate to an aggressive tumor phenotype. We show that HIF-2α localized to the cytoplasm in PPGLs. This subcompartmentalized protein expression differed between tumor subtypes, and strongly correlated to proliferation. Half of all sPGLs were metastatic at time of diagnosis. Cytoplasmic HIF-2α was strongly expressed in metastatic sPGLs and predicted poor outcome in this subgroup. We propose that higher cytoplasmic HIF-2α expression could serve as a useful clinical marker to differentiate paragangliomas from pheochromocytomas, and may help predict outcome in sPGL patients.
Collapse
Affiliation(s)
- Sinan Karakaya
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Sölvegatan 19, BMC B11, 223 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
| | - Lisa Gunnesson
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erik Elias
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Paula Martos-Salvo
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Sölvegatan 19, BMC B11, 223 84, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Lund University Cancer Center, Lund University, Lund, Sweden
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), 28029, Madrid, Spain
| | - Ola Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bo Wängberg
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Frida Abel
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Sven Påhlman
- Lund University Cancer Center, Lund University, Lund, Sweden
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Andreas Muth
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sofie Mohlin
- Division of Pediatrics, Department of Clinical Sciences, Lund University, Sölvegatan 19, BMC B11, 223 84, Lund, Sweden.
- Lund Stem Cell Center, Lund University, Lund, Sweden.
- Lund University Cancer Center, Lund University, Lund, Sweden.
| |
Collapse
|
7
|
Zethoven M, Martelotto L, Pattison A, Bowen B, Balachander S, Flynn A, Rossello FJ, Hogg A, Miller JA, Frysak Z, Grimmond S, Fishbein L, Tischler AS, Gill AJ, Hicks RJ, Dahia PLM, Clifton-Bligh R, Pacak K, Tothill RW. Single-nuclei and bulk-tissue gene-expression analysis of pheochromocytoma and paraganglioma links disease subtypes with tumor microenvironment. Nat Commun 2022; 13:6262. [PMID: 36271074 PMCID: PMC9587261 DOI: 10.1038/s41467-022-34011-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 10/10/2022] [Indexed: 01/13/2023] Open
Abstract
Pheochromocytomas (PC) and paragangliomas (PG) are rare neuroendocrine tumors associated with autonomic nerves. Here we use single-nuclei RNA-seq and bulk-tissue gene-expression data to characterize the cellular composition of PCPG and normal adrenal tissues, refine tumor gene-expression subtypes and make clinical and genotypic associations. We confirm seven PCPG gene-expression subtypes with significant genotype and clinical associations. Tumors with mutations in VHL, SDH-encoding genes (SDHx) or MAML3-fusions are characterized by hypoxia-inducible factor signaling and neoangiogenesis. PCPG have few infiltrating lymphocytes but abundant macrophages. While neoplastic cells transcriptionally resemble mature chromaffin cells, early chromaffin and neuroblast markers are also features of some PCPG subtypes. The gene-expression profile of metastatic SDHx-related PCPG indicates these tumors have elevated cellular proliferation and a lower number of non-neoplastic Schwann-cell-like cells, while GPR139 is a potential theranostic target. Our findings therefore clarify the diverse transcriptional programs and cellular composition of PCPG and identify biomarkers of potential clinical significance.
Collapse
Affiliation(s)
| | - Luciano Martelotto
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Pattison
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Blake Bowen
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Shiva Balachander
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Aidan Flynn
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Fernando J Rossello
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Annette Hogg
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Julie A Miller
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia.,Department of Surgery, Epworth Hospital, Richmond, VIC, Australia
| | - Zdenek Frysak
- 3rd Department of Internal Medicine - Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Sean Grimmond
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Lauren Fishbein
- Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, University of Colorado, Aurora, CO, USA
| | | | - Anthony J Gill
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia.,NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Rodney J Hicks
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Patricia L M Dahia
- Div. Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, USA
| | - Roderick Clifton-Bligh
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Richard W Tothill
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
8
|
Ben Amar D, Thoinet K, Villalard B, Imbaud O, Costechareyre C, Jarrosson L, Reynaud F, Novion Ducassou J, Couté Y, Brunet JF, Combaret V, Corradini N, Delloye-Bourgeois C, Castellani V. Environmental cues from neural crest derivatives act as metastatic triggers in an embryonic neuroblastoma model. Nat Commun 2022; 13:2549. [PMID: 35538114 PMCID: PMC9091272 DOI: 10.1038/s41467-022-30237-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 04/21/2022] [Indexed: 12/04/2022] Open
Abstract
Embryonic malignant transformation is concomitant to organogenesis, often affecting multipotent and migratory progenitors. While lineage relationships between malignant cells and their physiological counterparts are extensively investigated, the contribution of exogenous embryonic signals is not fully known. Neuroblastoma (NB) is a childhood malignancy of the peripheral nervous system arising from the embryonic trunk neural crest (NC) and characterized by heterogeneous and interconvertible tumor cell identities. Here, using experimental models mimicking the embryonic context coupled to proteomic and transcriptomic analyses, we show that signals released by embryonic sympathetic ganglia, including Olfactomedin-1, induce NB cells to shift from a noradrenergic to mesenchymal identity, and to activate a gene program promoting NB metastatic onset and dissemination. From this gene program, we extract a core signature specifically shared by metastatic cancers with NC origin. This reveals non-cell autonomous embryonic contributions regulating the plasticity of NB identities and setting pro-dissemination gene programs common to NC-derived cancers.
Collapse
Affiliation(s)
- Dounia Ben Amar
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | - Karine Thoinet
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | - Benjamin Villalard
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | - Olivier Imbaud
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | | | | | - Florie Reynaud
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | - Julia Novion Ducassou
- University Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, FR2048 38000, Grenoble, France
| | - Yohann Couté
- University Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, FR2048 38000, Grenoble, France
| | - Jean-François Brunet
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France
| | - Valérie Combaret
- Laboratory of Translational Research, Léon Bérard Centre, Lyon, France
| | - Nadège Corradini
- Departments of Oncology and Clinical Research, Centre Léon Berard and Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France
| | - Céline Delloye-Bourgeois
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller.
| | - Valérie Castellani
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller.
| |
Collapse
|
9
|
Cao J, Yang F, Zhou H, Fan D, Li H, Fan T, Sun P. Bone Marrow Mesenchymal Stem Cell (BMSC)-Exosomes Overexpressing miR-141 Inhibit the Malignant Biological Behavior of Glioma Cells via Wnt Signaling. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Our study explores whether BMSC-exosomes overexpressing miR-141 can regulate Wnt signal to inhibit the malignant biological behavior of glioma cells. Thirty healthy mice were selected to construct a glioma mouse model and assigned randomly into the control group, miR-141 NC group, and
miR-141 mimic group followed by analysis of cell proliferation, apoptosis, protein expression and mRNA expression by MTT method, flow cytometry, Western blot and RT-PCR methods. Compared with the other two groups, miR-141 mimic group showed reduced number of cell proliferation at 24 h and
48 h, decreased cell migration and invasion ability, and the increased cell apoptosis rate (P < 0.05). In miR-141 mimic group, the protein expression of miR-141 was the highest, while the protein expression of β-catenin, survivin and c-myc was the lowest (P <
0.05). In conclusion, BMSC-exosomes overexpressing miR-141 can inhibit the malignant biological behavior of GC cells possibly by inhibiting the activation of Wnt signaling pathway.
Collapse
Affiliation(s)
- Jing Cao
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, 071051, Hebei Province, China
| | - Fan Yang
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, 071051, Hebei Province, China
| | - Haiyan Zhou
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, 071051, Hebei Province, China
| | - Duojiao Fan
- Department of Science and Education, Baoding Second Hospital, Baoding City, 071051, Hebei Province, China
| | - Hengzhou Li
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, 071051, Hebei Province, China
| | - Tao Fan
- Department of Neurosurgery, Beijing Sanbo Brain Hospital of Capital Medical University, Beijing, 071051, China
| | - Peng Sun
- Department of Emergency Medicine, Baoding Second Hospital, Baoding City, 071051, Hebei Province, China
| |
Collapse
|
10
|
Wang Y, Chen D, Pang Y, Xu X, Guan X, Liu L. Value of Immunohistochemical Expression of Apelin, Succinate Dehydrogenase B, Chromogranin B, Human Epidermal Growth Factor Receptor-2, Contactin 4, and Succinyl-CoA Synthetase Subunit Beta in Differentiating Metastatic From Non-Metastatic Pheochromocytoma and Paraganglioma. Front Endocrinol (Lausanne) 2022; 13:882906. [PMID: 35574028 PMCID: PMC9096168 DOI: 10.3389/fendo.2022.882906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE We aimed to retrospectively collect pathologically identified pheochromocytoma and paraganglioma (PPGL) tumor tissues from our center and investigate the expression of apelin and succinyl-CoA synthetase subunit beta (SUCLG2), human epidermal growth factor receptor-2 (HER2 or ERBB-2), contactin 4 (CNTN4), chromogranin B (CHGB), and succinate dehydrogenase B (SDHB) in metastatic and non-metastatic PPGLs, for exploring their roles in the diagnosis of metastatic PPGLs. METHODS A total of 369 patients with pathologically and surgically confirmed PPGLs at Xiangya Hospital, Central South University, between June 2010 and June 2020 were retrospectively included. Sixty patients-12 patients with metastatic PPGLs and 48 patients with non-metastatic PPGLs-were selected through propensity score matching (1:4) to reduce the effect of PPGL type, sex, and age. We observed and quantified the expression of apelin, SDHB, CHGB, ERBB-2, CNTN4, and SUCLG2 in paraffin-embedded samples using immunohistochemical staining. RESULTS No significant differences were observed between the metastatic group and non-metastatic group with respect to the expression of CNTN4 and SUCLG2. The expression of apelin, SDHB, CHGB, and ERBB-2 was significantly different between the two groups. The expression of apelin, SDHB, and CHGB was significantly lower in the metastatic group than that in the non-metastatic group (P < 0.001). ERBB-2 expression was significantly higher in the metastatic group than in the non-metastatic group (P = 0.042). Kaplan-Meier analysis revealed that patients with negative expression of apelin, SDHB, and CHGB showed significantly lower metastasis-free survival than those with positive expression. Multivariate Cox analysis revealed that SDHB and CHGB levels were independently associated with metastasis-free survival. CONCLUSION The expression levels of apelin, CHGB, SDHB, and ERBB-2 may be predictive biomarkers for the diagnosis of metastatic PPGLs. Patients with negative expression of apelin, CHGB, and SDHB should be subjected to frequent postoperative follow-up procedures.
Collapse
Affiliation(s)
- Yong Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Danlei Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Yingxian Pang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaowen Xu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Guan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiao Guan, ; Longfei Liu,
| | - Longfei Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiao Guan, ; Longfei Liu,
| |
Collapse
|
11
|
Zhao Y, Gao Y, Xu X, Zhou J, Wang H. Multi-omics analysis of genomics, epigenomics and transcriptomics for molecular subtypes and core genes for lung adenocarcinoma. BMC Cancer 2021; 21:257. [PMID: 33750346 PMCID: PMC7942004 DOI: 10.1186/s12885-021-07888-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most frequently diagnosed histological subtype of lung cancer. Our purpose was to explore molecular subtypes and core genes for LUAD using multi-omics analysis. Methods Methylation, transcriptome, copy number variation (CNV), mutations and clinical feature information concerning LUAD were retrieved from The Cancer Genome Atlas Database (TCGA). Molecular subtypes were conducted via the “iClusterPlus” package in R, followed by Kaplan-Meier survival analysis. Correlation between iCluster subtypes and immune cells was analyzed. Core genes were screened out by integration of methylation, CNV and gene expression, which were externally validated by independent datasets. Results Two iCluster subtypes were conducted for LUAD. Patients in imprinting centre 1 (iC1) subtype had a poorer prognosis than those in iC2 subtype. Furthermore, iC2 subtype had a higher level of B cell infiltration than iC1 subtype. Two core genes including CNTN4 and RFTN1 were screened out, both of which had higher expression levels in iC2 subtype than iC1 subtype. There were distinct differences in CNV and methylation of them between two subtypes. After validation, low expression of CNTN4 and RFTN1 predicted poorer clinical outcomes for LUAD patients. Conclusion Our findings comprehensively analyzed genomics, epigenomics, and transcriptomics of LUAD, offering novel underlying molecular mechanisms for LUAD. Two multi-omics-based core genes (CNTN4 and RFTN1) could become potential therapeutic targets for LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07888-4.
Collapse
Affiliation(s)
- Yue Zhao
- Department II of Radiotherapy, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061110, Hebei, China.
| | - Yakun Gao
- Department of Ultrasound, Cangzhou Central Hospital, Cangzhou, 061110, Hebei, China
| | - Xiaodong Xu
- School of Clinical Medicine, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Jiwu Zhou
- Department II of Radiotherapy, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061110, Hebei, China
| | - He Wang
- Office of Educational Administration, Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, China.
| |
Collapse
|
12
|
Mulford AJ, Wing C, Dolan ME, Wheeler HE. Genetically regulated expression underlies cellular sensitivity to chemotherapy in diverse populations. Hum Mol Genet 2021; 30:305-317. [PMID: 33575800 DOI: 10.1093/hmg/ddab029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 11/14/2022] Open
Abstract
Most cancer chemotherapeutic agents are ineffective in a subset of patients; thus, it is important to consider the role of genetic variation in drug response. Lymphoblastoid cell lines (LCLs) in 1000 Genomes Project populations of diverse ancestries are a useful model for determining how genetic factors impact the variation in cytotoxicity. In our study, LCLs from three 1000 Genomes Project populations of diverse ancestries were previously treated with increasing concentrations of eight chemotherapeutic drugs, and cell growth inhibition was measured at each dose with half-maximal inhibitory concentration (IC50) or area under the dose-response curve (AUC) as our phenotype for each drug. We conducted both genome-wide association studies (GWAS) and transcriptome-wide association studies (TWAS) within and across ancestral populations. We identified four unique loci in GWAS and three genes in TWAS to be significantly associated with the chemotherapy-induced cytotoxicity within and across ancestral populations. In the etoposide TWAS, increased STARD5 predicted expression associated with decreased etoposide IC50 (P = 8.5 × 10-8). Functional studies in A549, a lung cancer cell line, revealed that knockdown of STARD5 expression resulted in the decreased sensitivity to etoposide following exposure for 72 (P = 0.033) and 96 h (P = 0.0001). By identifying loci and genes associated with cytotoxicity across ancestral populations, we strive to understand the genetic factors impacting the effectiveness of chemotherapy drugs and to contribute to the development of future cancer treatment.
Collapse
Affiliation(s)
- Ashley J Mulford
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA.,Program in Bioinformatics, Loyola University Chicago, Chicago, IL 60660, USA
| | - Claudia Wing
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - M Eileen Dolan
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Heather E Wheeler
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA.,Program in Bioinformatics, Loyola University Chicago, Chicago, IL 60660, USA
| |
Collapse
|
13
|
Zhang J, Cong R, Zhang Q, Zeng T, Song R, Meng X. Integrative analysis of ceRNA network and DNA methylation associated with gene expression in malignant pheochromocytomas: a study based on The Cancer Genome Atlas. Transl Androl Urol 2020; 9:344-354. [PMID: 32420140 PMCID: PMC7214974 DOI: 10.21037/tau.2020.01.29] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Competitive endogenous RNAs (ceRNAs) have revealed a new mechanism of interaction between RNAs. Epigenetic regulation in the gene expression dynamics has become increasingly important in malignant pheochromocytomas (PCCs). We performed an integrative analysis of ceRNA networks and DNA methylation to identify key biomarkers and contribute to the understanding of the molecular biological mechanisms of malignant PCCs. Methods Differentially expressed genes in malignant PCCs and controls were identified from The Cancer Genome Atlas database by using the Limma package in R (v3.4.4). An abnormal lncRNA-miRNA-mRNA ceRNA network was constructed for malignant PCCs, and function enrichment analysis was performed using the Database for Annotation, Visualization, and Integrated Discovery. For DNA methylation datasets, the methylation analysis package was used in identifying differential methylation genes, and potential prognostic genes were identified by Kaplan-Meier survival analysis. Results A total of 447 lncRNAs, 26 miRNAs, and 1,607 mRNAs were found to be differentially expressed in malignant PCCs as compared with those in normal samples. We then constructed an abnormal lncRNA-miRNA-mRNA ceRNA network for malignant PCCs. The network consisted of 12 lncRNAs, 6 miRNAs, and 220 mRNAs. Functional enrichment analysis showed that differentially expressed mRNAs were particularly enriched in the biological process, cellular component, and molecular function. Furthermore, four differentially expressed mRNAs from ceRNAs were identified through the cross-analysis of gene expression and DNA methylation profiles. LncRNA C9orf147 and 6 out of 220 mRNAs were indicated as prognostic biomarkers for patients with malignant PCCs (P<0.05). Conclusions Our research increases the understanding of the pathogenesis of malignant PCCs and offers potential genes as underlying therapeutic targets or prognostic biomarkers.
Collapse
Affiliation(s)
- Jiayi Zhang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Rong Cong
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qijie Zhang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Tengyue Zeng
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Rijin Song
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xianghu Meng
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
14
|
Molecular Profiling of Pheochromocytoma and Abdominal Paraganglioma Stratified by the PASS Algorithm Reveals Chromogranin B as Associated With Histologic Prediction of Malignant Behavior. Am J Surg Pathol 2020; 43:409-421. [PMID: 30451732 DOI: 10.1097/pas.0000000000001190] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pheochromocytomas (PCCs) and abdominal paragangliomas (PGLs), collectively abbreviated PPGL, are believed to exhibit malignant potential-but only subsets of cases will display full-blown malignant properties. The Pheochromocytoma of the Adrenal Gland Scaled Score (PASS) algorithm is a proposed histologic system to detect potential for aggressive behavior, but little is known regarding the coupling to underlying molecular genetics. In this study, a total of 92 PPGLs, previously characterized for susceptibility gene status and mRNA expressional profiles, were histologically assessed using the PASS criteria. A total of 32/92 PPGLs (35%) exhibited a PASS score ≥4, including all 8 cases with malignant behavior (7 with known metastases and 1 with extensively infiltrative local recurrence). Statistical analyzes between expressional data and clinical parameters as well as individual PASS criteria yielded significant associations to Chromogranin B (CHGB), BRCA2, HIST1H3B, BUB1B, and RET to name a few, and CHGB had the strongest correlation to both PASS and metastasis/local recurrence of all analyzed genes. Evident CHGB downregulation was observed in PPGLs with high PASS and overtly malignant behavior, and was also associated with shorter disease-related survival. This finding was validated using quantitative real-time polymerase chain reaction, in which CHGB expression correlated with both PASS and metastasis/local recurrence with consistent findings obtained in the TCGA cohort. Moreover, immunohistochemical analyses of subsets of tumors showed a correlation between high PASS scores and negative or weak CHGB protein expression. Patients with PPGLs obtaining high PASS scores postoperatively, also exhibited low preoperative plasma levels of CHGB. These data collectively point out CHGB as a possible preoperative and postoperative marker for PPGLs with potential for aggressive behavior.
Collapse
|
15
|
Lider S, Gheorghiu M. PHEOCHROMOCYTOMA-PARAGANGLIOMA: TRANSLATIONAL APPROACH FROM GENETICS TO CLINICAL NEUROENDOCRINOLOGY. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2019; 14:570-572. [PMID: 31149315 DOI: 10.4183/aeb.2018.570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the latest decade, translational medicine has played an important role. In neuroendocrine tumor field, genetic results are associated with clinical and paraclinical disease implications. This translational knowledge allows a faster action in some mutation types with aggressive outcome. In the following pages we will make a review about actualities in translational approach from genetics to clinical neuroendocrinology.
Collapse
Affiliation(s)
- S Lider
- "C.I. Parhon" National Institute of Endocrinology, Bucharest, Romania.,"C. Davila" University of Medicine and Pharmacy, Dept. of Endocrinology, Bucharest, Romania
| | - M Gheorghiu
- "C.I. Parhon" National Institute of Endocrinology, Bucharest, Romania.,"C. Davila" University of Medicine and Pharmacy, Dept. of Endocrinology, Bucharest, Romania
| |
Collapse
|
16
|
Suh YJ, Park JH, Bilegsaikhan SE, Lee DJ. Transcriptome Analysis Reveals Significant Differences in Gene Expression of Malignant Pheochromocytoma or Paraganglioma. Int J Endocrinol 2019; 2019:7014240. [PMID: 31205467 PMCID: PMC6530119 DOI: 10.1155/2019/7014240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/13/2019] [Accepted: 04/18/2019] [Indexed: 12/18/2022] Open
Abstract
Prediction of malignant behavior of pheochromocytoma (PC) or paraganglioma (PG) is of limited value. The Cancer Genome Atlas (TCGA) and the French 'Cortico et Médullosurrénale: les Tumeurs Endocrines' (COMETE) network in Paris (France) facilitate accurate differentiation of malignant PC/PG based on genetic information. Therefore, the objective of this transcriptome analysis is to identify the prognostic genes underlying the differentiation of malignant PC/PG in the TCGA and COMETE databases. TCGA carries data pertaining to multigenomic analysis of 173 PC/PG surgical resection samples while the COMETE cohort contains data involving 188 PC/PG surgical resection samples. Clinical information and mRNA expression datasets were downloaded from TCGA and COMETE databases. Based on eligibility criteria, 58 of 173 PC/PG samples in TCGA and 171 of 188 PC/PG samples collected by the COMETE network were selected. Using Ingenuity Pathway Analysis, the mRNA expression of malignant and benign PC/PG was compared. The 58 samples in TCGA included 11 malignant and 47 benign cases. Among the 171 samples obtained from the COMETE cohort, 19 were malignant and 152 were benign. A comparative analysis of the mRNA expression data of the two databases revealed that 11 up/downregulated pathways involved in malignant PC/PG were related to cancer signaling, metabolic alteration, and prominent mitosis, whereas 6 upregulated genes and 1 downregulated gene were significantly enriched in the functional annotation pathways. The TCGA and COMETE databases showed differences in mRNA expression associated with malignant and benign PC/PG. Improved recognition of prognostic genes facilitates the diagnosis and treatment of PC/PG.
Collapse
Affiliation(s)
- Yong Joon Suh
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Jung Ho Park
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Sanchir-Erdene Bilegsaikhan
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Dong Jin Lee
- Department of Otolaryngology-Head and Neck Surgery, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| |
Collapse
|
17
|
Oraibi OH, Wharry LI, Lynn AA, Chaudhry F, Jaume JC, Jun JY. LOCALLY INVASIVE PHEOCHROMOCYTOMA COMBINED WITH PRIMARY MALIGNANT ADRENAL LYMPHOMA. AACE Clin Case Rep 2018; 5:e124-e128. [PMID: 31967016 DOI: 10.4158/accr-2018-0221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/18/2018] [Indexed: 01/27/2023] Open
Abstract
Objective Pheochromocytoma (PHEO) combined with primary adrenal lymphoma is extremely rare. We describe a case of locally invasive PHEO combined with primary malignant lymphoma. Methods We provide a case description with biochemical analyses, imaging, and pathologic findings. Results A 79-year-old male presented with a 17-cm, complex mass in the left adrenal gland with non-contrast Hounsfield units of 100 and left para-aortic enlarged lymph nodes imaged by computed tomography. Biochemical evaluation showed plasma and 24-hour urinary normetanephrine significantly elevated about 22 times (about 13.5 times above the normal upper limit) while metanephrine levels were normal. With adequate perioperative preparation, en bloc resection of the left adrenal gland was performed. The pathology revealed a tumor consisting of B-cell lymphoma, a PHEO forming a large adrenal mass with soft tissue invasion with extensive sclerosis and a separate nodule with PHEO without sclerosis. Immunohistochemistry and in situ hybridization of the lymphoma were consistent with Epstein-Barr virus-positive, diffuse large B-cell lymphoma. Immunohistochemistry of the PHEO was positive for chromogranin, synaptophysin, and S100. The Ki67 index was 8.7% and PHEO of the adrenal gland scaled score was 8 (≥4 is considered potentially malignant). Conclusion To the best of our knowledge this is the first case demonstrating locally invasive and potentially malignant PHEO combined with primary malignant lymphoma in the same adrenal gland.
Collapse
|
18
|
Boosting support vector machines for cancer discrimination tasks. Comput Biol Med 2018; 101:236-249. [DOI: 10.1016/j.compbiomed.2018.08.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/31/2018] [Accepted: 08/04/2018] [Indexed: 01/17/2023]
|
19
|
Cho YY, Kwak MK, Lee SE, Ahn SH, Kim H, Suh S, Kim BJ, Song KH, Koh JM, Kim JH, Lee SH. A clinical prediction model to estimate the metastatic potential of pheochromocytoma/paraganglioma: ASES score. Surgery 2018; 164:511-517. [PMID: 29929757 DOI: 10.1016/j.surg.2018.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Malignant pheochromocytoma and paraganglioma can be defined only after the development of metastases in nonchromaffin tissues. There is no single clinical parameter that is sufficiently reliable to predict metastatic potential, so our goal was to develop a prediction model based on multiple clinical parameters. METHODS The baseline age, size, extra-adrenal location, secretory type score was calculated in a retrospective cohort study comprising 333 patients with pheochromocytoma and paraganglioma. In each patient, each variable for age ≤35 years, tumor size ≥ 6.0 cm, extra-adrenal, and norepinephrine-secretory type was coded as 1 point (otherwise 0 point); these points were summed to yield age, size, extra-adrenal location, secretory type score. RESULTS Metastases occurred in 23 of 333 patients (6.9%). Metastatic pheochromocytoma and paraganglioma was associated with age ≤35 years (hazard ratio [HR] 2.74, 95% confidence interval [95% CI] 1.19-6.35), tumor size ≥6.0 cm (HR 2.43, 95% CI 1.06-5.56), extra-adrenal location (HR 2.73, 95% confidence interval 1.10-7.40), and tumor producing only norepinephrine (HR 2.96, 95% CI 1.30-6.76). The area under the curve of the age, size, extra-adrenal location, secretory type score was 0.735. There was a significant difference in metastasis-free survival between participants with age, size, extra-adrenal location, secretory type score ≥2 and score <2 (P < .0001 by the log rank test). The negative predictive value of this system was 96.5% for a cutoff point of 2. CONCLUSION We developed a new prediction model, the age, size, extra-adrenal location, secretory type score, based on multiple clinical parameters to assess the metastatic potential of pheochromocytoma and paraganglioma.
Collapse
Affiliation(s)
- Yoon Young Cho
- Division of Endocrinology and Metabolism, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea; Institute of Health Sciences, Jinju, Korea
| | - Mi Kyung Kwak
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Eun Lee
- Division of Endocrinology and Metabolism, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seong Hee Ahn
- Division of Endocrinology and Metabolism, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Hyeonmok Kim
- Division of Endocrinology and Metabolism, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Sunghwan Suh
- Division of Endocrinology and Metabolism, Dong-A University Medical Center, Dong-A University College of Medicine, Busan, Korea and
| | - Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kee-Ho Song
- Division of Endocrinology and Metabolism, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hyeon Kim
- Division of Endocrinology and Metabolism, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|