1
|
He X, Geng Z, Zou G, Cui Z, Wang Y, Song J, Zhang J, Shao Y, Feng J, Wu Y, Liu T, Zhu X. Alpha-Synuclein Inhibition Promotes Erythropoiesis by Affecting Methylation Modifications of Fructose and Mannose Metabolism. Stem Cells Dev 2025; 34:85-98. [PMID: 39704132 DOI: 10.1089/scd.2024.0160] [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] [Indexed: 12/21/2024] Open
Abstract
Ninety-nine percent of alpha-synuclein (α-syn) in the human body is distributed in erythrocytes. However, the role that α-syn plays in erythropoiesis remains unclear. To determine the effect of α-syn on erythroid differentiation, the erythroid cells, derived from human CD34+ progenitors in the umbilical cord, were cultured in a system composed of a series of cytokines and harvested after 6 days. Our work showed α-syn inhibition-promoted erythropoiesis as characterized by altered activity of surface markers of erythroid development such as CD49d, CD36, and CD71; and different methylation status of GDP-D-mannose dehydratase, aldolase fructose-bisphosphate A, and sorbitol dehydrogenase, key enzymes involved in fructose and mannose metabolism. Reduced adenosine triphosphate and elevated lactic acid also suggested a shift in cellular metabolism from mitochondrial respiration to glycolysis. Our study revealed a previously unknown role for α-syn as a methylation regulator that alters the activity of key enzymes of the fructose and mannose metabolism, thus contributing to erythropoiesis.
Collapse
Affiliation(s)
- Xinrong He
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zixiang Geng
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gang Zou
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zeyu Cui
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Wang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiamin Song
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiye Shao
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingtao Feng
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuncheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoying Zhu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Esteban-Amo MJ, Jiménez-Cuadrado P, Serrano-Lorenzo P, de la Fuente MÁ, Simarro M. Succinate Dehydrogenase and Human Disease: Novel Insights into a Well-Known Enzyme. Biomedicines 2024; 12:2050. [PMID: 39335562 PMCID: PMC11429145 DOI: 10.3390/biomedicines12092050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Succinate dehydrogenase (also known as complex II) plays a dual role in respiration by catalyzing the oxidation of succinate to fumarate in the tricarboxylic acid (TCA) cycle and transferring electrons from succinate to ubiquinone in the mitochondrial electron transport chain (ETC). Owing to the privileged position of SDH/CII, its dysfunction leads to TCA cycle arrest and altered respiration. This review aims to elucidate the widely documented profound metabolic effects of SDH/CII deficiency, along with the newly unveiled survival mechanisms in SDH/CII-deficient cells. Such an understanding reveals exploitable vulnerabilities for strategic targeting, which is crucial for the development of novel and more precise therapies for primary mitochondrial diseases, as well as for familial and sporadic cancers associated with SDH/CII mutations.
Collapse
Affiliation(s)
- María J. Esteban-Amo
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (M.J.E.-A.); (P.J.-C.); (M.Á.d.l.F.)
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Patricia Jiménez-Cuadrado
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (M.J.E.-A.); (P.J.-C.); (M.Á.d.l.F.)
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Pablo Serrano-Lorenzo
- Mitochondrial Disorders Laboratory, Clinical Biochemistry Department, Hospital 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain;
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Á. de la Fuente
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (M.J.E.-A.); (P.J.-C.); (M.Á.d.l.F.)
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - María Simarro
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (M.J.E.-A.); (P.J.-C.); (M.Á.d.l.F.)
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| |
Collapse
|
3
|
Liu C, Zhou D, Yang K, Xu N, Peng J, Zhu Z. Research progress on the pathogenesis of the SDHB mutation and related diseases. Biomed Pharmacother 2023; 167:115500. [PMID: 37734265 DOI: 10.1016/j.biopha.2023.115500] [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: 06/12/2023] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
With the improvement of genetic testing technology in diseases in recent years, researchers have a more detailed and clear understanding of the source of cancers. Succinate dehydrogenase B (SDHB), a mitochondrial gene, is related to the metabolic activities of cells and tissues throughout the body. The mutations of SDHB have been found in pheochromocytoma, paraganglioma and other cancers, and is proved to affect the occurrence and progress of those cancers due to the important structural functions. The importance of SDHB is attracting more and more attention of researchers, however, reviews on the structure and function of SDHB, as well as on the mechanism of its carcinogenesis is inadequate. This paper reviews the relationship between SDHB mutations and related cancers, discusses the molecular mechanism of SDHB mutations that may lead to tumor formation, analyzes the mutation spectrum, structural domains, and penetrance of SDHB and sorts out some of the previously discovered diseases. For the patients with SDHB mutation, it is recommended that people in SDHB mutation families undergo regular genetic testing or SDHB immunohistochemistry (IHC). The purpose of this paper is hopefully to provide some reference and help for follow-up researches on SDHB.
Collapse
Affiliation(s)
- Chang Liu
- Ambulatory Surgical Center, First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, China
| | - Dayang Zhou
- Ambulatory Surgical Center, First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, China
| | - Kexin Yang
- Department of Surgical oncology, Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, 650118, China
| | - Ning Xu
- Ambulatory Surgical Center, First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, China
| | - Jibang Peng
- Department of Surgical oncology, First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, China
| | - Zhu Zhu
- Ambulatory Surgical Center, First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, China.
| |
Collapse
|
4
|
Yuan M, Feng Y, Zhao M, Xu T, Li L, Guo K, Hou D. Identification and verification of genes associated with hypoxia microenvironment in Alzheimer's disease. Sci Rep 2023; 13:16252. [PMID: 37759083 PMCID: PMC10533856 DOI: 10.1038/s41598-023-43595-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/26/2023] [Indexed: 09/29/2023] Open
Abstract
As the incidence of Alzheimer's disease (AD) increases year by year, more people begin to study this disease. In recent years, many studies on reactive oxygen species (ROS), neuroinflammation, autophagy, and other fields have confirmed that hypoxia is closely related to AD. However, no researchers have used bioinformatics methods to study the relationship between AD and hypoxia. Therefore, our study aimed to screen the role of hypoxia-related genes in AD and clarify their diagnostic significance. A total of 7681 differentially expressed genes (DEGs) were identified in GSE33000 by differential expression analysis and cluster analysis. Weighted gene co-expression network analysis (WGCNA) was used to detect 9 modules and 205 hub genes with high correlation coefficients. Next, machine learning algorithms were applied to 205 hub genes and four key genes were selected. Through the verification of external dataset and quantitative real-time PCR (qRT-PCR), the AD diagnostic model was established by ANTXR2, BDNF and NFKBIA. The bioinformatics analysis results suggest that hypoxia-related genes may increase the risk of AD. However, more in-depth studies are still needed to investigate their association, this article would guide the insights and directions for further research.
Collapse
Affiliation(s)
- Mingyang Yuan
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Yanjin Feng
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Mingri Zhao
- School of Life Sciences, Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, 410000, China
| | - Ting Xu
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Liuhong Li
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Ke Guo
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China
| | - Deren Hou
- The Third Xiangya Hospital, Department of Neurology, Central South University, Changsha, 410000, China.
| |
Collapse
|
5
|
Wu KK. Extracellular Succinate: A Physiological Messenger and a Pathological Trigger. Int J Mol Sci 2023; 24:11165. [PMID: 37446354 DOI: 10.3390/ijms241311165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
When tissues are under physiological stresses, such as vigorous exercise and cold exposure, skeletal muscle cells secrete succinate into the extracellular space for adaptation and survival. By contrast, environmental toxins and injurious agents induce cellular secretion of succinate to damage tissues, trigger inflammation, and induce tissue fibrosis. Extracellular succinate induces cellular changes and tissue adaptation or damage by ligating cell surface succinate receptor-1 (SUCNR-1) and activating downstream signaling pathways and transcriptional programs. Since SUCNR-1 mediates not only pathological processes but also physiological functions, targeting it for drug development is hampered by incomplete knowledge about the characteristics of its physiological vs. pathological actions. This review summarizes the current status of extracellular succinate in health and disease and discusses the underlying mechanisms and therapeutic implications.
Collapse
Affiliation(s)
- Kenneth K Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
- Institute of Biotechnology, College of Life Science, National Tsing-Hua University, Hsinchu 30013, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
6
|
S100A6 Protein-Expression and Function in Norm and Pathology. Int J Mol Sci 2023; 24:ijms24021341. [PMID: 36674873 PMCID: PMC9866648 DOI: 10.3390/ijms24021341] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
S100A6, also known as calcyclin, is a calcium-binding protein belonging to the S100 protein family. It was first identified and purified more than 30 years ago. Initial structural studies, focused mostly on the mode and affinity of Ca2+ binding and resolution of the resultant conformational changes, were soon complemented by research on its expression, localization and identification of binding partners. With time, the use of biophysical methods helped to resolve the structure and versatility of S100A6 complexes with some of its ligands. Meanwhile, it became clear that S100A6 expression was altered in various pathological states and correlated with the stage/progression of many diseases, including cancers, indicative of its important, and possibly causative, role in some of these diseases. This, in turn, prompted researchers to look for the mechanism of S100A6 action and to identify the intermediary signaling pathways and effectors. After all these years, our knowledge on various aspects of S100A6 biology is robust but still incomplete. The list of S100A6 ligands is growing all the time, as is our understanding of the physiological importance of these interactions. The present review summarizes available data concerning S100A6 expression/localization, interaction with intracellular and extracellular targets, involvement in Ca2+-dependent cellular processes and association with various pathologies.
Collapse
|
7
|
Martinelli S, Amore F, Canu L, Maggi M, Rapizzi E. Tumour microenvironment in pheochromocytoma and paraganglioma. Front Endocrinol (Lausanne) 2023; 14:1137456. [PMID: 37033265 PMCID: PMC10073672 DOI: 10.3389/fendo.2023.1137456] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Pheochromocytomas and Paragangliomas (Pheo/PGL) are rare catecholamine-producing tumours derived from adrenal medulla or from the extra-adrenal paraganglia respectively. Around 10-15% of Pheo/PGL develop metastatic forms and have a poor prognosis with a 37% of mortality rate at 5 years. These tumours have a strong genetic determinism, and the presence of succinate dehydrogenase B (SDHB) mutations are highly associated with metastatic forms. To date, no effective treatment is present for metastatic forms. In addition to cancer cells, the tumour microenvironment (TME) is also composed of non-neoplastic cells and non-cellular components, which are essential for tumour initiation and progression in multiple cancers, including Pheo/PGL. This review, for the first time, provides an overview of the roles of TME cells such as cancer-associated fibroblasts (CAFs) and tumour-associated macrophages (TAMs) on Pheo/PGL growth and progression. Moreover, the functions of the non-cellular components of the TME, among which the most representatives are growth factors, extracellular vesicles and extracellular matrix (ECM) are explored. The importance of succinate as an oncometabolite is emerging and since Pheo/PGL SDH mutated accumulate high levels of succinate, the role of succinate and of its receptor (SUCNR1) in the modulation of the carcinogenesis process is also analysed. Further understanding of the mechanism behind the complicated effects of TME on Pheo/PGL growth and spread could suggest novel therapeutic targets for further clinical treatments.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
- European Network for the Study of Adrenal Tumours (ENS@T) Center of Excellence, Florence, Italy
| | - Francesca Amore
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
- European Network for the Study of Adrenal Tumours (ENS@T) Center of Excellence, Florence, Italy
| | - Mario Maggi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
- European Network for the Study of Adrenal Tumours (ENS@T) Center of Excellence, Florence, Italy
| | - Elena Rapizzi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, Azienda Ospedaliera Universitaria (AOU) Careggi, Florence, Italy
- European Network for the Study of Adrenal Tumours (ENS@T) Center of Excellence, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- *Correspondence: Elena Rapizzi,
| |
Collapse
|
8
|
Kuo CC, Wu JY, Wu KK. Cancer-derived extracellular succinate: a driver of cancer metastasis. J Biomed Sci 2022; 29:93. [DOI: 10.1186/s12929-022-00878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractSuccinate is a tricarboxylic acid (TCA) cycle intermediate normally confined to the mitochondrial matrix. It is a substrate of succinate dehydrogenase (SDH). Mutation of SDH subunits (SDHD and SDHB) in hereditary tumors such as paraganglioma or reduction of SDHB expression in cancer results in matrix succinate accumulation which is transported to cytoplasma and secreted into the extracellular milieu. Excessive cytosolic succinate is known to stabilize hypoxia inducible factor-1α (HIF-1α) by inhibiting prolyl hydroxylase. Recent reports indicate that cancer-secreted succinate enhances cancer cell migration and promotes cancer metastasis by activating succinate receptor-1 (SUCNR-1)-mediated signaling and transcription pathways. Cancer-derived extracellular succinate enhances cancer cell and macrophage migration through SUCNR-1 → PI-3 K → HIF-1α pathway. Extracellular succinate induces tumor angiogenesis through SUCNR-1-mediated ERK1/2 and STAT3 activation resulting in upregulation of vascular endothelial growth factor (VEGF) expression. Succinate increases SUCNR-1 expression in cancer cells which is considered as a target for developing new anti-metastasis drugs. Furthermore, serum succinate which is elevated in cancer patients may be a theranostic biomarker for selecting patients for SUCNR-1 antagonist therapy.
Collapse
|
9
|
Bayley JP, Devilee P. Hypothesis: Why Different Types of SDH Gene Variants Cause Divergent Tumor Phenotypes. Genes (Basel) 2022; 13:genes13061025. [PMID: 35741787 PMCID: PMC9222429 DOI: 10.3390/genes13061025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022] Open
Abstract
Despite two decades of paraganglioma-pheochromocytoma research, the fundamental question of how the different succinate dehydrogenase (SDH)-related tumor phenotypes are initiated has remained unanswered. Here, we discuss two possible scenarios by which missense (hypomorphic alleles) or truncating (null alleles) SDH gene variants determine clinical phenotype. Dysfunctional SDH is a major source of reactive oxygen species (ROS) but ROS are inhibited by rising succinate levels. In scenario 1, we propose that SDH missense variants disrupt electron flow, causing elevated ROS levels that are toxic in sympathetic PPGL precursor cells but well controlled in oxygen-sensing parasympathetic paraganglion cells. We also suggest that SDHAF2 variants, solely associated with HNPGL, may cause the reversal of succinate dehydrogenase to fumarate reductase, producing very high ROS levels. In scenario 2, we propose a modified succinate threshold model of tumor initiation. Truncating SDH variants cause high succinate accumulation and likely initiate tumorigenesis via disruption of 2-oxoglutarate-dependent enzymes in both PPGL and HNPGL precursor tissues. We propose that missense variants (including SDHAF2) cause lower succinate accumulation and thus initiate tumorigenesis only in very metabolically active tissues such as parasympathetic paraganglia, which naturally show very high levels of succinate.
Collapse
Affiliation(s)
- Jean-Pierre Bayley
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
- Correspondence:
| | - Peter Devilee
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
10
|
Teicher BA, Morris J. Antibody-Drug Conjugate Targets, Drugs and Linkers. Curr Cancer Drug Targets 2022; 22:463-529. [PMID: 35209819 DOI: 10.2174/1568009622666220224110538] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/22/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
Antibody-drug conjugates offer the possibility of directing powerful cytotoxic agents to a malignant tumor while sparing normal tissue. The challenge is to select an antibody target expressed exclusively or at highly elevated levels on the surface of tumor cells and either not all or at low levels on normal cells. The current review explores 78 targets that have been explored as antibody-drug conjugate targets. Some of these targets have been abandoned, 9 or more are the targets of FDA-approved drugs, and most remain active clinical interest. Antibody-drug conjugates require potent cytotoxic drug payloads, several of these small molecules are discussed, as are the linkers between the protein component and small molecule components of the conjugates. Finally, conclusions regarding the elements for the successful antibody-drug conjugate are discussed.
Collapse
Affiliation(s)
- Beverly A Teicher
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| | - Joel Morris
- Developmental Therapeutics Program, DCTD, National Cancer Institute, Bethesda, MD 20892,United States
| |
Collapse
|
11
|
Tabebi M, Kumar Dutta R, Skoglund C, Söderkvist P, Gimm O. Loss of SDHB Induces a Metabolic Switch in the hPheo1 Cell Line toward Enhanced OXPHOS. Int J Mol Sci 2022; 23:560. [PMID: 35008989 PMCID: PMC8745660 DOI: 10.3390/ijms23010560] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Enzymes of tricarboxylic acid (TCA) have recently been recognized as tumor suppressors. Mutations in the SDHB subunit of succinate dehydrogenase (SDH) cause pheochromocytomas and paragangliomas (PCCs/PGLs) and predispose patients to malignant disease with poor prognosis. METHODS Using the human pheochromocytoma cell line (hPheo1), we knocked down SDHB gene expression using CRISPR-cas9 technology. RESULTS Microarray gene expression analysis showed that >500 differentially expressed gene targets, about 54%, were upregulated in response to SDHB knock down. Notably, genes involved in glycolysis, hypoxia, cell proliferation, and cell differentiation were up regulated, whereas genes involved in oxidative phosphorylation (OXPHOS) were downregulated. In vitro studies show that hPheo1 proliferation is not affected negatively and the cells that survive by shifting their metabolism to the use of glutamine as an alternative energy source and promote OXPHOS activity. Knock down of SDHB expression results in a significant increase in GLUD1 expression in hPheo1 cells cultured as monolayer or as 3D culture. Analysis of TCGA data confirms the enhancement of GLUD1 in SDHB mutated/low expressed PCCs/PGLs. CONCLUSIONS Our data suggest that the downregulation of SDHB in PCCs/PGLs results in increased GLUD1 expression and may represent a potential biomarker and therapeutic target in SDHB mutated tumors and SDHB loss of activity-dependent diseases.
Collapse
Affiliation(s)
- Mouna Tabebi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Ravi Kumar Dutta
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Camilla Skoglund
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Peter Söderkvist
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
- Clinical Genomics Linköping, Science for Life Laboratory, Linköping University, 581 83 Linköping, Sweden
| | - Oliver Gimm
- Department of Surgery and Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden;
| |
Collapse
|
12
|
Takács-Vellai K, Farkas Z, Ősz F, Stewart GW. Model systems in SDHx-related pheochromocytoma/paraganglioma. Cancer Metastasis Rev 2021; 40:1177-1201. [PMID: 34957538 PMCID: PMC8825606 DOI: 10.1007/s10555-021-10009-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/04/2021] [Indexed: 11/17/2022]
Abstract
Pheochromocytoma (PHEO) and paraganglioma (PGL) (together PPGL) are tumors with poor outcomes that arise from neuroendocrine cells in the adrenal gland, and sympathetic and parasympathetic ganglia outside the adrenal gland, respectively. Many follow germline mutations in genes coding for subunits of succinate dehydrogenase (SDH), a tetrameric enzyme in the tricarboxylic acid (TCA) cycle that both converts succinate to fumarate and participates in electron transport. Germline SDH subunit B (SDHB) mutations have a high metastatic potential. Herein, we review the spectrum of model organisms that have contributed hugely to our understanding of SDH dysfunction. In Saccharomyces cerevisiae (yeast), succinate accumulation inhibits alpha-ketoglutarate-dependent dioxygenase enzymes leading to DNA demethylation. In the worm Caenorhabditis elegans, mutated SDH creates developmental abnormalities, metabolic rewiring, an energy deficit and oxygen hypersensitivity (the latter is also found in Drosophila melanogaster). In the zebrafish Danio rerio, sdhb mutants display a shorter lifespan with defective energy metabolism. Recently, SDHB-deficient pheochromocytoma has been cultivated in xenografts and has generated cell lines, which can be traced back to a heterozygous SDHB-deficient rat. We propose that a combination of such models can be efficiently and effectively used in both pathophysiological studies and drug-screening projects in order to find novel strategies in PPGL treatment.
Collapse
Affiliation(s)
| | - Zsolt Farkas
- Department of Biological Anthropology, Eötvös Loránd University, Budapest, Hungary
| | - Fanni Ősz
- Department of Biological Anthropology, Eötvös Loránd University, Budapest, Hungary
| | - Gordon W Stewart
- Division of Medicine, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
13
|
Dona M, Lamers M, Rohde S, Gorissen M, Timmers HJLM. Targeting the Redox Balance Pathway Using Ascorbic Acid in sdhb Zebrafish Mutant Larvae. Cancers (Basel) 2021; 13:cancers13205124. [PMID: 34680273 PMCID: PMC8534273 DOI: 10.3390/cancers13205124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Thus far, no curative therapies are available for malignant SDHB-associated phaeochromocytomas and paragangliomas (PPGLs). Therapy development is severely hampered by the limited availability of suitable animal models. In this study, we investigated the potential of the sdhbrmc200 zebrafish model to study SDHB-associated PPGLs using a drug screening approach. One of the key features of cancer initiation and progression is redox imbalance. First, we identified increased reactive oxygen species levels in homozygous sdhbrmc200 larvae at baseline. Next, we tested the effect of anti- and pro-oxidant ascorbic acid (Vitamin C) on these larvae. We validated the sdhbrmc200 zebrafish model as a powerful drug screening tool to provide valuable insights into pathomechanisms, which may lead to novel therapeutic targets and therapy development in the future. Abstract Patients with mutations in the β-subunit of the succinate dehydrogenase (SDHB) have the highest risk to develop incurable malignant phaeochromocytomas and paragangliomas (PPGLs). Therapy development is hindered by limited possibilities to test new therapeutic strategies in vivo. One possible molecular mechanism of SDHB-associated tumorigenesis originates in an overproduction of reactive oxygen species (ROS) due to mitochondrial dysfunction. Ascorbic acid (Vitamin C) has already been shown to act as anti-cancer agent in several clinical trials for various types of cancer. In this study, the potential of the sdhbrmc200 zebrafish model to study SDHB-associated PPGLs using a drug screening approach was investigated. First, we identified increased basal ROS levels in homozygous sdhb larvae compared to heterozygous and wild-type siblings. Using a semi high-throughput drug screening, the effectiveness of different dosages of anti- and pro-oxidant Vitamin C were assessed to evaluate differences in survival, ROS levels, and locomotor activity. Low-dosage levels of Vitamin C induced a decrease of ROS levels but no significant effects on lifespan. In contrast, high-dosage levels of Vitamin C shortened the lifespan of the homozygous sdhbrmc200 larvae while not affecting the lifespan of heterozygous and wild-type siblings. These results validated the sdhbrmc200 zebrafish model as a powerful drug screening tool that may be used to identify novel therapeutic targets for SDHB-associated PPGLs.
Collapse
Affiliation(s)
- Margo Dona
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (M.L.); (S.R.); (H.J.L.M.T.)
- Correspondence:
| | - Maaike Lamers
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (M.L.); (S.R.); (H.J.L.M.T.)
| | - Svenja Rohde
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (M.L.); (S.R.); (H.J.L.M.T.)
| | - Marnix Gorissen
- Department of Animal Ecology and Physiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, 6525 AJ Nijmegen, The Netherlands;
| | - Henri J. L. M. Timmers
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (M.L.); (S.R.); (H.J.L.M.T.)
| |
Collapse
|
14
|
Yebra M, Bhargava S, Kumar A, Burgoyne AM, Tang CM, Yoon H, Banerjee S, Aguilera J, Cordes T, Sheth V, Noh S, Ustoy R, Li S, Advani SJ, Corless CL, Heinrich MC, Kurzrock R, Lippman SM, Fanta PT, Harismendy O, Metallo C, Sicklick JK. Establishment of Patient-Derived Succinate Dehydrogenase-Deficient Gastrointestinal Stromal Tumor Models for Predicting Therapeutic Response. Clin Cancer Res 2021; 28:187-200. [PMID: 34426440 DOI: 10.1158/1078-0432.ccr-21-2092] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract, with mutant succinate dehydrogenase (SDH) subunits (A-D) comprising less than 7.5% (i.e., 150-200/year) of new cases annually in the United States. Contrary to GISTs harboring KIT or PDGFRA mutations, SDH-mutant GISTs affect adolescents/young adults, often metastasize, and are frequently resistant to tyrosine kinase inhibitors (TKI). Lack of human models for any SDH-mutant tumors, including GIST, has limited molecular characterization and drug discovery. EXPERIMENTAL DESIGN We describe methods for establishing novel patient-derived SDH-mutant (mSDH) GIST models and interrogated the efficacy of temozolomide on these tumor models in vitro and in clinical trials of patients with mSDH GIST. RESULTS Molecular and metabolic characterization of our patient-derived mSDH GIST models revealed that these models recapitulate the transcriptional and metabolic hallmarks of parent tumors and SDH deficiency. We further demonstrate that temozolomide elicits DNA damage and apoptosis in our mSDH GIST models. Translating our in vitro discovery to the clinic, a cohort of patients with SDH-mutant GIST treated with temozolomide (n = 5) demonstrated a 40% objective response rate and 100% disease control rate, suggesting that temozolomide represents a promising therapy for this subset of GIST. CONCLUSIONS We report the first methods to establish patient-derived mSDH tumor models, which can be readily employed for understanding patient-specific tumor biology and treatment strategies. We also demonstrate that temozolomide is effective in patients with mSDH GIST who are refractory to existing chemotherapeutic drugs (namely, TKIs) in clinic for GISTs, bringing a promising treatment option for these patients to clinic.
Collapse
Affiliation(s)
- Mayra Yebra
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Shruti Bhargava
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Avi Kumar
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Adam M Burgoyne
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Hematology Oncology, University of California San Diego, San Diego, California
| | - Chih-Min Tang
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Hyunho Yoon
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Sudeep Banerjee
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Joseph Aguilera
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California
| | - Thekla Cordes
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Vipul Sheth
- Department of Radiology, Stanford University, Palo Alto, Stanford, California
| | - Sangkyu Noh
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Rowan Ustoy
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Sam Li
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California
| | - Sunil J Advani
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, California
| | | | - Michael C Heinrich
- Hematology/Medical Oncology, Portland VA Health Care System and OHSU Knight Cancer Institute, Portland, Oregon
| | - Razelle Kurzrock
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Hematology Oncology, University of California San Diego, San Diego, California.,Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, San Diego, California
| | - Scott M Lippman
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Hematology Oncology, University of California San Diego, San Diego, California.,Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, San Diego, California
| | - Paul T Fanta
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Hematology Oncology, University of California San Diego, San Diego, California.,Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, San Diego, California
| | - Olivier Harismendy
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Medicine, Division of Biomedical Informatics, University of California San Diego, San Diego, California
| | - Christian Metallo
- Moores Cancer Center, University of California San Diego, La Jolla, California.,Department of Bioengineering, University of California San Diego, La Jolla, California.,Diabetes and Endocrinology Research Center, University of California San Diego, La Jolla, California.,Institute of Engineering in Medicine, University of California San Diego, La Jolla, California
| | - Jason K Sicklick
- Moores Cancer Center, University of California San Diego, La Jolla, California. .,Department of Surgery, Division of Surgical Oncology, University of California San Diego, San Diego, California.,Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, San Diego, California
| |
Collapse
|
15
|
Wang Z, Chen H, Xue L, He W, Shu W, Wu H, Wang Z. High throughput proteomic and metabolic profiling identified target correction of metabolic abnormalities as a novel therapeutic approach in head and neck paraganglioma. Transl Oncol 2021; 14:101146. [PMID: 34118692 PMCID: PMC8193622 DOI: 10.1016/j.tranon.2021.101146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/22/2021] [Accepted: 06/01/2021] [Indexed: 01/02/2023] Open
Abstract
Head and neck paragangliomas (HNPGLs) are rare neoplasms that represent difficult treatment paradigms in neurotology. Germline mutations in genes encoding succinate dehydrogenase (SDH) are the cause of nearly all familial HNPGLs. However, the molecular mechanisms underlying tumorigenesis remain unclear. Mutational analysis identified 6 out of 14 HNPGLs harboring clinicopathologic SDH gene mutations. The SDHB gene was most frequently mutated in these patients, and western blot showed loss of SDHB protein in tumors with SDHB mutations. The paraganglioma cell line (PGL-626) was established from a sample that harbored a missense SDHB mutation (c.649C > T). Spectrometric analysis using tandem mass tags identified 151 proteins significantly differentially expressed in HNPGLs compared with normal nerves. Bioinformatics analyses confirmed the high level of enrichment of oxidative phosphorylation and metabolism pathways in HNPGLs. The mitochondrial complex subunits NDUFA2, NDUFA10, and NDUFA4, showed the most significantly increased expression and were localized predominantly in the cytoplasm of PGL-626 cells. The mitochondrial complex I inhibitor metformin exerted dose-dependent inhibitory effects on PGL-626 cells via cooperative down-regulation of NDUFA2, 4, and 10, with a significant decrease in the levels of reactive oxygen species and mitochondrial membrane potential. Further metabolomic analysis of PGL-626 cells showed that metabolites involved in central carbon metabolism in cancer and sphingolipid signaling pathways, pantothenate and CoA biosynthesis, and tryptophan and carbon metabolism were significantly altered after metformin treatment. Thus, this study provides insights into the molecular mechanisms underlying HNPGL tumorigenesis and identifies target correction of metabolic abnormalities as a novel therapeutic approach for this disease.
Collapse
Affiliation(s)
- Zhigang Wang
- Department of Otolaryngology Head & Neck Surgery, The Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639, Zhi-Zao-Ju Road, Shanghai 200011, China; Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Hongsai Chen
- Department of Otolaryngology Head & Neck Surgery, The Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639, Zhi-Zao-Ju Road, Shanghai 200011, China; Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Lu Xue
- Department of Otolaryngology Head & Neck Surgery, The Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639, Zhi-Zao-Ju Road, Shanghai 200011, China; Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Weiwei He
- Department of Otolaryngology Head & Neck Surgery, The Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639, Zhi-Zao-Ju Road, Shanghai 200011, China; Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Wenying Shu
- Department of Otolaryngology Head & Neck Surgery, The Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639, Zhi-Zao-Ju Road, Shanghai 200011, China; Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Hao Wu
- Department of Otolaryngology Head & Neck Surgery, The Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639, Zhi-Zao-Ju Road, Shanghai 200011, China; Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.
| | - Zhaoyan Wang
- Department of Otolaryngology Head & Neck Surgery, The Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639, Zhi-Zao-Ju Road, Shanghai 200011, China; Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.
| |
Collapse
|
16
|
Saxena N, Beraldi E, Fazli L, Somasekharan SP, Adomat H, Zhang F, Molokwu C, Gleave A, Nappi L, Nguyen K, Brar P, Nikesitch N, Wang Y, Collins C, Sorensen PH, Gleave M. Androgen receptor (AR) antagonism triggers acute succinate-mediated adaptive responses to reactivate AR signaling. EMBO Mol Med 2021; 13:e13427. [PMID: 33709547 PMCID: PMC8103094 DOI: 10.15252/emmm.202013427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 11/09/2022] Open
Abstract
Treatment-induced adaptive pathways converge to support androgen receptor (AR) reactivation and emergence of castration-resistant prostate cancer (PCa) after AR pathway inhibition (ARPI). We set out to explore poorly defined acute adaptive responses that orchestrate shifts in energy metabolism after ARPI and identified rapid changes in succinate dehydrogenase (SDH), a TCA cycle enzyme with well-known tumor suppressor activity. We show that AR directly regulates transcription of its catalytic subunits (SDHA, SDHB) via androgen response elements (AREs). ARPI acutely suppresses SDH activity, leading to accumulation of the oncometabolite, succinate. Succinate triggers calcium ions release from intracellular stores, which in turn phospho-activates the AR-cochaperone, Hsp27 via p-CaMKK2/p-AMPK/p-p38 axis to enhance AR protein stabilization and activity. Activation of this pathway was seen in tissue microarray analysis on prostatectomy tissues and patient-derived xenografts. This adaptive response is blocked by co-targeting AR with Hsp27 under both in vitro and in vivo studies, sensitizing PCa cells to ARPI treatments.
Collapse
Affiliation(s)
- Neetu Saxena
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Hans Adomat
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Fan Zhang
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Anna Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lucia Nappi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | - Pavn Brar
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Colin Collins
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Poul H Sorensen
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Matlac DM, Hadrava Vanova K, Bechmann N, Richter S, Folberth J, Ghayee HK, Ge GB, Abunimer L, Wesley R, Aherrahrou R, Dona M, Martínez-Montes ÁM, Calsina B, Merino MJ, Schwaninger M, Deen PMT, Zhuang Z, Neuzil J, Pacak K, Lehnert H, Fliedner SMJ. Succinate Mediates Tumorigenic Effects via Succinate Receptor 1: Potential for New Targeted Treatment Strategies in Succinate Dehydrogenase Deficient Paragangliomas. Front Endocrinol (Lausanne) 2021; 12:589451. [PMID: 33776908 PMCID: PMC7994772 DOI: 10.3389/fendo.2021.589451] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Paragangliomas and pheochromocytomas (PPGLs) are chromaffin tumors associated with severe catecholamine-induced morbidities. Surgical removal is often curative. However, complete resection may not be an option for patients with succinate dehydrogenase subunit A-D (SDHx) mutations. SDHx mutations are associated with a high risk for multiple recurrent, and metastatic PPGLs. Treatment options in these cases are limited and prognosis is dismal once metastases are present. Identification of new therapeutic targets and candidate drugs is thus urgently needed. Previously, we showed elevated expression of succinate receptor 1 (SUCNR1) in SDHB PPGLs and SDHD head and neck paragangliomas. Its ligand succinate has been reported to accumulate due to SDHx mutations. We thus hypothesize that autocrine stimulation of SUCNR1 plays a role in the pathogenesis of SDHx mutation-derived PPGLs. We confirmed elevated SUCNR1 expression in SDHx PPGLs and after SDHB knockout in progenitor cells derived from a human pheochromocytoma (hPheo1). Succinate significantly increased viability of SUCNR1-transfected PC12 and ERK pathway signaling compared to control cells. Candidate SUCNR1 inhibitors successfully reversed proliferative effects of succinate. Our data reveal an unrecognized oncometabolic function of succinate in SDHx PPGLs, providing a growth advantage via SUCNR1.
Collapse
Affiliation(s)
- Dieter M. Matlac
- Neuroendocrine Oncology and Metabolism, Medical Department I, Center of Brain, Behavior, and Metabolism, University Medical Center Schleswig-Holstein Lübeck, Lübeck, Germany
| | - Katerina Hadrava Vanova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Susan Richter
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Julica Folberth
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Hans K. Ghayee
- Department of Medicine, Division of Endocrinology, University of Florida and Malcom Randall VA Medical Center, Gainesville, FL, United States
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Luma Abunimer
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | | | - Redouane Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- Department of Biomedical Engineering, Centre for Public Health Genomics, University of Virginia, Charlottesville, VA, United States
| | - Margo Dona
- Division of Endocrinology 471, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ángel M. Martínez-Montes
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria J. Merino
- Laboratory of Surgical Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | | | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Hendrik Lehnert
- Neuroendocrine Oncology and Metabolism, Medical Department I, Center of Brain, Behavior, and Metabolism, University Medical Center Schleswig-Holstein Lübeck, Lübeck, Germany
| | - Stephanie M. J. Fliedner
- Neuroendocrine Oncology and Metabolism, Medical Department I, Center of Brain, Behavior, and Metabolism, University Medical Center Schleswig-Holstein Lübeck, Lübeck, Germany
- *Correspondence: Stephanie M. J. Fliedner,
| |
Collapse
|
18
|
Activation of RAS Signalling is Associated with Altered Cell Adhesion in Phaeochromocytoma. Int J Mol Sci 2020; 21:ijms21218072. [PMID: 33138083 PMCID: PMC7663737 DOI: 10.3390/ijms21218072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Phaeochromocytomas and paragangliomas (PPGLs) are neuroendocrine catecholamine-producing tumours that may progress into inoperable metastatic disease. Treatment options for metastatic disease are limited, indicating a need for functional studies to identify pharmacologically targetable pathophysiological mechanisms, which require biologically relevant experimental models. Recently, a human progenitor phaeochromocytoma cell line named “hPheo1” was established, but its genotype has not been characterised. Performing exome sequencing analysis, we identified a KIF1B T827I mutation, and the oncogenic NRAS Q61K mutation. While KIF1B mutations are recurring somatic events in PPGLs, NRAS mutations have hitherto not been detected in PPGLs. Therefore, we aimed to assess its implications for the hPheo1 cell line, and possible relevance for the pathophysiology of PPGLs. We found that transient downregulation of NRAS in hPheo1 led to elevated expression of genes associated with cell adhesion, and enhanced adhesion to hPheo1 cells’ extracellular matrix. Analyses of previously published mRNA data from two independent PPGL patient cohorts (212 tissue samples) revealed a subcluster of PPGLs featuring hyperactivated RAS pathway-signalling and under-expression of cell adhesion-related gene expression programs. Thus, we conclude that NRAS activity in hPheo1 decreases adhesion to their own extracellular matrix and mirrors a transcriptomic RAS-signalling-related phenomenon in PPGLs.
Collapse
|
19
|
Valcárcel LV, Torrano V, Tobalina L, Carracedo A, Planes FJ. rMTA: robust metabolic transformation analysis. Bioinformatics 2020; 35:4350-4355. [PMID: 30923806 DOI: 10.1093/bioinformatics/btz231] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/15/2019] [Accepted: 03/27/2019] [Indexed: 12/30/2022] Open
Abstract
MOTIVATION The development of computational tools exploiting -omics data and high-quality genome-scale metabolic networks for the identification of novel drug targets is a relevant topic in Systems Medicine. Metabolic Transformation Algorithm (MTA) is one of these tools, which aims to identify targets that transform a disease metabolic state back into a healthy state, with potential application in any disease where a clear metabolic alteration is observed. RESULTS Here, we present a robust extension to MTA (rMTA), which additionally incorporates a worst-case scenario analysis and minimization of metabolic adjustment to evaluate the beneficial effect of gene knockouts. We show that rMTA complements MTA in the different datasets analyzed (gene knockout perturbations in different organisms, Alzheimer's disease and prostate cancer), bringing a more accurate tool for predicting therapeutic targets. AVAILABILITY AND IMPLEMENTATION rMTA is freely available on The Cobra Toolbox: https://opencobra.github.io/cobratoolbox/latest/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Luis V Valcárcel
- Tecnun, University of Navarra, San Sebastián 20018, Spain
- Area de Hemato-Oncología, IDISNA, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Faculty of Medicine, Joint Research Centre for Computational Biomedicine, RWTH Aachen University, Aachen D-52074, Germany
| | - Verónica Torrano
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Faculty of Medicine, Joint Research Centre for Computational Biomedicine, RWTH Aachen University, Aachen D-52074, Germany
- CIC bioGUNE, Bizkaia Technology Park, Derio, Spain
| | - Luis Tobalina
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine, RWTH Aachen University, Aachen D-52074, Germany
| | - Arkaitz Carracedo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Faculty of Medicine, Joint Research Centre for Computational Biomedicine, RWTH Aachen University, Aachen D-52074, Germany
- CIC bioGUNE, Bizkaia Technology Park, Derio, Spain
- Ikerbasque, Basque foundation for science, Bilbao, Spain
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao E-48080, Spain
| | | |
Collapse
|
20
|
Abstract
The study of cancer metabolism has evolved vastly beyond the remit of tumour proliferation and survival with the identification of the role of 'oncometabolites' in tumorigenesis. Simply defined, oncometabolites are conventional metabolites that, when aberrantly accumulated, have pro-oncogenic functions. Their discovery has led researchers to revisit the Warburg hypothesis, first postulated in the 1950s, of aberrant metabolism as an aetiological determinant of cancer. As such, the identification of oncometabolites and their utilization in diagnostics and prognostics, as novel therapeutic targets and as biomarkers of disease, are areas of considerable interest in oncology. To date, fumarate, succinate, L-2-hydroxyglutarate (L-2-HG) and D-2-hydroxyglutarate (D-2-HG) have been characterized as bona fide oncometabolites. Extensive metabolic reprogramming occurs during tumour initiation and progression in renal cell carcinoma (RCC) and three oncometabolites - fumarate, succinate and L-2-HG - have been implicated in this disease process. All of these oncometabolites inhibit a superfamily of enzymes known as α-ketoglutarate-dependent dioxygenases, leading to epigenetic dysregulation and induction of pseudohypoxic phenotypes, and also have specific pro-oncogenic capabilities. Oncometabolites could potentially be exploited for the development of novel targeted therapies and as biomarkers of disease.
Collapse
Affiliation(s)
- Cissy Yong
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
21
|
Moosavi B, Zhu XL, Yang WC, Yang GF. Molecular pathogenesis of tumorigenesis caused by succinate dehydrogenase defect. Eur J Cell Biol 2020; 99:151057. [DOI: 10.1016/j.ejcb.2019.151057] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 09/19/2019] [Accepted: 10/29/2019] [Indexed: 12/14/2022] Open
|
22
|
Pepe S, Korbonits M, Iacovazzo D. Germline and mosaic mutations causing pituitary tumours: genetic and molecular aspects. J Endocrinol 2019; 240:R21-R45. [PMID: 30530903 DOI: 10.1530/joe-18-0446] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 12/24/2022]
Abstract
While 95% of pituitary adenomas arise sporadically without a known inheritable predisposing mutation, in about 5% of the cases they can arise in a familial setting, either isolated (familial isolated pituitary adenoma or FIPA) or as part of a syndrome. FIPA is caused, in 15-30% of all kindreds, by inactivating mutations in the AIP gene, encoding a co-chaperone with a vast array of interacting partners and causing most commonly growth hormone excess. While the mechanisms linking AIP with pituitary tumorigenesis have not been fully understood, they are likely to involve several pathways, including the cAMP-dependent protein kinase A pathway via defective G inhibitory protein signalling or altered interaction with phosphodiesterases. The cAMP pathway is also affected by other conditions predisposing to pituitary tumours, including X-linked acrogigantism caused by duplications of the GPR101 gene, encoding an orphan G stimulatory protein-coupled receptor. Activating mosaic mutations in the GNAS gene, coding for the Gα stimulatory protein, cause McCune-Albright syndrome, while inactivating mutations in the regulatory type 1α subunit of protein kinase A represent the most frequent genetic cause of Carney complex, a syndromic condition with multi-organ manifestations also involving the pituitary gland. In this review, we discuss the genetic and molecular aspects of isolated and syndromic familial pituitary adenomas due to germline or mosaic mutations, including those secondary to AIP and GPR101 mutations, multiple endocrine neoplasia type 1 and 4, Carney complex, McCune-Albright syndrome, DICER1 syndrome and mutations in the SDHx genes underlying the association of familial paragangliomas and phaeochromocytomas with pituitary adenomas.
Collapse
Affiliation(s)
- Sara Pepe
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Donato Iacovazzo
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
23
|
Primary fibroblast co-culture stimulates growth and metabolism in Sdhb-impaired mouse pheochromocytoma MTT cells. Cell Tissue Res 2018; 374:473-485. [PMID: 30159755 DOI: 10.1007/s00441-018-2907-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/07/2018] [Indexed: 12/28/2022]
Abstract
Pheochromocytomas and paragangliomas (PGLs) due to mutations of succinate dehydrogenase (SDH) B, a subunit of the SDH complex with a role in the Krebs cycle and the respiratory chain, tend to be larger at diagnosis and more prone to metastatic disease than other tumors. This presentation contrasts with the behavior of some cell line models of SDHB impairment, which show reduced growth compared to wild type. We hypothesize that reduced growth of SDHB-impaired monolayer culture models might reflect lack of support from sources within the tumor microenvironment. The present study therefore investigates how the microenvironment, modeled here by fibroblast co-culture, modulates cell metabolism, growth and invasion in an Sdhb-impaired mouse pheochromocytoma cell line. We employed two different constructs of short hairpin RNA to knockdown Sdhb and compared growth in a monolayer with and without fibroblast co-culture. Sdhb-silenced cells showed functional impairment of SDH with elevated succinate to fumarate ratio and decreased oxidative capacity. Cell growth was delayed with an increase in doubling time of 2 h or 20 h. Clonogenic cell survival and viability, on the other hand, were either unchanged or increased compared to control. In standard monolayer culture, no differences in pro-metastatic features were present. Co-culture with primary mouse fibroblast reversed the difference of proliferation between control and Sdhb knockdown but was unable to significantly influence invasiveness under these culture conditions. Metabolic studies identified that lactate secreted by fibroblasts was taken up preferentially by Sdhb-silenced cells. In summary, the present study identified a potential role for the tumor microenvironment in influencing phenotypic features of SDHB-mutated PGLs, providing a basis for the use of therapies targeted towards the tumor microenvironment.
Collapse
|
24
|
Yamanaka M, Shiga K, Fujiwara S, Mizuguchi Y, Yasuda S, Ishizawa K, Saiki Y, Higashi K, Ogawa T, Kimura N, Horii A. A Novel SDHB IVS2-2A>C Mutation Is Responsible for Hereditary Pheochromocytoma/Paraganglioma Syndrome. TOHOKU J EXP MED 2018; 245:99-105. [PMID: 29925701 DOI: 10.1620/tjem.245.99] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pheochromocytomas and paragangliomas are neuroendocrine tumors which arise from adrenal medulla, and sympathetic or parasympathetic nerves, respectively. Hereditary cases afflicted by both or either pheochromocytomas and paragangliomas have been reported: these are called hereditary pheochromocytoma/paraganglioma syndromes (HPPS). Many cases of HPPS are caused by mutations of one of the succinate dehydrogenase (SDH) genes; mainly SDHB and SDHD that encode subunits for the mitochondrial respiratory chain complex II. In this study, we investigated mutations of SDH genes in six HPPS patients from four Japanese pedigrees using peripheral blood lymphocytes (from one patient with pheochromocytoma and five patients with neck paraganglioma) and tumor tissues (from two patients with paraganglioma). Results showed that all of these pedigrees harbor germline mutations in one of the SDH genes. In two pedigrees, a novel IVS2-2A>C mutation in SDHB, at the acceptor-site in intron 2, was found, and the tumor RNA of the patient clearly showed frameshift caused by exon skipping. Each of the remaining two pedigrees harbors a reported missense mutation, R242H in SDHB or G106D in SDHD. Importantly, all these mutations are heterozygous in constitutional DNAs, and two-hit mutations were evident in tumor DNAs. We thus conclude that the newly identified IVS2-2A>C mutation in SDHB is responsible for HPPS. The novel mutation revealed by our study may contribute to improvement of clinical management for patients with HPPS.
Collapse
Affiliation(s)
- Mie Yamanaka
- Department of Molecular Pathology, Tohoku University School of Medicine.,Exploring-Germination-and-Growth Program for Young Scientists, Tohoku University
| | - Kiyoto Shiga
- Department of Otolaryngology and Head and Neck Surgery, Iwate Medical University School of Medicine
| | - Sho Fujiwara
- Department of Molecular Pathology, Tohoku University School of Medicine
| | | | - Sari Yasuda
- Department of Molecular Pathology, Tohoku University School of Medicine.,Exploring-Germination-and-Growth Program for Young Scientists, Tohoku University
| | - Kota Ishizawa
- Department of Molecular Pathology, Tohoku University School of Medicine
| | - Yuriko Saiki
- Department of Molecular Pathology, Tohoku University School of Medicine
| | - Kenjiro Higashi
- Department of Molecular Pathology, Tohoku University School of Medicine.,Department of Otolaryngology-Head and Neck Surgery, Tohoku University School of Medicine
| | - Takenori Ogawa
- Department of Molecular Pathology, Tohoku University School of Medicine.,Department of Otolaryngology-Head and Neck Surgery, Tohoku University School of Medicine
| | - Noriko Kimura
- Department of Clinical Research, Pathology Division, National Hospital Organization Hakodate National Hospital
| | - Akira Horii
- Department of Molecular Pathology, Tohoku University School of Medicine
| |
Collapse
|
25
|
Kluckova K, Tennant DA. Metabolic implications of hypoxia and pseudohypoxia in pheochromocytoma and paraganglioma. Cell Tissue Res 2018; 372:367-378. [PMID: 29450727 PMCID: PMC5915505 DOI: 10.1007/s00441-018-2801-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/17/2018] [Indexed: 12/13/2022]
Abstract
Hypoxia is a critical driver of cancer pathogenesis, directly inducing malignant phenotypes such as epithelial-mesenchymal transition, stem cell-like characteristics and metabolic transformation. However, hypoxia-associated phenotypes are often observed in cancer in the absence of hypoxia, a phenotype known as pseudohypoxia, which is very well documented in specific tumour types, including in paraganglioma/pheochromocytoma (PPGL). Approximately 40% of the PPGL tumours carry a germ line mutation in one of a number of susceptibility genes of which those that are found in succinate dehydrogenase (SDH) or in von Hippel-Lindau (VHL) genes manifest a strong pseudohypoxic phenotype. Mutations in SDH are oncogenic, forming tumours in a select subset of tissues, but the cause for this remains elusive. Although elevated succinate levels lead to increase in hypoxia-like signalling, there are other phenotypes that are being increasingly recognised in SDH-mutated PPGL, such as DNA hypermethylation. Further, recently unveiled changes in metabolic re-wiring of SDH-deficient cells might help to decipher cancer related roles of SDH in the future. In this review, we will discuss the various implications that the malfunctioning SDH can have and its impact on cancer development.
Collapse
Affiliation(s)
- Katarina Kluckova
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
26
|
Pang Y, Lu Y, Caisova V, Liu Y, Bullova P, Huynh TT, Zhou Y, Yu D, Frysak Z, Hartmann I, Taïeb D, Pacak K, Yang C. Targeting NAD +/PARP DNA Repair Pathway as a Novel Therapeutic Approach to SDHB-Mutated Cluster I Pheochromocytoma and Paraganglioma. Clin Cancer Res 2018; 24:3423-3432. [PMID: 29636359 DOI: 10.1158/1078-0432.ccr-17-3406] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/23/2018] [Accepted: 04/03/2018] [Indexed: 01/18/2023]
Abstract
Purpose: Cluster I pheochromocytomas and paragangliomas (PCPGs) tend to develop malignant transformation, tumor recurrence, and multiplicity. Transcriptomic profiling suggests that cluster I PCPGs and other related tumors exhibit distinctive changes in the tricarboxylic acid (TCA) cycle, the hypoxia signaling pathway, mitochondrial electron transport chain, and methylation status, suggesting that therapeutic regimen might be optimized by targeting these signature molecular pathways.Experimental Design: In the present study, we investigated the molecular signatures in clinical specimens from cluster I PCPGs in comparison with cluster II PCPGs that are related to kinase signaling and often present as benign tumors.Results: We found that cluster I PCPGs develop a dependency to mitochondrial complex I, evidenced by the upregulation of complex I components and enhanced NADH dehydrogenation. Alteration in mitochondrial function resulted in strengthened NAD+ metabolism, here considered as a key mechanism of chemoresistance, particularly, of succinate dehydrogenase subunit B (SDHB)-mutated cluster I PCPGs via the PARP1/BER DNA repair pathway. Combining a PARP inhibitor with temozolomide, a conventional chemotherapeutic agent, not only improved cytotoxicity but also reduced metastatic lesions, with prolonged overall survival of mice with SDHB knockdown PCPG allograft.Conclusions: In summary, our findings provide novel insights into an effective strategy for targeting cluster I PCPGs, especially those with SDHB mutations. Clin Cancer Res; 24(14); 3423-32. ©2018 AACR.
Collapse
Affiliation(s)
- Ying Pang
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Yanxin Lu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.,Basic Medical Science Department, Zunyi Medical College-Zhuhai Campus, Zhuhai, Guangdong, P.R. China
| | - Veronika Caisova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.,Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske 19 Budejovice, Czech Republic
| | - Yang Liu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Petra Bullova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.,Department of Molecular Medicine, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Thanh-Truc Huynh
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Yiqiang Zhou
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Di Yu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.,CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, P.R. China
| | - Zdenek Frysak
- 3rd Department of Internal Medicine, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Igor Hartmann
- Department of Urology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, Centre Européen de Rechercheen Imagerie Médicale, Aix-Marseille University, Marseille, France
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
27
|
Tseng PL, Wu WH, Hu TH, Chen CW, Cheng HC, Li CF, Tsai WH, Tsai HJ, Hsieh MC, Chuang JH, Chang WT. Decreased succinate dehydrogenase B in human hepatocellular carcinoma accelerates tumor malignancy by inducing the Warburg effect. Sci Rep 2018; 8:3081. [PMID: 29449614 PMCID: PMC5814459 DOI: 10.1038/s41598-018-21361-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/02/2018] [Indexed: 12/17/2022] Open
Abstract
Changes in TCA cycle enzymes or respiratory activity are possible mechanisms of aerobic glycolysis that contributes to tumor progression. To clarify whether the decrease of succinate dehydrogenase B (SDHB) alters energy metabolism, induces the Warburg effect and results in tumor malignancy, SDHB expression was examined and modulated in hepatocellular carcinoma (HCC) tissues and cells, respectively. SDHB level was often decreased in malignant HCC cells and tissues. Furthermore, the reduced SDHB expression was associated with advanced tumor stage and poor survival rate. Moreover, silencing of SDHB altered energy metabolism switched from aerobic respiration to glycolysis, resulted in the Warburg effect, and enhanced cell proliferation and motility. In contrast, the SDHB overexpression deregulated bioenergetic metabolism and decreased cell growth and migration. In mouse xenograft models, subcutaneous implantation and tail vein injection with SDHB knockdown cells resulted in a larger tumor volume and accelerated cancer metastasis, respectively. A mutation or decrease in SDHB induced the switch from aerobic respiration to glycolysis. This metabolic alteration was associated with tumor cell dedifferentiation, proliferation, motility and overall patient survival in HCC.
Collapse
Affiliation(s)
- Po-Lin Tseng
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, 302, Taiwan
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, 833, Taiwan
| | - Wei-Hsuan Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Tsung-Hui Hu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, 833, Taiwan
| | - Chih-Wei Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Surgery, Chi Mei Foundation Medical Center, Tainan, 710, Taiwan
- Department of Occupational Safety and Health/Institute of Industrial Safety and Disaster Prevention, College of Sustainable Environment, Chia Nan University of Pharmacy and Science, Tainan, 717, Taiwan
| | - Hung-Chi Cheng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Chien-Feng Li
- Department of Pathology, Chi Mei Foundation Medical Center, Tainan, 710, Taiwan
| | - Wen-Hui Tsai
- Department of Pediatrics, Chi Mei Foundation Medical Center, Tainan, 710, Taiwan
| | - Hui-Ju Tsai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Meng-Che Hsieh
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Jiin-Haur Chuang
- Department of Pediatric Surgery, Kaohsiung Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung, 833, Taiwan
| | - Wen-Tsan Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
28
|
Xiao Z, Liu S, Ai F, Chen X, Li X, Liu R, Ren W, Zhang X, Shu P, Zhang D. SDHB downregulation facilitates the proliferation and invasion of colorectal cancer through AMPK functions excluding those involved in the modulation of aerobic glycolysis. Exp Ther Med 2017; 15:864-872. [PMID: 29399091 PMCID: PMC5772827 DOI: 10.3892/etm.2017.5482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/19/2017] [Indexed: 02/01/2023] Open
Abstract
Loss-of-function of succinate dehydrogenase-B (SDHB) is a predisposing factor of aerobic glycolysis and cancer progression. Adenosine monophosphate activated protein kinase (AMPK) is involved in the regulation of aerobic glycolysis and the diverse hallmarks of cancer. The present study investigated whether AMPK mediated the regulatory effects of SDHB in aerobic glycolysis and cancer growth. The expression of SDHB and AMPK in colorectal cancer (CRC) and normal tissues was assessed by western blotting. HT-29 CRC cells were used to establish in vitro models of ectopic overexpression and knockdown of SDHB. SDHB was downregulated, while AMPK and phosphorylated-AMPK (Thr172) were upregulated in CRC tissues. Experiments involving the loss- or gain-of-function of SDHB, revealed that this protein negatively regulated AMPK by influencing its expression and activity. However, SDHB and AMPK were identified to suppress lactic acid production in CRC cells, indicating that each had an inhibitory effect on aerobic glycolysis. Therefore, the regulation of aerobic glycolysis by SDHB is unlikely to be mediated via AMPK. SDHB knockdown promoted the viability, migration and invasion of HT-29 cells, whereas inhibition of AMPK demonstrated the opposite effect. SDHB overexpression impaired cell migration and invasion, and this effect was reversed following AMPK activation. These results indicate that AMPK may mediate the effects of SDHB in CRC cell proliferation and migration. In conclusion, SDHB downregulation in CRC cells may increase AMPK activity, which may subsequently facilitate the proliferation and invasion of these cancer cells. However, the regulation of aerobic glycolysis by SDHB may be independent of AMPK. Further studies are warranted to elucidate the mechanism by which SDHB regulates aerobic glycolysis.
Collapse
Affiliation(s)
- Zhiming Xiao
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shaojun Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Feiyan Ai
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, Hunan 410013, P.R. China
| | - Xiong Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Xiayu Li
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, Hunan 410013, P.R. China
| | - Rui Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Weiguo Ren
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, Hunan 410013, P.R. China
| | - Xuemei Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, Hunan 410013, P.R. China
| | - Peng Shu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, Hunan 410013, P.R. China
| | - Decai Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
29
|
Taïeb D, Pacak K. New Insights into the Nuclear Imaging Phenotypes of Cluster 1 Pheochromocytoma and Paraganglioma. Trends Endocrinol Metab 2017; 28:807-817. [PMID: 28867159 PMCID: PMC5673583 DOI: 10.1016/j.tem.2017.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 08/05/2017] [Accepted: 08/07/2017] [Indexed: 12/11/2022]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) belong to the family of neural crest cell-derived neoplasms. In up to 70% of cases they are associated with germline and somatic mutations in 15 well-characterized PPGL driver or fusion genes. PPGLs can be grouped into three main clusters, where cluster 1 includes PPGLs characterized by a pseudohypoxic signature. Although cluster 1 tumors share several common features, they exhibit unique behaviors. We present here unique insights into the imaging phenotypes of cluster 1 PPGLs based on glucose uptake, catecholamine metabolism, and somatostatin receptor expression. Recent data suggest that succinate is a major player in the imaging phenotype of succinate dehydrogenase-deficient PPGLs. This review emphasizes the emerging stromal cell-succinate interaction and highlights new perspectives in PPGL theranostics.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, European Center for Research in Medical Imaging (CERIMED), Aix-Marseille University, Marseille, France.
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
30
|
Liu S, Xiao Z, Ai F, Liu F, Chen X, Cao K, Ren W, Zhang X, Shu P, Zhang D. miR-142-5p promotes development of colorectal cancer through targeting SDHB and facilitating generation of aerobic glycolysis. Biomed Pharmacother 2017. [PMID: 28622713 DOI: 10.1016/j.biopha.2017.05.134] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Aberrant expression of miRNAs contributes to the development of human malignancies. A recent study revealed that miR-142-5p is increased in the serum of colorectal cancer (CRC) patients compared to health people. Using starBase v2.0, we found that succinate dehydrogenase-B (SDHB) is a potential target of miR-142-5p, while SDHB is negatively correlated to cancer development through regulating energetic metabolism. Based on these information, this study further examined the expression profiles of miR-142-5p and SDHB in CRC tissues and cell lines using PCR and Western blotting. Transfection experiment and luciferase assay were performed to identify relationship between miR-142-5p and SDHB. Oxygen intake, glucose consumption and production of lactic acid were used to evaluate the influence on energetic metabolism. CRC growth and proliferation were assessed by in vitro and in vivo studies. Results showed that miR-142-5p was up-regulated in CRC, but SDHB was down-regulated. SDHB was confirmed as a target of miR-142-5p, and decreased SDHB in CRC was result from the abnormal up-regulation of miR-142-5p. Lose of SDHB by miR-142-5p inhibited oxygen intake by CRC cells, but increased glucose consumption and lactate production. These suggest miR-142-5p up-regulation in CRC probably facilitates generation of aerobic glycolysis by reducing SDHB. miR-142-5p promoted proliferation and colony formation of CRC, but inhibited apoptosis. SDHB overexpression abrogated these effect of miR-142-5p, which indicates that SDHB depletion mediates tumor-promoting actions of miR-142-5p. This study added novel insight into the CRC development regulated by miR-142-5p. It may be a promising therapy target in the future molecular therapy.
Collapse
Affiliation(s)
- Shaojun Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China
| | - Zhiming Xiao
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China
| | - Feiyan Ai
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410013 Hunan, China
| | - Fen Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410013 Hunan, China
| | - Xiong Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China
| | - Ke Cao
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Weiguo Ren
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410013 Hunan, China
| | - Xuemei Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410013 Hunan, China
| | - Peng Shu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410013 Hunan, China
| | - Decai Zhang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha 410013 Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410013 Hunan, China.
| |
Collapse
|
31
|
Sharma S, Wang J, Cortes Gomez E, Taggart RT, Baysal BE. Mitochondrial complex II regulates a distinct oxygen sensing mechanism in monocytes. Hum Mol Genet 2017; 26:1328-1339. [PMID: 28204537 DOI: 10.1093/hmg/ddx041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/25/2017] [Indexed: 01/28/2023] Open
Abstract
Mutations in mitochondrial complex II (succinate dehydrogenase; SDH) genes predispose to paraganglioma tumors that show constitutive activation of hypoxia responses. We recently showed that SDHB mRNAs in hypoxic monocytes gain a stop codon mutation by APOBEC3A-mediated C-to-U RNA editing. Here, we test the hypothesis that inhibition of complex II facilitates hypoxic gene expression in monocytes using an integrative experimental approach. By RNA sequencing, we show that specific inhibition of complex II by atpenin A5 in normoxic conditions mimics hypoxia and induces hypoxic transcripts as well as APOBEC3A-mediated RNA editing in human monocytes. Myxothiazol, a complex III inhibitor, has similar effects in normoxic monocytes. Atpenin A5 partially inhibits oxygen consumption, and neither hypoxia nor atpenin A5 in normoxia robustly stabilizes hypoxia-inducible factor (HIF)-1α in primary monocytes. Several earlier studies in transformed cell lines suggested that normoxic stabilization of HIF-1α explains the persistent expression of hypoxic genes upon complex II inactivation. On the contrary, we find that atpenin A5 antagonizes the stabilization of HIF-1α and reduces hypoxic gene expression in transformed cell lines. Accordingly, compound germline heterozygosity of mouse Sdhb/Sdhc/Sdhd null alleles blunts chronic hypoxia-induced increases in hemoglobin levels, an adaptive response mainly regulated by HIF-2α. In contrast, atpenin A5 or myxothiazol does not reduce hypoxia-induced gene expression or RNA editing in monocytes. These results reveal a novel role for mitochondrial respiratory inhibition in induction of the hypoxic transcriptome in monocytes and suggest that inhibition of complex II activates a distinct hypoxia signaling pathway in a cell-type specific manner.
Collapse
Affiliation(s)
| | - Jianming Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | | | |
Collapse
|
32
|
Chai T, Cui F, Mu P, Yang Y, Xu N, Yin Z, Jia Q, Yang S, Qiu J, Wang C. Enantio-alteration of gene transcription associated with bioconcentration in adult zebrafish (Danio rerio) exposed to chiral PCB149. Sci Rep 2016; 6:19478. [PMID: 26786282 PMCID: PMC4726444 DOI: 10.1038/srep19478] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/14/2015] [Indexed: 02/07/2023] Open
Abstract
Enantioselective enrichment of chiral PCB149 (2,2’,3,4’,5’,6-hexachlorobiphenyl) was analysed in adult zebrafish (Danio rerio) exposed to the racemate, (−)-PCB149, and (+)-PCB149. Greater enrichment of (−)-PCB149 compared to (+) PCB149 was observed following 0.5 ng/L exposure; however, as the exposure time and concentration increased, racemic enrichment was observed in adult fish exposed to the racemate. No biotransformation between the two isomers was observed in fish exposed to single enantiomers. When zebrafish were exposed to different forms of chiral PCB149, enantioselective expression of genes associated with polychlorinated biphenyls (PCBs) was observed in brain and liver tissues and enantioselective correlations between bioconcentration and target gene expression levels were observed in brain and liver tissues. The strong positive correlations between expression levels of target genes (alox5a and alox12) and PCB149 bioconcentration suggest that prolonged exposure to the racemate of chiral PCB149 may result in inflammation-associated diseases. Prolonged exposure to (−)-PCB149 may also affect metabolic pathways such as dehydrogenation and methylation in the brain tissues of adult zebrafish. Hepatic expression levels of genes related to the antioxidant system were significantly negatively correlated with bioconcentration following exposure to (+)-PCB149.
Collapse
Affiliation(s)
- Tingting Chai
- Institute of Quality Standards &Testing Technology for Agro-Products, Key Laboratory of Agro-product Quality and Safety, Chinese Academy of Agricultural Sciences; Key Laboratory of Agri-food Quality and Safety, Ministry of Agriculture, Beijing 100081, China.,College of Science, China Agricultural University, Beijing 100193, China
| | - Feng Cui
- College of Science, China Agricultural University, Beijing 100193, China
| | - Pengqian Mu
- Institute of Quality Standards &Testing Technology for Agro-Products, Key Laboratory of Agro-product Quality and Safety, Chinese Academy of Agricultural Sciences; Key Laboratory of Agri-food Quality and Safety, Ministry of Agriculture, Beijing 100081, China
| | - Yang Yang
- College of Science, China Agricultural University, Beijing 100193, China
| | - Nana Xu
- Institute of Quality Standards &Testing Technology for Agro-Products, Key Laboratory of Agro-product Quality and Safety, Chinese Academy of Agricultural Sciences; Key Laboratory of Agri-food Quality and Safety, Ministry of Agriculture, Beijing 100081, China
| | - Zhiqiang Yin
- Institute of Quality Standards &Testing Technology for Agro-Products, Key Laboratory of Agro-product Quality and Safety, Chinese Academy of Agricultural Sciences; Key Laboratory of Agri-food Quality and Safety, Ministry of Agriculture, Beijing 100081, China
| | - Qi Jia
- Institute of Quality Standards &Testing Technology for Agro-Products, Key Laboratory of Agro-product Quality and Safety, Chinese Academy of Agricultural Sciences; Key Laboratory of Agri-food Quality and Safety, Ministry of Agriculture, Beijing 100081, China
| | - Shuming Yang
- Institute of Quality Standards &Testing Technology for Agro-Products, Key Laboratory of Agro-product Quality and Safety, Chinese Academy of Agricultural Sciences; Key Laboratory of Agri-food Quality and Safety, Ministry of Agriculture, Beijing 100081, China
| | - Jing Qiu
- Institute of Quality Standards &Testing Technology for Agro-Products, Key Laboratory of Agro-product Quality and Safety, Chinese Academy of Agricultural Sciences; Key Laboratory of Agri-food Quality and Safety, Ministry of Agriculture, Beijing 100081, China
| | - Chengju Wang
- College of Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
33
|
Loss of SDHB Elevates Catecholamine Synthesis and Secretion Depending on ROS Production and HIF Stabilization. Neurochem Res 2015; 41:696-706. [PMID: 26620190 DOI: 10.1007/s11064-015-1738-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 10/10/2015] [Indexed: 12/30/2022]
Abstract
Germline mutations in genes encoding succinate dehydrogenase subunits are associated with the development of familial pheochromocytomas and paragangliomas [hereditary paraganglioma/pheochromocytoma syndrome (HPPS)]. In particular, a mutation in succinate dehydrogenase subunit B (SDHB) is highly associated with abdominal paraganglioma and subsequent distant metastasis (malignant paraganglioma), indicating the importance of SDHB genetic testing. The discovery of HPPS suggests an association among genetic mitochondrial defects, tumor development, and catecholamine oversecretion. To investigate this association, we transfected pheochromocytoma cells (PC12) with SDHB-specific siRNA. SDHB silencing virtually abolished complex II activity, demonstrating the utility of this in vitro model for investigating the pseudo-hypoxic drive hypothesis. Lack of complex II activity resulting from RNA interference of SDHB increased tyrosine hydroxylase (TH; the rate-limiting enzyme in catecholamine biosynthesis) activity and catecholamine secretion. Reduced apoptosis was observed accompanied by Bcl-2 accumulation in PC12 cells, consistent with the phenotypes of paragangliomas with SDHB mutations. In addition, SDHB silencing increased reactive oxygen species (ROS) production and nuclear HIF1α stabilization under normoxic conditions. Furthermore, phenotypes induced by complex II activity knockdown were abolished by pretreatment with N-acetyl cysteine (an ROS scavenger) and by prior HIF1α knockdown, indicating an ROS- and HIF1α-dependent mechanism. Our results indicate that increased ROS may act as signal transduction messengers that induce HIF1α stabilization and may be necessary for the pseudo-hypoxic states observed in our experimental model. To our knowledge, this is the first study demonstrating that pseudo-hypoxic states resulting from SDHB knockdown are associated with increased TH activity and catecholamine oversecretion.
Collapse
|
34
|
Baysal BE, Maher ER. 15 YEARS OF PARAGANGLIOMA: Genetics and mechanism of pheochromocytoma-paraganglioma syndromes characterized by germline SDHB and SDHD mutations. Endocr Relat Cancer 2015; 22:T71-82. [PMID: 26113606 DOI: 10.1530/erc-15-0226] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/25/2015] [Indexed: 12/29/2022]
Abstract
Pheochromocytomas and paragangliomas (PPGL) are rare neuroendocrine neoplasms that derive from small paraganglionic tissues which are located from skull base to the pelvic floor. Genetic predisposition plays an important role in development of PPGLs. Since the discovery of first mutations in the succinate dehydrogenase D (SDHD) gene, which encodes the smallest subunit of mitochondrial complex II (SDH), genetic studies have revealed a major role for mutations in SDH subunit genes, primarily in SDHB and SDHD, in predisposition to both familial and non-familial PPGLs. SDH-mutated PPGLs show robust expression of hypoxia induced genes, and genomic and histone hypermethylation. These effects occur in part through succinate-mediated inhibition of α-ketoglutarate-dependent dioxygenases. However, details of mechanisms by which SDH mutations activate hypoxic pathways and trigger subsequent neoplastic transformation remain poorly understood. Here, we present a brief review of the genetic and mechanistic aspects of SDH-mutated PPGLs.
Collapse
Affiliation(s)
- Bora E Baysal
- Department of PathologyRoswell Park Cancer Institute, Buffalo, New York 14263, USADepartment of Medical GeneticsCambridge NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Eamonn R Maher
- Department of PathologyRoswell Park Cancer Institute, Buffalo, New York 14263, USADepartment of Medical GeneticsCambridge NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
35
|
Low-Intensity Pulsed Ultrasound Improves the Functional Properties of Cardiac Mesoangioblasts. Stem Cell Rev Rep 2015. [DOI: 10.1007/s12015-015-9608-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
36
|
Her YF, Maher LJ. Succinate Dehydrogenase Loss in Familial Paraganglioma: Biochemistry, Genetics, and Epigenetics. Int J Endocrinol 2015; 2015:296167. [PMID: 26294907 PMCID: PMC4532907 DOI: 10.1155/2015/296167] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/05/2015] [Indexed: 01/14/2023] Open
Abstract
It is counterintuitive that metabolic defects reducing ATP production can cause, rather than protect from, cancer. Yet this is precisely the case for familial paraganglioma, a form of neuroendocrine malignancy caused by loss of succinate dehydrogenase in the tricarboxylic acid cycle. Here we review biochemical, genetic, and epigenetic considerations in succinate dehydrogenase loss and present leading models and mysteries associated with this fascinating and important tumor.
Collapse
Affiliation(s)
- Yeng F. Her
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | - L. James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
- *L. James Maher III:
| |
Collapse
|
37
|
Shiwa T, Oki K, Yoneda M, Arihiro K, Ohno H, Kishimoto R, Kohno N. A Patient with an Extra-adrenal Pheochromocytoma and Germ-line SDHB Mutation Accompanied by an Atypical Meningioma. Intern Med 2015; 54:2355-60. [PMID: 26370861 DOI: 10.2169/internalmedicine.54.4663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The gene succinate dehydrogenase subunit B (SDHB) encodes a protein comprising part of the mitochondrial complex II, which links the Krebs cycle and the electron-transport chain. Heterozygous germ-line SDHB mutations causes familial pheochromocytoma-paraganglioma syndrome and has also been linked to gastrointestinal stromal tumors, as well as renal cell carcinomas. We herein report a patient with a germ-line SDHB mutation who presented with an atypical meningioma that was identified as originating from a somatic SDHB mutation. The 41-year-old man, who had a surgical history of extra-adrenal pheochromocytoma at 23 years of age, recently developed gait disorder and hypertension. At the radiological examination, a tumor was detected in the cervical spinal cord at the C6-7 intervertebral level. The pathological findings of the isolated tumor were atypical meningioma assessed as grade II according to the World Health Organization criteria. Inherited neoplasia syndrome was suspected because of the patient's history of early-onset extra-adrenal pheochromocytoma and the development of meningioma. We therefore performed molecular genetic analyses. A direct sequence analysis revealed a heterozygous germ-line frameshift mutation in SDHB, specifically an 11-nucleotide deletion, c.305-315delCAATGAACATC, in exon 4, resulting in a frameshift p.A102EfsX12. Additionally, the sequence analysis of the tumor DNA revealed only a mutated allele with a frameshift mutation in the germ-line SDHB. Our findings suggest that SDHB plays an important role in the pathogenesis of meningiomas as well as pheochromocytomas. Therefore, a differential diagnosis for metastatic pheochromocytoma and other new onset tumors, including meningioma, particularly in patients with germ-line SDHB mutations and a previous history of pheochromocytoma should be carefully made.
Collapse
Affiliation(s)
- Tsuguka Shiwa
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Chen L, Liu T, Zhang S, Zhou J, Wang Y, Di W. Succinate dehydrogenase subunit B inhibits the AMPK-HIF-1α pathway in human ovarian cancer in vitro. J Ovarian Res 2014; 7:115. [PMID: 25491408 PMCID: PMC4279696 DOI: 10.1186/s13048-014-0115-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 11/27/2014] [Indexed: 11/10/2022] Open
Abstract
Background Ovarian carcinoma is one of the most common gynecological cancers with high mortality rates. Numerous evidences demonstrate that cancer cells undergo metabolic abnormality during tumorigenesis in tumor microenvironment and further facilitate tumor progression. Succinate dehydrogenase (SDH or Complex II) is one of the important enzymes in the tricarboxylic acid (TCA) cycle. Succinate dehydrogenase subunit B (SDHB) gene, which encodes one of the four subunits of SDH, has been recognized as a tumor suppressor. However the role of SDHB in ovarian cancer is still unclear. Methods Using the SDHB specific siRNA and overexpression plasmid, the expression of SDHB was silenced and conversely induced in ovarian cancer cell lines SKOV3 and A2780, respectively. The possible role of SDHB in ovarian cancer was investigated in vitro, using proliferation, migration and invasion assays. To explore the mechanism, proliferation and migration related proteins such as Bcl-2, cleaved caspase 3, p-ERK, MMP-2, and p-FAK were examined by western blot. P-P38, p-AMPKα, and HIF-1α were also examined by western blot. CoCl2 was used to induce HIF-1α expression in SKOV3 and A2780 cells. Results SDHB silencing promoted cell proliferation, invasion, and migration, but inhibited apoptosis of SKOV3 and A2780 cells. In contrast, overexpression of SDHB inhibited cell proliferation, invasion, migration, and promoted apoptosis in SKOV3 cells. It was observed that up-regulation of Bcl-2 and MMP-2, activation of p-P38, p-ERK, and p-FAK, inhibition of cleaved caspase 3 in SDHB-silenced cells. Meanwhile, decreased Bcl-2 and MMP-2, inhibition of p-P38, p-ERK, and p-FAK, activation of cleaved caspase 3 were shown in SDHB-overexpressed SKOV3 cells. HIF-1α, an essential factor in tumor progression, was up-regulated in SDHB-silenced cells with the activation of p-AMPKα and down-regulated in SDHB-overexpressed cancer cells with the decreased p-AMPKα. And SDHB was proved to be decreased due to upregulation of HIF-1α expression in CoCl2-treated cancer cells. Conclusions Our results firstly revealed that SDHB played a key role in cell proliferation, invasion, migration, and apoptosis of human ovarian carcinoma via AMPK-HIF-1α pathway. SDHB-overexpression might be a new approach to inhibit tumor progression in human ovarian carcinoma. Electronic supplementary material The online version of this article (doi:10.1186/s13048-014-0115-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lilan Chen
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China. .,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China.
| | - Ting Liu
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China. .,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China.
| | - Shu Zhang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China. .,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China.
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China. .,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China.
| | - Yunfei Wang
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China. .,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China.
| | - Wen Di
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China. .,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, P. R. China.
| |
Collapse
|
39
|
Abstract
Losses in the succinate dehydrogenase (SDH) complex characterize 20% to 30% of extra-adrenal paragangliomas and 7% to 8% of gastric GISTs, and rare renal cell carcinomas. This loss is reflected as lack of the normally ubiquitous immunohistochemical expression of the SDH subunit B (SDHB). In paragangliomas, SDHB loss correlates with homozygous loss of any of the SDH subunits, typically by loss-of-function mutations. The occurrence of SDHB losses in other epithelial malignancies is unknown. In this study, we immunohistochemically examined 2258 epithelial, mostly malignant neoplasms including common carcinomas of all sites. Among renal cell carcinomas, SDHB loss was observed in 4 of 711 cases (0.6%), including a patient with an SDHB-deficient GIST. Histologically, the SDHB-negative renal carcinomas varied. There was 1 clear cell carcinoma with a high nuclear grade, 1 papillary carcinoma type 2, 1 unclassified carcinoma with a glandular pattern, and 1 oncocytoid low-grade carcinoma as previously described for SDHB-negative renal carcinoma. None of these patients was known to have paragangliomas or had loss of SDHA expression in the tumor. Three of these patients had metastases at presentation (2 in the adrenal, 1 in the retroperitoneal lymph nodes). There were no cases with SDHB loss among 64 renal oncocytomas. SDHB losses were not seen in other carcinomas, except in 1 prostatic adenocarcinoma (1/57), 1 lymphoepithelial carcinoma of the stomach, and 1 (1/40) seminoma. On the basis of this study, SDHB losses occur in 0.6% of renal cell carcinomas and extremely rarely in other carcinomas. Some of these renal carcinomas may be clinically aggressive. The clinical significance and molecular genetics of these SDHB-negative tumors requires further study.
Collapse
|
40
|
Hwang MS, Rohlena J, Dong LF, Neuzil J, Grimm S. Powerhouse down: Complex II dissociation in the respiratory chain. Mitochondrion 2014; 19 Pt A:20-8. [PMID: 24933571 DOI: 10.1016/j.mito.2014.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 05/28/2014] [Accepted: 06/04/2014] [Indexed: 12/12/2022]
Abstract
Complex II of the respiratory chain (RC) recently emerged as a prominent regulator of cell death. In both cancer cells as well as neurodegenerative diseases, mutations in subunits have been found along with other genetic alterations indirectly affecting this complex. Anticancer compounds were developed that target complex II and cause cell death in a tumor-specific way. Our mechanistic understanding of how complex II is activated for cell death induction has recently been made clearer in recent studies, the results of which are covered in this review. This protein assembly is specifically activated for cell death via the dissociation of its SDHA and SDHB subunits from the membrane-anchoring proteins through pH change or mitochondrial Ca(2+) influx. The SDH activity contained in the SDHA/SDHB subcomplex remains intact and then generates, in an uncontrolled fashion, excessive amounts of reactive oxygen species (ROS) for cell death. Future studies on this mitochondrial complex will further elucidate it as a target for cancer treatments and reveal its role as a nexus for many diverse stimuli in cell death signaling.
Collapse
Affiliation(s)
- Ming-Shih Hwang
- Division of Experimental Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Jakub Rohlena
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic
| | - Lan-Feng Dong
- School of Medical Science, Griffith Health Institute, Griffith University, Southport Qld 4222, Australia
| | - Jiri Neuzil
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic; School of Medical Science, Griffith Health Institute, Griffith University, Southport Qld 4222, Australia
| | - Stefan Grimm
- Division of Experimental Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
41
|
Miettinen M, Lasota J. Succinate dehydrogenase deficient gastrointestinal stromal tumors (GISTs) - a review. Int J Biochem Cell Biol 2014; 53:514-9. [PMID: 24886695 DOI: 10.1016/j.biocel.2014.05.033] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/20/2014] [Accepted: 05/21/2014] [Indexed: 02/07/2023]
Abstract
Loss of function of the succinate dehydrogenase complex characterizes a rare group of human tumors including some gastrointestinal stromal tumors, paragangliomas, renal carcinomas, and pituitary adenomas, and these can all be characterized as SDH-deficient tumors. Approximately 7.5% of gastric gastrointestinal stromal tumors are SDH-deficient and not driven by KIT/PDGFRA mutations, as are most other GISTs. The occurrence of SDH-deficient GISTs is restricted to stomach, and they typically occur in children and young adults representing a spectrum of clinical behavior from indolent to progressive. Slow progression is a common feature even after metastatic spread has taken place, and many patients live years with metastases. SDH-deficient GISTs have characteristic morphologic features including multinodular gastric wall involvement, often multiple separate tumors, common lymphovascular invasion, and occasional lymph node metastases. Diagnostic is the loss of succinate dehydrogenase subunit B (SDHB) from the tumor cells and this can be practically assessed by immunohistochemistry. SDHA is lost in cases associated with SDHA mutations. Approximately half of the patients have SDH subunit gene mutations, often germline and most commonly A (30%), and B, C or D (together 20%), with both alleles inactivated in the tumor cells according to the classic tumor suppressor gene model. Half of the cases are not associated with SDH-mutations and epigenetic silencing of the SDH complex is the possible pathogenesis. Extensive genomic methylation has been observed in these tumors, which is in contrast with other GISTs. SDH-loss causes succinate accumulation and activation of pseudohypoxia signaling via overexpression of HIF-proteins. Activation of insulin-like growth factor 1-signaling is also typical of these tumors. SDH-deficient GISTs are a unique group of GISTs with an energy metabolism defect as the key oncogenic mechanism. This article is part of a Directed Issue entitled: Rare Cancers.
Collapse
Affiliation(s)
- Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA.
| | - Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
42
|
|
43
|
Vadakara J, von Mehren M. Gastrointestinal stromal tumors: management of metastatic disease and emerging therapies. Hematol Oncol Clin North Am 2013; 27:905-20. [PMID: 24093167 PMCID: PMC3792495 DOI: 10.1016/j.hoc.2013.07.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastrointestinal stromal tumors (GIST) are the most common mesenchymal tumors of the gastrointestinal tract. Before the advent of tyrosine kinase inhibitors (TKIs) there were few treatment options available to patients with metastatic GIST. Surgery was the mainstay of treatment and the prognosis was dismal. With the advent of imatinib and second-line TKIs the prognosis of metastatic GIST has improved dramatically; however, there is still a need for therapies for patients with disease refractory to TKI therapy. Newer agents are under investigation and may have promise. This article discusses the current standard of care in terms of standard and investigational pharmacotherapy in the management of metastatic GIST.
Collapse
Affiliation(s)
- Joseph Vadakara
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111
| | - Margaret von Mehren
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111
| |
Collapse
|
44
|
Deblois G, St-Pierre J, Giguère V. The PGC-1/ERR signaling axis in cancer. Oncogene 2013; 32:3483-90. [PMID: 23208510 DOI: 10.1038/onc.2012.529] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/15/2012] [Accepted: 10/15/2012] [Indexed: 12/20/2022]
Abstract
Proliferating cells need to produce a large amount of energy and, at the same time, need to maintain a constant supply of biosynthetic precursors of macromolecules that are used as building blocks for generating new cells. Indeed, many cancer cells undergo a switch from mitochondrial to glycolytic metabolism and display a truncated tricarboxylic acid cycle to match these specific metabolic requirements of proliferation. Understanding the mechanisms by which cancer cells reprogram various metabolic pathways to satisfy their unique bioenergetic requirements has become an active field of research. Concomitantly, it has emerged that members of a family of orphan nuclear receptors known as the estrogen-related receptors (ERRs), working in concert with members of the PPARγ coactivator (PGC)-1 family, act as central transcriptional regulators of metabolic gene networks involved in maintaining energy homeostasis in normal cells. Recent studies have suggested that the PGC-1/ERR transcriptional axis is also important in the metabolic reprogramming of cancer cells. This review focuses on the functional integration of the PGC-1/ERR axis with known oncogenes and the observation that modulation of the activity of this axis can have both pro- and anti-proliferative properties.
Collapse
Affiliation(s)
- G Deblois
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
45
|
Belinsky MG, Rink L, von Mehren M. Succinate dehydrogenase deficiency in pediatric and adult gastrointestinal stromal tumors. Front Oncol 2013; 3:117. [PMID: 23730622 PMCID: PMC3656383 DOI: 10.3389/fonc.2013.00117] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 04/26/2013] [Indexed: 12/18/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) in adults are generally driven by somatic gain-of-function mutations in KIT or PDGFRA, and biological therapies targeted to these receptor tyrosine kinases comprise part of the treatment regimen for metastatic and inoperable GISTs. A minority (10-15%) of GISTs in adults, along with ∼85% of pediatric GISTs, lacks oncogenic mutations in KIT and PDGFRA. Not surprisingly these wild type (WT) GISTs respond poorly to kinase inhibitor therapy. A subset of WT GISTs shares a set of distinguishing clinical and pathological features, and a flurry of recent reports has convincingly demonstrated shared molecular characteristics. These GISTs have a distinct transcriptional profile including over-expression of the insulin-like growth factor-1 receptor, and exhibit deficiency in the succinate dehydrogenase (SDH) enzyme complex. The latter is often but not always linked to bi-allelic inactivation of SDH subunit genes, particularly SDHA. This review will summarize the molecular, pathological, and clinical connections that link this group of SDH-deficient neoplasms, and offer a view toward understanding the underlying biology of the disease and the therapeutic challenges implicit to this biology.
Collapse
Affiliation(s)
- Martin G. Belinsky
- Department of Medical Oncology, Fox Chase Cancer CenterPhiladelphia, PA, USA
| | - Lori Rink
- Department of Medical Oncology, Fox Chase Cancer CenterPhiladelphia, PA, USA
| | - Margaret von Mehren
- Department of Medical Oncology, Fox Chase Cancer CenterPhiladelphia, PA, USA
| |
Collapse
|
46
|
Grimm S. Respiratory chain complex II as general sensor for apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2013; 1827:565-72. [DOI: 10.1016/j.bbabio.2012.09.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/09/2012] [Accepted: 09/10/2012] [Indexed: 01/05/2023]
|
47
|
Kiss NB, Muth A, Andreasson A, Juhlin CC, Geli J, Bäckdahl M, Höög A, Wängberg B, Nilsson O, Ahlman H, Larsson C. Acquired hypermethylation of the P16INK4A promoter in abdominal paraganglioma: relation to adverse tumor phenotype and predisposing mutation. Endocr Relat Cancer 2013; 20:65-78. [PMID: 23154831 PMCID: PMC3573842 DOI: 10.1530/erc-12-0267] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recurrent alterations in promoter methylation of tumor suppressor genes (TSGs) and LINE1 (L1RE1) repeat elements were previously reported in pheochromocytoma and abdominal paraganglioma. This study was undertaken to explore CpG methylation abnormalities in an extended tumor panel and assess possible relationships between metastatic disease and mutation status. CpG methylation was quantified by bisulfite pyrosequencing for selected TSG promoters and LINE1 repeats. Methylation indices above normal reference were observed for DCR2 (TNFRSF10D), CDH1, P16 (CDKN2A), RARB, and RASSF1A. Z-scores for overall TSG, and individual TSG methylation levels, but not LINE1, were significantly correlated with metastatic disease, paraganglioma, disease predisposition, or outcome. Most strikingly, P16 hypermethylation was strongly associated with SDHB mutation as opposed to RET/MEN2, VHL/VHL, or NF1-related disease. Parallel analyses of constitutional, tumor, and metastasis DNA implicate an order of events where constitutional SDHB mutations are followed by TSG hypermethylation and 1p loss in primary tumors, later transferred to metastatic tissue. In the combined material, P16 hypermethylation was prevalent in SDHB-mutated samples and was associated with short disease-related survival. The findings verify the previously reported importance of P16 and other TSG hypermethylation in an independent tumor series. Furthermore, a constitutional SDHB mutation is proposed to predispose for an epigenetic tumor phenotype occurring before the emanation of clinically recognized malignancy.
Collapse
Affiliation(s)
- Nimrod B Kiss
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Giubellino A, Bullova P, Nölting S, Turkova H, Powers JF, Liu Q, Guichard S, Tischler AS, Grossman AB, Pacak K. Combined inhibition of mTORC1 and mTORC2 signaling pathways is a promising therapeutic option in inhibiting pheochromocytoma tumor growth: in vitro and in vivo studies in female athymic nude mice. Endocrinology 2013; 154:646-55. [PMID: 23307788 PMCID: PMC3548182 DOI: 10.1210/en.2012-1854] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Several lines of evidence, including the recent discovery of novel susceptibility genes, point out an important role for the mammalian target of rapamycin (mTOR) signaling pathway in the development of pheochromocytoma. Analyzing a set of pheochromocytomas from patients with different genetic backgrounds, we observed and confirmed a significant overexpression of key mTOR complex (mTORC) signaling mediators. Using selective ATP-competitive inhibitors targeting both mTORC1 and mTORC2, we significantly arrested the in vitro cell proliferation and blocked migration of pheochromocytoma cells as a result of the pharmacological suppression of the Akt/mTOR signaling pathway. Moreover, AZD8055, a selective ATP-competitive dual mTORC1/2 small molecular inhibitor, significantly reduced the tumor burden in a model of metastatic pheochromocytoma using female athymic nude mice. This study suggests that targeting both mTORC1 and mTORC2 is a potentially rewarding strategy and supports the application of selective inhibitors in combinatorial drug regimens for metastatic pheochromocytoma.
Collapse
Affiliation(s)
- Alessio Giubellino
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Wojtovich AP, Smith CO, Haynes CM, Nehrke KW, Brookes PS. Physiological consequences of complex II inhibition for aging, disease, and the mKATP channel. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2013; 1827:598-611. [PMID: 23291191 DOI: 10.1016/j.bbabio.2012.12.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/14/2012] [Accepted: 12/17/2012] [Indexed: 12/21/2022]
Abstract
In recent years, it has become apparent that there exist several roles for respiratory complex II beyond metabolism. These include: (i) succinate signaling, (ii) reactive oxygen species (ROS) generation, (iii) ischemic preconditioning, (iv) various disease states and aging, and (v) a role in the function of the mitochondrial ATP-sensitive K(+) (mKATP) channel. This review will address the involvement of complex II in each of these areas, with a focus on how complex II regulates or may be involved in the assembly of the mKATP. This article is part of a Special Issue entitled: Respiratory complex II: Role in cellular physiology and disease.
Collapse
Affiliation(s)
- Andrew P Wojtovich
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | | |
Collapse
|
50
|
An iTRAQ-based mitoproteomics approach for profiling the nephrotoxicity mechanisms of ochratoxin A in HEK 293 cells. J Proteomics 2013; 78:398-415. [DOI: 10.1016/j.jprot.2012.10.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 09/28/2012] [Accepted: 10/11/2012] [Indexed: 01/05/2023]
|