201
|
Hypoxia-Inducible Factor 1 and Mitochondria: An Intimate Connection. Biomolecules 2022; 13:biom13010050. [PMID: 36671435 PMCID: PMC9855368 DOI: 10.3390/biom13010050] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
The general objective of the review is to explain the interaction between HIF-1 and mitochondria. On the one hand, this review describes the effects of HIF-1 on mitochondrial structure, including quantity, distribution, and morphology, as well as on mitochondrial metabolism and respiratory function. On the other hand, various factors, including mitochondrial activation of enzymes, the respiratory chain, complex and decoupling proteins, affect the stability and activity of HIF-1. It is possible to develop future molecular therapeutic interventions by understanding the interrelationships between HIF-1 and mitochondria.
Collapse
|
202
|
Vohwinkel CU, Burns N, Coit E, Yuan X, Vladar EK, Sul C, Schmidt EP, Carmeliet P, Stenmark K, Nozik ES, Tuder RM, Eltzschig HK. HIF1A-dependent induction of alveolar epithelial PFKFB3 dampens acute lung injury. JCI Insight 2022; 7:e157855. [PMID: 36326834 PMCID: PMC9869967 DOI: 10.1172/jci.insight.157855] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Acute lung injury (ALI) is a severe form of lung inflammation causing acute respiratory distress syndrome in patients. ALI pathogenesis is closely linked to uncontrolled alveolar inflammation. We hypothesize that specific enzymes of the glycolytic pathway could function as key regulators of alveolar inflammation. Therefore, we screened isolated alveolar epithelia from mice exposed to ALI induced by injurious ventilation to assess their metabolic responses. These studies pointed us toward a selective role for isoform 3 of the 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB3). Pharmacologic inhibition or genetic deletion of Pfkfb3 in alveolar epithelia (Pfkfb3loxP/loxP SPC-ER-Cre+ mice) was associated with profound increases in ALI during injurious mechanical ventilation or acid instillation. Studies in genetic models linked Pfkfb3 expression and function to Hif1a. Not only did intratracheal pyruvate instillation reconstitute Pfkfb3loxP/loxP or Hif1aloxP/loxP SPC-ER-Cre+ mice, but pyruvate was also effective in ALI treatment of wild-type mice. Finally, proof-of-principle studies in human lung biopsies demonstrated increased PFKFB3 staining in injured lungs and colocalized PFKFB3 to alveolar epithelia. These studies reveal a specific role for PFKFB3 in counterbalancing alveolar inflammation and lay the groundwork for novel metabolic therapeutic approaches during ALI.
Collapse
Affiliation(s)
- Christine U. Vohwinkel
- Cardio Vascular Pulmonary Research Lab and
- Section of Critical Care Medicine, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Nana Burns
- Cardio Vascular Pulmonary Research Lab and
- Section of Critical Care Medicine, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Ethan Coit
- Cardio Vascular Pulmonary Research Lab and
- Section of Critical Care Medicine, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center Houston, Houston, Texas, USA
| | - Eszter K. Vladar
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Christina Sul
- Section of Critical Care Medicine, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Eric P. Schmidt
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Kurt Stenmark
- Cardio Vascular Pulmonary Research Lab and
- Section of Critical Care Medicine, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Eva S. Nozik
- Cardio Vascular Pulmonary Research Lab and
- Section of Critical Care Medicine, Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Rubin M. Tuder
- Cardio Vascular Pulmonary Research Lab and
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center Houston, Houston, Texas, USA
| |
Collapse
|
203
|
Dai X, Zhou Y, Han F, Li J. Succinylation and redox status in cancer cells. Front Oncol 2022; 12:1081712. [PMID: 36605449 PMCID: PMC9807787 DOI: 10.3389/fonc.2022.1081712] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
Succinylation is a post-translational modification (PTM) event that associates metabolic reprogramming with various pathological disorders including cancers via transferring a succinyl group to a residue of the target protein in an enzymic or non-enzymic manner. With our incremental knowledge on the roles of PTM played in tumor initiation and progression, relatively little has been focused on succinylation and its clinical implications. By delineating the associations of succinylation with cancer hallmarks, we identify the, in general, promotive roles of succinylation in manifesting cancer hallmarks, and conceptualize two working modes of succinylation in driving oncogenic signaling, i.e., via altering the structure and charge of target proteins towards enhanced stability and activity. We also characterize succinylation as a reflection of cellular redox homeostatic status and metabolic state, and bring forth the possible use of hyper-succinylated genome for early cancer diagnosis or disease progression indication. In addition, we propose redox modulation tools such as cold atmospheric plasma as a promising intervention approach against tumor cells and cancer stemness via targeting the redox homeostatic environment cells established under a pathological condition such as hypoxia. Taken together, we emphasize the central role of succinylation in bridging the gap between cellular metabolism and redox status, and its clinical relevance as a mark for cancer diagnosis as well as a target in onco-therapeutics.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China,*Correspondence: Xiaofeng Dai, ; Jitian Li,
| | - Yanyan Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fei Han
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China,*Correspondence: Xiaofeng Dai, ; Jitian Li,
| |
Collapse
|
204
|
Wang S, Kang Y, Wang R, Deng J, Yu Y, Yu J, Wang J. Emerging Roles of NDUFS8 Located in Mitochondrial Complex I in Different Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248754. [PMID: 36557887 PMCID: PMC9783039 DOI: 10.3390/molecules27248754] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
NADH:ubiquinone oxidoreductase core subunit S8 (NDUFS8) is an essential core subunit and component of the iron-sulfur (FeS) fragment of mitochondrial complex I directly involved in the electron transfer process and energy metabolism. Pathogenic variants of the NDUFS8 are relevant to infantile-onset and severe diseases, including Leigh syndrome, cancer, and diabetes mellitus. With over 1000 nuclear genes potentially causing a mitochondrial disorder, the current diagnostic approach requires targeted molecular analysis, guided by a combination of clinical and biochemical features. Currently, there are only several studies on pathogenic variants of the NDUFS8 in Leigh syndrome, and a lack of literature on its precise mechanism in cancer and diabetes mellitus exists. Therefore, NDUFS8-related diseases should be extensively explored and precisely diagnosed at the molecular level with the application of next-generation sequencing technologies. A more distinct comprehension will be needed to shed light on NDUFS8 and its related diseases for further research. In this review, a comprehensive summary of the current knowledge about NDUFS8 structural function, its pathogenic mutations in Leigh syndrome, as well as its underlying roles in cancer and diabetes mellitus is provided, offering potential pathogenesis, progress, and therapeutic target of different diseases. We also put forward some problems and solutions for the following investigations.
Collapse
Affiliation(s)
- Sifan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; (S.W.); (Y.K.); (R.W.); (J.D.); (Y.Y.)
- Department of Pathology, School of Basic Medicine, Central South University, Changsha 410008, China
- Xiangya School of Medicine, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yuanbo Kang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; (S.W.); (Y.K.); (R.W.); (J.D.); (Y.Y.)
- Department of Pathology, School of Basic Medicine, Central South University, Changsha 410008, China
- Xiangya School of Medicine, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ruifeng Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; (S.W.); (Y.K.); (R.W.); (J.D.); (Y.Y.)
- Department of Pathology, School of Basic Medicine, Central South University, Changsha 410008, China
| | - Junqi Deng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; (S.W.); (Y.K.); (R.W.); (J.D.); (Y.Y.)
- Department of Pathology, School of Basic Medicine, Central South University, Changsha 410008, China
| | - Yupei Yu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; (S.W.); (Y.K.); (R.W.); (J.D.); (Y.Y.)
- Department of Pathology, School of Basic Medicine, Central South University, Changsha 410008, China
| | - Jun Yu
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410008, China
- Correspondence: (J.Y.); (J.W.); Tel./Fax: +86-731-84805411 (J.W.)
| | - Junpu Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; (S.W.); (Y.K.); (R.W.); (J.D.); (Y.Y.)
- Department of Pathology, School of Basic Medicine, Central South University, Changsha 410008, China
- Xiangya School of Medicine, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Correspondence: (J.Y.); (J.W.); Tel./Fax: +86-731-84805411 (J.W.)
| |
Collapse
|
205
|
Kocianova E, Piatrikova V, Golias T. Revisiting the Warburg Effect with Focus on Lactate. Cancers (Basel) 2022; 14:cancers14246028. [PMID: 36551514 PMCID: PMC9776395 DOI: 10.3390/cancers14246028] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Rewired metabolism is acknowledged as one of the drivers of tumor growth. As a result, aerobic glycolysis, or the Warburg effect, is a feature of many cancers. Increased glucose uptake and glycolysis provide intermediates for anabolic reactions necessary for cancer cell proliferation while contributing sufficient energy. However, the accompanying increased lactate production, seemingly wasting glucose carbon, was originally explained only by the need to regenerate NAD+ for successive rounds of glycolysis by the lactate dehydrogenase (LDH) reaction in the cytosol. After the discovery of a mitochondrial LDH isoform, lactate oxidation entered the picture, and lactate was recognized as an important oxidative fuel. It has also been revealed that lactate serves a variety of signaling functions and helps cells adapt to the new environment. Here, we discuss recent findings on lactate metabolism and signaling in cancer while attempting to explain why the Warburg effect is adopted by cancer cells.
Collapse
Affiliation(s)
- Eva Kocianova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 84505 Bratislava, Slovakia
| | - Viktoria Piatrikova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 84505 Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 84215 Bratislava, Slovakia
| | - Tereza Golias
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, 84505 Bratislava, Slovakia
- Correspondence:
| |
Collapse
|
206
|
Kidney tumors associated with germline mutations of FH and SDHB show a CpG island methylator phenotype (CIMP). PLoS One 2022; 17:e0278108. [PMID: 36455002 PMCID: PMC9714951 DOI: 10.1371/journal.pone.0278108] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/10/2022] [Indexed: 12/03/2022] Open
Abstract
Germline mutations within the Krebs cycle enzyme genes fumarate hydratase (FH) or succinate dehydrogenase (SDHB, SDHC, SDHD) are associated with an increased risk of aggressive and early metastasizing variants of renal cell carcinoma (RCC). These RCCs express significantly increased levels of intracellular fumarate or succinate that inhibit 2-oxoglutarate-dependent dioxygenases, such as the TET enzymes that regulate DNA methylation. This study evaluated the genome-wide methylation profiles of 34 RCCs from patients with RCC susceptibility syndromes and 11 associated normal samples using the Illumina HumanMethylation450 BeadChip. All the HLRCC (FH mutated) and SDHB-RCC (SDHB mutated) tumors demonstrated a distinct CpG island methylator phenotype (CIMP). HLRCC tumors demonstrated an extensive and relatively uniform level of hypermethylation that showed some correlation with tumor size. SDHB-RCC demonstrated a lesser and more varied pattern of hypermethylation that overlapped in part with the HLRCC hypermethylation. Combined methylation and mRNA expression analysis of the HLRCC tumors demonstrated hypermethylation and transcription downregulation of genes associated with the HIF pathway, HIF3A and CITED4, the WNT pathway, SFRP1, and epithelial-to-mesenchymal transition and MYC expression, OVOL1. These observations were confirmed in the TCGA CIMP-RCC tumors. A selected panel of probes could identify the CIMP tumors and differentiate between HLRCC and SDHB-RCC tumors. This panel accurately detected all CIMP-RCC tumors within the TCGA RCC cohort, identifying them as HLRCC -like, and could potentially be used to create a liquid biopsy-based screening tool. The CIMP signature in these aggressive tumors could provide both a useful biomarker for diagnosis and a target for novel therapies.
Collapse
|
207
|
Atallah R, Olschewski A, Heinemann A. Succinate at the Crossroad of Metabolism and Angiogenesis: Roles of SDH, HIF1α and SUCNR1. Biomedicines 2022; 10:3089. [PMID: 36551845 PMCID: PMC9775124 DOI: 10.3390/biomedicines10123089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Angiogenesis is an essential process by which new blood vessels develop from existing ones. While adequate angiogenesis is a physiological process during, for example, tissue repair, insufficient and excessive angiogenesis stands on the pathological side. Fine balance between pro- and anti-angiogenic factors in the tissue environment regulates angiogenesis. Identification of these factors and how they function is a pressing topic to develop angiogenesis-targeted therapeutics. During the last decade, exciting data highlighted non-metabolic functions of intermediates of the mitochondrial Krebs cycle including succinate. Among these functions is the contribution of succinate to angiogenesis in various contexts and through different mechanisms. As the concept of targeting metabolism to treat a wide range of diseases is rising, in this review we summarize the mechanisms by which succinate regulates angiogenesis in normal and pathological settings. Gaining a comprehensive insight into how this metabolite functions as an angiogenic signal will provide a useful approach to understand diseases with aberrant or excessive angiogenic background, and may provide strategies to tackle them.
Collapse
Affiliation(s)
- Reham Atallah
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Akos Heinemann
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
208
|
Germanova E, Khmil N, Pavlik L, Mikheeva I, Mironova G, Lukyanova L. The Role of Mitochondrial Enzymes, Succinate-Coupled Signaling Pathways and Mitochondrial Ultrastructure in the Formation of Urgent Adaptation to Acute Hypoxia in the Myocardium. Int J Mol Sci 2022; 23:ijms232214248. [PMID: 36430733 PMCID: PMC9696391 DOI: 10.3390/ijms232214248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/21/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
The effect of a single one-hour exposure to three modes of hypobaric hypoxia (HBH) differed in the content of O2 in inhaled air (FiO2-14%, 10%, 8%) in the development of mitochondrial-dependent adaptive processes in the myocardium was studied in vivo. The following parameters have been examined: (a) an urgent reaction of catalytic subunits of mitochondrial enzymes (NDUFV2, SDHA, Cyt b, COX2, ATP5A) in the myocardium as an indicator of the state of the respiratory chain electron transport function; (b) an urgent activation of signaling pathways dependent on GPR91, HIF-1α and VEGF, allowing us to assess their role in the formation of urgent mechanisms of adaptation to hypoxia in the myocardium; (c) changes in the ultrastructure of three subpopulations of myocardial mitochondria under these conditions. The studies were conducted on two rat phenotypes: rats with low resistance (LR) and high resistance (HR) to hypoxia. The adaptive and compensatory role of the mitochondrial complex II (MC II) in maintaining the electron transport and energy function of the myocardium in a wide range of reduced O2 concentrations in the initial period of hypoxic exposure has been established. The features of urgent reciprocal regulatory interaction of NAD- and FAD-dependent oxidation pathways in myocardial mitochondria under these conditions have been revealed. The data indicating the participation of GPR91, HIF-1a and VEGF in this process have been obtained. The ultrastructure of the mitochondrial subpopulations in the myocardium of LR and HR rats differed in normoxic conditions and reacted differently to hypoxia of varying severity. The parameters studied together are highly informative indicators of the quality of cardiac activity and metabolic biomarkers of urgent adaptation in various hypoxic conditions.
Collapse
Affiliation(s)
- Elita Germanova
- Institute of General Pathology and Pathophysiology, 8 Baltijskaya Str., Moscow 125315, Russia
| | - Natalya Khmil
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Lyubov Pavlik
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Irina Mikheeva
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Galina Mironova
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
- Correspondence: (G.M.); (L.L.)
| | - Ludmila Lukyanova
- Institute of General Pathology and Pathophysiology, 8 Baltijskaya Str., Moscow 125315, Russia
- Correspondence: (G.M.); (L.L.)
| |
Collapse
|
209
|
Webster BR, Gopal N, Ball MW. Tumorigenesis Mechanisms Found in Hereditary Renal Cell Carcinoma: A Review. Genes (Basel) 2022; 13:2122. [PMID: 36421797 PMCID: PMC9690265 DOI: 10.3390/genes13112122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 09/29/2023] Open
Abstract
Renal cell carcinoma is a heterogenous cancer composed of an increasing number of unique subtypes each with their own cellular and tumor behavior. The study of hereditary renal cell carcinoma, which composes just 5% of all types of tumor cases, has allowed for the elucidation of subtype-specific tumorigenesis mechanisms that can also be applied to their sporadic counterparts. This review will focus on the major forms of hereditary renal cell carcinoma and the genetic alterations contributing to their tumorigenesis, including von Hippel Lindau syndrome, Hereditary Papillary Renal Cell Carcinoma, Succinate Dehydrogenase-Deficient Renal Cell Carcinoma, Hereditary Leiomyomatosis and Renal Cell Carcinoma, BRCA Associated Protein 1 Tumor Predisposition Syndrome, Tuberous Sclerosis, Birt-Hogg-Dubé Syndrome and Translocation RCC. The mechanisms for tumorigenesis described in this review are beginning to be exploited via the utilization of novel targets to treat renal cell carcinoma in a subtype-specific fashion.
Collapse
Affiliation(s)
| | | | - Mark W. Ball
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA
| |
Collapse
|
210
|
Yoon S, Shin M, Shim J. Inter-organ regulation by the brain in Drosophila development and physiology. J Neurogenet 2022:1-13. [DOI: 10.1080/01677063.2022.2137162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Sunggyu Yoon
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Mingyu Shin
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Jiwon Shim
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Science, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
211
|
Kuhn AR, van Bilsen M. Oncometabolism: A Paradigm for the Metabolic Remodeling of the Failing Heart. Int J Mol Sci 2022; 23:ijms232213902. [PMID: 36430377 PMCID: PMC9699042 DOI: 10.3390/ijms232213902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Heart failure is associated with profound alterations in cardiac intermediary metabolism. One of the prevailing hypotheses is that metabolic remodeling leads to a mismatch between cardiac energy (ATP) production and demand, thereby impairing cardiac function. However, even after decades of research, the relevance of metabolic remodeling in the pathogenesis of heart failure has remained elusive. Here we propose that cardiac metabolic remodeling should be looked upon from more perspectives than the mere production of ATP needed for cardiac contraction and relaxation. Recently, advances in cancer research have revealed that the metabolic rewiring of cancer cells, often coined as oncometabolism, directly impacts cellular phenotype and function. Accordingly, it is well feasible that the rewiring of cardiac cellular metabolism during the development of heart failure serves similar functions. In this review, we reflect on the influence of principal metabolic pathways on cellular phenotype as originally described in cancer cells and discuss their potential relevance for cardiac pathogenesis. We discuss current knowledge of metabolism-driven phenotypical alterations in the different cell types of the heart and evaluate their impact on cardiac pathogenesis and therapy.
Collapse
|
212
|
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
|
213
|
Yamaguchi Y, Yokoyama M, Takemoto A, Nakamura Y, Fukuda S, Uehara S, Tanaka H, Yoshida S, Matsuoka Y, Fujii Y. Succinate dehydrogenase-deficient malignant paraganglioma complicated by succinate dehydrogenase-deficient renal cell carcinoma. IJU Case Rep 2022; 5:480-483. [PMID: 36341179 PMCID: PMC9626355 DOI: 10.1002/iju5.12520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 07/26/2022] [Indexed: 10/17/2023] Open
Abstract
INTRODUCTION SDH Gene mutation is known to be a common cause of pheochromocytoma/paraganglioma and renal cell carcinoma. Here, we report a case of succinate dehydrogenase B-deficient paraganglioma, which has a high risk of metastasis and recurrence, complicated by succinate dehydrogenase-deficient renal cell carcinoma, which is rare and accounts for approximately 0.1% of all renal cell carcinomas. CASE PRESENTATION A 50-year-old man underwent en bloc resection of a retroperitoneal tumor and the right kidney for retroperitoneal paraganglioma and right renal tumor. Both tumors showed negative expressions of succinate dehydrogenase B in immunostaining. The patient was diagnosed with succinate dehydrogenase-deficient paraganglioma and succinate dehydrogenase-deficient renal cell carcinoma. Seventeen months later, retroperitoneal lymphadenectomy revealed lymph node metastasis of the paraganglioma. Deletion of the SDHB gene was revealed by genome sequencing of the lymph node. CONCLUSION This is the first reported case of synchronously diagnosed succinate dehydrogenase-deficient paraganglioma and succinate dehydrogenase-deficient renal cell carcinoma.
Collapse
Affiliation(s)
| | - Minato Yokoyama
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Akira Takemoto
- Bioresource Research CenterTokyo Medical and Dental UniversityTokyoJapan
| | - Yuki Nakamura
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Shohei Fukuda
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Sho Uehara
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Hajime Tanaka
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Soichiro Yoshida
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Yoh Matsuoka
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Yasuhisa Fujii
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
214
|
Park D, Lim G, Yoon SJ, Yi HS, Choi DW. The role of immunomodulatory metabolites in shaping the inflammatory response of macrophages. BMB Rep 2022; 55:519-527. [PMID: 36195564 PMCID: PMC9712705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 12/14/2022] Open
Abstract
Macrophage activation has long been implicated in a myriad of human pathophysiology, particularly in the context of the dysregulated capacities of an unleashing intracellular or/and extracellular inflammatory response. A growing number of studies have functionally coupled the macrophages' inflammatory capacities with dynamic metabolic reprogramming which occurs during activation, albeit the results have been mostly interpreted through classic metabolism point of view; macrophages take advantage of the rewired metabolism as a source of energy and for biosynthetic precursors. However, a specific subset of metabolic products, namely immune-modulatory metabolites, has recently emerged as significant regulatory signals which control inflammatory responses in macrophages and the relevant extracellular milieu. In this review, we introduce recently highlighted immuno-modulatory metabolites, with the aim of understanding their physiological and pathological relevance in the macrophage inflammatory response. [BMB Reports 2022; 55(11): 519-527].
Collapse
Affiliation(s)
- Doyoung Park
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 35015, Korea
| | - Gyumin Lim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 35015, Korea
| | - Sung-Jin Yoon
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Hyon-Seung Yi
- Department of Internal Medicine and Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Dong Wook Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 35015, Korea
| |
Collapse
|
215
|
Kobayashi H. Recent advances in understanding the metabolic plasticity of ovarian cancer: A systematic review. Heliyon 2022; 8:e11487. [PMID: 36406733 PMCID: PMC9668530 DOI: 10.1016/j.heliyon.2022.e11487] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/03/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a gynecologic malignancy with a poor prognosis due to resistance to first-line chemotherapeutic agents. Some cancer cells are primarily dependent on glycolysis, but others favor mitochondrial oxidative phosphorylation (OXPHOS) over glycolysis. Changes in metabolic reprogramming have been reported to be involved in cancer cell survival. In this review, we summarize the metabolic profiles (e.g., metabolic heterogeneity, plasticity, and reprogramming) and adaptation to the dynamic tumor microenvironment and discuss potential novel therapeutic strategies. A literature search was performed between January 2000 and March 2022 in the PubMed and Google Scholar databases using a combination of specific terms. Ovarian cancer cells, including cancer stem cells, depend on glycolysis, OXPHOS, or both for survival. Several environmental stresses, such as nutrient starvation or glucose deprivation, hypoxic stress, acidification, and excessive reactive oxygen species (ROS) generation, reprogram the metabolic pathways to adapt. The interaction between tumors and adjacent stromal cells allows cancer cells to enhance mitochondrial energy metabolism. The metabolic reprogramming varies depending on genomic and epigenetic alterations of metabolism-related genes and the metabolic environment. Developing accurate and non-invasive methods for early identification of metabolic alterations could facilitate optimal cancer diagnosis and treatment. Cancer metabolism research has entered an exciting era where novel strategies targeting metabolic profiling will become more innovative.
Collapse
|
216
|
Paul S, Ghosh S, Kumar S. Tumor glycolysis, an essential sweet tooth of tumor cells. Semin Cancer Biol 2022; 86:1216-1230. [PMID: 36330953 DOI: 10.1016/j.semcancer.2022.09.007] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Cancer cells undergo metabolic alterations to meet the immense demand for energy, building blocks, and redox potential. Tumors show glucose-avid and lactate-secreting behavior even in the presence of oxygen, a process known as aerobic glycolysis. Glycolysis is the backbone of cancer cell metabolism, and cancer cells have evolved various mechanisms to enhance it. Glucose metabolism is intertwined with other metabolic pathways, making cancer metabolism diverse and heterogeneous, where glycolysis plays a central role. Oncogenic signaling accelerates the metabolic activities of glycolytic enzymes, mainly by enhancing their expression or by post-translational modifications. Aerobic glycolysis ferments glucose into lactate which supports tumor growth and metastasis by various mechanisms. Herein, we focused on tumor glycolysis, especially its interactions with the pentose phosphate pathway, glutamine metabolism, one-carbon metabolism, and mitochondrial oxidation. Further, we describe the role and regulation of key glycolytic enzymes in cancer. We summarize the role of lactate, an end product of glycolysis, in tumor growth, and the metabolic adaptations during metastasis. Lastly, we briefly discuss limitations and future directions to improve our understanding of glucose metabolism in cancer.
Collapse
Affiliation(s)
- Sumana Paul
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, 400076 Mumbai, India
| | - Saikat Ghosh
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Sushil Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, 400076 Mumbai, India.
| |
Collapse
|
217
|
Park D, Lim G, Yoon SJ, Yi HS, Choi DW. The role of immunomodulatory metabolites in shaping the inflammatory response of macrophages. BMB Rep 2022; 55:519-527. [PMID: 36195564 PMCID: PMC9712705 DOI: 10.5483/bmbrep.2022.55.11.128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 07/20/2023] Open
Abstract
Macrophage activation has long been implicated in a myriad of human pathophysiology, particularly in the context of the dysregulated capacities of an unleashing intracellular or/and extracellular inflammatory response. A growing number of studies have functionally coupled the macrophages' inflammatory capacities with dynamic metabolic reprogramming which occurs during activation, albeit the results have been mostly interpreted through classic metabolism point of view; macrophages take advantage of the rewired metabolism as a source of energy and for biosynthetic precursors. However, a specific subset of metabolic products, namely immune-modulatory metabolites, has recently emerged as significant regulatory signals which control inflammatory responses in macrophages and the relevant extracellular milieu. In this review, we introduce recently highlighted immuno-modulatory metabolites, with the aim of understanding their physiological and pathological relevance in the macrophage inflammatory response. [BMB Reports 2022; 55(11): 519-527].
Collapse
Affiliation(s)
- Doyoung Park
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 35015, Korea
| | - Gyumin Lim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 35015, Korea
| | - Sung-Jin Yoon
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Hyon-Seung Yi
- Department of Internal Medicine and Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Dong Wook Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 35015, Korea
| |
Collapse
|
218
|
Shi Y, Ding L, Mo C, Luo Y, Huang S, Cai S, Xia Y, Zhang X. Bladder paraganglioma, gastrointestinal stromal tumor, and SDHB germline mutation in a patient with Carney-Stratakis syndrome: A case report and literature review. Front Oncol 2022; 12:1030092. [PMID: 36387130 PMCID: PMC9650230 DOI: 10.3389/fonc.2022.1030092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/13/2022] [Indexed: 12/02/2022] Open
Abstract
Background Carney-Stratakis syndrome (CSS) is a rare dyad of paraganglioma (PGL)/pheochromocytoma (PHEO) and gastrointestinal stromal tumor (GIST). PGLs are neuroendocrine tumors of neural crest origin which are mostly found in the head, neck, and retroperitoneal space. GISTs are the most common mesenchymal tumors of the digestive tract, usually caused by KIT/PDGFRA mutations. Here, we reported a case of CSS with unusual bladder PGL and succinate dehydrogenase (SDH) deficient GIST due to a germline mutation in SDH-subunit B (SDHB) gene. Case presentation A 39-year-old female patient initially diagnosed with gastric GIST and isolated pelvic metastasis was eventually found to be CSS with bladder PGL and SDH-deficient GIST after surgery. This patient underwent resection of gastric and bladder tumors, and postoperative pathology confirmed the diagnosis of CSS. According to the next-generation sequencing (NGS), the patient carried a germline mutation in the SDHB gene, which was the cause of the disorder. The patient had no tumor recurrence with regular follow-up in 10 months. Conclusions CSS is an autosomal genetic disorder with no gender difference in incidence, and PGLs are more frequent than GISTs. SDH germline mutation is the molecular biological mechanism of CSS while the most common type is SDHB mutation. The unique mechanism of tumorigenesis including hypoxia and hypermethylation caused by SDH deficiency renders target therapy with tyrosine kinase inhibitors ineffective, therefore complete surgical resection is the optimal treatment in the absence of tumor metastases.
Collapse
Affiliation(s)
- Yihang Shi
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li Ding
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chengqiang Mo
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanji Luo
- Department of Urology Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shaoqing Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shirong Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanzhe Xia
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xinhua Zhang, ; Yanzhe Xia,
| | - Xinhua Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xinhua Zhang, ; Yanzhe Xia,
| |
Collapse
|
219
|
Zethoven M, Martelotto L, Pattison A, Bowen B, Balachander S, Flynn A, Rossello FJ, Hogg A, Miller JA, Frysak Z, Grimmond S, Fishbein L, Tischler AS, Gill AJ, Hicks RJ, Dahia PLM, Clifton-Bligh R, Pacak K, Tothill RW. Single-nuclei and bulk-tissue gene-expression analysis of pheochromocytoma and paraganglioma links disease subtypes with tumor microenvironment. Nat Commun 2022; 13:6262. [PMID: 36271074 PMCID: PMC9587261 DOI: 10.1038/s41467-022-34011-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 10/10/2022] [Indexed: 01/13/2023] Open
Abstract
Pheochromocytomas (PC) and paragangliomas (PG) are rare neuroendocrine tumors associated with autonomic nerves. Here we use single-nuclei RNA-seq and bulk-tissue gene-expression data to characterize the cellular composition of PCPG and normal adrenal tissues, refine tumor gene-expression subtypes and make clinical and genotypic associations. We confirm seven PCPG gene-expression subtypes with significant genotype and clinical associations. Tumors with mutations in VHL, SDH-encoding genes (SDHx) or MAML3-fusions are characterized by hypoxia-inducible factor signaling and neoangiogenesis. PCPG have few infiltrating lymphocytes but abundant macrophages. While neoplastic cells transcriptionally resemble mature chromaffin cells, early chromaffin and neuroblast markers are also features of some PCPG subtypes. The gene-expression profile of metastatic SDHx-related PCPG indicates these tumors have elevated cellular proliferation and a lower number of non-neoplastic Schwann-cell-like cells, while GPR139 is a potential theranostic target. Our findings therefore clarify the diverse transcriptional programs and cellular composition of PCPG and identify biomarkers of potential clinical significance.
Collapse
Affiliation(s)
| | - Luciano Martelotto
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Pattison
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Blake Bowen
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Shiva Balachander
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Aidan Flynn
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Fernando J Rossello
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Annette Hogg
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Julie A Miller
- Department of Surgery, Royal Melbourne Hospital, Parkville, VIC, Australia.,Department of Surgery, Epworth Hospital, Richmond, VIC, Australia
| | - Zdenek Frysak
- 3rd Department of Internal Medicine - Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Sean Grimmond
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Lauren Fishbein
- Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, University of Colorado, Aurora, CO, USA
| | | | - Anthony J Gill
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia.,NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Rodney J Hicks
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Patricia L M Dahia
- Div. Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, USA
| | - Roderick Clifton-Bligh
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Richard W Tothill
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
220
|
Goretzki A, Lin YJ, Zimmermann J, Rainer H, Junker AC, Wolfheimer S, Vieths S, Scheurer S, Schülke S. Role of Glycolysis and Fatty Acid Synthesis in the Activation and T Cell-Modulating Potential of Dendritic Cells Stimulated with a TLR5-Ligand Allergen Fusion Protein. Int J Mol Sci 2022; 23:ijms232012695. [PMID: 36293550 PMCID: PMC9604253 DOI: 10.3390/ijms232012695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022] Open
Abstract
Trained immune responses, based on metabolic and epigenetic changes in innate immune cells, are de facto innate immune memory and, therefore, are of great interest in vaccine development. In previous studies, the recombinant fusion protein rFlaA:Betv1, combining the adjuvant and toll-like receptor (TLR)5-ligand flagellin (FlaA) and the major birch pollen allergen Bet v 1 into a single molecule, significantly suppressed allergic sensitization in vivo while also changing the metabolism of myeloid dendritic cells (mDCs). Within this study, the immune-metabolic effects of rFlaA:Betv1 during mDC activation were elucidated. In line with results for other well-characterized TLR-ligands, rFlaA:Betv1 increased glycolysis while suppressing oxidative phosphorylation to different extents, making rFlaA:Betv1 a suitable model to study the immune-metabolic effects of TLR-adjuvanted vaccines. In vitro pretreatment of mDCs with cerulenin (inhibitor of fatty acid biosynthesis) led to a decrease in both rFlaA:Betv1-induced anti-inflammatory cytokine Interleukin (IL) 10 and T helper cell type (TH) 1-related cytokine IL-12p70, while the pro-inflammatory cytokine IL 1β was unaffected. Interestingly, pretreatment with the glutaminase inhibitor BPTES resulted in an increase in IL-1β, but decreased IL-12p70 secretion while leaving IL-10 unchanged. Inhibition of the glycolytic enzyme hexokinase-2 by 2-deoxyglucose led to a decrease in all investigated cytokines (IL-10, IL-12p70, and IL-1β). Inhibitors of mitochondrial respiration had no effect on rFlaA:Betv1-induced IL-10 level, but either enhanced the secretion of IL-1β (oligomycin) or decreased IL-12p70 (antimycin A). In extracellular flux measurements, mDCs showed a strongly enhanced glycolysis after rFlaA:Betv1 stimulation, which was slightly increased after respiratory shutdown using antimycin A. rFlaA:Betv1-stimulated mDCs secreted directly antimicrobial substances in a mTOR- and fatty acid metabolism-dependent manner. In co-cultures of rFlaA:Betv1-stimulated mDCs with CD4+ T cells, the suppression of Bet v 1-specific TH2 responses was shown to depend on fatty acid synthesis. The effector function of rFlaA:Betv1-activated mDCs mainly relies on glycolysis, with fatty acid synthesis also significantly contributing to rFlaA:Betv1-mediated cytokine secretion, the production of antimicrobial molecules, and the modulation of T cell responses.
Collapse
|
221
|
Aird R, Wills J, Roby KF, Bénézech C, Stimson RH, Wabitsch M, Pollard JW, Finch A, Michailidou Z. Hypoxia-driven metabolic reprogramming of adipocytes fuels cancer cell proliferation. Front Endocrinol (Lausanne) 2022; 13:989523. [PMID: 36329893 PMCID: PMC9623062 DOI: 10.3389/fendo.2022.989523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/15/2022] [Indexed: 12/05/2022] Open
Abstract
Objective Obesity increases the risk of certain cancers, especially tumours that reside close to adipose tissue (breast and ovarian metastasis in the omentum). The obesogenic and tumour micro-environment share a common pathogenic feature, oxygen deprivation (hypoxia). Here we test how hypoxia changes the metabolome of adipocytes to assist cancer cell growth. Methods Human and mouse breast and ovarian cancer cell lines were co-cultured with human and mouse adipocytes respectively under normoxia or hypoxia. Proliferation and lipid uptake in cancer cells were measured by commercial assays. Metabolite changes under normoxia or hypoxia were measured in the media of human adipocytes by targeted LC/MS. Results Hypoxic cancer-conditioned media increased lipolysis in both human and mouse adipocytes. This led to increased transfer of lipids to cancer cells and consequent increased proliferation under hypoxia. These effects were dependent on HIF1α expression in adipocytes, as mouse adipocytes lacking HIF1α showed blunted responses under hypoxic conditions. Targeted metabolomics of the human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes media revealed that culture with hypoxic-conditioned media from non-malignant mammary epithelial cells (MCF10A) can alter the adipocyte metabolome and drive proliferation of the non-malignant cells. Conclusion Here, we show that hypoxia in the adipose-tumour microenvironment is the driving force of the lipid uptake in both mammary and ovarian cancer cells. Hypoxia can modify the adipocyte metabolome towards accelerated lipolysis, glucose deprivation and reduced ketosis. These metabolic shifts in adipocytes could assist both mammary epithelial and cancer cells to bypass the inhibitory effects of hypoxia on proliferation and thrive.
Collapse
Affiliation(s)
- R. Aird
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, Edinburgh University, Edinburgh, United Kingdom
| | - J. Wills
- MRC Institute of Genetics and Molecular Medicine, Edinburgh University, Edinburgh, United Kingdom
| | - K. F. Roby
- University of Kansas Medical Center, Kansas City, Kansas, KS, United States
| | - C. Bénézech
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, Edinburgh University, Edinburgh, United Kingdom
| | - R. H. Stimson
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, Edinburgh University, Edinburgh, United Kingdom
| | - M. Wabitsch
- University Medical Center Department of Pediatrics and Adolescent Medicine, Ulm, Germany
| | - J. W. Pollard
- Medical Research Council (MRC) Centre for Reproductive Health, Edinburgh University, Edinburgh, United Kingdom
| | - A. Finch
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Z. Michailidou
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, Edinburgh University, Edinburgh, United Kingdom
| |
Collapse
|
222
|
Wang X, Xu B, Du J, Xia J, Lei G, Zhou C, Hu J, Zhang Y, Chen S, Shao F, Yang J, Li Y. Characterization of pyruvate metabolism and citric acid cycle patterns predicts response to immunotherapeutic and ferroptosis in gastric cancer. Cancer Cell Int 2022; 22:317. [PMID: 36229828 PMCID: PMC9563156 DOI: 10.1186/s12935-022-02739-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/28/2022] [Indexed: 11/27/2022] Open
Abstract
Background Gastric cancer is one of the most common malignancies of the digestive system with a high lethal rate. Studies have shown that inherited and acquired mutations in pyruvate metabolism and citric acid cycle (P-CA) enzymes are involved in tumorigenesis and tumor development. However, it is unclear how different P-CA patterns affect the tumor microenvironment (TME), which is critical for cancer progression. Methods This study mainly concentrated on investigating the role of the P-CA patterns in multicellular immune cell infiltration of GC TME. First, the expression levels of P-CA regulators were profiled in GC samples from The Cancer Genome Atlas and Gene Expression Omnibus cohorts to construct a consensus clustering analysis and identify three distinct P-CA clusters. GSVA was conducted to reveal the different biological processes in three P-CA clusters. Subsequently, 1127 cluster-related differentially expressed genes were identified, and prognostic-related genes were screened using univariate Cox regression analysis. A scoring system was then set up to quantify the P-CA gene signature and further evaluate the response of the patients to the immunotherapy. Results We found that GC patients in the high P-CA score group had a higher tumor mutational burden, higher microsatellite instability, and better prognosis. The opposite was observed in the low P-CA score group. Interestingly, we demonstrated P-CA gene cluster could predict the sensitivity to immunotherapy and ferroptosis-induced therapy. Conclusion Collectively, the P-CA gene signature in this study exhibits potential roles in the tumor microenvironment and predicts the response to immunotherapeutic. The identification of these P-CA patterns may significantly accelerate the strategic development of immunotherapy for GC. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02739-z.
Collapse
Affiliation(s)
- Xu Wang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, 610072, Sichuan, China.,Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
| | - Bing Xu
- Department of Clinical Laboratory, Hangzhou Women's Hospital, Hangzhou, 310005, Zhejiang, China
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
| | - Guojie Lei
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
| | - Chaoting Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
| | - Jiayu Hu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
| | - Yinhao Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
| | - Sufeng Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
| | - Fangchun Shao
- Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China.
| | - Jiyun Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, 610072, Sichuan, China.
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
223
|
Pladevall-Morera D, Zylicz JJ. Chromatin as a sensor of metabolic changes during early development. Front Cell Dev Biol 2022; 10:1014498. [PMID: 36299478 PMCID: PMC9588933 DOI: 10.3389/fcell.2022.1014498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular metabolism is a complex network of biochemical reactions fueling development with energy and biomass; however, it can also shape the cellular epigenome. Indeed, some intermediates of metabolic reactions exert a non-canonical function by acting as co-factors, substrates or inhibitors of chromatin modifying enzymes. Therefore, fluctuating availability of such molecules has the potential to regulate the epigenetic landscape. Thanks to this functional coupling, chromatin can act as a sensor of metabolic changes and thus impact cell fate. Growing evidence suggest that both metabolic and epigenetic reprogramming are crucial for ensuring a successful embryo development from the zygote until gastrulation. In this review, we provide an overview of the complex relationship between metabolism and epigenetics in regulating the early stages of mammalian embryo development. We report on recent breakthroughs in uncovering the non-canonical functions of metabolism especially when re-localized to the nucleus. In addition, we identify the challenges and outline future perspectives to advance the novel field of epi-metabolomics especially in the context of early development.
Collapse
Affiliation(s)
| | - Jan J. Zylicz
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
224
|
Krycer JR, Nayler SP. A Survey of the Metabolic Landscape of the Developing Cerebellum at Single-Cell Resolution. CEREBELLUM (LONDON, ENGLAND) 2022; 21:838-850. [PMID: 35767214 DOI: 10.1007/s12311-022-01415-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
The use of cell-culture models to investigate development and disease of the cerebellum is a recent advance, facilitated by the discovery that patterning of precursors is capable of giving rise to cells with specific neuronal identity. Pluripotent stem cell-derived organoids, which exhibit self-organisational characteristics reminiscent of early cerebellar tissue, present a number of challenges including recapitulation of conditions resembling the mature brain. An understanding of the processes driving fetal and postnatal maturation is required to reproduce these conditions in vitro and advance the capability of the system to model adult-onset disease. A key tool for achieving this is single-cell RNA sequencing, which enables visualisation of key transcriptional features of subpopulations comprising tissues. Here, we explore and compare available single-cell RNA sequencing data derived from the developing human cerebellum and its synthetic, in vitro counterpart (stem cell-derived cerebellar organoids). We focus on performing a qualitative assessment of the expression of key metabolic pathway genes, given recent findings exemplifying tissue-specific metabolic activity, including hypoxia and metabolic shifts associated with neuronal expansion. Signatures indicative of known cell type-specific metabolic differences, such as the astrocyte-neuron lactate shuttle and glutamate-glutamine cycle were evident at a transcriptional level. Cerebellar tissue and cerebellar organoids showed a number of behavioural similarities, including HIF1 signalling, which may serve to drive expansion of granule cell progenitors in both settings. We further highlight numerous differences between cultured organoids and native tissue which may provide clarity on the state of metabolic state following differentiation of organoids, providing the future framework to test and further hypotheses regarding promoting maturation. Overall, this analysis provides insight into understanding the state of in vitro models of the cerebellum, a critical factor required for modelling susceptibility of various cell types to cerebellar disease.
Collapse
Affiliation(s)
- James R Krycer
- Queensland Institute of Medical Research Berghofer Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Sam P Nayler
- Queensland Institute of Medical Research Berghofer Research Institute, Herston, QLD, Australia.
| |
Collapse
|
225
|
Loughrey PB, Roncaroli F, Healy E, Weir P, Basetti M, Casey RT, Hunter SJ, Korbonits M. Succinate dehydrogenase and MYC-associated factor X mutations in pituitary neuroendocrine tumours. Endocr Relat Cancer 2022; 29:R157-R172. [PMID: 35938916 PMCID: PMC9513646 DOI: 10.1530/erc-22-0157] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/05/2022] [Indexed: 11/28/2022]
Abstract
Pituitary neuroendocrine tumours (PitNETs) associated with paragangliomas or phaeochromocytomas are rare. SDHx variants are estimated to be associated with 0.3-1.8% of PitNETs. Only a few case reports have documented the association with MAX variants. Prolactinomas are the most common PitNETs occurring in patients with SDHx variants, followed by somatotrophinomas, clinically non-functioning tumours and corticotrophinomas. One pituitary carcinoma has been described. SDHC, SDHB and SDHA mutations are inherited in an autosomal dominant fashion and tumorigenesis seems to adhere to Knudson's two-hit hypothesis. SDHD and SDHAF2 mutations most commonly have paternal inheritance. Immunohistochemistry for SDHB or MAX and loss of heterozygosity analysis can support the assessment of pathogenicity of the variants. Metabolomics is promising in the diagnosis of SDHx-related disease. Future research should aim to further clarify the role of SDHx and MAX variants or other genes in the molecular pathogenesis of PitNETs, including pseudohypoxic and kinase signalling pathways along with elucidating epigenetic mechanisms to predict tumour behaviour.
Collapse
Affiliation(s)
- Paul Benjamin Loughrey
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast, UK
- Regional Centre for Endocrinology and Diabetes, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Division of Neuroscience and Experimental Psychology, School of Medicine, Manchester University, Manchester, UK
| | - Estelle Healy
- Department of Cellular Pathology, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
| | - Philip Weir
- Department of Neurosurgery, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
| | - Madhu Basetti
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Ruth T Casey
- Department of Endocrinology, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| | - Steven J Hunter
- Regional Centre for Endocrinology and Diabetes, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
226
|
He Y, Huang B, Yang Y, Song W, Fan Y, Zhang L, Liu G. MicroRNA‐16‐5p exacerbates sepsis by upregulating aerobic glycolysis via SIRT3‐SDHA axis. Cell Biol Int 2022; 46:2207-2219. [DOI: 10.1002/cbin.11908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/26/2022] [Accepted: 09/03/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Yue‐Xian He
- Department of Pediatrics The Fifth Affiliated Hospital of Zunyi Medical University Zhuhai Guangdong People's Republic of China
- Department of Pediatrics The First Affiliated Hospital of Jinan University Guangzhou Guangdong People's Republic of China
| | - Bo‐Lun Huang
- Department of PICU Guangzhou Women and Children's Medical Center Guangzhou Guangdong People's Republic of China
| | - Yi‐Yu Yang
- Department of PICU Guangzhou Women and Children's Medical Center Guangzhou Guangdong People's Republic of China
| | - Wen‐Xiu Song
- Department of Pediatrics The Fifth Affiliated Hospital of Zunyi Medical University Zhuhai Guangdong People's Republic of China
| | - Yong‐Bo Fan
- Department of Pediatrics The Fifth Affiliated Hospital of Zunyi Medical University Zhuhai Guangdong People's Republic of China
| | - Li‐Mei Zhang
- Department of Pediatrics The Fifth Affiliated Hospital of Zunyi Medical University Zhuhai Guangdong People's Republic of China
| | - Guo‐Sheng Liu
- Department of Pediatrics The First Affiliated Hospital of Jinan University Guangzhou Guangdong People's Republic of China
| |
Collapse
|
227
|
Mitochondrial Regulation of the Hypoxia-Inducible Factor in the Development of Pulmonary Hypertension. J Clin Med 2022; 11:jcm11175219. [PMID: 36079149 PMCID: PMC9457092 DOI: 10.3390/jcm11175219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary hypertension (PH) is a severe progressive lung disorder characterized by pulmonary vasoconstriction and vascular remodeling, culminating in right-sided heart failure and increased mortality. Data from animal models and human subjects demonstrated that hypoxia-inducible factor (HIF)-related signaling is essential in the progression of PH. This review summarizes the regulatory pathways and mechanisms of HIF-mediated signaling, emphasizing the role of mitochondria in HIF regulation and PH pathogenesis. We also try to determine the potential to therapeutically target the components of the HIF system for the management of PH.
Collapse
|
228
|
Wang M, Pang Y, Guo Y, Tian L, Liu Y, Shen C, Liu M, Meng Y, Cai Z, Wang Y, Zhao W. Metabolic reprogramming: A novel therapeutic target in diabetic kidney disease. Front Pharmacol 2022; 13:970601. [PMID: 36120335 PMCID: PMC9479190 DOI: 10.3389/fphar.2022.970601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic kidney disease (DKD) is one of the most common microvascular complications of diabetes mellitus. However, the pathological mechanisms contributing to DKD are multifactorial and poorly understood. Diabetes is characterized by metabolic disorders that can bring about a series of changes in energy metabolism. As the most energy-consuming organs secondary only to the heart, the kidneys must maintain energy homeostasis. Aberrations in energy metabolism can lead to cellular dysfunction or even death. Metabolic reprogramming, a shift from mitochondrial oxidative phosphorylation to glycolysis and its side branches, is thought to play a critical role in the development and progression of DKD. This review focuses on the current knowledge about metabolic reprogramming and the role it plays in DKD development. The underlying etiologies, pathological damages in the involved cells, and potential molecular regulators of metabolic alterations are also discussed. Understanding the role of metabolic reprogramming in DKD may provide novel therapeutic approaches to delay its progression to end-stage renal disease.
Collapse
|
229
|
Lee MH, Sanders L, Kumar R, Hernandez-Saavedra D, Yun X, Ford JA, Perez MJ, Mickael C, Gandjeva A, Koyanagi DE, Harral JW, Irwin DC, Kassa B, Eckel RH, Shimoda LA, Graham BB, Tuder RM. Contribution of fatty acid oxidation to the pathogenesis of pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2022; 323:L355-L371. [PMID: 35763400 PMCID: PMC9448289 DOI: 10.1152/ajplung.00039.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/05/2022] [Accepted: 06/25/2022] [Indexed: 11/22/2022] Open
Abstract
Dysregulated metabolism characterizes both animal and human forms of pulmonary hypertension (PH). Enzymes involved in fatty acid metabolism have previously not been assessed in human pulmonary arteries affected by pulmonary arterial hypertension (PAH), and how inhibition of fatty acid oxidation (FAO) may attenuate PH remains unclear. Fatty acid metabolism gene transcription was quantified in laser-dissected pulmonary arteries from 10 explanted lungs with advanced PAH (5 idiopathic, 5 associated with systemic sclerosis), and 5 donors without lung diseases. Effects of oxfenicine, a FAO inhibitor, on female Sugen 5416-chronic hypoxia (SuHx) rats were studied in vivo using right heart catheterization, and ex vivo using perfused lungs and pulmonary artery ring segments. The impact of pharmacologic (oxfenicine) and genetic (carnitine palmitoyltransferase 1a heterozygosity) FAO suppression was additionally probed in mouse models of Schistosoma and hypoxia-induced PH. Potential mechanisms underlying FAO-induced PH pathogenesis were examined by quantifying ATP and mitochondrial mass in oxfenicine-treated SuHx pulmonary arterial cells, and by assessing pulmonary arterial macrophage infiltration with immunohistochemistry. We found upregulated pulmonary arterial transcription of 26 and 13 FAO genes in idiopathic and systemic sclerosis-associated PAH, respectively. In addition to promoting de-remodeling of pulmonary arteries in SuHx rats, oxfenicine attenuated endothelin-1-induced vasoconstriction. FAO inhibition also conferred modest benefit in the two mouse models of PH. Oxfenicine increased mitochondrial mass in cultured rat pulmonary arterial cells, and decreased the density of perivascular macrophage infiltration in pulmonary arteries of treated SuHx rats. In summary, FAO inhibition attenuated experimental PH, and may be beneficial in human PAH.
Collapse
Affiliation(s)
- Michael H Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Linda Sanders
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Rahul Kumar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Daniel Hernandez-Saavedra
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Joshay A Ford
- University of Colorado School of Medicine, Aurora, Colorado
| | - Mario J Perez
- Department of Psychiatry, University of Colorado, Aurora, Colorado
| | - Claudia Mickael
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Aneta Gandjeva
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Daniel E Koyanagi
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Julie W Harral
- Cardiovascular Pulmonary Research Laboratory, Department of Pediatrics and Medicine, University of Colorado, Aurora, Colorado
| | - David C Irwin
- Cardiovascular Pulmonary Research Laboratory, Department of Pediatrics and Medicine, University of Colorado, Aurora, Colorado
| | - Biruk Kassa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Robert H Eckel
- Division of Endocrinology, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Brian B Graham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Rubin M Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
230
|
Yoo A, Tang C, Zucker M, Fitzgerald K, DiNatale RG, Rappold PM, Weiss K, Freeman B, Lee CH, Schultz N, Motzer R, Russo P, Coleman J, Reuter VE, Chen YB, Carlo MI, Gill AJ, Kotecha RR, Hakimi AA, Reznik E. Genomic and Metabolic Hallmarks of SDH- and FH-deficient Renal Cell Carcinomas. Eur Urol Focus 2022; 8:1278-1288. [PMID: 35288096 PMCID: PMC9464266 DOI: 10.1016/j.euf.2021.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/09/2021] [Accepted: 12/01/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Succinate dehydrogenase-deficient and fumarate hydratase-deficient renal cell carcinomas (SDHRCC and FHRCC) are rare kidney cancers driven by loss of TCA cycle enzymes. OBJECTIVE To define and compare the genomic and metabolomic hallmarks of SDHRCC and FHRCC. DESIGN, SETTING, AND PARTICIPANTS We analyzed SDHRCC and FHRCC tumors with either immunohistochemical evidence of loss of protein expression or genomically confirmed biallelic inactivation of SDHA/B/C/D/AF2 or FH. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Somatic alterations were identified using clinical pipelines, with allele-specific copy number alterations (CNAs) identified using FACETS. Mass spectrometry-based metabolomic profiling was performed on available SDHRCC and FHRCC tumors. RESULTS AND LIMITATIONS Tumors were analyzed for 42 patients (25 FHRCC, 17 SDHRCC). In the germline analysis, 16/17 SDHRCCs harbored a germline alteration in SDHB, whereas only 17/22 FHRCCs had pathogenic germline FH variants. SDHRCCs had a lower mutation burden (p = 0.02) and CNA burden (p = 0.0002) than FHRCCs. All SDHRCCs presented with deletion of chromosome 1p (overlapping SDHB), whereas FHRCCs demonstrated high but not ubiquitous loss of 1q (FH locus). Both SDHRCCs and FHRCCs exhibited significant idiopathic accumulation of the metabolite guanine. FHRCC tumors had elevated levels of urea cycle metabolites (argininosuccinate, citrulline, and fumarate), whereas SDHRCC tumors had elevation of numerous acylcarnitines. These characteristic metabolic changes allowed identification of a previously unrecognized SDH-deficient RCC. CONCLUSIONS Despite sharing similar genetic etiology, SDHRCC and FHRCC represent distinct molecular entities with unique genetic and metabolic abnormalities. PATIENT SUMMARY Kidney cancers driven by loss of the gene encoding either the succinate dehydrogenase or fumarate hydratase enzyme are rare. We sought to define and compare the genetic and metabolic features of these cancer entities.
Collapse
Affiliation(s)
- Angela Yoo
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA,SUNY Downstate Health Sciences University, Brooklyn, NY, USA,Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cerise Tang
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Mark Zucker
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelly Fitzgerald
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renzo G. DiNatale
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Phillip M. Rappold
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kate Weiss
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Freeman
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chung-Han Lee
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nikolaus Schultz
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Motzer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul Russo
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan Coleman
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Victor E. Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria I. Carlo
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anthony J. Gill
- Sydney Medical School, University of Sydney, Sydney, Australia,Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St. Leonards, Australia,NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St. Leonards, Australia
| | - Ritesh R. Kotecha
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Corresponding authors. Memorial Sloan Kettering Cancer Center, New York, NY, USA. (R.R. Kotecha), (A. Ari Hakimi), (E. Reznik)
| | - A. Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Corresponding authors. Memorial Sloan Kettering Cancer Center, New York, NY, USA. (R.R. Kotecha), (A. Ari Hakimi), (E. Reznik)
| | - Ed Reznik
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Corresponding authors. Memorial Sloan Kettering Cancer Center, New York, NY, USA. (R.R. Kotecha), (A. Ari Hakimi), (E. Reznik)
| |
Collapse
|
231
|
Ding D, Zhang J, Du B, Wang X, Hou L, Guo S, Chen B, Kang L. Non-canonical function of an Hif-1α splice variant contributes to the sustained flight of locusts. eLife 2022; 11:74554. [PMID: 36039636 PMCID: PMC9427102 DOI: 10.7554/elife.74554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 07/11/2022] [Indexed: 12/30/2022] Open
Abstract
The hypoxia inducible factor (Hif) pathway is functionally conserved across metazoans in modulating cellular adaptations to hypoxia. However, the functions of this pathway under aerobic physiological conditions are rarely investigated. Here, we show that Hif-1α2, a locust Hif-1α isoform, does not induce canonical hypoxic responses but functions as a specific regulator of locust flight, which is a completely aerobic physiological process. Two Hif-1α splice variants were identified in locusts, a ubiquitously expressed Hif-1α1 and a muscle-predominantly expressed Hif-1α2. Hif-1α1 that induces typical hypoxic responses upon hypoxia exposure remains inactive during flight. By contrast, the expression of Hif-1α2, which lacks C-terminal transactivation domain, is less sensitive to oxygen tension but induced extensively by flying. Hif-1α2 regulates physiological processes involved in glucose metabolism and antioxidation during flight and sustains flight endurance by maintaining redox homeostasis through upregulating the production of a reactive oxygen species (ROS) quencher, DJ-1. Overall, this study reveals a novel Hif-mediated mechanism underlying prolonged aerobic physiological activity.
Collapse
Affiliation(s)
- Ding Ding
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jie Zhang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Baozhen Du
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Xuanzhao Wang
- School of Life Science, Hebei University, Baoding, China
| | - Li Hou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Siyuan Guo
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Bing Chen
- School of Life Science, Hebei University, Baoding, China
| | - Le Kang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China.,School of Life Science, Hebei University, Baoding, China
| |
Collapse
|
232
|
Hanse EA, Kong M. A happy cell stays home: When metabolic stress creates epigenetic advantages in the tumor microenvironment. Front Oncol 2022; 12:962928. [PMID: 36091163 PMCID: PMC9459228 DOI: 10.3389/fonc.2022.962928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
A paradox of fast-proliferating tumor cells is that they deplete extracellular nutrients that often results in a nutrient poor microenvironment in vivo. Having a better understanding of the adaptation mechanisms cells exhibit in response to metabolic stress will open new therapeutic windows targeting the tumor’s extreme nutrient microenvironment. Glutamine is one of the most depleted amino acids in the tumor core and here, we provide insight into how important glutamine and its downstream by-product, α-ketoglutarate (αKG), are to communicating information about the nutrient environment. This communication is key in the cell’s ability to foster adaptation. We highlight the epigenetic changes brought on when αKG concentrations are altered in cancer and discuss how depriving cells of glutamine may lead to cancer cell de-differentiation and the ability to grow and thrive in foreign environments. When we starve cells, they adapt to survive. Those survival “skills” allow them to go out looking for other places to live and metastasize. We further examine current challenges to modelling the metabolic tumor microenvironment in the laboratory and discuss strategies that consider current findings to target the tumor’s poor nutrient microenvironment.
Collapse
|
233
|
Heinz A, Nonnenmacher Y, Henne A, Khalil MA, Bejkollari K, Dostert C, Hosseini S, Goldmann O, He W, Palorini R, Verschueren C, Korte M, Chiaradonna F, Medina E, Brenner D, Hiller K. Itaconate controls its own synthesis via feedback-inhibition of reverse TCA cycle activity at IDH2. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166530. [PMID: 36038039 DOI: 10.1016/j.bbadis.2022.166530] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022]
Abstract
Macrophages undergo extensive metabolic reprogramming during classical pro-inflammatory polarization (M1-like). The accumulation of itaconate has been recognized as both a consequence and mediator of the inflammatory response. In this study we first examined the specific functions of itaconate inside fractionated mitochondria. We show that M1 macrophages produce itaconate de novo via aconitase decarboxylase 1 (ACOD1) inside mitochondria. The carbon for this reaction is not only supplied by oxidative TCA cycling, but also through the reductive carboxylation of α-ketoglutarate by isocitrate dehydrogenase (IDH). While macrophages are capable of sustaining a certain degree of itaconate production during hypoxia by augmenting the activity of IDH-dependent reductive carboxylation, we demonstrate that sufficient itaconate synthesis requires a balance of reductive and oxidative TCA cycle metabolism in mouse macrophages. In comparison, human macrophages increase itaconate accumulation under hypoxic conditions by augmenting reductive carboxylation activity. We further demonstrated that itaconate attenuates reductive carboxylation at IDH2, restricting its own production and the accumulation of the immunomodulatory metabolites citrate and 2-hydroxyglutarate. In line with this, reductive carboxylation is enhanced in ACOD1-depleted macrophages. Mechanistically, the inhibition of IDH2 by itaconate is linked to the alteration of the mitochondrial NADP+/NADPH ratio and competitive succinate dehydrogenase inhibition. Taken together, our findings extend the current model of TCA cycle reprogramming during pro-inflammatory macrophage activation and identified novel regulatory properties of itaconate.
Collapse
Affiliation(s)
- Alexander Heinz
- Department for Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Yannic Nonnenmacher
- Department for Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Antonia Henne
- Department for Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Michelle-Amirah Khalil
- Department for Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Ketlin Bejkollari
- Department for Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Catherine Dostert
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Germany
| | - Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Wei He
- Department for Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Roberta Palorini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, Milan, Italy
| | - Charlène Verschueren
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Martin Korte
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Germany; Neuroinflammation and Neurodegeneration Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ferdinando Chiaradonna
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, Milan, Italy
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology and Genetics, Luxembourg Centre for System Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Karsten Hiller
- Department for Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany.
| |
Collapse
|
234
|
Jiang M, Chen ZG, Li H, Zhang TT, Yang MJ, Peng XX, Peng B. Succinate and inosine coordinate innate immune response to bacterial infection. PLoS Pathog 2022; 18:e1010796. [PMID: 36026499 PMCID: PMC9455851 DOI: 10.1371/journal.ppat.1010796] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 09/08/2022] [Accepted: 08/04/2022] [Indexed: 12/17/2022] Open
Abstract
Macrophages restrict bacterial infection partly by stimulating phagocytosis and partly by stimulating release of cytokines and complement components. Here, we treat macrophages with LPS and a bacterial pathogen, and demonstrate that expression of cytokine IL-1β and bacterial phagocytosis increase to a transient peak 8 to 12 h post-treatment, while expression of complement component 3 (C3) continues to rise for 24 h post-treatment. Metabolomic analysis suggests a correlation between the cellular concentrations of succinate and IL-1β and of inosine and C3. This may involve a regulatory feedback mechanism, whereby succinate stimulates and inosine inhibits HIF-1α through their competitive interactions with prolyl hydroxylase. Furthermore, increased level of inosine in LPS-stimulated macrophages is linked to accumulation of adenosine monophosphate and that exogenous inosine improves the survival of bacterial pathogen-infected mice and tilapia. The implications of these data suggests potential therapeutic tools to prevent, manage or treat bacterial infections.
Collapse
Affiliation(s)
- Ming Jiang
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Higher Education Mega Center, Guangzhou, People’s Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, People’s Republic of China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, People’s Republic of China
| | - Zhuang-gui Chen
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Higher Education Mega Center, Guangzhou, People’s Republic of China
| | - Hui Li
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Higher Education Mega Center, Guangzhou, People’s Republic of China
| | - Tian-tuo Zhang
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Higher Education Mega Center, Guangzhou, People’s Republic of China
| | - Man-jun Yang
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Higher Education Mega Center, Guangzhou, People’s Republic of China
| | - Xuan-xian Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Higher Education Mega Center, Guangzhou, People’s Republic of China
| | - Bo Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Higher Education Mega Center, Guangzhou, People’s Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, People’s Republic of China
- * E-mail:
| |
Collapse
|
235
|
Coope A, Ghanameh Z, Kingston O, Sheridan CM, Barrett-Jolley R, Phelan MM, Oldershaw RA. 1H NMR Metabolite Monitoring during the Differentiation of Human Induced Pluripotent Stem Cells Provides New Insights into the Molecular Events That Regulate Embryonic Chondrogenesis. Int J Mol Sci 2022; 23:ijms23169266. [PMID: 36012540 PMCID: PMC9409419 DOI: 10.3390/ijms23169266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
The integration of cell metabolism with signalling pathways, transcription factor networks and epigenetic mediators is critical in coordinating molecular and cellular events during embryogenesis. Induced pluripotent stem cells (IPSCs) are an established model for embryogenesis, germ layer specification and cell lineage differentiation, advancing the study of human embryonic development and the translation of innovations in drug discovery, disease modelling and cell-based therapies. The metabolic regulation of IPSC pluripotency is mediated by balancing glycolysis and oxidative phosphorylation, but there is a paucity of data regarding the influence of individual metabolite changes during cell lineage differentiation. We used 1H NMR metabolite fingerprinting and footprinting to monitor metabolite levels as IPSCs are directed in a three-stage protocol through primitive streak/mesendoderm, mesoderm and chondrogenic populations. Metabolite changes were associated with central metabolism, with aerobic glycolysis predominant in IPSC, elevated oxidative phosphorylation during differentiation and fatty acid oxidation and ketone body use in chondrogenic cells. Metabolites were also implicated in the epigenetic regulation of pluripotency, cell signalling and biosynthetic pathways. Our results show that 1H NMR metabolomics is an effective tool for monitoring metabolite changes during the differentiation of pluripotent cells with implications on optimising media and environmental parameters for the study of embryogenesis and translational applications.
Collapse
Affiliation(s)
- Ashley Coope
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
- Clinical Directorate Professional Services, Aintree University Hospital, Liverpool University Hospitals NHS Foundation Trust, Lower Lane, Liverpool L9 7AL, UK
| | - Zain Ghanameh
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Olivia Kingston
- Department of Eye and Vision Sciences, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Carl M. Sheridan
- Department of Eye and Vision Sciences, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Richard Barrett-Jolley
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Marie M. Phelan
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Biosciences Building, Crown Street, Liverpool L7 7BE, UK
- High Field NMR Facility, Liverpool Shared Research Facilities (LIV-SRF), Faculty of Health and Life Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - Rachel A. Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
- Correspondence:
| |
Collapse
|
236
|
Provenzano A, Chetta M, De Filpo G, Cantini G, La Barbera A, Nesi G, Santi R, Martinelli S, Rapizzi E, Luconi M, Maggi M, Mannelli M, Ercolino T, Canu L. Novel Germline PHD2 Variant in a Metastatic Pheochromocytoma and Chronic Myeloid Leukemia, but in the Absence of Polycythemia. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58081113. [PMID: 36013579 PMCID: PMC9416477 DOI: 10.3390/medicina58081113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022]
Abstract
Background: Pheochromocytoma (Pheo) and paraganglioma (PGL) are rare tumors, mostly resulting from pathogenic variants of predisposing genes, with a genetic contribution that now stands at around 70%. Germline variants account for approximately 40%, while the remaining 30% is attributable to somatic variants. Objective: This study aimed to describe a new PHD2 (EGLN1) variant in a patient affected by metastatic Pheo and chronic myeloid leukemia (CML) without polycythemia and to emphasize the need to adopt a comprehensive next-generation sequencing (NGS) panel. Methods: Genetic analysis was carried out by NGS. This analysis was initially performed using a panel of genes known for tumor predisposition (EGLN1, EPAS1, FH, KIF1Bβ, MAX, NF1, RET, SDHA, SDHAF2, SDHB, SDHC, SDHD, TMEM127, and VHL), followed initially by SNP-CGH array, to exclude the presence of the pathogenic Copy Number Variants (CNVs) and the loss of heterozygosity (LOH) and subsequently by whole exome sequencing (WES) comparative sequence analysis of the DNA extracted from tumor fragments and peripheral blood. Results: We found a novel germline PHD2 (EGLN1) gene variant, c.153G>A, p.W51*, in a patient affected by metastatic Pheo and chronic myeloid leukemia (CML) in the absence of polycythemia. Conclusions: According to the latest guidelines, it is mandatory to perform genetic analysis in all Pheo/PGL cases regardless of phenotype. In patients with metastatic disease and no evidence of polycythemia, we propose testing for PHD2 (EGLN1) gene variants. A possible correlation between PHD2 (EGLN1) pathogenic variants and CML clinical course should be considered.
Collapse
Affiliation(s)
- Aldesia Provenzano
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Massimiliano Chetta
- Medical Genetics, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Cardarelli, Padiglione, 80131 Naples, Italy
| | - Giuseppina De Filpo
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Giulia Cantini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
| | - Andrea La Barbera
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Gabriella Nesi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Raffaella Santi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Serena Martinelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
| | - Elena Rapizzi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
| | - Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
| | - Mario Maggi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
- Endocrinology Unit, Azienda Ospedaliera-Universitaria Careggi, 50139 Florence, Italy
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
| | - Tonino Ercolino
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
- Endocrinology Unit, Azienda Ospedaliera-Universitaria Careggi, 50139 Florence, Italy
| | - Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
- Endocrinology Unit, Azienda Ospedaliera-Universitaria Careggi, 50139 Florence, Italy
- Correspondence:
| |
Collapse
|
237
|
Gudgeon N, Munford H, Bishop EL, Hill J, Fulton-Ward T, Bending D, Roberts J, Tennant DA, Dimeloe S. Succinate uptake by T cells suppresses their effector function via inhibition of mitochondrial glucose oxidation. Cell Rep 2022; 40:111193. [PMID: 35977513 PMCID: PMC9638018 DOI: 10.1016/j.celrep.2022.111193] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/26/2022] [Accepted: 07/20/2022] [Indexed: 11/03/2022] Open
Abstract
Succinate dehydrogenase (SDH) loss-of-function mutations drive succinate accumulation in tumor microenvironments, for example in the neuroendocrine tumors pheochromocytoma (PC) and paraganglioma (PG). Control of innate immune cell activity by succinate is described, but effects on T cells have not been interrogated. Here we report that exposure of human CD4+ and CD8+ T cells to tumor-associated succinate concentrations suppresses degranulation and cytokine secretion, including of the key anti-tumor cytokine interferon-γ (IFN-γ). Mechanistically, this is associated with succinate uptake-partly via the monocarboxylate transporter 1 (MCT1)-inhibition of succinyl coenzyme A synthetase activity and impaired glucose flux through the tricarboxylic acid cycle. Consistently, pharmacological and genetic interventions restoring glucose oxidation rescue T cell function. Tumor RNA-sequencing data from patients with PC and PG reveal profound suppression of IFN-γ-induced genes in SDH-deficient tumors compared with those with other mutations, supporting a role for succinate in modulating the anti-tumor immune response in vivo.
Collapse
Affiliation(s)
- Nancy Gudgeon
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Haydn Munford
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Emma L Bishop
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - James Hill
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Taylor Fulton-Ward
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - David Bending
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jennie Roberts
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sarah Dimeloe
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
238
|
Bahiraii S, Brenner M, Yan F, Weckwerth W, Heiss EH. Sulforaphane diminishes moonlighting of pyruvate kinase M2 and interleukin 1β expression in M1 (LPS) macrophages. Front Immunol 2022; 13:935692. [PMID: 35983049 PMCID: PMC9380596 DOI: 10.3389/fimmu.2022.935692] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Murine macrophages activated by the Toll-like receptor 4 agonist lipopolysaccharide (LPS) polarize to the M1 type by inducing proinflammatory marker proteins and changing their energy metabolism to increased aerobic glycolysis and reduced respiration. We here show that the aliphatic isothiocyanate sulforaphane (Sfn) diminishes M1 marker expression (IL-1β, IL-6, TNF-α, iNOS, NO, and ROS) and leads to highly energetic cells characterized by both high glycolytic and high respiratory activity as assessed by extracellular flux analysis. Focusing on a potential connection between high glycolytic activity and low IL-1β expression in M1 (LPS/Sfn) macrophages, we reveal that Sfn impedes the moonlighting function of pyruvate kinase M2 (PKM2) in M1 macrophages. Sfn limits mono/dimerization and nuclear residence of PKM2 accompanied by reduced HIF-1α levels, Stat3 phosphorylation at tyrosine 705, and IL-1β expression while preserving high levels of cytosolic PKM2 tetramer with high glycolytic enzyme activity. Sfn prevents glutathionylation of PKM2 in LPS-stimulated macrophages which may account for the reduced loss of PKM2 tetramer. Overall, we uncover PKM2 as a novel affected hub within the anti-inflammatory activity profile of Sfn.
Collapse
Affiliation(s)
- Sheyda Bahiraii
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences (VDS PhaNuSpo), University of Vienna, Vienna, Austria
| | - Martin Brenner
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences (VDS PhaNuSpo), University of Vienna, Vienna, Austria
- Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Fangfang Yan
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wolfram Weckwerth
- Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
- Molecular Systems Biology (MOSYS), Department of Functional and Evolutionary Ecology, University of Vienna, Vienna, Austria
| | - Elke H. Heiss
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
239
|
Moriconi C, Dzieciatkowska M, Roy M, D'Alessandro A, Roingeard P, Lee JY, Gibb DR, Tredicine M, McGill MA, Qiu A, La Carpia F, Francis RO, Hod EA, Thomas T, Picard M, Akpan IJ, Luckey CJ, Zimring JC, Spitalnik SL, Hudson KE. Retention of functional mitochondria in mature red blood cells from patients with sickle cell disease. Br J Haematol 2022; 198:574-586. [PMID: 35670632 PMCID: PMC9329257 DOI: 10.1111/bjh.18287] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/26/2022] [Accepted: 05/17/2022] [Indexed: 01/07/2023]
Abstract
Sickle cell disease (SCD) is an inherited blood disorder characterized by sickled red blood cells (RBCs), which are more sensitive to haemolysis and can contribute to disease pathophysiology. Although treatment of SCD can include RBC transfusion, patients with SCD have high rates of alloimmunization. We hypothesized that RBCs from patients with SCD have functionally active mitochondria and can elicit a type 1 interferon response. We evaluated blood samples from more than 100 patients with SCD and found elevated frequencies of mitochondria in reticulocytes and mature RBCs, as compared to healthy blood donors. The presence of mitochondria in mature RBCs was confirmed by flow cytometry, electron microscopy, and proteomic analysis. The mitochondria in mature RBCs were metabolically competent, as determined by enzymatic activities and elevated levels of mitochondria-derived metabolites. Metabolically-active mitochondria in RBCs may increase oxidative stress, which could facilitate and/or exacerbate SCD complications. Coculture of mitochondria-positive RBCs with neutrophils induced production of type 1 interferons, which are known to increase RBC alloimmunization rates. These data demonstrate that mitochondria retained in mature RBCs are functional and can elicit immune responses, suggesting that inappropriate retention of mitochondria in RBCs may play an underappreciated role in SCD complications and be an RBC alloimmunization risk factor.
Collapse
Affiliation(s)
- Chiara Moriconi
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Micaela Roy
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, Colorado, USA
| | - Philippe Roingeard
- INSERM U1259 and Electron Microscopy Facility, Université de Tours and CHRU de Tours, Tours, France
| | - June Young Lee
- Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David R Gibb
- Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maria Tredicine
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marlon A McGill
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York City, New York, USA
| | - Annie Qiu
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Francesca La Carpia
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Richard O Francis
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Eldad A Hod
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Tiffany Thomas
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York City, New York, USA
| | - Imo J Akpan
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, New York City, New York, USA
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - James C Zimring
- University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Carter Immunology Center, University of Virginia, Charlottesville, Virginia, USA
| | - Steven L Spitalnik
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Krystalyn E Hudson
- Laboratory of Transfusion Biology, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, New York, USA
| |
Collapse
|
240
|
Glycolysis and the Pentose Phosphate Pathway Promote LPS-Induced NOX2 Oxidase- and IFN-β-Dependent Inflammation in Macrophages. Antioxidants (Basel) 2022; 11:antiox11081488. [PMID: 36009206 PMCID: PMC9405479 DOI: 10.3390/antiox11081488] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages undergo a metabolic switch from oxidative phosphorylation to glycolysis when exposed to gram-negative bacterial lipopolysaccharide (LPS), which modulates antibacterial host defence mechanisms. Here, we show that LPS treatment of macrophages increased the classical oxidative burst response via the NADPH oxidase (NOX) 2 enzyme, which was blocked by 2-deoxyglucose (2-DG) inhibition of glycolysis. The inhibition of the pentose phosphate pathway with 6-aminonicotinamide (6-AN) also suppressed the LPS-induced increase in NOX2 activity and was associated with a significant reduction in the mRNA expression of NOX2 and its organizer protein p47phox. Notably, the LPS-dependent enhancement in NOX2 oxidase activity was independent of both succinate and mitochondrial reactive oxygen species (ROS) production. LPS also increased type I IFN-β expression, which was suppressed by 2-DG and 6-AN and, therefore, is dependent on glycolysis and the pentose phosphate pathway. The type I IFN-β response to LPS was also inhibited by apocynin pre-treatment, suggesting that NOX2-derived ROS promotes the TLR4-induced response to LPS. Moreover, recombinant IFN-β increased NOX2 oxidase-dependent ROS production, as well as NOX2 and p47phox expression. Our findings identify a previously undescribed molecular mechanism where both glycolysis and the pentose phosphate pathway are required to promote LPS-induced inflammation in macrophages.
Collapse
|
241
|
Baysal BE, Alahmari AA, Rodrick TC, Tabaczynski D, Curtin L, Seshadri M, Jones DR, Sexton S. Succinate dehydrogenase inversely regulates red cell distribution width and healthy lifespan in chronically hypoxic mice. JCI Insight 2022; 7:158737. [PMID: 35881479 PMCID: PMC9536274 DOI: 10.1172/jci.insight.158737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
Increased red cell distribution width (RDW), which measures erythrocyte volume (MCV) variability (anisocytosis), has been linked to early mortality in many diseases and in older adults through unknown mechanisms. Hypoxic stress has been proposed as a potential mechanism. However, experimental models to investigate the link between increased RDW and reduced survival are lacking. Here, we show that lifelong hypobaric hypoxia (~10% O2) increases erythrocyte numbers, hemoglobin and RDW, while reducing longevity in male mice. Compound heterozygous knockout (chKO) mutations in succinate dehydrogenase (Sdh; mitochondrial complex II) genes Sdhb, Sdhc and Sdhd reduce Sdh subunit protein levels, RDW, and increase healthy lifespan compared to wild-type (WT) mice in chronic hypoxia. RDW-SD, a direct measure of MCV variability, and the standard deviation of MCV (1SD-RDW) show the most statistically significant reductions in Sdh hKO mice. Tissue metabolomic profiling of 147 common metabolites shows the largest increase in succinate with elevated succinate to fumarate and succinate to oxoglutarate (2-ketoglutarate) ratios in Sdh hKO mice. These results demonstrate that mitochondrial complex II level is an underlying determinant of both RDW and healthy lifespan in hypoxia, and suggest that therapeutic targeting of Sdh might reduce high RDW-associated clinical mortality in hypoxic diseases.
Collapse
Affiliation(s)
- Bora E Baysal
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, United States of America
| | - Abdulrahman A Alahmari
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, United States of America
| | - Tori C Rodrick
- Metabolomics Core Resource Laboratory, NYU Langone Health, New York, United States of America
| | - Debra Tabaczynski
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, United States of America
| | - Leslie Curtin
- Laboratory Animal Shared Resources, Roswell Park Comprehensive Cancer Center, Buffalo, United States of America
| | - Mukund Seshadri
- Department of Dentistry and Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, United States of America
| | - Drew R Jones
- Metabolomics Core Resource Laboratory, NYU Langone Health, New York, United States of America
| | - Sandra Sexton
- Laboratory Animal Shared Resources, Roswell Park Comprehensive Cancer Center, Buffalo, United States of America
| |
Collapse
|
242
|
Bénit P, Goncalves J, El Khoury R, Rak M, Favier J, Gimenez-Roqueplo AP, Rustin P. Succinate Dehydrogenase, Succinate, and Superoxides: A Genetic, Epigenetic, Metabolic, Environmental Explosive Crossroad. Biomedicines 2022; 10:1788. [PMID: 35892689 PMCID: PMC9394281 DOI: 10.3390/biomedicines10081788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Research focused on succinate dehydrogenase (SDH) and its substrate, succinate, culminated in the 1950s accompanying the rapid development of research dedicated to bioenergetics and intermediary metabolism. This allowed researchers to uncover the implication of SDH in both the mitochondrial respiratory chain and the Krebs cycle. Nowadays, this theme is experiencing a real revival following the discovery of the role of SDH and succinate in a subset of tumors and cancers in humans. The aim of this review is to enlighten the many questions yet unanswered, ranging from fundamental to clinically oriented aspects, up to the danger of the current use of SDH as a target for a subclass of pesticides.
Collapse
Affiliation(s)
- Paule Bénit
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| | - Judith Goncalves
- Paris Centre de Recherche Cardiovasculaire (PARCC), Inserm, Université Paris Cité, F-75015 Paris, France; (J.G.); (J.F.)
| | - Riyad El Khoury
- Department of Pathology and Laboratory Medicine, Neuromuscular Diagnostic Laboratory, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
| | - Malgorzata Rak
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| | - Judith Favier
- Paris Centre de Recherche Cardiovasculaire (PARCC), Inserm, Université Paris Cité, F-75015 Paris, France; (J.G.); (J.F.)
| | - Anne-Paule Gimenez-Roqueplo
- Département de Médecine Génomique des Tumeurs et des Cancers, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, F-75015 Paris, France;
| | - Pierre Rustin
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (P.B.); (M.R.)
| |
Collapse
|
243
|
Smith ALM, Whitehall JC, Greaves LC. Mitochondrial
DNA
mutations in aging and cancer. Mol Oncol 2022; 16:3276-3294. [PMID: 35842901 PMCID: PMC9490137 DOI: 10.1002/1878-0261.13291] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/18/2022] [Accepted: 07/15/2022] [Indexed: 11/09/2022] Open
Abstract
Advancing age is a major risk factor for malignant transformation and the development of cancer. As such, over 50% of neoplasms occur in individuals over the age of 70. The pathologies of both ageing and cancer have been characterized by respective groups of molecular hallmarks, and while some features are divergent between the two pathologies, several are shared. Perturbed mitochondrial function is one such common hallmark, and this observation therefore suggests that mitochondrial alterations may be of significance in age‐related cancer development. There is now considerable evidence documenting the accumulation of somatic mitochondrial DNA (mtDNA) mutations in ageing human postmitotic and replicative tissues. Similarly, mutations of the mitochondrial genome have been reported in human cancers for decades. The plethora of functions in which mitochondria partake, such as oxidative phosphorylation, redox balance, apoptosis and numerous biosynthetic pathways, manifests a variety of ways in which alterations in mtDNA may contribute to tumour growth. However, the specific mechanisms by which mtDNA mutations contribute to tumour progression remain elusive and often contradictory. This review aims to consolidate current knowledge and describe future direction within the field.
Collapse
Affiliation(s)
- Anna LM Smith
- Wellcome Centre for Mitochondrial Research, Biosciences Institute Newcastle University Newcastle Upon Tyne NE2 4HH UK
| | - Julia C Whitehall
- Wellcome Centre for Mitochondrial Research, Biosciences Institute Newcastle University Newcastle Upon Tyne NE2 4HH UK
| | - Laura C Greaves
- Wellcome Centre for Mitochondrial Research, Biosciences Institute Newcastle University Newcastle Upon Tyne NE2 4HH UK
| |
Collapse
|
244
|
Role of succinic acid in the regulation of sepsis. Int Immunopharmacol 2022; 110:109065. [PMID: 35853278 DOI: 10.1016/j.intimp.2022.109065] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/13/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022]
Abstract
Sepsis is a life-threatening disease characterized by a defensive response to damage. The immune response in patients with sepsis is overenhanced in the early stages and suppressed in the later stages, leading to poor prognosis. Metabolic reprogramming and epigenetic changes play a role in sepsis. Metabolic intermediates such as elevated succinic acid levels are significantly altered in patients with sepsis. Succinic acid, a metabolic intermediate of the tricarboxylic acid cycle, participates in energy supply and plays a role in metabolic reprogramming. Simultaneously, as an epigenetic regulator, it participates in gene transcription, translation, and post-translational modifications. It also participates in the inflammatory response, hypoxia, and the production of reactive oxygen species via endocrine and paracrine pathways. In this review, we have discussed the effects of succinic acid on sepsis and its therapeutic potential.
Collapse
|
245
|
Sanchez M, Hamel D, Bajon E, Duhamel F, Bhosle VK, Zhu T, Rivera JC, Dabouz R, Nadeau-Vallée M, Sitaras N, Tremblay DÉ, Omri S, Habelrih T, Rouget R, Hou X, Gobeil F, Joyal JS, Sapieha P, Mitchell G, Ribeiro-Da-Silva A, Mohammad Nezhady MA, Chemtob S. The Succinate Receptor SUCNR1 Resides at the Endoplasmic Reticulum and Relocates to the Plasma Membrane in Hypoxic Conditions. Cells 2022; 11:2185. [PMID: 35883628 PMCID: PMC9321536 DOI: 10.3390/cells11142185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 12/24/2022] Open
Abstract
The GPCR SUCNR1/GPR91 exerts proangiogenesis upon stimulation with the Krebs cycle metabolite succinate. GPCR signaling depends on the surrounding environment and intracellular localization through location bias. Here, we show by microscopy and by cell fractionation that in neurons, SUCNR1 resides at the endoplasmic reticulum (ER), while being fully functional, as shown by calcium release and the induction of the expression of the proangiogenic gene for VEGFA. ER localization was found to depend upon N-glycosylation, particularly at position N8; the nonglycosylated mutant receptor localizes at the plasma membrane shuttled by RAB11. This SUCNR1 glycosylation is physiologically regulated, so that during hypoxic conditions, SUCNR1 is deglycosylated and relocates to the plasma membrane. Downstream signal transduction of SUCNR1 was found to activate the prostaglandin synthesis pathway through direct interaction with COX-2 at the ER; pharmacologic antagonism of the PGE2 EP4 receptor (localized at the nucleus) was found to prevent VEGFA expression. Concordantly, restoring the expression of SUCNR1 in the retina of SUCNR1-null mice renormalized vascularization; this effect is markedly diminished after transfection of the plasma membrane-localized SUCNR1 N8A mutant, emphasizing that ER localization of the succinate receptor is necessary for proper vascularization. These findings uncover an unprecedented physiologic process where GPCR resides at the ER for signaling function.
Collapse
Affiliation(s)
- Melanie Sanchez
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - David Hamel
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Emmanuel Bajon
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - François Duhamel
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Vikrant K. Bhosle
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
- Cell Biology Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
| | - Tang Zhu
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Jose Carlos Rivera
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Rabah Dabouz
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Mathieu Nadeau-Vallée
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Nicholas Sitaras
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - David-Étienne Tremblay
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Samy Omri
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Tiffany Habelrih
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Raphael Rouget
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Xin Hou
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Fernand Gobeil
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Grant Mitchell
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Alfredo Ribeiro-Da-Silva
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Mohammad Ali Mohammad Nezhady
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Program of Molecular Biology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Research Center-CHU Ste-Justine, Departments of Pediatrics, Ophthalmology, and Pharmacology, Faculty of Medicine, Université de Montréal, 3175, Chemin Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
- Research Center-CHU Ste-Justine, Departments of Pediatrics, Ophthalmology, and Pharmacology, Faculty of Medicine, Université de Montréal, 3175, Chemin Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| |
Collapse
|
246
|
Adnan A, Raju S, Kumar R, Basu S. An Appraisal and Update of Fluorodeoxyglucose and Non-Fluorodeoxyglucose-PET Tracers in Thyroid and Non-Thyroid Endocrine Neoplasms. PET Clin 2022; 17:343-367. [PMID: 35717097 DOI: 10.1016/j.cpet.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Endocrine neoplasms and malignancies are a diverse group of tumors with varied clinical, histopathologic, and functional features. These tumors vary from sporadic to hereditary, isolated entities to multiple neoplastic syndromes, functioning and non functioning tumors, unifocal locally invasive, and advanced to multifocal tumors with disseminated distant metastases. The presence of various specific biomarkers and specific receptor targets serves as valuable tools for diagnosis, prognosis, and management. PET-CT with FDG and a multitude of novel and specific radiotracers towards specific therapeutic targets mandates personalization of their use, so as to ensure maximum clinical benefit in the management of these neoplasms.
Collapse
Affiliation(s)
- Aadil Adnan
- Radiation Medicine Centre (B.A.R.C), Tata Memorial Centre Annexe, Parel, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Shobhana Raju
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sandip Basu
- Radiation Medicine Centre (B.A.R.C), Tata Memorial Centre Annexe, Parel, Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
247
|
Pardella E, Ippolito L, Giannoni E, Chiarugi P. Nutritional and metabolic signalling through GPCRs. FEBS Lett 2022; 596:2364-2381. [PMID: 35776088 DOI: 10.1002/1873-3468.14441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/11/2022]
Abstract
Deregulated metabolism is a well-known feature of several challenging diseases, including diabetes, obesity and cancer. Besides their important role as intracellular bioenergetic molecules, dietary nutrients and metabolic intermediates are released in the extracellular environment. As such, they may achieve unconventional roles as hormone-like molecules by activating cell-surface G-protein-coupled receptors (GPCRs) that regulate several pathophysiological processes. In this review, we provide an insight into the role of lactate, succinate, fatty acids, amino acids, ketogenesis-derived and β-oxidation-derived intermediates as extracellular signalling molecules. Moreover, the mechanisms by which their cognate metabolite-sensing GPCRs integrate nutritional and metabolic signals with specific intracellular pathways will be described. A better comprehension of these aspects is of fundamental importance to identify GPCRs as novel druggable targets.
Collapse
Affiliation(s)
- Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| |
Collapse
|
248
|
Singh V, Singh R, Kushwaha R, Verma SP, Tripathi AK, Mahdi AA. The Molecular Role of HIF1α Is Elucidated in Chronic Myeloid Leukemia. Front Oncol 2022; 12:912942. [PMID: 35847841 PMCID: PMC9279726 DOI: 10.3389/fonc.2022.912942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic myeloid leukemia (CML) is potentially fatal blood cancer, but there is an unmet need to discover novel molecular biomarkers. The hypothesis of this study aimed to elucidate the relationship of HIF1α with the redox system, Krebs cycles, notch1, and other regulatory proteins to better understand the pathophysiology and clinical relevance in chronic myeloid leukemia (CML) patients, as the molecular mechanism of this axis is still not clear. This study included CML patient samples (n = 60; 60: blood; 10: bone marrow tissues) and compared them with healthy controls (n = 20; blood). Clinical diagnosis confirmed on bone marrow aspiration, marrow trephine biopsy, and BCR/ABL1 translocation. Cases were subclassified into chronic, accelerated, and blast crises as per WHO guidelines. Molecular experiments included redox parameters, DNA fragmentation, Krebs cycle metabolites, and gene expression by RT-PCR/Western blot/LC-MS, PPI (STRING), Pearson correlation, and ROC curve analysis. Here, our findings show that p210/p190BCR/ABL1 translocation is common in all blast crisis phases of CML. Redox factor/Krebs oncometabolite concentrations were high, leading to upregulation and stabilization of HIF1α. HIF1α leads to the pathogenesis in CML cells by upregulating their downstream genes (Notch 2/4/Ikaros/SIRT1/Foxo-3a/p53, etc.). Whereas, downregulated ubiquitin proteasomal and apoptotic factors in CML pateints, can trigger degradation of HIF1α through proline hydroxylation. However, HIF1α showed a negative corelation with the notch1 pathway. Notch1 plays a tumor-suppressive role in CML and might have the potential to be used as a diagnostic marker along with other factors in CML patients. The outcome also revealed that oxidant treatment could not be effective in augmentation with conventional therapy because CML cells can enhance the levels of antioxidants for their survival. HIF1α might be a novel therapeutic target other than BCR/ABL1 translocation.
Collapse
Affiliation(s)
- Vivek Singh
- Department of Biochemistry, King George’s Medical University, Lucknow, India
| | - Ranjana Singh
- Department of Biochemistry, King George’s Medical University, Lucknow, India
- *Correspondence: Ranjana Singh, ;
| | - Rashmi Kushwaha
- Department of Pathology, King George’s Medical University, Lucknow, India
| | | | - Anil Kumar Tripathi
- Department of Clinical Hematology, King George’s Medical University, Lucknow, India
| | - Abbas Ali Mahdi
- Department of Biochemistry, King George’s Medical University, Lucknow, India
| |
Collapse
|
249
|
Joshi A, Ito T, Picard D, Neckers L. The Mitochondrial HSP90 Paralog TRAP1: Structural Dynamics, Interactome, Role in Metabolic Regulation, and Inhibitors. Biomolecules 2022; 12:biom12070880. [PMID: 35883436 PMCID: PMC9312948 DOI: 10.3390/biom12070880] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
The HSP90 paralog TRAP1 was discovered more than 20 years ago; yet, a detailed understanding of the function of this mitochondrial molecular chaperone remains elusive. The dispensable nature of TRAP1 in vitro and in vivo further complicates an understanding of its role in mitochondrial biology. TRAP1 is more homologous to the bacterial HSP90, HtpG, than to eukaryotic HSP90. Lacking co-chaperones, the unique structural features of TRAP1 likely regulate its temperature-sensitive ATPase activity and shed light on the alternative mechanisms driving the chaperone’s nucleotide-dependent cycle in a defined environment whose physiological temperature approaches 50 °C. TRAP1 appears to be an important bioregulator of mitochondrial respiration, mediating the balance between oxidative phosphorylation and glycolysis, while at the same time promoting mitochondrial homeostasis and displaying cytoprotective activity. Inactivation/loss of TRAP1 has been observed in several neurodegenerative diseases while TRAP1 expression is reported to be elevated in multiple cancers and, as with HSP90, evidence of addiction to TRAP1 has been observed. In this review, we summarize what is currently known about this unique HSP90 paralog and why a better understanding of TRAP1 structure, function, and regulation is likely to enhance our understanding of the mechanistic basis of mitochondrial homeostasis.
Collapse
Affiliation(s)
- Abhinav Joshi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA; (A.J.); (T.I.)
| | - Takeshi Ito
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA; (A.J.); (T.I.)
| | - Didier Picard
- Department of Molecular and Cellular Biology, Université de Genève, Sciences III, 30 Quai Ernest-Ansermet, CH-1211 Geneva, Switzerland;
| | - Len Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA; (A.J.); (T.I.)
- Correspondence: ; Tel.: +1-240-858-3918
| |
Collapse
|
250
|
Yuan T, Zhou T, Qian M, Du J, Liu Y, Wang J, Li Y, Fan G, Yan F, Dai X, Li X, Wu Y, Dong X, He Q, Zhu H, Yang B. SDHA/B reduction promotes hepatocellular carcinoma by facilitating the deNEDDylation of cullin1 and stabilizing YAP/TAZ. Hepatology 2022. [PMID: 35713976 DOI: 10.1002/hep.32621] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Succinate dehydrogenase enzyme (SDH) is frequently diminished in samples from patients with hepatocellular carcinoma (HCC), and SDH reduction is associated with elevated succinate level and poor prognosis in patients with HCC. However, the underlying mechanisms of how impaired SDH activity promotes HCC remain unclear. APPROACH AND RESULTS In this study, we observed remarkable downregulations of SDH subunits A and B (SDHA/B) in chronic liver injury-induced murine HCC models and patient samples. Subsequent RNA sequencing, hematoxylin and eosin staining, and immunohistochemistry analyses of HCC samples revealed that Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) were significantly upregulated in HCC, with their levels inversely correlating with that of SDHA/B. YAP/TAZ stability was greatly enhanced in SDHA/B-depleted HCC cells along with accumulation of succinate. Further mechanistic analyses demonstrated that impaired activity of SDHA/B resulted in succinate accumulation, which facilitated the deNEDDylation of cullin1 and therefore disrupted the E3 ubiquitin ligase SCFβ-TrCP complex, consequently leading to YAP/TAZ stabilization and activation in HCC cells. The accelerated in vitro cell proliferation and in vivo tumor growth caused by SDHA/B reduction or succinate exposure were largely dependent on the aberrant activation of YAP/TAZ. CONCLUSIONS Our study demonstrated that SDHA/B reduction promotes HCC proliferation by preventing the proteasomal degradation of YAP/TAZ through modulating cullin1 NEDDylation, thus binding SDH-deficient HCC cells to YAP/TAZ pathway and rendering these cells vulnerable to YAP/TAZ inhibition. Our findings warrant further investigation on the therapeutic effects of targeting YAP/TAZ in patients with HCC displaying reduced SDHA/B or elevated succinate levels.
Collapse
Affiliation(s)
- Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Tianyi Zhou
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meijia Qian
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jiamin Du
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yue Liu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jia'er Wang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yonghao Li
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Guanghan Fan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fangjie Yan
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyang Dai
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
| | - Xiawei Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yulian Wu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center of Zhejiang University, Hangzhou, China
| | - Xin Dong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China.,Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center of Zhejiang University, Hangzhou, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center of Zhejiang University, Hangzhou, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|