1
|
Griffith CM, Conrotte JF, Paydar P, Xie X, Heins-Marroquin U, Gavotto F, Jäger C, Ellens KW, Linster CL. CLYBL averts vitamin B 12 depletion by repairing malyl-CoA. Nat Chem Biol 2025:10.1038/s41589-025-01857-9. [PMID: 40108300 DOI: 10.1038/s41589-025-01857-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
Citrate lyase beta-like protein (CLYBL) is a ubiquitously expressed mammalian enzyme known for its role in the degradation of itaconate, a bactericidal immunometabolite produced in activated macrophages. The association of CLYBL loss of function with reduced circulating vitamin B12 levels was proposed to result from inhibition of the B12-dependent enzyme methylmalonyl-CoA mutase by itaconyl-CoA. The discrepancy between the highly inducible and locally confined production of itaconate and the broad expression profile of CLYBL across tissues suggested a role for this enzyme beyond itaconate catabolism. Here we discover that CLYBL additionally functions as a metabolite repair enzyme for malyl-CoA, a side product of promiscuous citric acid cycle enzymes. We found that CLYBL knockout cells, accumulating malyl-CoA but not itaconyl-CoA, show decreased levels of adenosylcobalamin and that malyl-CoA is a more potent inhibitor of methylmalonyl-CoA mutase than itaconyl-CoA. Our work thus suggests that malyl-CoA plays a role in the B12 deficiency observed in individuals with CLYBL loss of function.
Collapse
Affiliation(s)
- Corey M Griffith
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jean-François Conrotte
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Parisa Paydar
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Xinqiang Xie
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ursula Heins-Marroquin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Floriane Gavotto
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Christian Jäger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Kenneth W Ellens
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Carole L Linster
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
2
|
Denley MCS, Straub MS, Marcionelli G, Güra MA, Penton D, Delvendahl I, Poms M, Vekeriotaite B, Cherkaoui S, Conte F, von Meyenn F, Froese DS, Baumgartner MR. Mitochondrial dysfunction drives a neuronal exhaustion phenotype in methylmalonic aciduria. Commun Biol 2025; 8:410. [PMID: 40069408 PMCID: PMC11897345 DOI: 10.1038/s42003-025-07828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 02/26/2025] [Indexed: 03/15/2025] Open
Abstract
Methylmalonic aciduria (MMA) is an inborn error of metabolism resulting in loss of function of the enzyme methylmalonyl-CoA mutase (MMUT). Despite acute and persistent neurological symptoms, the pathogenesis of MMA in the central nervous system is poorly understood, which has contributed to a dearth of effective brain specific treatments. Here we utilised patient-derived induced pluripotent stem cells and in vitro differentiation to generate a human neuronal model of MMA. We reveal strong evidence of mitochondrial dysfunction caused by deficiency of MMUT in patient neurons. By employing patch-clamp electrophysiology, targeted metabolomics, and bulk transcriptomics, we expose an altered state of excitability, which is exacerbated by application of dimethyl-2-oxoglutarate, and we suggest may be connected to metabolic rewiring. Our work provides first evidence of mitochondrial driven neuronal dysfunction in MMA, which through our comprehensive characterisation of this paradigmatic model, enables first steps to identifying effective therapies.
Collapse
Affiliation(s)
- Matthew C S Denley
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Monique S Straub
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Giulio Marcionelli
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Miriam A Güra
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - David Penton
- Electrophysiology Core Facility, University of Zurich, Zurich, CH-8057, Switzerland
| | - Igor Delvendahl
- Department of Molecular Life Sciences, University of Zurich, Zurich, CH-8057, Switzerland
| | - Martin Poms
- Clinical Chemistry and Biochemistry and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Beata Vekeriotaite
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, CH-8603, Switzerland
| | - Sarah Cherkaoui
- Pediatric Cancer Metabolism Laboratory, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland
| | - Federica Conte
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud, University Medical Center, Nijmegen, 6525 GA, Netherlands
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, CH-8603, Switzerland
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland.
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, CH-8032, Switzerland.
| |
Collapse
|
3
|
Yap S, Gasperini S, Matsumoto S, Feillet F. Role of carglumic acid in the long-term management of propionic and methylmalonic acidurias. Orphanet J Rare Dis 2024; 19:464. [PMID: 39695809 DOI: 10.1186/s13023-024-03468-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Propionic aciduria (PA) and methylmalonic aciduria (MMA) are rare inherited disorders caused by defects in the propionate metabolic pathway. PA due to propionyl coenzyme A carboxylase deficiency results in accumulation of propionic acid, while in MMA, deficiency in methylmalonyl coenzyme A mutase leads to accumulation of methylmalonic acid. Hyperammonemia is related to a secondary deficiency of N-acetylglutamate (NAG), the activator of carbamoyl phosphate synthetase 1, which is an irreversible rate-limiting enzyme in the urea cycle. Carglumic acid (CGA) is a synthetic structural analog of human NAG and is approved for the treatment of patients with hyperammonemia due to PA or MMA. CGA is well tolerated and its use in normalizing ammonia levels during acute hyperammonemic episodes in patients with PA and MMA is well established. This expert opinion analyzed clinical evidence for CGA and discussed its place, along with other management strategies, in the long-term management of PA or MMA. A literature search of PubMed was undertaken to identify publications related to the chronic use of CGA, transplantation, dietary management, ammonia scavengers, and gene therapy for treatment of patients with PA or MMA. The authors selected the most relevant studies for inclusion. Four clinical studies, one single center case series, and three case reports show that CGA is safe and effective in the chronic treatment of PA and MMA. In particular, the addition of CGA is associated with a reduction in hyperammonemic decompensation episodes and admission to hospital, compared with conventional dietary treatment alone. Current treatment guidelines and recommendations include the use of CGA mainly in acute decompensation, however, lag in considering the benefits of long-term CGA treatment on clinical and biochemical outcomes in patients with PA or MMA. CGA is safe and effective in the chronic treatment of PA and MMA and may help to resolve some of the issues associated with other strategies used to treat these disorders. Thus, CGA appears to have potential for the chronic management of patients with PA and MMA and should be recommended for inclusion in the chronic treatment of these disorders.
Collapse
Affiliation(s)
- Sufin Yap
- Department of Inherited Metabolic Diseases, Sheffield Children's Hospital, Sheffield Children's NHS Foundation Trust, Western Bank, Sheffield, S10 2TH, UK.
| | - Serena Gasperini
- Metabolic Rare Disease Unit "Fondazione Mariani", Pediatric Department, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Shirou Matsumoto
- Department of Neonatology, Kumamoto University, Honjo 1-1-1, Chu-oh-ku, Kumamoto, Japan
| | - François Feillet
- Pediatric Unit, Reference Center for Inborn Errors of Metabolism, University Hospital of Nancy, INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, Nancy, France
| |
Collapse
|
4
|
Buoso C, Seifert M, Lang M, Griffith CM, Talavera Andújar B, Castelo Rueda MP, Fischer C, Doerrier C, Talasz H, Zanon A, Pramstaller PP, Schymanski EL, Pichler I, Weiss G. Dopamine‑iron homeostasis interaction rescues mitochondrial fitness in Parkinson's disease. Neurobiol Dis 2024; 196:106506. [PMID: 38648865 DOI: 10.1016/j.nbd.2024.106506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Imbalances of iron and dopamine metabolism along with mitochondrial dysfunction have been linked to the pathogenesis of Parkinson's disease (PD). We have previously suggested a direct link between iron homeostasis and dopamine metabolism, as dopamine can increase cellular uptake of iron into macrophages thereby promoting oxidative stress responses. In this study, we investigated the interplay between iron, dopamine, and mitochondrial activity in neuroblastoma SH-SY5Y cells and human induced pluripotent stem cell (hiPSC)-derived dopaminergic neurons differentiated from a healthy control and a PD patient with a mutation in the α-synuclein (SNCA) gene. In SH-SY5Y cells, dopamine treatment resulted in increased expression of the transmembrane iron transporters transferrin receptor 1 (TFR1), ferroportin (FPN), and mitoferrin2 (MFRN2) and intracellular iron accumulation, suggesting that dopamine may promote iron uptake. Furthermore, dopamine supplementation led to reduced mitochondrial fitness including decreased mitochondrial respiration, increased cytochrome c control efficiency, reduced mtDNA copy number and citrate synthase activity, increased oxidative stress and impaired aconitase activity. In dopaminergic neurons derived from a healthy control individual, dopamine showed comparable effects as observed in SH-SY5Y cells. The hiPSC-derived PD neurons harboring an endogenous SNCA mutation demonstrated altered mitochondrial iron homeostasis, reduced mitochondrial capacity along with increased oxidative stress and alterations of tricarboxylic acid cycle linked metabolic pathways compared with control neurons. Importantly, dopamine treatment of PD neurons promoted a rescue effect by increasing mitochondrial respiration, activating antioxidant stress response, and normalizing altered metabolite levels linked to mitochondrial function. These observations provide evidence that dopamine affects iron homeostasis, intracellular stress responses and mitochondrial function in healthy cells, while dopamine supplementation can restore the disturbed regulatory network in PD cells.
Collapse
Affiliation(s)
- Chiara Buoso
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy; Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Martin Lang
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy
| | - Corey M Griffith
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Begoña Talavera Andújar
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | | | - Christine Fischer
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Heribert Talasz
- Institute of Medical Biochemistry, Protein Core Facility, Biocenter Innsbruck, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | | | - Emma L Schymanski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy.
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
5
|
Shen G, Liu J, Yang H, Xie N, Yang Y. mRNA therapies: Pioneering a new era in rare genetic disease treatment. J Control Release 2024; 369:696-721. [PMID: 38580137 DOI: 10.1016/j.jconrel.2024.03.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024]
Abstract
Rare genetic diseases, often referred to as orphan diseases due to their low prevalence and limited treatment options, have long posed significant challenges to our medical system. In recent years, Messenger RNA (mRNA) therapy has emerged as a highly promising treatment approach for various diseases caused by genetic mutations. Chemically modified mRNA is introduced into cells using carriers like lipid-based nanoparticles (LNPs), producing functional proteins that compensate for genetic deficiencies. Given the advantages of precise dosing, biocompatibility, transient expression, and minimal risk of genomic integration, mRNA therapies can safely and effectively correct genetic defects in rare diseases and improve symptoms. Currently, dozens of mRNA drugs targeting rare diseases are undergoing clinical trials. This comprehensive review summarizes the progress of mRNA therapy in treating rare genetic diseases. It introduces the development, molecular design, and delivery systems of mRNA therapy, highlighting their research progress in rare genetic diseases based on protein replacement and gene editing. The review also summarizes research progress in various rare disease models and clinical trials. Additionally, it discusses the challenges and future prospects of mRNA therapy. Researchers are encouraged to join this field and collaborate to advance the clinical translation of mRNA therapy, bringing hope to patients with rare genetic diseases.
Collapse
Affiliation(s)
- Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanmei Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| | - Yang Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Hou X, Chen Y, Li X, Gu X, Dong W, Shi J, Ji S. Protein succinylation: regulating metabolism and beyond. Front Nutr 2024; 11:1336057. [PMID: 38379549 PMCID: PMC10876795 DOI: 10.3389/fnut.2024.1336057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/22/2024] [Indexed: 02/22/2024] Open
Abstract
Modifications of protein post-translation are critical modulatory processes, which alters target protein biological activity,function and/or location, even involved in pathogenesis of some diseases. So far, there are at least 16 types of post-translation modifications identified, particularly through recent mass spectrometry analysis. Among them, succinylation (Ksuc) on protein lysine residues causes a variety of biological changes. Succinylation of proteins contributes to many cellular processes such as proliferation, growth, differentiation, metabolism and even tumorigenesis. Mechanically, Succinylation leads to conformation alteration of chromatin or remodeling. As a result, transcription/expression of target genes is changed accordingly. Recent research indicated that succinylation mainly contributes to metabolism modulations, from gene expression of metabolic enzymes to their activity modulation. In this review, we will conclude roles of succinylation in metabolic regulation of glucose, fat, amino acids and related metabolic disease launched by aberrant succinylation. Our goal is to stimulate extra attention to these still not well researched perhaps important succinylation modification on proteins and cell processes.
Collapse
Affiliation(s)
- Xiaoli Hou
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, China
| | - Yiqiu Chen
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, China
| | - Xiao Li
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, China
| | - Xianliang Gu
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, China
| | - Weixia Dong
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, China
| | - Jie Shi
- Zhoukou Vocational and Technical College, Zhoukou, China
| | - Shaoping Ji
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, China
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng, China
| |
Collapse
|
7
|
Hosseindoust A, Choi Y, Ha S, Tajudeen H, Mun J, Kinara E, Kim Y, Kim J. Anti-Bordetella bronchiseptica effects of targeted bacteriophages via microbiome and metabolic mediated mechanisms. Sci Rep 2023; 13:21755. [PMID: 38066337 PMCID: PMC10709636 DOI: 10.1038/s41598-023-49248-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Bordetella bronchiseptica poses a significant challenge in the context of respiratory infections, particularly in weanling pigs. In this study, we investigated the impact of a novel targeted bacteriophage in controlling B. bronchiseptica challenge (BBC) in an experimental design involving five distinct treatment groups: NC (no challenge), PC (BBC challenge), BF (108 pfu bacteriophage/kg diet + BBC), BN (2 × 107 pfu/day bacteriophage by nasal spray + BBC), and AT (antibiotic + BBC). The experiment was conducted for 2 weeks. The highest turbinate score was observed in the PC. The BF treatment showed higher plasma IL (interleukine)-1β and IL-6 compared with the BN and AT treatments. Plasma concentrations of IL-1β were increased in the BF pigs compared with the BN, AT, and NC. Among the BBC groups, the PC treatment exhibited a higher abundance of Staphylococcus. aureus and B. bronchiseptica in the lung. A lower S. aureus, Streptococcus. suis, and B. bronchiseptica colonization was detected in the AT compared with the BF and BN treatments. The BF showed lower plasma zonulin compared with the BN and AT. A higher plasma concentration of superoxide dismutase was observed in the BF and AT compared with PC and BN. The BN influenced the glycine, serine-threonine metabolism; glycerolipid metabolism; glyoxylate-dicarboxylate metabolism; and arachidonic acid metabolism compared with the NC. In conclusion, nasal-sprayed bacteriophage effectively controlled B. bronchiseptica infection, however, their efficiency was lower than the antibiotic.
Collapse
Affiliation(s)
- Abdolreza Hosseindoust
- Department of Animal Industry Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - YoHan Choi
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - SangHun Ha
- Department of Animal Industry Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Habeeb Tajudeen
- Department of Animal Industry Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - JunYoung Mun
- Department of Animal Industry Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Elick Kinara
- Department of Animal Industry Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - YoungIn Kim
- CTC Bio, Inc., Seoul, 138-858, Republic of Korea
| | - JinSoo Kim
- Department of Animal Industry Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
8
|
Feaver RE, Bowers MS, Cole BK, Hoang S, Lawson MJ, Taylor J, LaMoreaux BD, Zhao L, Henke BR, Johns BA, Nyborg AC, Wamhoff BR, Figler RA. Human cardiovascular disease model predicts xanthine oxidase inhibitor cardiovascular risk. PLoS One 2023; 18:e0291330. [PMID: 37682977 PMCID: PMC10490929 DOI: 10.1371/journal.pone.0291330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Some health concerns are often not identified until late into clinical development of drugs, which can place participants and patients at significant risk. For example, the United States Food and Drug Administration (FDA) labeled the xanthine oxidase inhibitor febuxostat with a"boxed" warning regarding an increased risk of cardiovascular death, and this safety risk was only identified during Phase 3b clinical trials after its approval. Thus, better preclinical assessment of drug efficacy and safety are needed to accurately evaluate candidate drug risk earlier in discovery and development. This study explored whether an in vitro vascular model incorporating human vascular cells and hemodynamics could be used to differentiate the potential cardiovascular risk associated with molecules that have similar on-target mechanisms of action. We compared the transcriptomic responses induced by febuxostat and other xanthine oxidase inhibitors to a database of 111 different compounds profiled in the human vascular model. Of the 111 compounds in the database, 107 are clinical-stage and 33 are FDA-labelled for increased cardiovascular risk. Febuxostat induces pathway-level regulation that has high similarity to the set of drugs FDA-labelled for increased cardiovascular risk. These results were replicated with a febuxostat analog, but not another structurally distinct xanthine oxidase inhibitor that does not confer cardiovascular risk. Together, these data suggest that the FDA warning for febuxostat stems from the chemical structure of the medication itself, rather than the target, xanthine oxidase. Importantly, these data indicate that cardiovascular risk can be evaluated in this in vitro human vascular model, which may facilitate understanding the drug candidate safety profile earlier in discovery and development.
Collapse
Affiliation(s)
- Ryan E. Feaver
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - M. Scott Bowers
- Horizon Therapeutics plc, Deerfield, Illinois, United States of America
| | - Banumathi K. Cole
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Steve Hoang
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Mark J. Lawson
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Justin Taylor
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | | | - Lin Zhao
- Horizon Therapeutics plc, Deerfield, Illinois, United States of America
| | - Brad R. Henke
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Brian A. Johns
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Andrew C. Nyborg
- Horizon Therapeutics plc, Deerfield, Illinois, United States of America
| | - Brian R. Wamhoff
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Robert A. Figler
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| |
Collapse
|
9
|
Schumann A, Brutsche M, Havermans M, Grünert SC, Kölker S, Groß O, Hannibal L, Spiekerkoetter U. The impact of metabolic stressors on mitochondrial homeostasis in a renal epithelial cell model of methylmalonic aciduria. Sci Rep 2023; 13:7677. [PMID: 37169781 PMCID: PMC10175303 DOI: 10.1038/s41598-023-34373-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023] Open
Abstract
Methylmalonic aciduria (MMA-uria) is caused by deficiency of the mitochondrial enzyme methylmalonyl-CoA mutase (MUT). MUT deficiency hampers energy generation from specific amino acids, odd-chain fatty acids and cholesterol. Chronic kidney disease (CKD) is a well-known long-term complication. We exposed human renal epithelial cells from healthy controls and MMA-uria patients to different culture conditions (normal treatment (NT), high protein (HP) and isoleucine/valine (I/V)) to test the effect of metabolic stressors on renal mitochondrial energy metabolism. Creatinine levels were increased and antioxidant stress defense was severely comprised in MMA-uria cells. Alterations in mitochondrial homeostasis were observed. Changes in tricarboxylic acid cycle metabolites and impaired energy generation from fatty acid oxidation were detected. Methylcitrate as potentially toxic, disease-specific metabolite was increased by HP and I/V load. Mitophagy was disabled in MMA-uria cells, while autophagy was highly active particularly under HP and I/V conditions. Mitochondrial dynamics were shifted towards fission. Sirtuin1, a stress-resistance protein, was down-regulated by HP and I/V exposure in MMA-uria cells. Taken together, both interventions aggravated metabolic fingerprints observed in MMA-uria cells at baseline. The results point to protein toxicity in MMA-uria and lead to a better understanding, how the accumulating, potentially toxic organic acids might trigger CKD.
Collapse
Affiliation(s)
- Anke Schumann
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Mathildenstr. 1, 79106, Freiburg, Germany.
| | - Marion Brutsche
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Mathildenstr. 1, 79106, Freiburg, Germany
| | - Monique Havermans
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sarah C Grünert
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Mathildenstr. 1, 79106, Freiburg, Germany
| | - Stefan Kölker
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Luciana Hannibal
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Laboratory of Clinical Biochemistry and Metabolism, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Ute Spiekerkoetter
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Mathildenstr. 1, 79106, Freiburg, Germany
| |
Collapse
|
10
|
Decreased propionyl-CoA metabolism facilitates metabolic reprogramming and promotes hepatocellular carcinoma. J Hepatol 2023; 78:627-642. [PMID: 36462680 DOI: 10.1016/j.jhep.2022.11.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Alterations of multiple metabolites characterize distinct features of metabolic reprograming in hepatocellular carcinoma (HCC). However, the role of most metabolites, including propionyl-CoA (Pro-CoA), in metabolic reprogramming and hepatocarcinogenesis remains elusive. In this study, we aimed to dissect how Pro-CoA metabolism affects these processes. METHODS TCGA data and HCC samples were used to analyze ALDH6A1-mediated Pro-CoA metabolism and its correlation with HCC. Multiple metabolites were assayed by targeted mass spectrometry. The role of ALDH6A1-generated Pro-CoA in HCC was evaluated in HCC cell lines as well as xenograft nude mouse models and primary liver cancer mouse models. Non-targeted metabolomic and targeted energy metabolomic analyses, as well as multiple biochemical assays, were performed. RESULTS Decreases in Pro-CoA and its derivative propionyl-L-carnitine due to ALDH6A1 downregulation were tightly associated with HCC. Functionally, ALDH6A1-mediated Pro-CoA metabolism suppressed HCC proliferation in vitro and impaired hepatocarcinogenesis in mice. The aldehyde dehydrogenase activity was indispensable for this function of ALDH6A1, while Pro-CoA carboxylases antagonized ALDH6A1 function by eliminating Pro-CoA. Mechanistically, ALDH6A1 caused a signature enrichment of central carbon metabolism in cancer and impaired energy metabolism: ALDH6A1-generated Pro-CoA suppressed citrate synthase activity, which subsequently reduced tricarboxylic acid cycle flux, impaired mitochondrial respiration and membrane potential, and decreased ATP production. Moreover, Pro-CoA metabolism generated 2-methylcitric acid, which mimicked the inhibitory effect of Pro-CoA on citrate synthase and dampened mitochondrial respiration and HCC proliferation. CONCLUSIONS The decline of ALDH6A1-mediated Pro-CoA metabolism contributes to metabolic remodeling and facilitates hepatocarcinogenesis. Pro-CoA, propionyl-L-carnitine and 2-methylcitric acid may serve as novel metabolic biomarkers for the diagnosis and treatment of HCC. Pro-CoA metabolism may provide potential targets for development of novel strategies against HCC. IMPACT AND IMPLICATIONS Our study presents new insights on the role of propionyl-CoA metabolism in metabolic reprogramming and hepatocarcinogenesis. This work has uncovered potential diagnostic and predictive biomarkers, which could be used by physicians to improve clinical practice and may also serve as targets for the development of therapeutic strategies against HCC.
Collapse
|
11
|
Povero D. Novel oncometabolites and metabolic checkpoints involved in hepatocellular carcinoma development. J Hepatol 2023; 78:463-466. [PMID: 36640958 PMCID: PMC10174048 DOI: 10.1016/j.jhep.2023.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/02/2023] [Indexed: 01/13/2023]
Affiliation(s)
- Davide Povero
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, 55905, MN, USA.
| |
Collapse
|
12
|
Head PE, Venditti CP. Anaplerosis in action. Nat Metab 2023; 5:5-7. [PMID: 36717753 PMCID: PMC10181862 DOI: 10.1038/s42255-022-00724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Investigation of multi-omic changes and their effects on regulation of metabolic pathways confirm anaplerotic deficiencies in methylmalonic acidaemia, strengthening the need for future therapies aimed at replenishing intermediates of the tricarboxylic acid cycle.
Collapse
Affiliation(s)
- PamelaSara E Head
- National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD, USA
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles P Venditti
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
13
|
Billington CJ, Chapman KA, Leon E, Meltzer BW, Berger SI, Olson M, Figler RA, Hoang SA, Wanxing C, Wamhoff BR, Collado MS, Cusmano‐Ozog K. Genomic and biochemical analysis of repeatedly observed variants in DBT in individuals with maple syrup urine disease of Central American ancestry. Am J Med Genet A 2022; 188:2738-2749. [PMID: 35799415 PMCID: PMC9542135 DOI: 10.1002/ajmg.a.62893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 01/25/2023]
Abstract
Maple syrup urine disease (MSUD) is an intoxication-type inherited metabolic disorder in which hyperleucinemia leads to brain swelling and death without treatment. MSUD is caused by branched-chain alpha-ketoacid dehydrogenase deficiency due to biallelic loss of the protein products from the genes BCKDHA, BCKDHB, or DBT, while a distinct but related condition is caused by loss of DLD. In this case series, eleven individuals with MSUD caused by two pathogenic variants in DBT are presented. All eleven individuals have a deletion of exon 2 (delEx2, NM_001918.3:c.48_171del); six individuals are homozygous and five individuals are compound heterozygous with a novel missense variant (NM_001918.5:c.916 T > C [p.Ser306Pro]) confirmed to be in trans. Western Blot indicates decreased amount of protein product in delEx2;c.916 T > C liver cells and absence of protein product in delEx2 homozygous hepatocytes. Ultrahigh performance liquid chromatography-tandem mass spectrometry demonstrates an accumulation of branched-chain amino acids and alpha-ketoacids in explanted hepatocytes. Individuals with these variants have a neonatal-onset, non-thiamine-responsive, classical form of MSUD. Strikingly, the entire cohort is derived from families who immigrated to the Washington, DC, metro area from Honduras or El Salvador suggesting the possibility of a founder effect.
Collapse
Affiliation(s)
- Charles J. Billington
- Children's National Rare Disease InstituteWashingtonDistrict of ColumbiaUSA
- Department of PediatricsUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Eyby Leon
- Children's National Rare Disease InstituteWashingtonDistrict of ColumbiaUSA
| | - Beatrix W. Meltzer
- Laboratory Medicine, Children's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Seth I. Berger
- Children's National Rare Disease InstituteWashingtonDistrict of ColumbiaUSA
| | - Matthew Olson
- HemoShear Therapeutics, Inc.CharlottesvilleVirginiaUSA
| | | | | | - Cui Wanxing
- Georgetown University HospitalWashingtonDistrict of ColumbiaUSA
| | | | | | | |
Collapse
|
14
|
Head PE, Myung S, Chen Y, Schneller JL, Wang C, Duncan N, Hoffman P, Chang D, Gebremariam A, Gucek M, Manoli I, Venditti CP. Aberrant methylmalonylation underlies methylmalonic acidemia and is attenuated by an engineered sirtuin. Sci Transl Med 2022; 14:eabn4772. [PMID: 35613279 PMCID: PMC10468269 DOI: 10.1126/scitranslmed.abn4772] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organic acidemias such as methylmalonic acidemia (MMA) are a group of inborn errors of metabolism that typically arise from defects in the catabolism of amino and fatty acids. Accretion of acyl-CoA species is postulated to underlie disease pathophysiology, but the mechanism(s) remain unknown. Here, we surveyed hepatic explants from patients with MMA and unaffected donors, in parallel with samples from various mouse models of methylmalonyl-CoA mutase deficiency. We found a widespread posttranslational modification, methylmalonylation, that inhibited enzymes in the urea cycle and glycine cleavage pathway in MMA. Biochemical studies and mouse genetics established that sirtuin 5 (SIRT5) controlled the metabolism of MMA-related posttranslational modifications. SIRT5 was engineered to resist acylation-driven inhibition via lysine to arginine mutagenesis. The modified SIRT5 was used to create an adeno-associated viral 8 (AAV8) vector and systemically delivered to mutant and control mice. Gene therapy ameliorated hyperammonemia and reduced global methylmalonylation in the MMA mice.
Collapse
Affiliation(s)
- PamelaSara E. Head
- National Institute of General Medical Sciences, NIH, 45 Center Drive MSC 6200 Bethesda, MD, 20892-6200 USA
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - Sangho Myung
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - Yong Chen
- National Heart Lung and Blood Institute, NIH, Building 31, 31 Center Drive Bethesda, MD 20892, USA
| | - Jessica L. Schneller
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - Cindy Wang
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - Nicholas Duncan
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - Pauline Hoffman
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - David Chang
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - Abigael Gebremariam
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - Marjan Gucek
- National Heart Lung and Blood Institute, NIH, Building 31, 31 Center Drive Bethesda, MD 20892, USA
| | - Irini Manoli
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| | - Charles P. Venditti
- National Human Genome Research Institute, NIH, Bethesda, MD, 10 Center Drive Building 10, Room 7S257 Bethesda, MD 20892, USA
| |
Collapse
|
15
|
Zhao C, Wang Y, Yang H, Wang S, Tang MC, Cyr D, Parente F, Allard P, Waters P, Furtos A, Yang G, Mitchell GA. Propionic acidemia in mice: Liver acyl-CoA levels and clinical course. Mol Genet Metab 2022; 135:47-55. [PMID: 34896004 DOI: 10.1016/j.ymgme.2021.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 12/11/2022]
Abstract
Propionic acidemia (PA) is a severe autosomal recessive metabolic disease caused by deficiency of propionyl-CoA carboxylase (PCC). We studied PA transgenic (Pat) mice that lack endogenous PCC but express a hypoactive human PCCA cDNA, permitting their survival. Pat cohorts followed from 3 to 20 weeks of age showed growth failure and lethal crises of lethargy and hyperammonemia, commoner in males (27/50, 54%) than in females (11/52, 21%) and occurring mainly in Pat mice with the most severe growth deficiency. Groups of Pat mice were studied under basal conditions (P-Ba mice) and during acute crises (P-Ac). Plasma acylcarnitines in P-Ba mice, compared to controls, showed markedly elevated C3- and low C2-carnitine, with a further decrease in C2-carnitine in P-Ac mice. These clinical and biochemical findings resemble those of human PA patients. Liver acyl-CoA measurements showed that propionyl-CoA was a minor species in controls (propionyl-CoA/acetyl-CoA ratio, 0.09). In contrast, in P-Ba liver the ratio was 1.4 and in P-Ac liver, 13, with concurrent reductions of the levels of acetyl-CoA and other acyl-CoAs. Plasma ammonia levels in control, P-Ba and P-Ac mice were 109 ± 10, 311 ± 48 and 551 ± 61 μmol/L respectively. Four-week administration to Pat mice, of carglumate (N-carbamyl-L-glutamic acid), an analogue of N-carbamylglutamate, the product of the only acyl-CoA-requiring reaction directly related to the urea cycle, was associated with increased food consumption, improved growth and absence of fatal crises. Pat mice showed many similarities to human PA patients and provide a useful model for studying tissue pathophysiology and treatment outcomes.
Collapse
Affiliation(s)
- Chen Zhao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A & F University, Yangling, Shaanxi, China; Medical Genetics Service, Department of Pediatrics and Research Center, CHU Sainte-Justine and Université de Montréal, Montreal, Quebec, Canada
| | - Youlin Wang
- Medical Genetics Service, Department of Pediatrics and Research Center, CHU Sainte-Justine and Université de Montréal, Montreal, Quebec, Canada
| | - Hao Yang
- Medical Genetics Service, Department of Pediatrics and Research Center, CHU Sainte-Justine and Université de Montréal, Montreal, Quebec, Canada
| | - Shupei Wang
- Medical Genetics Service, Department of Pediatrics and Research Center, CHU Sainte-Justine and Université de Montréal, Montreal, Quebec, Canada
| | | | - Denis Cyr
- Medical Genetics Service, Department of Laboratory Medicine, CHU Sherbrooke and Department of Pediatrics, Université de Sherbrooke, Quebec, Canada
| | - Fabienne Parente
- Biochemical Genetics Laboratory, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Pierre Allard
- Biochemical Genetics Laboratory, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Paula Waters
- Medical Genetics Service, Department of Laboratory Medicine, CHU Sherbrooke and Department of Pediatrics, Université de Sherbrooke, Quebec, Canada
| | - Alexandra Furtos
- Département de Chimie, Université de Montréal, Montreal, Quebec, Canada
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A & F University, Yangling, Shaanxi, China.
| | - Grant A Mitchell
- Medical Genetics Service, Department of Pediatrics and Research Center, CHU Sainte-Justine and Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
16
|
Wang D, Guo J, Liu T, Zhou X, Yang Z, Shi C, Wang W, Li R, Zhang Y, Junzhang, Yan J, Zhu X, Li Y, Gong M, Cui Y, Wu X. Plasma metabolomics-based reveals the treatment mechanism of ShenGui capsule for application to coronary heart disease in a rat model. Anal Biochem 2021; 642:114480. [PMID: 34813769 DOI: 10.1016/j.ab.2021.114480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022]
Abstract
Shen Gui capsule (SGC) has been demonstrated to have a significant treatment effect for coronary heart disease (CHD). Nevertheless, the holistic therapeutic mechanism of SGC in vivo remain poorly interpreted. We aimed to systematically explore the preventive effect and mechanism of SGC on CHD rats using plasma metabolomics strategy. Rat CHD model was established by left anterior descending coronary artery ligation (LAD). Echocardiography, histological analyses of the myocardium and biochemical assays on serum were used to confirm the successful establishment of the CHD model and therapeutic effects of SGC. Then, UHPLC-MS/MS-based plasma metabolomics was combined with multivariate data analysis to screen potential pharmaco biomarkers associated with SGC treatment in the LAD-induced rat CHD model. After SGC treatment, 12 abnormal metabolites considered as potiential pharmaco biomarkers recovered to near normal levels. These biomarkers were involved in several metabolic pathways, including fat and protein metabolism, phenylalanine metabolism, neuroactive ligand-receptor interaction, androgen receptor signaling pathway, and estrone metabolism.These results suggested that SGC achieves therapeutic action on CHD via regulating various aspects of the body such as energy metabolism, neurological disturbances and inflammation, and thus plays a significant role in the treatment of CHD and its complications. The study is useful to systematically understand and analyze the mechanism of SGC's "multipie pathways, multiple levels, multiple targets" prevention and treatment of CHD.
Collapse
Affiliation(s)
- Dan Wang
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China; Department of Pharmacy, Chu Hisen-I Memorial Hospital, Tianjin Medical University, Tianjin, 300134, China
| | - Jialin Guo
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China; Department of Pharmacy, Tianjin Chest Hospital, Tianjin, 300211, China
| | - Tiantian Liu
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xinfeng Zhou
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Zijun Yang
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Chang Shi
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Weiting Wang
- Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, China
| | - Rongshan Li
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yanwen Zhang
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Junzhang
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Jiuxing Yan
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xuehui Zhu
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Ying Li
- Tianjin Neurological Institute, Tianjin Medical University, Tianjin, 300052, China
| | - Min Gong
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yan Cui
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Xiaohui Wu
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
17
|
Goedeke L, Canfrán-Duque A, Rotllan N, Chaube B, Thompson BM, Lee RG, Cline GW, McDonald JG, Shulman GI, Lasunción MA, Suárez Y, Fernández-Hernando C. MMAB promotes negative feedback control of cholesterol homeostasis. Nat Commun 2021; 12:6448. [PMID: 34750386 PMCID: PMC8575900 DOI: 10.1038/s41467-021-26787-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/14/2021] [Indexed: 12/21/2022] Open
Abstract
Intricate regulatory networks govern the net balance of cholesterol biosynthesis, uptake and efflux; however, the mechanisms surrounding cholesterol homeostasis remain incompletely understood. Here, we develop an integrative genomic strategy to detect regulators of LDLR activity and identify 250 genes whose knockdown affects LDL-cholesterol uptake and whose expression is modulated by intracellular cholesterol levels in human hepatic cells. From these hits, we focus on MMAB, an enzyme which catalyzes the conversion of vitamin B12 to adenosylcobalamin, and whose expression has previously been linked with altered levels of circulating cholesterol in humans. We demonstrate that hepatic levels of MMAB are modulated by dietary and cellular cholesterol levels through SREBP2, the master transcriptional regulator of cholesterol homeostasis. Knockdown of MMAB decreases intracellular cholesterol levels and augments SREBP2-mediated gene expression and LDL-cholesterol uptake in human and mouse hepatic cell lines. Reductions in total sterol content were attributed to increased intracellular levels of propionic and methylmalonic acid and subsequent inhibition of HMGCR activity and cholesterol biosynthesis. Moreover, mice treated with antisense inhibitors of MMAB display a significant reduction in hepatic HMGCR activity, hepatic sterol content and increased expression of SREBP2-mediated genes. Collectively, these findings reveal an unexpected role for the adenosylcobalamin pathway in regulating LDLR expression and identify MMAB as an additional control point by which cholesterol biosynthesis is regulated by its end product.
Collapse
Affiliation(s)
- Leigh Goedeke
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Alberto Canfrán-Duque
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Balkrishna Chaube
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Bonne M Thompson
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Richard G Lee
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Carlsbad, CA, 92010, USA
| | - Gary W Cline
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Miguel A Lasunción
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS) and CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
18
|
Schumann A, Belche V, Schaller K, Grünert SC, Kaech A, Baumgartner MR, Kölker S, Hannibal L, Spiekerkoetter U. Mitochondrial damage in renal epithelial cells is potentiated by protein exposure in propionic aciduria. J Inherit Metab Dis 2021; 44:1330-1342. [PMID: 34297429 DOI: 10.1002/jimd.12419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/23/2022]
Abstract
Propionic aciduria (PA) is caused by deficiency of the mitochondrial enzyme propionyl-CoA carboxylase (PCC). Due to inefficient propionate catabolism patients are endangered by life-threatening ketoacidotic crisis. Protein and amino acid restriction are major therapeutic pillars. However, long-term complications like neurological deterioration and cardiac abnormalities cannot be prevented. Chronic kidney disease (CKD), which is a well-known characteristic of methylmalonic aciduria two enzymatic steps downstream from PCC, has been recognized as a novel late-onset complication in PA. The pathophysiology of CKD in PA is unclear. We investigated mitochondrial structure and metabolism in human renal tubular cells of healthy controls and PA patients. The cells were exposed to either standard cell culture conditions (NT), high protein (HP) or high concentrations of isoleucine and valine (I/V). Mitochondrial morphology changed to condensed, fractured morphology in PA cells irrespective of the cell culture medium. HP and I/V exposure, however, potentiated oxidative stress in PA cells. Mitochondrial mass was enriched in PA cells, and further increased by HP and I/V exposure suggesting a need for compensation. Alterations in the tricarboxylic acid cycle intermediates and accumulation of medium- and long-chain acylcarnitines pointed to altered mitochondrial energy metabolism. Mitophagy was silenced while autophagy as cellular defense mechanisms was highly active in PA cells. The data demonstrate that PA is associated with renal mitochondrial damage which is aggravated by protein and I/V load. Preservation of mitochondrial energy homeostasis in renal cells may be a potential future therapeutic target.
Collapse
Affiliation(s)
- Anke Schumann
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Véronique Belche
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Kristin Schaller
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Sarah C Grünert
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Stefan Kölker
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Luciana Hannibal
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Laboratory of Clinical Biochemistry and Metabolism, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Ute Spiekerkoetter
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
19
|
Luciani A, Denley MCS, Govers LP, Sorrentino V, Froese DS. Mitochondrial disease, mitophagy, and cellular distress in methylmalonic acidemia. Cell Mol Life Sci 2021; 78:6851-6867. [PMID: 34524466 PMCID: PMC8558192 DOI: 10.1007/s00018-021-03934-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/18/2021] [Accepted: 08/30/2021] [Indexed: 01/09/2023]
Abstract
Mitochondria—the intracellular powerhouse in which nutrients are converted into energy in the form of ATP or heat—are highly dynamic, double-membraned organelles that harness a plethora of cellular functions that sustain energy metabolism and homeostasis. Exciting new discoveries now indicate that the maintenance of this ever changing and functionally pleiotropic organelle is particularly relevant in terminally differentiated cells that are highly dependent on aerobic metabolism. Given the central role in maintaining metabolic and physiological homeostasis, dysregulation of the mitochondrial network might therefore confer a potentially devastating vulnerability to high-energy requiring cell types, contributing to a broad variety of hereditary and acquired diseases. In this Review, we highlight the biological functions of mitochondria-localized enzymes from the perspective of understanding—and potentially reversing—the pathophysiology of inherited disorders affecting the homeostasis of the mitochondrial network and cellular metabolism. Using methylmalonic acidemia as a paradigm of complex mitochondrial dysfunction, we discuss how mitochondrial directed-signaling circuitries govern the homeostasis and physiology of specialized cell types and how these may be disturbed in disease. This Review also provides a critical analysis of affected tissues, potential molecular mechanisms, and novel cellular and animal models of methylmalonic acidemia which are being used to develop new therapeutic options for this disease. These insights might ultimately lead to new therapeutics, not only for methylmalonic acidemia, but also for other currently intractable mitochondrial diseases, potentially transforming our ability to regulate homeostasis and health.
Collapse
Affiliation(s)
- Alessandro Luciani
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8032, Zurich, Switzerland.
| | - Matthew C S Denley
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, 8032, Zurich, Switzerland
| | - Larissa P Govers
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8032, Zurich, Switzerland
| | - Vincenzo Sorrentino
- Department of Musculo-Skeletal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1015, Lausanne, Switzerland.
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, 8032, Zurich, Switzerland.
| |
Collapse
|
20
|
Wang C, Liu Y, Zhang X, Wang H, Cui Y, Zhi X, Zheng J, Wang N, Shu J, Li D, Cai C. Phenotypic and genotypic analysis of children with methylmalonic academia: A single-center study in China and a recent literature review. Clin Chim Acta 2021; 522:14-22. [PMID: 34389282 DOI: 10.1016/j.cca.2021.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Methylmalonic acidemia (MMA) is a rare inherited metabolic disease caused by methylmalonyl-CoA deficiency or cobalamin metabolism disorder. It is mainly inherited in autosomal recessive mode. According to whether combined with homocysteinemia and the causative genes, it can be divided into many different subtypes. Early diagnosis and early treatment can significantly improve the prognosis. METHODS The children with MMA diagnosed in Tianjin Children's Hospital from 2012 to 2020 were collected. All the children underwent comprehensive physical and laboratory examinations. The metabolites in blood and urine were screened by mass spectrometry. Sanger sequencing, Next-generation sequencing and methylation detection were used for gene detection. RESULTS The detection rate of MMA was 0.20% in children with high-risk of inherited metabolic diseases. The three most common clinical phenotypes of children with MMA were respiratory / metabolic acidosis, global developmental delay and anemia, which were found in 36.00%, 33.33% and 30.67% of children respectively. The most common mutations of MMACHC gene in children with cblC were c.609G > A, c.658_660delAAG and c.80A > G, with frequencies of 34.09%, 13.64% and 13.64%, respectively. CONCLUSIONS This research expands the study of phenotype and genotype of MMA in Chinese population, and can provide reference for clinical diagnosis and treatment of MMA.
Collapse
Affiliation(s)
- Chao Wang
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, China
| | - Yang Liu
- Department of Neonatology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China
| | - Xinjie Zhang
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, China
| | - Hong Wang
- Department of Neurology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China
| | - Yaqiong Cui
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, China
| | - Xiufang Zhi
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, China
| | - Jie Zheng
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, China
| | - Ning Wang
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, China
| | - Jianbo Shu
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, China.
| | - Dong Li
- Department of Neonatology, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China
| | - Chunquan Cai
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, China
| |
Collapse
|
21
|
Armstrong AJ, Collado MS, Henke BR, Olson MW, Hoang SA, Hamilton CA, Pourtaheri TD, Chapman KA, Summar MM, Johns BA, Wamhoff BR, Reardon JE, Figler RA. A novel small molecule approach for the treatment of propionic and methylmalonic acidemias. Mol Genet Metab 2021; 133:71-82. [PMID: 33741272 PMCID: PMC9109253 DOI: 10.1016/j.ymgme.2021.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/12/2021] [Accepted: 03/02/2021] [Indexed: 12/16/2022]
Abstract
Propionic Acidemia (PA) and Methylmalonic Acidemia (MMA) are inborn errors of metabolism affecting the catabolism of valine, isoleucine, methionine, threonine and odd-chain fatty acids. These are multi-organ disorders caused by the enzymatic deficiency of propionyl-CoA carboxylase (PCC) or methylmalonyl-CoA mutase (MUT), resulting in the accumulation of propionyl-coenzyme A (P-CoA) and methylmalonyl-CoA (M-CoA in MMA only). Primary metabolites of these CoA esters include 2-methylcitric acid (MCA), propionyl-carnitine (C3), and 3-hydroxypropionic acid, which are detectable in both PA and MMA, and methylmalonic acid, which is detectable in MMA patients only (Chapman et al., 2012). We deployed liver cell-based models that utilized PA and MMA patient-derived primary hepatocytes to validate a small molecule therapy for PA and MMA patients. The small molecule, HST5040, resulted in a dose-dependent reduction in the levels of P-CoA, M-CoA (in MMA) and the disease-relevant biomarkers C3, MCA, and methylmalonic acid (in MMA). A putative working model of how HST5040 reduces the P-CoA and its derived metabolites involves the conversion of HST5040 to HST5040-CoA driving the redistribution of free and conjugated CoA pools, resulting in the differential reduction of the aberrantly high P-CoA and M-CoA. The reduction of P-CoA and M-CoA, either by slowing production (due to increased demands on the free CoA (CoASH) pool) or enhancing clearance (to replenish the CoASH pool), results in a net decrease in the CoA-derived metabolites (C3, MCA and MMA (MMA only)). A Phase 2 study in PA and MMA patients will be initiated in the United States.
Collapse
Affiliation(s)
| | | | - Brad R Henke
- HemoShear Therapeutics, Inc., Charlottesville, VA, USA
| | | | | | | | | | | | | | - Brian A Johns
- HemoShear Therapeutics, Inc., Charlottesville, VA, USA
| | | | | | | |
Collapse
|
22
|
Armstrong AJ, Henke BR, Collado MS, Taylor JM, Pourtaheri TD, Dillberger JE, Roper TD, Wamhoff BR, Olson MW, Figler RA, Hoang SA, Reardon JE, Johns BA. Identification of 2,2-Dimethylbutanoic Acid (HST5040), a Clinical Development Candidate for the Treatment of Propionic Acidemia and Methylmalonic Acidemia. J Med Chem 2021; 64:5037-5048. [PMID: 33848153 DOI: 10.1021/acs.jmedchem.1c00124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Propionic acidemia (PA) and methylmalonic acidemia (MMA) are rare autosomal recessive disorders of propionyl-CoA (P-CoA) catabolism, caused by a deficiency in the enzymes P-CoA carboxylase and methylmalonyl-CoA (M-CoA) mutase, respectively. PA and MMA are classified as intoxication-type inborn errors of metabolism because the intramitochondrial accumulation of P-CoA, M-CoA, and other metabolites results in secondary inhibition of multiple pathways of intermediary metabolism, leading to organ dysfunction and failure. Herein, we describe the structure-activity relationships of a series of short-chain carboxylic acids which reduce disease-related metabolites in PA and MMA primary hepatocyte disease models. These studies culminated in the identification of 2,2-dimethylbutanoic acid (10, HST5040) as a clinical candidate for the treatment of PA and MMA. Additionally, we describe the in vitro and in vivo absorption, distribution, metabolism, and excretion profile of HST5040, data from preclinical studies, and the synthesis of the sodium salt of HST5040 for clinical trials.
Collapse
Affiliation(s)
- Allison J Armstrong
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Brad R Henke
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Maria Sol Collado
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Justin M Taylor
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Taylor D Pourtaheri
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - John E Dillberger
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Thomas D Roper
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Brian R Wamhoff
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Matthew W Olson
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Robert A Figler
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Stephen A Hoang
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - John E Reardon
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| | - Brian A Johns
- HemoShear Therapeutics Inc., 501 Locust Avenue, Charlottesville, Virginia 22902, United States
| |
Collapse
|
23
|
Validation of a multicellular tumor microenvironment system for modeling patient tumor biology and drug response. Sci Rep 2021; 11:5535. [PMID: 33692370 PMCID: PMC7946945 DOI: 10.1038/s41598-021-84612-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Lung cancer rates are rising globally and non-small cell lung cancer (NSCLC) has a five year survival rate of only 24%. Unfortunately, the development of drugs to treat cancer is severely hampered by the inefficiency of translating pre-clinical studies into clinical benefit. Thus, we sought to apply a tumor microenvironment system (TMES) to NSCLC. Using microvascular endothelial cells, lung cancer derived fibroblasts, and NSCLC tumor cells in the presence of in vivo tumor-derived hemodynamic flow and transport, we demonstrate that the TMES generates an in-vivo like biological state and predicts drug response to EGFR inhibitors. Transcriptomic and proteomic profiling indicate that the TMES recapitulates the in vivo and patient molecular biological state providing a mechanistic rationale for the predictive nature of the TMES. This work further validates the TMES for modeling patient tumor biology and drug response indicating utility of the TMES as a predictive tool for drug discovery and development and potential for use as a system for patient avatars.
Collapse
|