1
|
Fernández AC, Estrella J, Oglesbee D, Larson AA, Van Hove JLK. The clinical utility in hospital-wide use of growth differentiation factor 15 as a biomarker for mitochondrial DNA-related disorders. J Inherit Metab Dis 2025; 48:e12821. [PMID: 39582258 DOI: 10.1002/jimd.12821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/14/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024]
Abstract
Clinical recognition of primary mitochondrial disorders (PMD) is difficult due to the clinical and genetic heterogeneity. Whereas lactate has low sensitivity and specificity, in structured clinical studies growth differentiation factor 15 (GDF15) has shown promise with elevations in mitochondrial DNA (mtDNA)-related PMD, but its specificity has been questioned. In a tertiary care hospital-wide study, medical records were retrospectively reviewed from 418 cases where GDF15 levels were obtained by clinicians. Patients were classified into patients with PMD due to mtDNA-related defects (mtDNA maintenance, mtDNA deletions, and mtDNA-encoded tRNA variants), PMD due to structural defects or other nuclear causes, and in non-mitochondrial disease. Patients with liver disease or systemic critical illness were excluded. GDF15 was assayed in a clinical laboratory with a cutoff of 750 ng/L. There were 38 mtDNA-related PMD (GDF15 >750 pg/mL in 76%), 35 other nuclear DNA-encoded PMD or structural subunits (31% elevated GDF15), 309 non-mitochondrial disorders (13% elevated GDF15). Based on the highest Youden J-index, the optimal cut-off value to identify these target mtDNA-related disorders was 815 pg/mL, with sensitivity 76%, specificity 88%, positive predictive value of 41% and negative predictive value of 97%. At this optimized cutoff level, mtDNA-encoded PMD patients had elevated GDF15 in 76%, nuclear DNA-encoded PMD in 26%, and non-mitochondrial disorders in 11% of patients. Thus, in a real-life clinical setting, after excluding abnormal liver function and critical illness, GDF15 had good clinical utility increasing the odds at predicting mtDNA-related primary mitochondrial disorders 14-fold, but not for structural or other nuclear-encoded primary mitochondrial disorders.
Collapse
Affiliation(s)
- Andrea Cortés Fernández
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, Colorado, USA
| | - Jane Estrella
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, Colorado, USA
- Department of Diagnostic Genomics, PathWest Laboratory Medicine WA, Nedlands, Western Australia, Australia
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic School of Medicine, Rochester, Minnesota, USA
| | - Austin A Larson
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, Colorado, USA
| | - Johan L K Van Hove
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
2
|
Kanemaru E, Ichinose F. Essential role of sulfide oxidation in brain health and neurological disorders. Pharmacol Ther 2024; 266:108787. [PMID: 39719173 DOI: 10.1016/j.pharmthera.2024.108787] [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: 07/11/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Hydrogen sulfide (H2S) is an environmental hazard well known for its neurotoxicity. In mammalian cells, H2S is predominantly generated by transsulfuration pathway enzymes. In addition, H2S produced by gut microbiome significantly contributes to the total sulfide burden in the body. Although low levels of H2S is believed to exert various physiological functions such as neurotransmission and vasomotor control, elevated levels of H2S inhibit the activity of cytochrome c oxidase (i.e., mitochondrial complex IV), thereby impairing oxidative phosphorylation. To protect the electron transport chain from respiratory poisoning by H2S, the compound is actively oxidized to form persulfides and polysulfides by a mitochondrial resident sulfide oxidation pathway. The reaction, catalyzed by sulfide:quinone oxidoreductase (SQOR), is the initial and critical step in sulfide oxidation. The persulfide species are subsequently oxidized to sulfite, thiosulfate, and sulfate by persulfide dioxygenase (ETHE1 or SDO), thiosulfate sulfurtransferase (TST), and sulfite oxidase (SUOX). While SQOR is abundantly expressed in the colon, liver, lung, and skeletal muscle, its expression is notably low in the brains of most mammals. Consequently, the brain's limited capacity to oxidize H2S renders it particularly sensitive to the deleterious effects of H2S accumulation. Impaired sulfide oxidation can lead to fatal encephalopathy, and the overproduction of H2S has been implicated in the developmental delays observed in Down syndrome. Our recent findings indicate that the brain's limited capacity to oxidize sulfide exacerbates its sensitivity to oxygen deprivation. The beneficial effects of sulfide oxidation are likely to be mediated not only by the detoxification of H2S but also by the formation of persulfide, which exerts cytoprotective effects through multiple mechanisms. Therefore, pharmacological agents designed to scavenge H2S and/or enhance persulfide levels may offer therapeutic potential against neurological disorders characterized by impaired or insufficient sulfide oxidation or excessive H2S production.
Collapse
Affiliation(s)
- Eiki Kanemaru
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
3
|
Pizzamiglio C, Stefanetti RJ, McFarland R, Thomas N, Ransley G, Hugerth M, Grönberg A, Serrano SS, Elmér E, Hanna MG, Hansson MJ, Gorman GS, Pitceathly RDS. Optimizing rare disorder trials: a phase 1a/1b randomized study of KL1333 in adults with mitochondrial disease. Brain 2024:awae308. [PMID: 39657714 DOI: 10.1093/brain/awae308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/27/2024] [Accepted: 09/08/2024] [Indexed: 12/12/2024] Open
Abstract
Over the past two decades there has been increased interest in orphan drug development for rare diseases. However, hurdles to clinical trial design for these disorders remain. This phase 1a/1b study addressed several challenges, while evaluating the safety and tolerability of the novel oral molecule KL1333 in healthy volunteers and subjects with primary mitochondrial disease. KL1333 aims to normalize the NAD+:NADH ratio that is critical for ATP production. The trial incorporated innovative design elements with potential translatability to other rare diseases including patient involvement, adaptive design and exploratory objectives, all of which have subsequently informed the protocol of an ongoing phase 2, pivotal efficacy study of KL1333. Results indicate KL1333 is safe and well tolerated, with dose-dependent gastrointestinal side effects, and validate potential novel outcome measures in primary mitochondrial disease including the 30-s Sit to Stand, and the patient-reported fatigue scales. Importantly, the data from the trial support efficacy of KL1333 based on improvements in fatigue and functional strength and endurance. Furthermore, the study highlights the value in using phase 1 studies to capture data that helps optimize later phase efficacy trial design.
Collapse
Affiliation(s)
- Chiara Pizzamiglio
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Renae J Stefanetti
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle BRC, NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle BRC, NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Naomi Thomas
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle BRC, NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - George Ransley
- Leonard Wolfson Experimental Neurology Centre, University College London Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | | | | | - Sonia Simon Serrano
- Abliva AB, SE-223 81 Lund, Sweden
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| | - Eskil Elmér
- Abliva AB, SE-223 81 Lund, Sweden
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| | - Michael G Hanna
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Magnus J Hansson
- Abliva AB, SE-223 81 Lund, Sweden
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| | - Gráinne S Gorman
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle BRC, NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London WC1N 3BG, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| |
Collapse
|
4
|
Gao R, Gu L, Zuo W, Wang P. Long-term prognostic factors and outcomes in mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes: a clinical and biochemical marker analysis. Front Neurol 2024; 15:1491283. [PMID: 39697439 PMCID: PMC11652343 DOI: 10.3389/fneur.2024.1491283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/05/2024] [Indexed: 12/20/2024] Open
Abstract
Background MELAS (Mitochondrial Encephalomyopathy, Lactic Acidosis, and Stroke-like episodes) is a common subtype of mitochondrial encephalomyopathy. However, few studies have explored the relationship between biochemical markers and prognosis. This study aimed to explore the relationship between clinical and biochemical markers and prognosis of patients with MELAS. Methods This was a retrospective single-center study. A total of 39 MELAS patients were followed for an average of 7.3 ± 4.7 (range 1-21 years). All patients underwent detailed demographic registration, neurological examinations, biochemical and mitochondrial DNA analyses, muscle biopsy. Throughout the follow-up period, the modified Rankin Scale (mRS) scores, recurrent strokes rates, and mortality were tracked. Results All patients initially presented with stroke-like episodes. Of the 39 subjects who were followed, 8 died, primarily due to acute stroke-like episodes and status epilepticus. Univariate analysis showed a higher risk of mortality in patients with severe lactate elevation compared to those with normal and mildly elevated levels (OR = 5.714, 95% CI 1.086-30.071, p = 0.040). While the absence of anemia was associated with a lower risk of death compared to those with anemia (OR = 0.175, 95% CI 0.033-0.921, p = 0.040). In multivariate analysis, severe lactate elevation (OR = 7.279, 95% CI 1.102-48.086, p = 0.039) and anemia (OR = 0.137, 95% CI 0.021-0.908, p = 0.039) were identified as independent predictors of mortality. MRS scores were categorized as follows: 41% of patients scored 0 to 2, 38.5% scored 3 to 5, and 20.5% had a score of 6 or had died. There was a positive correlation between lactic acid levels and MRS scores (r = 0.460, p = 0.003). In contrast, hemoglobin levels were negatively correlated with MRS scores (r = -0.375, p = 0.015). Furthermore, a positive correlation was observed between MRS scores and the frequency of stroke-like episodes (r = 0.280, p = 0.042). Conclusion Our study found that the majority of patients with MELAS had poor clinical outcomes. Anemia and significantly increased lactate levels were identified as indicators of poor prognosis in MELAS. Early intervention may lead to improvements in clinical outcomes.
Collapse
Affiliation(s)
| | | | | | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Nankai University, Tianjin, China
| |
Collapse
|
5
|
Wu D, Yang S, Yuan C, Zhang K, Tan J, Guan K, Zeng H, Huang C. Targeting purine metabolism-related enzymes for therapeutic intervention: A review from molecular mechanism to therapeutic breakthrough. Int J Biol Macromol 2024; 282:136828. [PMID: 39447802 DOI: 10.1016/j.ijbiomac.2024.136828] [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: 04/23/2024] [Revised: 10/02/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Purines are ancient metabolites with established and emerging metabolic and non-metabolic signaling attributes. The expression of purine metabolism-related genes is frequently activated in human malignancies, correlating with increased cancer aggressiveness and chemoresistance. Importantly, under certain stimulating conditions, the purine biosynthetic enzymes can assemble into a metabolon called "purinosomes" to enhance purine flux. Current evidence suggests that purine flux is regulated by a complex circuit that encompasses transcriptional, post-translational, metabolic, and association-dependent regulatory mechanisms. Furthermore, purines within the tumor microenvironment modulate cancer immunity through signaling mediated by purinergic receptors. The deregulation of purine metabolism has significant metabolic consequences, particularly hyperuricemia. Herbal-based therapeutics have emerged as valuable pharmacological interventions for the treatment of hyperuricemia by inhibiting the activity of hepatic XOD, modulating the expression of renal urate transporters, and suppressing inflammatory responses. This review summarizes recent advancements in the understanding of purine metabolism in clinically relevant malignancies and metabolic disorders. Additionally, we discuss the role of herbal interventions and the interaction between the host and gut microbiota in the regulation of purine homeostasis. This information will fuel the innovation of therapeutic strategies that target the disease-associated rewiring of purine metabolism for therapeutic applications.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Shengqiang Yang
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Chenyang Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Jiachen Tan
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China.
| | - Hong Zeng
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
6
|
Huang Q, Monzel AS, Rausser S, Haahr R, Devine J, Liu CC, Kelly C, Thompson E, Kurade M, Michelson J, Li S, Engelstad K, Tanji K, Lauriola V, Wang T, Wang S, Marsland AL, Kaufman BA, St-Onge MP, Sloan R, Juster RP, Gouspillou G, Hirano M, Picard M, Trumpff C. The Energetic Stress Marker GDF15 is Induced by Acute Psychosocial Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590241. [PMID: 38659958 PMCID: PMC11042343 DOI: 10.1101/2024.04.19.590241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
GDF15 (growth differentiation factor 15) is a marker of cellular and mitochondrial energetic stress linked to physical-mental illness, aging, and mortality. Here, we describe the psychobiological regulation of plasma and saliva GDF15 in four human studies including 3,599 samples from 148 healthy individuals. We report two main observations establishing GDF15 as a novel tractable biomarker of psychosocial stress. 1) In two experimental laboratory studies, socio-evaluative stress rapidly elevates GDF15 and lactate, two molecular markers of energetic/reductive stress. 2) Similar to other stress-related metabolic hormones, we also find that saliva GDF15 exhibit a robust awakening response, being highest at the time of waking up and declining by ~42-92% within 30-45 minutes. These data position GDF15 as a dynamic biomarker of psychosocial stress accessible in human blood and saliva, pointing towards a shared psychobiological pathway linking mental and mitochondrial energetic stress. These foundational observations open the door to large-scale studies using GDF15 to non-invasively probe how acute psychosocial factors promote cellular and mitochondrial and energetic stress contributing to the stress-disease cascade across the lifespan.
Collapse
Affiliation(s)
- Qiuhan Huang
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S. Monzel
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Shannon Rausser
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Rachel Haahr
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Devine
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Cynthia C. Liu
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Catherine Kelly
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth Thompson
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Mangesh Kurade
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeremy Michelson
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Shufang Li
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| | - Kris Engelstad
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| | - Kurenai Tanji
- Department of pathology and cell biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Vincenzo Lauriola
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Tian Wang
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Shuang Wang
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Anna L Marsland
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brett A Kaufman
- Department of Medicine, Division of Cardiology, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA United States
| | - Marie-Pierre St-Onge
- Division of General Medicine and Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, USA
| | - Richard Sloan
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Robert-Paul Juster
- Department of Psychiatry and Addiction, University of Montreal, Montreal, QC, Canada
| | - Gilles Gouspillou
- Research Institute of the McGill University Health Centre, Department of Critical Care, Montréal, QC, Canada
| | - Michio Hirano
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
7
|
Zheng JY, Ji XY, Zhao AQ, Sun FY, Liu LF, Xin GZ. Mass Spectrometry Probe Combined with Machine Learning to Capture the Relationship between Metabolites and Mitochondrial Complex Activity at the Whole-Cell Level. Anal Chem 2024; 96:18195-18203. [PMID: 39484990 DOI: 10.1021/acs.analchem.4c04376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Mitochondrial complex activity controls a multitude of physiological processes by regulating the cellular metabolism. Current methods for evaluating mitochondrial complex activity mainly focus on single metabolic reactions within mitochondria. These methods often require fresh samples in large quantities for mitochondria purification or intact mitochondrial membranes for real-time monitoring. Confronting these limitations, we shifted the analytical perspective toward interactive metabolic networks at the whole-cell level to reflect mitochondrial complex activity. To this end, we compiled a panel of mitochondrial respiratory chain-mapped metabolites (MRCMs), whose perturbations theoretically provide an overall reflection on mitochondrial complex activity. By introducing N-dimethyl-p-phenylenediamine and N-methyl-p-phenylenediamine as a pair of mass spectrometry probes, an ultraperformance liquid chromatography-tandem mass spectrometry method with high sensitivity (LLOQ as low as 0.2 fmol) was developed to obtain accurate quantitative data of MRCMs. Machine learning was then combined to capture the relationship between MRCMs and mitochondrial complex activity. Using Complex I as a proof-of-concept, we identified NADH, alanine, and phosphoenolpyruvate as metabolites associated with Complex I activity based on the whole-cell level. The effectiveness of using their concentrations to reflect Complex I activity was further validated in external data sets. Hence, by capturing the relationship between metabolites and mitochondrial complex activity at the whole-cell level, this study explores a novel analytical paradigm for the interrogation of mitochondrial complex activity, offering a favorable complement to existing methods particularly when sample quantities, type, and treatment timeliness pose challenges. More importantly, it shifts the focus from individual metabolic reactions within mitochondria to a more comprehensive view of an interactive metabolic network, which should serve as a promising direction for future research into the functional architecture between mitochondrial complexes and metabolites.
Collapse
Affiliation(s)
- Jia-Yi Zheng
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Xiao-Yuan Ji
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - An-Qi Zhao
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Fang-Yuan Sun
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Li-Fang Liu
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Gui-Zhong Xin
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| |
Collapse
|
8
|
Berra L, Medeiros KJ, Marrazzo F, Patel S, Imber D, Rezoagli E, Yu B, Sonny A, Bittner EA, Fisher D, Chipman D, Sharma R, Shah H, Gray BE, Harris NS, Ichinose F, Mootha VK. Feasibility of Delivering 5-Day Normobaric Hypoxia Breathing in a Hospital Setting. Respir Care 2024; 69:1400-1408. [PMID: 39079724 PMCID: PMC11549621 DOI: 10.4187/respcare.11928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
BACKGROUND Beneficial effects of breathing at [Formula: see text] < 0.21 on disease outcomes have been reported in previous preclinical and clinical studies. However, the safety and intra-hospital feasibility of breathing hypoxic gas for 5 d have not been established. In this study, we examined the physiologic effects of breathing a gas mixture with [Formula: see text] as low as 0.11 in 5 healthy volunteers. METHODS All 5 subjects completed the study, spending 5 consecutive days in a hypoxic tent, where the ambient oxygen level was lowered in a stepwise manner over 5 d, from [Formula: see text] of 0.16 on the first day to [Formula: see text] of 0.11 on the fifth day of the study. All the subjects returned to an environment at room air on the sixth day. The subjects' [Formula: see text], heart rate, and breathing frequency were continuously recorded, along with daily blood sampling, neurologic evaluations, transthoracic echocardiography, and mental status assessments. RESULTS Breathing hypoxia concentration dependently caused profound physiologic changes, including decreased [Formula: see text] and increased heart rate. At [Formula: see text] of 0.14, the mean [Formula: see text] was 92%; at [Formula: see text] of 0.13, the mean [Formula: see text] was 93%; at [Formula: see text] of 0.12, the mean [Formula: see text] was 88%; at [Formula: see text] of 0.11, the mean [Formula: see text] was 85%; and, finally, at an [Formula: see text] of 0.21, the mean [Formula: see text] was 98%. These changes were accompanied by increased erythropoietin levels and reticulocyte counts in blood. All 5 subjects concluded the study with no adverse events. No subjects exhibited signs of mental status changes or pulmonary hypertension. CONCLUSIONS Results of the current physiologic study suggests that, within a hospital setting, delivering [Formula: see text] as low as 0.11 is feasible and safe in healthy subjects, and provides the foundation for future studies in which therapeutic effects of hypoxia breathing are tested.
Collapse
Affiliation(s)
- Lorenzo Berra
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts.
| | - Kyle J Medeiros
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Francesco Marrazzo
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Sarvagna Patel
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - David Imber
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Emanuele Rezoagli
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Binglan Yu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Abraham Sonny
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Edward A Bittner
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Daniel Fisher
- Department of Respiratory Care, Massachusetts General Hospital, Boston, Massachusetts
| | - Daniel Chipman
- Department of Respiratory Care, Massachusetts General Hospital, Boston, Massachusetts
| | - Rohit Sharma
- Harvard Medical School, Boston, Massachusetts. Department of Systems Biology, Massachusetts General Hospital, Boston, Massachusetts. Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Hardik Shah
- Harvard Medical School, Boston, Massachusetts. Department of Systems Biology, Massachusetts General Hospital, Boston, Massachusetts. Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts. Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Brianna E Gray
- Translational and Clinical Research Centers, Massachusetts General Hospital, Boston, Massachusetts
| | - N Stuart Harris
- Harvard Medical School, Boston, Massachusetts. Division of Wilderness Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - Vamsi K Mootha
- Harvard Medical School, Boston, Massachusetts. Department of Systems Biology, Massachusetts General Hospital, Boston, Massachusetts. Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
9
|
Correia SP, Moedas MF, Taylor LS, Naess K, Lim AZ, McFarland R, Kazior Z, Rumyantseva A, Wibom R, Engvall M, Bruhn H, Lesko N, Végvári Á, Käll L, Trost M, Alston CL, Freyer C, Taylor RW, Wedell A, Wredenberg A. Quantitative proteomics of patient fibroblasts reveal biomarkers and diagnostic signatures of mitochondrial disease. JCI Insight 2024; 9:e178645. [PMID: 39288270 PMCID: PMC11530135 DOI: 10.1172/jci.insight.178645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUNDMitochondrial diseases belong to the group of inborn errors of metabolism (IEM), with a prevalence of 1 in 2,000-5,000 individuals. They are the most common form of IEM, but, despite advances in next-generation sequencing technologies, almost half of the patients are left genetically undiagnosed.METHODSWe investigated a cohort of 61 patients with defined mitochondrial disease to improve diagnostics, identify biomarkers, and correlate metabolic pathways to specific disease groups. Clinical presentations were structured using human phenotype ontology terms, and mass spectrometry-based proteomics was performed on primary fibroblasts. Additionally, we integrated 6 patients carrying variants of uncertain significance (VUS) to test proteomics as a diagnostic expansion.RESULTSProteomic profiles from patient samples could be classified according to their biochemical and genetic characteristics, with the expression of 5 proteins (GPX4, MORF4L1, MOXD1, MSRA, and TMED9) correlating with the disease cohort, thus acting as putative biomarkers. Pathway analysis showed a deregulation of inflammatory and mitochondrial stress responses. This included the upregulation of glycosphingolipid metabolism and mitochondrial protein import, as well as the downregulation of arachidonic acid metabolism. Furthermore, we could assign pathogenicity to a VUS in MRPS23 by demonstrating the loss of associated mitochondrial ribosome subunits.CONCLUSIONWe established mass spectrometry-based proteomics on patient fibroblasts as a viable and versatile tool for diagnosing patients with mitochondrial disease.FUNDINGThe NovoNordisk Foundation, Knut and Alice Wallenberg Foundation, Wellcome Centre for Mitochondrial Research, UK Medical Research Council, and the UK NHS Highly Specialised Service for Rare Mitochondrial Disorders of Adults and Children.
Collapse
Affiliation(s)
- Sandrina P. Correia
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Marco F. Moedas
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Lucie S. Taylor
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Karin Naess
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Albert Z. Lim
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Robert McFarland
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Zuzanna Kazior
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anastasia Rumyantseva
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rolf Wibom
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Engvall
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Helene Bruhn
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Nicole Lesko
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ákos Végvári
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Lukas Käll
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden
| | - Matthias Trost
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Laboratory for Biomedical Mass Spectrometry, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Charlotte L. Alston
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Christoph Freyer
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Robert W. Taylor
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Anna Wedell
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Wredenberg
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Su X, Cheung CYY, Zhong J, Ru Y, Fong CHY, Lee CH, Liu Y, Cheung CKY, Lam KSL, Xu A, Cai Z. Ten metabolites-based algorithm predicts the future development of type 2 diabetes in Chinese. J Adv Res 2024; 64:131-142. [PMID: 38030128 PMCID: PMC11464468 DOI: 10.1016/j.jare.2023.11.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023] Open
Abstract
INTRODUCTION Type 2 diabetes (T2D) is a heterogeneous metabolic disease with large variations in the relative contributions of insulin resistance and β-cell dysfunction across different glucose tolerance subgroups and ethnicities. A more precise yet feasible approach to categorize risk preceding T2D onset is urgently needed. This study aimed to identify potential metabolic biomarkers that could contribute to the development of T2D and investigate whether their impact on T2D is mediated through insulin resistance and β-cell dysfunction. METHODS A non-targeted metabolomic analysis was performed in plasma samples of 196 incident T2D cases and 196 age- and sex-matched non-T2D controls recruited from a long-term prospective Chinese community-based cohort with a follow-up period of ∼ 16 years. RESULTS Metabolic profiles revealed profound perturbation of metabolomes before T2D onset. Overall metabolic shifts were strongly associated with insulin resistance rather than β-cell dysfunction. In addition, 188 out of the 578 annotated metabolites were associated with insulin resistance. Bi-directional mediation analysis revealed putative causal relationships among the metabolites, insulin resistance and T2D risk. We built a machine-learning based prediction model, integrating the conventional clinical risk factors (age, BMI, TyG index and 2hG) and 10 metabolites (acetyl-tryptophan, kynurenine, γ-glutamyl-phenylalanine, DG(18:2/22:6), DG(38:7), LPI(18:2), LPC(P-16:0), LPC(P-18:1), LPC(P-20:0) and LPE(P-20:0)) (AUROC = 0.894, 5.6% improvement comparing to the conventional clinical risk model), that successfully predicts the development of T2D. CONCLUSIONS Our findings support the notion that the metabolic changes resulting from insulin resistance, rather than β-cell dysfunction, are the primary drivers of T2D in Chinese adults. Metabolomes as a valuable phenotype hold potential clinical utility in the prediction of T2D.
Collapse
Affiliation(s)
- Xiuli Su
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Chloe Y Y Cheung
- Department of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Junda Zhong
- Department of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Yi Ru
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Carol H Y Fong
- Department of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Chi-Ho Lee
- Department of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Yan Liu
- Department of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Cynthia K Y Cheung
- Department of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Karen S L Lam
- Department of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.
| | - Aimin Xu
- Department of Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China.
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
11
|
Kelly C, Trumpff C, Acosta C, Assuras S, Baker J, Basarrate S, Behnke A, Bo K, Bobba-Alves N, Champagne FA, Conklin Q, Cross M, De Jager P, Engelstad K, Epel E, Franklin SG, Hirano M, Huang Q, Junker A, Juster RP, Kapri D, Kirschbaum C, Kurade M, Lauriola V, Li S, Liu CC, Liu G, McEwen B, McGill MA, McIntyre K, Monzel AS, Michelson J, Prather AA, Puterman E, Rosales XQ, Shapiro PA, Shire D, Slavich GM, Sloan RP, Smith JLM, Spann M, Spicer J, Sturm G, Tepler S, de Schotten MT, Wager TD, Picard M. A platform to map the mind-mitochondria connection and the hallmarks of psychobiology: the MiSBIE study. Trends Endocrinol Metab 2024; 35:884-901. [PMID: 39389809 PMCID: PMC11555495 DOI: 10.1016/j.tem.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 10/12/2024]
Abstract
Health emerges from coordinated psychobiological processes powered by mitochondrial energy transformation. But how do mitochondria regulate the multisystem responses that shape resilience and disease risk across the lifespan? The Mitochondrial Stress, Brain Imaging, and Epigenetics (MiSBIE) study was established to address this question and determine how mitochondria influence the interconnected neuroendocrine, immune, metabolic, cardiovascular, cognitive, and emotional systems among individuals spanning the spectrum of mitochondrial energy transformation capacity, including participants with rare mitochondrial DNA (mtDNA) lesions causing mitochondrial diseases (MitoDs). This interdisciplinary effort is expected to generate new insights into the pathophysiology of MitoDs, provide a foundation to develop novel biomarkers of human health, and integrate our fragmented knowledge of bioenergetic, brain-body, and mind-mitochondria processes relevant to medicine and public health.
Collapse
Affiliation(s)
- Catherine Kelly
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Carlos Acosta
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephanie Assuras
- Department of Clinical Neuropsychology, Division of Cognitive Neuroscience, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Baker
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Sophia Basarrate
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Alexander Behnke
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA; Clinical and Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany
| | - Ke Bo
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Natalia Bobba-Alves
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Quinn Conklin
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Marissa Cross
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Philip De Jager
- Center for Translational and Computational Neuroimmunology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kris Engelstad
- H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia Translational Neuroscience Initiative, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Elissa Epel
- Weill Institute for Neurosciences, Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Soah G Franklin
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Michio Hirano
- H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia Translational Neuroscience Initiative, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Qiuhan Huang
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Alex Junker
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Robert-Paul Juster
- Department of Psychiatry and Addiction, University of Montreal, Montreal, Quebec, Canada
| | - Darshana Kapri
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Clemens Kirschbaum
- Faculty of Psychology, Institute of Biopsychology, Technical University Dresden, Dresden, Germany
| | - Mangesh Kurade
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Vincenzo Lauriola
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Shufang Li
- H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia Translational Neuroscience Initiative, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Cynthia C Liu
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Grace Liu
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Bruce McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Marlon A McGill
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Kathleen McIntyre
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S Monzel
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeremy Michelson
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Aric A Prather
- Weill Institute for Neurosciences, Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Eli Puterman
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiomara Q Rosales
- H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia Translational Neuroscience Initiative, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Peter A Shapiro
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA; Consultation-Liaison Psychiatry, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - David Shire
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Richard P Sloan
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Janell L M Smith
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Marisa Spann
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Julie Spicer
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel Sturm
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Sophia Tepler
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behavior Laboratory, Paris, France; Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA University of Bordeaux, Bordeaux, France
| | - Tor D Wager
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA; H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia Translational Neuroscience Initiative, Department of Neurology, Columbia University Medical Center, New York, NY, USA; Robert N. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
12
|
Shaulson ED, Cohen AA, Picard M. The brain-body energy conservation model of aging. NATURE AGING 2024; 4:1354-1371. [PMID: 39379694 DOI: 10.1038/s43587-024-00716-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 09/04/2024] [Indexed: 10/10/2024]
Abstract
Aging involves seemingly paradoxical changes in energy metabolism. Molecular damage accumulation increases cellular energy expenditure, yet whole-body energy expenditure remains stable or decreases with age. We resolve this apparent contradiction by positioning the brain as the mediator and broker in the organismal energy economy. As somatic tissues accumulate damage over time, costly intracellular stress responses are activated, causing aging or senescent cells to secrete cytokines that convey increased cellular energy demand (hypermetabolism) to the brain. To conserve energy in the face of a shrinking energy budget, the brain deploys energy conservation responses, which suppress low-priority processes, producing fatigue, physical inactivity, blunted sensory capacities, immune alterations and endocrine 'deficits'. We term this cascade the brain-body energy conservation (BEC) model of aging. The BEC outlines (1) the energetic cost of cellular aging, (2) how brain perception of senescence-associated hypermetabolism may drive the phenotypic manifestations of aging and (3) energetic principles underlying the modifiability of aging trajectories by stressors and geroscience interventions.
Collapse
Affiliation(s)
- Evan D Shaulson
- Department of Psychiatry, Division of Behavioral Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Alan A Cohen
- Robert N. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- Robert N. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA.
- Department of Neurology, H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia Translational Neuroscience Initiative, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
13
|
Hou M, Yuan J, Dong X, Wang Y, Yang S, Gao J. Engineering Oxygen-Independent NADH Oxidase Integrated with Electrocatalytic FAD Cofactor Regeneration. JACS AU 2024; 4:3581-3592. [PMID: 39328752 PMCID: PMC11423319 DOI: 10.1021/jacsau.4c00528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/28/2024]
Abstract
An electrochemically mediated enzyme process for nicotinamide adenine dinucleotide (NADH) oxidation and biosensing has been developed in which the oxygen-dependent activities of wild-type NADH oxidase are replaced by electrochemical regeneration of the flavin adenine dinucleotide (FAD) cofactor in the active site. Consequently, the present bioelectrocatalysis does not rely on a continuous oxygen supply through bubbling air or pure oxygen in biosynthetic applications, which reduces enzyme stability. The coupled electrochemical and enzymatic catalysis is achieved through a combination of enzyme immobilization on the electrode and electrochemical oxidation of FADH2 in the active site mediated by the electron transfer mediator ferrocene carboxylic acid (FcCA). Furthermore, to minimize the effect of dissolved oxygen when the electrocatalytic process is exposed to air, we successfully designed mutations at the Leu40 and Cys42 sites of Leuconostoc mesenteroides (LmNOx) to block the oxygen passage into the active site and to eliminate the native FAD cofactor regeneration half-reaction. The engineered enzymes, whose activities are significantly reduced or inactive in solution, are electrocatalytically active toward conversion of NADH to NAD+, demonstrating successful FAD cofactor regeneration in the active site via electrochemistry. Finally, we developed two highly responsive electrochemical biosensors for NADH detection which has a superior substrate specific to standard detectors using metal electrodes, and comparable detection range and detection limit (1-3 μM).
Collapse
Affiliation(s)
- Mengjie Hou
- School
of Chemical Biology and Biotechnology, Peking
University Shenzhen Graduate School, Shenzhen 518055, China
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Shenzhen 518055, China
| | - Jing Yuan
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Shenzhen 518055, China
| | - Xinyu Dong
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Shenzhen 518055, China
| | - Yingjie Wang
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Shenzhen 518055, China
| | - Shihe Yang
- School
of Advanced Materials, Peking University
Shenzhen Graduate School, Shenzhen 518055, China
- Institute
of Biomedical Engineering, Shenzhen Bay
Laboratory, Shenzhen 518055, China
| | - Jiali Gao
- School
of Chemical Biology and Biotechnology, Peking
University Shenzhen Graduate School, Shenzhen 518055, China
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Shenzhen 518055, China
- Department
of Chemistry and Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
14
|
Löding S, Antti H, Sjöberg RL, Melin B, Björkblom B. Blood based metabolic markers of glioma from pre-diagnosis to surgery. Sci Rep 2024; 14:20680. [PMID: 39237693 PMCID: PMC11377417 DOI: 10.1038/s41598-024-71375-6] [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: 05/24/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
Gliomas are highly complex and metabolically active brain tumors associated with poor prognosis. Recent reports have found altered levels of blood metabolites during early tumor development, suggesting that tumor development could be detected several years before clinical manifestation. In this study, we performed metabolite analyses of blood samples collected from healthy controls and future glioma patients, up to eight years before glioma diagnosis, and on the day of glioma surgery. We discovered that metabolites related to early glioma development were associated with an increased energy turnover, as highlighted by elevated levels of TCA-related metabolites such as fumarate, malate, lactate and pyruvate in pre-diagnostic cases. We also found that metabolites related to glioma progression at surgery were primarily high levels of amino acids and metabolites of amino acid catabolism, with elevated levels of 11 amino acids and two branched-chain alpha-ketoacids, ketoleucine and ketoisoleucine. High amino acid turnover in glioma tumor tissue is currently utilized for PET imaging, diagnosis and delineation of tumor margins. By examining blood-based metabolic progression patterns towards disease onset, we demonstrate that this high amino acid turnover is also detectable in a simple blood sample. These findings provide additional insight of metabolic alterations during glioma development and progression.
Collapse
Affiliation(s)
- Sebastian Löding
- Department of Chemistry, Umeå University, Linnaeus väg 10, 901 87, Umeå, Sweden.
| | - Henrik Antti
- Department of Chemistry, Umeå University, Linnaeus väg 10, 901 87, Umeå, Sweden
| | - Rickard L Sjöberg
- Department of Clinical Science, Neurosciences, Umeå University, 901 85, Umeå, Sweden
| | - Beatrice Melin
- Department of Diagnostics and Intervention, Oncology, Umeå University, 901 87, Umeå, Sweden
| | - Benny Björkblom
- Department of Chemistry, Umeå University, Linnaeus väg 10, 901 87, Umeå, Sweden.
| |
Collapse
|
15
|
Wadsworth BJ, Leiwe M, Minogue EA, Cunha PP, Engman V, Brombach C, Asvestis C, Sah-Teli SK, Marklund E, Karppinen P, Ruas JL, Rundqvist H, Lanner JT, Johnson RS. A 2-hydroxybutyrate-mediated feedback loop regulates muscular fatigue. eLife 2024; 12:RP92707. [PMID: 39226092 PMCID: PMC11371357 DOI: 10.7554/elife.92707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Several metabolites have been shown to have independent and at times unexpected biological effects outside of their metabolic pathways. These include succinate, lactate, fumarate, and 2-hydroxyglutarate. 2-Hydroxybutyrate (2HB) is a byproduct of endogenous cysteine synthesis, produced during periods of cellular stress. 2HB rises acutely after exercise; it also rises during infection and is also chronically increased in a number of metabolic disorders. We show here that 2HB inhibits branched-chain aminotransferase enzymes, which in turn triggers a SIRT4-dependent shift in the compartmental abundance of protein ADP-ribosylation. The 2HB-induced decrease in nuclear protein ADP-ribosylation leads to a C/EBPβ-mediated transcriptional response in the branched-chain amino acid degradation pathway. This response to 2HB exposure leads to an improved oxidative capacity in vitro. We found that repeated injection with 2HB can replicate the improvement to oxidative capacity that occurs following exercise training. Together, we show that 2-HB regulates fundamental aspects of skeletal muscle metabolism.
Collapse
Affiliation(s)
- Brennan J Wadsworth
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Marina Leiwe
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Eleanor A Minogue
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| | - Pedro P Cunha
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| | - Viktor Engman
- Department of Physiology and Pharmacology, Karolinska InstituteStockholmSweden
| | - Carolin Brombach
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Christos Asvestis
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Shiv K Sah-Teli
- Faculty of Medical Biochemistry and Molecular Biology, University of OuluOuluFinland
- Department of Biochemistry, University of CambridgeCambridgeUnited Kingdom
| | - Emilia Marklund
- Department of Cell and Molecular Biology, Karolinska InstituteStockholmSweden
| | - Peppi Karppinen
- Faculty of Medical Biochemistry and Molecular Biology, University of OuluOuluFinland
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska InstituteStockholmSweden
| | - Helene Rundqvist
- Department of Laboratory Medicine, Karolinska InstitutetStockholmSweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska InstituteStockholmSweden
| | - Randall S Johnson
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
16
|
Li K, Liu P, Wang X, Zheng Z, Liu M, Ye J, Zhu L. Causal role of gut microbiota, serum metabolites, immunophenotypes in myocarditis: a mendelian randomization study. Front Genet 2024; 15:1382502. [PMID: 39280093 PMCID: PMC11392795 DOI: 10.3389/fgene.2024.1382502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Background The intricate relationship among gut microbiota, serum metabolites, and immunophenotypes may significantly impact myocarditis. However, direct causal links between these domains and myocarditis are not well understood. Methods The study performed Mendelian randomization (MR) analysis using genetic data from public sources. Exposure data included 211 gut microbiota, 486 serum metabolites, and 731 immunophenotypes from Mibiogen, the Metabolomics GWAS server, and GWAS catalog databases. Single nucleotide polymorphisms (SNPs) were selected as instrumental variables based on established criteria. Myocarditis data from GWAS (427,911 participants, 24, 180, 570 SNPs) were used as the outcome variable. MR analysis was conducted using Inverse Variance Weighting (IVW), with Cochran's Q test for heterogeneity and Egger's intercept to assess horizontal pleiotropy. Results 9 gut microbiota, 10 serum metabolites, and 2 immunophenotypes were negatively associated with myocarditis risk. In contrast, 5 gut microbiota, 12 serum metabolites, and 7 immunophenotypes were positively associated with myocarditis risk (all, P < 0.05). Sensitivity analyses confirmed the stability of these results. Conclusion This MR study suggests that gut microbiota, serum metabolites, and immunophenotypes may causally influence myocarditis risk. These findings provide genetic evidence for myocarditis etiology and could inform future precision prevention and treatment strategies.
Collapse
Affiliation(s)
- Kaiyuan Li
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, China
- Department of Cardiovascular Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Peng Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiuqi Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhipeng Zheng
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, China
- Department of Cardiovascular Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Miao Liu
- Department of Cardiovascular Medicine, Center Hospital of Shandong First Medical University, Jinan, China
| | - Jun Ye
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, China
- Department of Cardiovascular Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Li Zhu
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, China
- Department of Cardiovascular Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| |
Collapse
|
17
|
Molenaars M, Mir H, Alvarez SW, Arivazhagan L, Rosselot C, Zhan D, Park CY, Garcia-Ocana A, Schmidt AM, Possemato R. Acute inhibition of iron-sulfur cluster biosynthesis disrupts metabolic flexibility in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608291. [PMID: 39229169 PMCID: PMC11370322 DOI: 10.1101/2024.08.19.608291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Iron-sulfur clusters (ISCs) are cell-essential cofactors present in ∼60 proteins including subunits of OXPHOS complexes I-III, DNA polymerases, and iron-sensing proteins. Dysfunctions in ISC biosynthesis are associated with anemias, neurodegenerative disorders, and metabolic diseases. To assess consequences of acute ISC inhibition in a whole body setting, we developed a mouse model in which key ISC biosynthetic enzyme NFS1 can be acutely and reversibly suppressed. Contrary to in vitro ISC inhibition and pharmacological OXPHOS suppression, global NFS1 inhibition rapidly enhances lipid utilization and decreases adiposity without affecting caloric intake and physical activity. ISC proteins decrease, including key proteins involved in OXPHOS (SDHB), lipoic acid synthesis (LIAS), and insulin mRNA processing (CDKAL1), causing acute metabolic inflexibility. Age-related metabolic changes decelerate loss of adiposity substantially prolonged survival of mice with NFS1 inhibition. Thus, the observation that ISC metabolism impacts organismal fuel choice will aid in understanding the mechanisms underlying ISC diseases with increased risk for diabetes. Graphical abstract Highlights Acute ISC inhibition leads to rapid loss of adiposity in miceMulti-metabolic pathway disruption upon ISC deficiency blocks energy storageNfs1 inhibition induces glucose dyshomeostasis due to ISC deficiency in β-cellsEnergy distress caused by inhibition of ISC synthesis is attenuated in aged mice.
Collapse
|
18
|
He JL, Zhao YW, Yang JL, Ju JM, Ye BQ, Huang JY, Huang ZH, Zhao WY, Zeng WF, Xia M, Liu Y. Enhanced interactions among gut mycobiomes with the deterioration of glycemic control. MED 2024; 5:909-925.e7. [PMID: 38670112 DOI: 10.1016/j.medj.2024.03.023] [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: 11/23/2023] [Revised: 02/06/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND The gut mycobiome is closely linked to health and disease; however, its role in the progression of type 2 diabetes mellitus (T2DM) remains obscure. Here, a multi-omics approach was employed to explore the role of intestinal fungi in the deterioration of glycemic control. METHODS 350 participants without hypoglycemic therapies were invited for a standard oral glucose tolerance test to determine their status of glycemic control. The gut mycobiome was identified through internal transcribed spacer sequencing, host genetics were determined by genotyping array, and plasma metabolites were measured with untargeted liquid chromatography mass spectrometry. FINDINGS The richness of fungi was higher, whereas its dissimilarity was markedly lower, in participants with T2DM. Moreover, the diversity and composition of fungi were closely associated with insulin sensitivity and pancreatic β-cell functions. With the exacerbation of glycemic control, the co-occurrence network among fungus taxa became increasingly complex, and the complexity of the interaction network was inversely associated with insulin sensitivity. Mendelian randomization analysis further demonstrated that the Archaeorhizomycetes class, Fusarium genus, and Neoascochyta genus were causally linked to impaired glucose metabolism. Furthermore, integrative analysis with metabolomics showed that increased 4-hydroxy-2-oxoglutaric acid, ketoleucine, lysophosphatidylcholine (20:3/0:0), and N-lactoyl-phenylalanine, but decreased lysophosphatidylcholine (O-18:2), functioned as key molecules linking the adverse effect of Fusarium genus on insulin sensitivity. CONCLUSIONS Our study uncovers a strong association between disturbance in gut fungi and the progression of T2DM and highlights the potential of targeting the gut mycobiome for the management of T2DM. FUNDINGS This study was supported by MOST and NSFC of China.
Collapse
Affiliation(s)
- Jia-Lin He
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ya-Wen Zhao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jia-Lu Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jing-Meng Ju
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Bing-Qi Ye
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jing-Yi Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhi-Hao Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wan-Ying Zhao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wei-Feng Zeng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China.
| | - Yan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China.
| |
Collapse
|
19
|
Tamaroff J, Nguyen S, Wilson NE, Stefanovski D, Xiao R, Scattergood T, Capiola C, Schur GM, Dunn J, Dedio A, Wade K, Shah H, Sharma R, Mootha VK, Kelly A, Lin KY, Lynch DR, Reddy R, Rickels MR, McCormack SE. Insulin sensitivity and insulin secretion in adults with Friedreich's Ataxia: the role of skeletal muscle. J Clin Endocrinol Metab 2024:dgae545. [PMID: 39109797 DOI: 10.1210/clinem/dgae545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/28/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Friedreich's Ataxia (FRDA) is a multi-system disorder caused by frataxin deficiency. FRDA-related diabetes mellitus (DM) is common. Frataxin supports skeletal muscle mitochondrial oxidative phosphorylation (OXPHOS) capacity, a mediator of insulin sensitivity. Our objective was to test the association between skeletal muscle health and insulin sensitivity and secretion in adults with FRDA without DM. METHODS Case-control study (NCT02920671). Glucose and insulin metabolism (stable-isotope oral glucose tolerance tests), body composition (dual-energy x-ray absorptiometry), physical activity (self-report), and skeletal muscle OXPHOS capacity (creatine chemical exchange saturation transfer MRI) were assessed. RESULTS Participants included 11 individuals with FRDA (4 female), median age 27y (IQR 23, 39), BMI 26.9kg/m2 (24.1, 29.4), and 24 controls (11 female), 29y (26, 39), 24.4kg/m2 (21.8, 27.0). Fasting glucose was higher in FRDA (91 vs. 83mg/dL (5.0 vs. 4.6mmol/L), p<0.05). Individuals with FRDA had lower insulin sensitivity (WBISI 2.8 vs. 5.3, p<0.01), higher post-prandial insulin secretion (insulin secretory rate iAUC 30-180 minutes, 24,652 vs. 17,858, p<0.05), and more suppressed post-prandial endogenous glucose production (-0.9% vs. 26.9% of fasting EGP, p<0.05). In regression analyses, lower OXPHOS and inactivity explained some of the difference in insulin sensitivity. More visceral fat contributed to lower insulin sensitivity independent of FRDA. Insulin secretion accounting for sensitivity (disposition index) was not different. CONCLUSIONS Lower mitochondrial OXPHOS capacity, inactivity, and visceral adiposity contribute to lower insulin sensitivity in FRDA. Higher insulin secretion appears compensatory, and when inadequate, could herald DM. Further studies are needed to determine if muscle- or adipose-focused interventions could delay FRDA-related DM.
Collapse
Affiliation(s)
- Jaclyn Tamaroff
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
- Division of Pediatric Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| | - Sara Nguyen
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Neil E Wilson
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Darko Stefanovski
- New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA
| | - Rui Xiao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA
| | - Theresa Scattergood
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Christopher Capiola
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Gayatri Maria Schur
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
- Medical Scientist Training Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Julia Dunn
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Anna Dedio
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Kristin Wade
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Hardik Shah
- Howard Hughes Medical Institute, Department of Molecular Biology
- Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Systems Biology, Harvard Medical School, Boston, MA
- Broad Institute, Cambridge, MA
- Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL
| | - Rohit Sharma
- Howard Hughes Medical Institute, Department of Molecular Biology
- Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Systems Biology, Harvard Medical School, Boston, MA
- Broad Institute, Cambridge, MA
| | - Vamsi K Mootha
- Howard Hughes Medical Institute, Department of Molecular Biology
- Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Systems Biology, Harvard Medical School, Boston, MA
- Broad Institute, Cambridge, MA
| | - Andrea Kelly
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Kimberly Y Lin
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Division of Pediatric Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - David R Lynch
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Ravinder Reddy
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Michael R Rickels
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Shana E McCormack
- Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
20
|
Xiao S, Li VL, Long JZ. Lac-Phe (N-lactoyl-phenylalanine). Trends Endocrinol Metab 2024; 35:758-759. [PMID: 39137723 PMCID: PMC11446501 DOI: 10.1016/j.tem.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 08/15/2024]
Affiliation(s)
- Shuke Xiao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Veronica L Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA; Department of Chemistry, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; The Phil and Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
21
|
Hinton A, Claypool SM, Neikirk K, Senoo N, Wanjalla CN, Kirabo A, Williams CR. Mitochondrial Structure and Function in Human Heart Failure. Circ Res 2024; 135:372-396. [PMID: 38963864 PMCID: PMC11225798 DOI: 10.1161/circresaha.124.323800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Despite clinical and scientific advancements, heart failure is the major cause of morbidity and mortality worldwide. Both mitochondrial dysfunction and inflammation contribute to the development and progression of heart failure. Although inflammation is crucial to reparative healing following acute cardiomyocyte injury, chronic inflammation damages the heart, impairs function, and decreases cardiac output. Mitochondria, which comprise one third of cardiomyocyte volume, may prove a potential therapeutic target for heart failure. Known primarily for energy production, mitochondria are also involved in other processes including calcium homeostasis and the regulation of cellular apoptosis. Mitochondrial function is closely related to morphology, which alters through mitochondrial dynamics, thus ensuring that the energy needs of the cell are met. However, in heart failure, changes in substrate use lead to mitochondrial dysfunction and impaired myocyte function. This review discusses mitochondrial and cristae dynamics, including the role of the mitochondria contact site and cristae organizing system complex in mitochondrial ultrastructure changes. Additionally, this review covers the role of mitochondria-endoplasmic reticulum contact sites, mitochondrial communication via nanotunnels, and altered metabolite production during heart failure. We highlight these often-neglected factors and promising clinical mitochondrial targets for heart failure.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Steven M. Claypool
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Nanami Senoo
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH (C.R.W.)
| |
Collapse
|
22
|
Aguilar K, Jakubek P, Zorzano A, Wieckowski MR. Primary mitochondrial diseases: The intertwined pathophysiology of bioenergetic dysregulation, oxidative stress and neuroinflammation. Eur J Clin Invest 2024; 54:e14217. [PMID: 38644687 DOI: 10.1111/eci.14217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/23/2024]
Abstract
OBJECTIVES AND SCOPE Primary mitochondrial diseases (PMDs) are rare genetic disorders resulting from mutations in genes crucial for effective oxidative phosphorylation (OXPHOS) that can affect mitochondrial function. In this review, we examine the bioenergetic alterations and oxidative stress observed in cellular models of primary mitochondrial diseases (PMDs), shedding light on the intricate complexity between mitochondrial dysfunction and cellular pathology. We explore the diverse cellular models utilized to study PMDs, including patient-derived fibroblasts, induced pluripotent stem cells (iPSCs) and cybrids. Moreover, we also emphasize the connection between oxidative stress and neuroinflammation. INSIGHTS The central nervous system (CNS) is particularly vulnerable to mitochondrial dysfunction due to its dependence on aerobic metabolism and the correct functioning of OXPHOS. Similar to other neurodegenerative diseases affecting the CNS, individuals with PMDs exhibit several neuroinflammatory hallmarks alongside neurodegeneration, a pattern also extensively observed in mouse models of mitochondrial diseases. Based on histopathological analysis of postmortem human brain tissue and findings in mouse models of PMDs, we posit that neuroinflammation is not merely a consequence of neurodegeneration but a potential pathogenic mechanism for disease progression that deserves further investigation. This recognition may pave the way for novel therapeutic strategies for this group of devastating diseases that currently lack effective treatments. SUMMARY In summary, this review provides a comprehensive overview of bioenergetic alterations and redox imbalance in cellular models of PMDs while underscoring the significance of neuroinflammation as a potential driver in disease progression.
Collapse
Affiliation(s)
- Kevin Aguilar
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Patrycja Jakubek
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| |
Collapse
|
23
|
Walker MA, Li S, Livak KJ, Karaa A, Wu CJ, Mootha VK. T cell activation contributes to purifying selection against the MELAS-associated m.3243A>G pathogenic variant in blood. J Inherit Metab Dis 2024; 47:757-765. [PMID: 38499449 PMCID: PMC11251844 DOI: 10.1002/jimd.12726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/28/2023] [Accepted: 02/20/2024] [Indexed: 03/20/2024]
Abstract
T cells have been shown to maintain a lower percentage (heteroplasmy) of the pathogenic m.3243A>G variant (MT-TL1, associated with maternally inherited diabetes and deafness [MIDD] and mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes [MELAS]). The mechanism(s) underlying this purifying selection, however, remain unknown. Here we report that purified patient memory CD4+ T cells have lower bulk m.3243A>G heteroplasmy compared to naïve CD4+ T cells. In vitro activation of naïve CD4+ m.3243A>G patient T cells results in lower bulk m.3243A>G heteroplasmy after proliferation. Finally, m.3243A>G patient T cell receptor repertoire sequencing reveals relative oligoclonality compared to controls. These data support a role for T cell activation in peripheral, purifying selection against high m.3243A>G heteroplasmy T cells at the level of the cell, in a likely cell-autonomous fashion.
Collapse
Affiliation(s)
- Melissa A Walker
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Howard Hughes Medical Institute and the Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Shuqiang Li
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Amel Karaa
- Department of Pediatrics, Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Catherine J Wu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Vamsi K Mootha
- Howard Hughes Medical Institute and the Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
van Wegberg AMJ, van der Weerd JC, Engelke UFH, Coene KLM, Jahja R, Bakker SJL, Huijbregts SCJ, Wevers RA, Heiner-Fokkema MR, van Spronsen FJ. The clinical relevance of novel biomarkers as outcome parameter in adults with phenylketonuria. J Inherit Metab Dis 2024; 47:624-635. [PMID: 38556470 DOI: 10.1002/jimd.12732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024]
Abstract
Recent studies in PKU patients identified alternative biomarkers in blood using untargeted metabolomics. To test the added clinical value of these novel biomarkers, targeted metabolomics of 11 PKU biomarkers (phenylalanine, glutamyl-phenylalanine, glutamyl-glutamyl-phenylalanine, N-lactoyl-phenylalanine, N-acetyl-phenylalanine, the dipeptides phenylalanyl-phenylalanine and phenylalanyl-leucine, phenylalanine-hexose conjugate, phenyllactate, phenylpyruvate, and phenylacetate) was performed in stored serum samples of the well-defined PKU patient-COBESO cohort and a healthy control group. Serum samples of 35 PKU adults and 20 healthy age- and sex-matched controls were analyzed using ultra-high performance liquid chromatography quadrupole time-of-flight mass spectrometry. Group differences were tested using the Mann-Whitney U test. Multiple linear regression analyses were performed with these biomarkers as predictors of (neuro-)cognitive functions working memory, sustained attention, inhibitory control, and mental health. Compared to healthy controls, phenylalanine, glutamyl-phenylalanine, N-lactoyl-phenylalanine, N-acetyl-phenylalanine, phenylalanine-hexose conjugate, phenyllactate, phenylpyruvate, and phenylacetate were significant elevated in PKU adults (p < 0.001). The remaining three were below limit of detection in PKU and controls. Both phenylalanine and N-lactoyl-phenylalanine were associated with DSM-VI Attention deficit/hyperactivity (R2 = 0.195, p = 0.039 and R2 = 0.335, p = 0.002, respectively) of the ASR questionnaire. In addition, N-lactoyl-phenylalanine showed significant associations with ASR DSM-VI avoidant personality (R2 = 0.265, p = 0.010), internalizing (R2 = 0.192, p = 0.046) and externalizing problems (R2 = 0.217, p = 0.029) of the ASR questionnaire and multiple aspects of the MS2D and FI tests, reflecting working memory with R2 between 0.178 (p = 0.048) and 0.204 (p = 0.033). Even though the strength of the models was not considered strong, N-lactoyl-phenylalanine outperformed phenylalanine in its association with working memory and mental health outcomes.
Collapse
Affiliation(s)
- A M J van Wegberg
- Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, The Netherlands
| | - J C van der Weerd
- Department of Laboratory Medicine, Laboratory of Metabolic Diseases, University of Groningen, University Medical Centre Groningen, The Netherlands
| | - U F H Engelke
- Department of Human Genetics, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - K L M Coene
- Laboratory of Clinical Chemistry and Hematology, Máxima Medical Centre, Veldhoven, The Netherlands
| | - R Jahja
- Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, The Netherlands
| | - S J L Bakker
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, The Netherlands
| | - S C J Huijbregts
- Department of Clinical Child and Adolescent Studies-Neurodevelopmental Disorders, Faculty of Social Sciences, Leiden University, Leiden, The Netherlands
| | - R A Wevers
- Department of Human Genetics, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M R Heiner-Fokkema
- Department of Laboratory Medicine, Laboratory of Metabolic Diseases, University of Groningen, University Medical Centre Groningen, The Netherlands
| | - F J van Spronsen
- Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, The Netherlands
| |
Collapse
|
25
|
Liufu T, Zhao X, Yu M, Xie Z, Meng L, Lv H, Zhang W, Yuan Y, Xing G, Deng J, Wang Z. Multiomics analysis reveals serine catabolism as a potential therapeutic target for MELAS. FASEB J 2024; 38:e23742. [PMID: 38865203 DOI: 10.1096/fj.202302286rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/19/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024]
Abstract
Mitochondrial disease is a devastating genetic disorder, with mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) and m.3243A>G being the most common phenotype and genotype, respectively. The treatment for MELAS patients is still less effective. Here, we performed transcriptomic and proteomic analysis in muscle tissue of MELAS patients, and discovered that the expression of molecules involved in serine catabolism were significantly upregulated, and serine hydroxymethyltransferase 2 (SHMT2) increased significantly in both the mRNA and protein levels. The SHMT2 protein level was also increased in myoblasts with m.3243A>G mutation, which was transdifferentiated from patients derived fibroblasts, accompanying with the decreased nicotinamide adenine dinucleotide (NAD+)/reduced NAD+ (NADH) ratio and cell viability. After treating with SHMT2 inhibitor (SHIN1), the NAD+/NADH ratio and cell viability in MELAS myoblasts increased significantly. Taken together, our study indicates that enhanced serine catabolism plays an important role in the pathogenesis of MELAS and that SHIN1 can be a potential small molecule for the treatment of this disease.
Collapse
Affiliation(s)
- Tongling Liufu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Xutong Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhiying Xie
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Lingchao Meng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - He Lv
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| | - Guogang Xing
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| |
Collapse
|
26
|
Khaghani F, Hemmati M, Ebrahimi M, Salmaninejad A. Emerging Multi-omic Approaches to the Molecular Diagnosis of Mitochondrial Disease and Available Strategies for Treatment and Prevention. Curr Genomics 2024; 25:358-379. [PMID: 39323625 PMCID: PMC11420563 DOI: 10.2174/0113892029308327240612110334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 09/27/2024] Open
Abstract
Mitochondria are semi-autonomous organelles present in several copies within most cells in the human body that are controlled by the precise collaboration of mitochondrial DNA (mtDNA) and nuclear DNA (nDNA) encoding mitochondrial proteins. They play important roles in numerous metabolic pathways, such as the synthesis of adenosine triphosphate (ATP), the predominant energy substrate of the cell generated through oxidative phosphorylation (OXPHOS), intracellular calcium homeostasis, metabolite biosynthesis, aging, cell cycles, and so forth. Previous studies revealed that dysfunction of these multi-functional organelles, which may arise due to mutations in either the nuclear or mitochondrial genome, leads to a diverse group of clinically and genetically heterogeneous disorders. These diseases include neurodegenerative and metabolic disorders as well as cardiac and skeletal myopathies in both adults and newborns. The plethora of phenotypes and defects displayed leads to challenges in the diagnosis and treatment of mitochondrial diseases. In this regard, the related literature proposed several diagnostic options, such as high throughput mitochondrial genomics and omics technologies, as well as numerous therapeutic options, such as pharmacological approaches, manipulating the mitochondrial genome, increasing the mitochondria content of the affected cells, and recently mitochondrial diseases transmission prevention. Therefore, the present article attempted to review the latest advances and challenges in diagnostic and therapeutic options for mitochondrial diseases.
Collapse
Affiliation(s)
- Faeze Khaghani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
- Medical Genetic Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboobeh Hemmati
- Medical Genetic Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Ebrahimi
- Department of Animal Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Arash Salmaninejad
- Medical Genetic Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Regenerative Medicine, Organ Procurement and Transplantation Multi-Disciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
27
|
Kanemaru E, Shimoda K, Marutani E, Morita M, Miranda M, Miyazaki Y, Sinow C, Sharma R, Dong F, Bloch DB, Akaike T, Ichinose F. Exclusion of sulfide:quinone oxidoreductase from mitochondria causes Leigh-like disease in mice by impairing sulfide metabolism. J Clin Invest 2024; 134:e170994. [PMID: 38870029 PMCID: PMC11290971 DOI: 10.1172/jci170994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/11/2024] [Indexed: 06/15/2024] Open
Abstract
Leigh syndrome is the most common inherited mitochondrial disease in children and is often fatal within the first few years of life. In 2020, mutations in the gene encoding sulfide:quinone oxidoreductase (SQOR), a mitochondrial protein, were identified as a cause of Leigh syndrome. Here, we report that mice with a mutation in the gene encoding SQOR (SqorΔN/ΔN mice), which prevented SQOR from entering mitochondria, had clinical and pathological manifestations of Leigh syndrome. SqorΔN/ΔN mice had increased blood lactate levels that were associated with markedly decreased complex IV activity and increased hydrogen sulfide (H2S) levels. Because H2S is produced by both gut microbiota and host tissue, we tested whether metronidazole (a broad-spectrum antibiotic) or a sulfur-restricted diet rescues SqorΔN/ΔN mice from developing Leigh syndrome. Daily treatment with metronidazole alleviated increased H2S levels, normalized complex IV activity and blood lactate levels, and prolonged the survival of SqorΔN/ΔN mice. Similarly, a sulfur-restricted diet normalized blood lactate levels and inhibited the development of Leigh syndrome. Taken together, these observations suggest that mitochondrial SQOR is essential to prevent systemic accumulation of H2S. Metronidazole administration and a sulfur-restricted diet may be therapeutic approaches to treatment of patients with Leigh syndrome caused by mutations in SQOR.
Collapse
Affiliation(s)
- Eiki Kanemaru
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kakeru Shimoda
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eizo Marutani
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Maria Miranda
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yusuke Miyazaki
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Claire Sinow
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rohit Sharma
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Fangcong Dong
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Donald B. Bloch
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Wu K, Shieh JS, Qin L, Guo JJ. Mitochondrial mechanisms in the pathogenesis of chronic inflammatory musculoskeletal disorders. Cell Biosci 2024; 14:76. [PMID: 38849951 PMCID: PMC11162051 DOI: 10.1186/s13578-024-01259-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
Chronic inflammatory musculoskeletal disorders characterized by prolonged muscle inflammation, resulting in enduring pain and diminished functionality, pose significant challenges for the patients. Emerging scientific evidence points to mitochondrial malfunction as a pivotal factor contributing to these ailments. Mitochondria play a critical role in powering skeletal muscle activity, but in the context of persistent inflammation, disruptions in their quantity, configuration, and performance have been well-documented. Various disturbances, encompassing alterations in mitochondrial dynamics (such as fission and fusion), calcium regulation, oxidative stress, biogenesis, and the process of mitophagy, are believed to play a central role in the progression of these disorders. Additionally, unfolded protein responses and the accumulation of fatty acids within muscle cells may adversely affect the internal milieu, impairing the equilibrium of mitochondrial functioning. The structural discrepancies between different mitochondrial subsets namely, intramyofibrillar and subsarcolemmal mitochondria likely impact their metabolic capabilities and susceptibility to inflammatory influences. The release of signals from damaged mitochondria is known to incite inflammatory responses. Intriguingly, migrasomes and extracellular vesicles serve as vehicles for intercellular transfer of mitochondria, aiding in the removal of impaired mitochondria and regulation of inflammation. Viral infections have been implicated in inducing stress on mitochondria. Prolonged dysfunction of these vital organelles sustains oxidative harm, metabolic irregularities, and heightened cytokine release, impeding the body's ability to repair tissues. This review provides a comprehensive analysis of advancements in understanding changes in the intracellular environment, mitochondrial architecture and distribution, biogenesis, dynamics, autophagy, oxidative stress, cytokines associated with mitochondria, vesicular structures, and associated membranes in the context of chronic inflammatory musculoskeletal disorders. Strategies targeting key elements regulating mitochondrial quality exhibit promise in the restoration of mitochondrial function, alleviation of inflammation, and enhancement of overall outcomes.
Collapse
Affiliation(s)
- Kailun Wu
- Department of Orthopedics, The Fourth Affiliated Hospital of Soochow University/Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People's Republic of China
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Ju-Sheng Shieh
- Department of Periodontology, School of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 11490, Taiwan
| | - Ling Qin
- Musculoskeletal Research Laboratory of the Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - Jiong Jiong Guo
- Department of Orthopedics and Sports Medicine, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.
- MOE China-Europe Sports Medicine Belt and Road Joint Laboratory, Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
29
|
Long X, Liu M, Nan Y, Chen Q, Xiao Z, Xiang Y, Ying X, Sun J, Huang Q, Ai K. Revitalizing Ancient Mitochondria with Nano-Strategies: Mitochondria-Remedying Nanodrugs Concentrate on Disease Control. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308239. [PMID: 38224339 DOI: 10.1002/adma.202308239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Mitochondria, widely known as the energy factories of eukaryotic cells, have a myriad of vital functions across diverse cellular processes. Dysfunctions within mitochondria serve as catalysts for various diseases, prompting widespread cellular demise. Mounting research on remedying damaged mitochondria indicates that mitochondria constitute a valuable target for therapeutic intervention against diseases. But the less clinical practice and lower recovery rate imply the limitation of traditional drugs, which need a further breakthrough. Nanotechnology has approached favorable regiospecific biodistribution and high efficacy by capitalizing on excellent nanomaterials and targeting drug delivery. Mitochondria-remedying nanodrugs have achieved ideal therapeutic effects. This review elucidates the significance of mitochondria in various cells and organs, while also compiling mortality data for related diseases. Correspondingly, nanodrug-mediate therapeutic strategies and applicable mitochondria-remedying nanodrugs in disease are detailed, with a full understanding of the roles of mitochondria dysfunction and the advantages of nanodrugs. In addition, the future challenges and directions are widely discussed. In conclusion, this review provides comprehensive insights into the design and development of mitochondria-remedying nanodrugs, aiming to help scientists who desire to extend their research fields and engage in this interdisciplinary subject.
Collapse
Affiliation(s)
- Xingyu Long
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, 750002, P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Yuting Xiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Xiaohong Ying
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Jian Sun
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, P. R. China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410078, P. R. China
| |
Collapse
|
30
|
Pan X, Heacock ML, Abdulaziz EN, Violante S, Zuckerman AL, Shrestha N, Yao C, Goodman RP, Cross JR, Cracan V. A genetically encoded tool to increase cellular NADH/NAD + ratio in living cells. Nat Chem Biol 2024; 20:594-604. [PMID: 37884806 PMCID: PMC11045668 DOI: 10.1038/s41589-023-01460-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
Impaired redox metabolism is a key contributor to the etiology of many diseases, including primary mitochondrial disorders, cancer, neurodegeneration and aging. However, mechanistic studies of redox imbalance remain challenging due to limited strategies that can perturb redox metabolism in various cellular or organismal backgrounds. Most studies involving impaired redox metabolism have focused on oxidative stress; consequently, less is known about the settings where there is an overabundance of NADH reducing equivalents, termed reductive stress. Here we introduce a soluble transhydrogenase from Escherichia coli (EcSTH) as a novel genetically encoded tool to promote reductive stress in living cells. When expressed in mammalian cells, EcSTH, and a mitochondrially targeted version (mitoEcSTH), robustly elevated the NADH/NAD+ ratio in a compartment-specific manner. Using this tool, we determined that metabolic and transcriptomic signatures of the NADH reductive stress are cellular background specific. Collectively, our novel genetically encoded tool represents an orthogonal strategy to promote reductive stress.
Collapse
Affiliation(s)
- Xingxiu Pan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
| | - Mina L Heacock
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Calibr, The Scripps Research Institute, La Jolla, CA, USA
| | - Evana N Abdulaziz
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Process Development Associate, Amgen, Thousand Oaks, CA, USA
| | - Sara Violante
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Austin L Zuckerman
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
- Program in Mathematics and Science Education, University of California San Diego, San Diego, CA, USA
- Program in Mathematics and Science Education, San Diego State University, San Diego, USA
| | - Nirajan Shrestha
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Canglin Yao
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
| | - Russell P Goodman
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Valentin Cracan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA.
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
31
|
Gervasoni J, Primiano A, Cicchinelli M, Santucci L, Servidei S, Urbani A, Primiano G, Iavarone F. Mitochondrial Biomarkers in the Omics Era: A Clinical-Pathophysiological Perspective. Int J Mol Sci 2024; 25:4855. [PMID: 38732076 PMCID: PMC11084339 DOI: 10.3390/ijms25094855] [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: 03/27/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Mitochondrial diseases (MDs) affect 4300 individuals, with different ages of presentation and manifestation in any organ. How defects in mitochondria can cause such a diverse range of human diseases remains poorly understood. In recent years, several published research articles regarding the metabolic and protein profiles of these neurogenetic disorders have helped shed light on the pathogenetic mechanisms. By investigating different pathways in MDs, often with the aim of identifying disease biomarkers, it is possible to identify molecular processes underlying the disease. In this perspective, omics technologies such as proteomics and metabolomics considered in this review, can support unresolved mitochondrial questions, helping to improve outcomes for patients.
Collapse
Affiliation(s)
- Jacopo Gervasoni
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
| | - Aniello Primiano
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
| | - Michela Cicchinelli
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy;
| | - Lavinia Santucci
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
| | - Serenella Servidei
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Andrea Urbani
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy;
| | - Guido Primiano
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Federica Iavarone
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy;
| |
Collapse
|
32
|
Smith KK, Moreira JD, Wilson CR, Padera JO, Lamason AN, Xue L, Gopal DM, Flynn DB, Fetterman JL. A systematic review on the biochemical threshold of mitochondrial genetic variants. Genome Res 2024; 34:341-365. [PMID: 38627095 PMCID: PMC11067886 DOI: 10.1101/gr.278200.123] [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: 06/19/2023] [Accepted: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Mitochondrial DNA (mtDNA) variants cause a range of diseases from severe pediatric syndromes to aging-related conditions. The percentage of mtDNA copies carrying a pathogenic variant, variant allele frequency (VAF), must reach a threshold before a biochemical defect occurs, termed the biochemical threshold. Whether the often-cited biochemical threshold of >60% VAF is similar across mtDNA variants and cell types is unclear. In our systematic review, we sought to identify the biochemical threshold of mtDNA variants in relation to VAF by human tissue/cell type. We used controlled vocabulary terms to identify articles measuring oxidative phosphorylation (OXPHOS) complex activities in relation to VAF. We identified 76 eligible publications, describing 69, 12, 16, and 49 cases for complexes I, III, IV, and V, respectively. Few studies evaluated OXPHOS activities in diverse tissue types, likely reflective of clinical access. A number of cases with similar VAFs for the same pathogenic variant had varying degrees of residual activity of the affected complex, alluding to the presence of modifying variants. Tissues and cells with VAFs <60% associated with low complex activities were described, suggesting the possibility of a biochemical threshold of <60%. Using Kendall rank correlation tests, the VAF of the m.8993T > G variant correlated with complex V activity in skeletal muscle (τ = -0.58, P = 0.01, n = 13); however, no correlation was observed in fibroblasts (P = 0.7, n = 9). Our systematic review highlights the need to investigate the biochemical threshold over a wider range of VAFs in disease-relevant cell types to better define the biochemical threshold for specific mtDNA variants.
Collapse
Affiliation(s)
- Karan K Smith
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Jesse D Moreira
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
- Programs in Human Physiology, Department of Health Sciences, Boston University Sargent College, Boston, Massachusetts 02215, USA
| | - Callum R Wilson
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - June O Padera
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Ashlee N Lamason
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Liying Xue
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Deepa M Gopal
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - David B Flynn
- Medical Sciences and Education, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Jessica L Fetterman
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA;
| |
Collapse
|
33
|
Scott B, Day EA, O'Brien KL, Scanlan J, Cromwell G, Scannail AN, McDonnell ME, Finlay DK, Lynch L. Metformin and feeding increase levels of the appetite-suppressing metabolite Lac-Phe in humans. Nat Metab 2024; 6:651-658. [PMID: 38499765 PMCID: PMC11052712 DOI: 10.1038/s42255-024-01018-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/22/2024] [Indexed: 03/20/2024]
Abstract
Metformin, a widely used first-line treatment for type 2 diabetes (T2D), is known to reduce blood glucose levels and suppress appetite. Here we report a significant elevation of the appetite-suppressing metabolite N-lactoyl phenylalanine (Lac-Phe) in the blood of individuals treated with metformin across seven observational and interventional studies. Furthermore, Lac-Phe levels were found to rise in response to acute metformin administration and post-prandially in patients with T2D or in metabolically healthy volunteers.
Collapse
Affiliation(s)
- Barry Scott
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Emily A Day
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Katie L O'Brien
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - John Scanlan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Grace Cromwell
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Aine Ni Scannail
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marie E McDonnell
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David K Finlay
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
- Division of Endocrinology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Rogers RS, Sharma R, Shah HB, Skinner OS, Guo XA, Panda A, Gupta R, Durham TJ, Shaughnessy KB, Mayers JR, Hibbert KA, Baron RM, Thompson BT, Mootha VK. Circulating N-lactoyl-amino acids and N-formyl-methionine reflect mitochondrial dysfunction and predict mortality in septic shock. Metabolomics 2024; 20:36. [PMID: 38446263 PMCID: PMC10917846 DOI: 10.1007/s11306-024-02089-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/11/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Sepsis is a highly morbid condition characterized by multi-organ dysfunction resulting from dysregulated inflammation in response to acute infection. Mitochondrial dysfunction may contribute to sepsis pathogenesis, but quantifying mitochondrial dysfunction remains challenging. OBJECTIVE To assess the extent to which circulating markers of mitochondrial dysfunction are increased in septic shock, and their relationship to severity and mortality. METHODS We performed both full-scan and targeted (known markers of genetic mitochondrial disease) metabolomics on plasma to determine markers of mitochondrial dysfunction which distinguish subjects with septic shock (n = 42) from cardiogenic shock without infection (n = 19), bacteremia without sepsis (n = 18), and ambulatory controls (n = 19) - the latter three being conditions in which mitochondrial function, proxied by peripheral oxygen consumption, is presumed intact. RESULTS Nine metabolites were significantly increased in septic shock compared to all three comparator groups. This list includes N-formyl-L-methionine (f-Met), a marker of dysregulated mitochondrial protein translation, and N-lactoyl-phenylalanine (lac-Phe), representative of the N-lactoyl-amino acids (lac-AAs), which are elevated in plasma of patients with monogenic mitochondrial disease. Compared to lactate, the clinical biomarker used to define septic shock, there was greater separation between survivors and non-survivors of septic shock for both f-Met and the lac-AAs measured within 24 h of ICU admission. Additionally, tryptophan was the one metabolite significantly decreased in septic shock compared to all other groups, while its breakdown product kynurenate was one of the 9 significantly increased. CONCLUSION Future studies which validate the measurement of lac-AAs and f-Met in conjunction with lactate could define a sepsis subtype characterized by mitochondrial dysfunction.
Collapse
Affiliation(s)
- Robert S Rogers
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute, Cambridge, MA, USA.
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, MA, USA.
| | - Rohit Sharma
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Hardik B Shah
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Owen S Skinner
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | | | | | - Rahul Gupta
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Timothy J Durham
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Kelsey B Shaughnessy
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, MA, USA
| | - Jared R Mayers
- Division of Pulmonary and Critical Care, Brigham & Women's Hospital, Boston, MA, USA
| | - Kathryn A Hibbert
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care, Brigham & Women's Hospital, Boston, MA, USA
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, MA, USA
| | - Vamsi K Mootha
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute, Cambridge, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
35
|
Ishima T, Kimura N, Kobayashi M, Nagai R, Osaka H, Aizawa K. A Simple, Fast, Sensitive LC-MS/MS Method to Quantify NAD(H) in Biological Samples: Plasma NAD(H) Measurement to Monitor Brain Pathophysiology. Int J Mol Sci 2024; 25:2325. [PMID: 38397001 PMCID: PMC10888655 DOI: 10.3390/ijms25042325] [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: 01/19/2024] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a cofactor in redox reactions and an essential mediator of energy metabolism. The redox balance between NAD+ and NADH affects various diseases, cell differentiation, and aging, and in recent years there has been a growing need for measurement techniques with improved accuracy. However, NAD(H) measurements, representing both NAD+ and NADH, have been limited by the compound's properties. We achieved highly sensitive simultaneous measurement of NAD+ and NADH under non-ion pairing, mobile phase conditions of water, or methanol containing 5 mM ammonium acetate. These were achieved using a simple pre-treatment and 7-min analysis time. Use of the stable isotope 13C5-NAD+ as an internal standard enabled validation close to BMV criteria and demonstrated the robustness of NAD(H) determination. Measurements using this method showed that brain NAD(H) levels correlate strongly with plasma NAD(H) levels in the same mouse, indicating that NAD(H) concentrations in brain tissue are reflected in plasma. As NAD(H) is involved in various neurodegenerative diseases and cerebral ischemia, as well as brain diseases such as mitochondrial myopathies, monitoring changes in NADH levels in plasma after drug administration will be useful for development of future diagnostics and therapeutics.
Collapse
Affiliation(s)
- Tamaki Ishima
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan; (T.I.); (N.K.)
| | - Natsuka Kimura
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan; (T.I.); (N.K.)
| | - Mizuki Kobayashi
- Department of Pediatrics, Jichi Medical University, Shimotsuke 329-0498, Japan; (M.K.); (H.O.)
| | - Ryozo Nagai
- Jichi Medical University, Shimotsuke 329-0498, Japan;
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Shimotsuke 329-0498, Japan; (M.K.); (H.O.)
| | - Kenichi Aizawa
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan; (T.I.); (N.K.)
- Clinical Pharmacology Center, Jichi Medical University Hospital, Shimotsuke 329-0498, Japan
- Division of Translational Research, Clinical Research Center, Jichi Medical University Hospital, Shimotsuke 329-0498, Japan
| |
Collapse
|
36
|
Sercel AJ, Sturm G, Gallagher D, St-Onge MP, Kempes CP, Pontzer H, Hirano M, Picard M. Hypermetabolism and energetic constraints in mitochondrial disorders. Nat Metab 2024; 6:192-195. [PMID: 38337097 DOI: 10.1038/s42255-023-00968-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Affiliation(s)
- Alexander J Sercel
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, USA
| | - Gabriel Sturm
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Dympna Gallagher
- Department of Medicine, Columbia University Irving Medical Center, New York, USA
| | - Marie-Pierre St-Onge
- Division of General Medicine and Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, USA
| | | | - Herman Pontzer
- Department of Evolutionary Anthropology, Duke University, North Carolina, USA
- Duke Global Health Institute, Duke University, North Carolina, USA
| | - Michio Hirano
- Department of Neurology, H. Houston Merritt Center, Columbia University Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, USA
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, USA.
- Department of Neurology, H. Houston Merritt Center, Columbia University Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, USA.
- New York State Psychiatric Institute, New York, USA.
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA.
| |
Collapse
|
37
|
Kemperman RH, Ganetzky RD, Master SR. Development and validation of a multiplexed LC-MS/MS ketone body assay for clinical diagnostics. J Mass Spectrom Adv Clin Lab 2024; 31:49-58. [PMID: 38375486 PMCID: PMC10874984 DOI: 10.1016/j.jmsacl.2024.01.004] [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: 08/21/2023] [Revised: 12/27/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Objectives Ketone bodies (KBs) serve as important energy sources that spare glucose, providing the primary energy for cardiac muscle, skeletal muscle during aerobic exercise, and the brain during periods of catabolism. The levels and relationships between the KBs are critical indicators of metabolic health and disease. However, challenges in separating isomeric KBs and concerns about sample stability have previously limited their clinical measurement. Methods A novel 6.5-minute liquid chromatography-mass spectrometry-based assay was developed, enabling the precise measurement of alpha-, beta- and gamma-hydroxybutyrate, beta-hydroxyisobutyrate, and acetoacetate. This method was fully validated for human serum and plasma samples by investigating extraction efficiency, matrix effects, accuracy, recovery, intra- and inter-precision, linearity, lower limit of quantitation (LLOQ), carryover, specificity, stability, and more. From 107 normal samples, reference ranges were established for all analytes and the beta-hydroxybutyrate/acetoacetate ratio. Results All five analytes were adequately separated chromatographically. An extraction efficiency between 80 and 120 % was observed for all KBs. Accuracy was evaluated through spike and recovery using 10 random patient samples, with an average recovery of 85-115 % for all KBs and a coefficient of variation of ≤ 3 %. Coefficients of variation for intra- and inter-day imprecision were < 5 %, and the total imprecision was < 10 %. No significant interferences were observed. Specimens remained stable for up to 6 h on ice or 2 h at room temperature. Conclusions The developed method is highly sensitive and robust. It has been validated for use with human serum and plasma, overcoming stability concerns and providing a reliable and efficient quantitative estimation of ketone bodies.
Collapse
Affiliation(s)
| | - Rebecca D. Ganetzky
- Children’s Hospital of Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, PA, United States
| | - Stephen R. Master
- Children’s Hospital of Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, PA, United States
| |
Collapse
|
38
|
Gropman AL, Uittenbogaard MN, Chiaramello AE. Challenges and opportunities to bridge translational to clinical research for personalized mitochondrial medicine. Neurotherapeutics 2024; 21:e00311. [PMID: 38266483 PMCID: PMC10903101 DOI: 10.1016/j.neurot.2023.e00311] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024] Open
Abstract
Mitochondrial disorders are a group of rare and heterogeneous genetic diseases characterized by dysfunctional mitochondria leading to deficient adenosine triphosphate synthesis and chronic energy deficit in patients. The majority of these patients exhibit a wide range of phenotypic manifestations targeting several organ systems, making their clinical diagnosis and management challenging. Bridging translational to clinical research is crucial for improving the early diagnosis and prognosis of these intractable mitochondrial disorders and for discovering novel therapeutic drug candidates and modalities. This review provides the current state of clinical testing in mitochondrial disorders, discusses the challenges and opportunities for converting basic discoveries into clinical settings, explores the most suited patient-centric approaches to harness the extraordinary heterogeneity among patients affected by the same primary mitochondrial disorder, and describes the current outlook of clinical trials.
Collapse
Affiliation(s)
- Andrea L Gropman
- Children's National Medical Center, Division of Neurogenetics and Neurodevelopmental Pediatrics, Washington, DC 20010, USA
| | - Martine N Uittenbogaard
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Anne E Chiaramello
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|
39
|
Van Hove JL, Friederich MW, Strode DK, Van Hove RA, Miller KR, Sharma R, Shah H, Estrella J, Gabel L, Horslen S, Kohli R, Lovell MA, Miethke AG, Molleston JP, Romero R, Squires JE, Alonso EM, Guthery SL, Kamath BM, Loomes KM, Rosenthal P, Mysore KR, Cavallo LA, Valentino PL, Magee JC, Sundaram SS, Sokol RJ. Protein biomarkers GDF15 and FGF21 to differentiate mitochondrial hepatopathies from other pediatric liver diseases. Hepatol Commun 2024; 8:e0361. [PMID: 38180987 PMCID: PMC10781130 DOI: 10.1097/hc9.0000000000000361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/17/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Mitochondrial hepatopathies (MHs) are primary mitochondrial genetic disorders that can present as childhood liver disease. No recognized biomarkers discriminate MH from other childhood liver diseases. The protein biomarkers growth differentiation factor 15 (GDF15) and fibroblast growth factor 21 (FGF21) differentiate mitochondrial myopathies from other myopathies. We evaluated these biomarkers to determine if they discriminate MH from other liver diseases in children. METHODS Serum biomarkers were measured in 36 children with MH (17 had a genetic diagnosis); 38 each with biliary atresia, α1-antitrypsin deficiency, and Alagille syndrome; 20 with NASH; and 186 controls. RESULTS GDF15 levels compared to controls were mildly elevated in patients with α1-antitrypsin deficiency, Alagille syndrome, and biliary atresia-young subgroup, but markedly elevated in MH (p<0.001). FGF21 levels were mildly elevated in NASH and markedly elevated in MH (p<0.001). Both biomarkers were higher in patients with MH with a known genetic cause but were similar in acute and chronic presentations. Both markers had a strong performance to identify MH with a molecular diagnosis with the AUC for GDF15 0.93±0.04 and for FGF21 0.90±0.06. Simultaneous elevation of both markers >98th percentile of controls identified genetically confirmed MH with a sensitivity of 88% and specificity of 96%. In MH, independent predictors of survival without requiring liver transplantation were international normalized ratio and either GDF15 or FGF21 levels, with levels <2000 ng/L predicting survival without liver transplantation (p<0.01). CONCLUSIONS GDF15 and FGF21 are significantly higher in children with MH compared to other childhood liver diseases and controls and, when combined, were predictive of MH and had prognostic implications.
Collapse
Affiliation(s)
- Johan L.K. Van Hove
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Marisa W. Friederich
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Dana K. Strode
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Roxanne A. Van Hove
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Kristen R. Miller
- Section of Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Rohit Sharma
- Department of Molecular Biology and Department of Medicine, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Hardik Shah
- Department of Molecular Biology and Department of Medicine, Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jane Estrella
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Neurosciences, University of California San Diego, San Diego, California, USA
| | - Linda Gabel
- Department of Pathology and Laboratory Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Simon Horslen
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Mark A. Lovell
- Department of Pathology and Laboratory Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Alexander G. Miethke
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jean P. Molleston
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Indiana University and Riley Hospital for Children, Indianapolis, Indiana, USA
| | - Rene Romero
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, Georgia, USA
| | - James E. Squires
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Estella M. Alonso
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Ann and Robert H. Lurie Children’s Hospital, Chicago, Illinois, USA
| | - Stephen L. Guthery
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Spencer F. Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
- Intermountain Primary Children’s Hospital, University of Utah, Salt Lake City, Utah, USA
| | - Binita M. Kamath
- Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Kathleen M. Loomes
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Philip Rosenthal
- Departments of Pediatrics and Surgery, University of California San Francisco, San Francisco, California, USA
| | - Krupa R. Mysore
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, USA
| | - Laurel A. Cavallo
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas, USA
| | - Pamela L. Valentino
- Division of Gastroenterology and Hepatology, Seattle Children’s Hospital, University of Washington, Seattle, Washington, USA
| | - John C. Magee
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Shikha S. Sundaram
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Ronald J. Sokol
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
40
|
Suciu I, Delp J, Gutbier S, Suess J, Henschke L, Celardo I, Mayer TU, Amelio I, Leist M. Definition of the Neurotoxicity-Associated Metabolic Signature Triggered by Berberine and Other Respiratory Chain Inhibitors. Antioxidants (Basel) 2023; 13:49. [PMID: 38247474 PMCID: PMC10812665 DOI: 10.3390/antiox13010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
To characterize the hits from a phenotypic neurotoxicity screen, we obtained transcriptomics data for valinomycin, diethylstilbestrol, colchicine, rotenone, 1-methyl-4-phenylpyridinium (MPP), carbaryl and berberine (Ber). For all compounds, the concentration triggering neurite degeneration correlated with the onset of gene expression changes. The mechanistically diverse toxicants caused similar patterns of gene regulation: the responses were dominated by cell de-differentiation and a triggering of canonical stress response pathways driven by ATF4 and NRF2. To obtain more detailed and specific information on the modes-of-action, the effects on energy metabolism (respiration and glycolysis) were measured. Ber, rotenone and MPP inhibited the mitochondrial respiratory chain and they shared complex I as the target. This group of toxicants was further evaluated by metabolomics under experimental conditions that did not deplete ATP. Ber (204 changed metabolites) showed similar effects as MPP and rotenone. The overall metabolic situation was characterized by oxidative stress, an over-abundance of NADH (>1000% increase) and a re-routing of metabolism in order to dispose of the nitrogen resulting from increased amino acid turnover. This unique overall pattern led to the accumulation of metabolites known as biomarkers of neurodegeneration (saccharopine, aminoadipate and branched-chain ketoacids). These findings suggest that neurotoxicity of mitochondrial inhibitors may result from an ensemble of metabolic changes rather than from a simple ATP depletion. The combi-omics approach used here provided richer and more specific MoA data than the more common transcriptomics analysis alone. As Ber, a human drug and food supplement, mimicked closely the mode-of-action of known neurotoxicants, its potential hazard requires further investigation.
Collapse
Affiliation(s)
- Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Graduate School of Chemical Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Johannes Delp
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Simon Gutbier
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Julian Suess
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Lars Henschke
- Graduate School of Chemical Biology, University of Konstanz, 78464 Konstanz, Germany
- Department of Molecular Genetics, University of Konstanz, 78464 Konstanz, Germany
| | - Ivana Celardo
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Thomas U. Mayer
- Department of Molecular Genetics, University of Konstanz, 78464 Konstanz, Germany
| | - Ivano Amelio
- Division for Systems Toxicology, Department of Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| |
Collapse
|
41
|
Pan LA, Naviaux JC, Wang L, Li K, Monk JM, Lingampelly SS, Segreti AM, Bloom K, Vockley J, Tarnopolsky MA, Finegold DN, Peters DG, Naviaux RK. Metabolic features of treatment-refractory major depressive disorder with suicidal ideation. Transl Psychiatry 2023; 13:393. [PMID: 38097555 PMCID: PMC10721812 DOI: 10.1038/s41398-023-02696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/18/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
Peripheral blood metabolomics was used to gain chemical insight into the biology of treatment-refractory Major Depressive Disorder with suicidal ideation, and to identify individualized differences for personalized care. The study cohort consisted of 99 patients with treatment-refractory major depressive disorder and suicidal ideation (trMDD-SI n = 52 females and 47 males) and 94 age- and sex-matched healthy controls (n = 48 females and 46 males). The median age was 29 years (IQR 22-42). Targeted, broad-spectrum metabolomics measured 448 metabolites. Fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) were measured as biomarkers of mitochondrial dysfunction. The diagnostic accuracy of plasma metabolomics was over 90% (95%CI: 0.80-1.0) by area under the receiver operator characteristic (AUROC) curve analysis. Over 55% of the metabolic impact in males and 75% in females came from abnormalities in lipids. Modified purines and pyrimidines from tRNA, rRNA, and mRNA turnover were increased in the trMDD-SI group. FGF21 was increased in both males and females. Increased lactate, glutamate, and saccharopine, and decreased cystine provided evidence of reductive stress. Seventy-five percent of the metabolomic abnormalities found were individualized. Personalized deficiencies in CoQ10, flavin adenine dinucleotide (FAD), citrulline, lutein, carnitine, or folate were found. Pathways regulated by mitochondrial function dominated the metabolic signature. Peripheral blood metabolomics identified mitochondrial dysfunction and reductive stress as common denominators in suicidal ideation associated with treatment-refractory major depressive disorder. Individualized metabolic differences were found that may help with personalized management.
Collapse
Affiliation(s)
- Lisa A Pan
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Panomics Mental Health Initiative, Pittsburgh, PA, USA.
- New Hope Molecular, LLC, Pittsburgh, PA, USA.
- New Hope Molecular, LLC, 750 Washington Rd, Suite 19, Pittsburgh, PA, 15228, USA.
| | - Jane C Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Jonathan M Monk
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Sai Sachin Lingampelly
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Anna Maria Segreti
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kaitlyn Bloom
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jerry Vockley
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - David N Finegold
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Panomics Mental Health Initiative, Pittsburgh, PA, USA
- New Hope Molecular, LLC, Pittsburgh, PA, USA
| | - David G Peters
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Panomics Mental Health Initiative, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Pediatrics, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, USA.
| |
Collapse
|
42
|
Hao M, Qin Y, Li Y, Tang Y, Ma Z, Tan J, Jin L, Wang F, Gong X. Metabolome subtyping reveals multi-omics characteristics and biological heterogeneity in major psychiatric disorders. Psychiatry Res 2023; 330:115605. [PMID: 38006718 DOI: 10.1016/j.psychres.2023.115605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/27/2023]
Abstract
Growing evidence suggests that major psychiatric disorders (MPDs) share common etiologies and pathological processes. However, the diagnosis is currently based on descriptive symptoms, which ignores the underlying pathogenesis and hinders the development of clinical treatments. This highlights the urgency of characterizing molecular biomarkers and establishing objective diagnoses of MPDs. Here, we collected untargeted metabolomics, proteomics and DNA methylation data of 327 patients with MPDs, 131 individuals with genetic high risk and 146 healthy controls to explore the multi-omics characteristics of MPDs. First, differential metabolites (DMs) were identified and we classified MPD patients into 3 subtypes based on DMs. The subtypes showed distinct metabolomics, proteomics and DNA methylation signatures. Specifically, one subtype showed dysregulation of complement and coagulation proteins, while the DNA methylation showed abnormalities in chemical synapses and autophagy. Integrative analysis in metabolic pathways identified the important roles of the citrate cycle, sphingolipid metabolism and amino acid metabolism. Finally, we constructed prediction models based on the metabolites and proteomics that successfully captured the risks of MPD patients. Our study established molecular subtypes of MPDs and elucidated their biological heterogeneity through a multi-omics investigation. These results facilitate the understanding of pathological mechanisms and promote the diagnosis and prevention of MPDs.
Collapse
Affiliation(s)
- Meng Hao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China; Zhangjiang Fudan International Innovation Center, Fudan Zhangjiang Institute, Obstetrics and Gynecology Hospital, Human Phenome Institute, Fudan University, China
| | - Yue Qin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China; Zhangjiang Fudan International Innovation Center, Fudan Zhangjiang Institute, Obstetrics and Gynecology Hospital, Human Phenome Institute, Fudan University, China
| | - Yi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China; Zhangjiang Fudan International Innovation Center, Fudan Zhangjiang Institute, Obstetrics and Gynecology Hospital, Human Phenome Institute, Fudan University, China; International Human Phenome Institutes, Shanghai, China
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zehan Ma
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jingze Tan
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China; Zhangjiang Fudan International Innovation Center, Fudan Zhangjiang Institute, Obstetrics and Gynecology Hospital, Human Phenome Institute, Fudan University, China; International Human Phenome Institutes, Shanghai, China
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China; Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China.
| | - Xiaohong Gong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
43
|
Shastry A, Dunham-Snary K. Metabolomics and mitochondrial dysfunction in cardiometabolic disease. Life Sci 2023; 333:122137. [PMID: 37788764 DOI: 10.1016/j.lfs.2023.122137] [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: 08/01/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
Circulating metabolites are indicators of systemic metabolic dysfunction and can be detected through contemporary techniques in metabolomics. These metabolites are involved in numerous mitochondrial metabolic processes including glycolysis, fatty acid β-oxidation, and amino acid catabolism, and changes in the abundance of these metabolites is implicated in the pathogenesis of cardiometabolic diseases (CMDs). Epigenetic regulation and direct metabolite-protein interactions modulate metabolism, both within cells and in the circulation. Dysfunction of multiple mitochondrial components stemming from mitochondrial DNA mutations are implicated in disease pathogenesis. This review will summarize the current state of knowledge regarding: i) the interactions between metabolites found within the mitochondrial environment during CMDs, ii) various metabolites' effects on cellular and systemic function, iii) how harnessing the power of metabolomic analyses represents the next frontier of precision medicine, and iv) how these concepts integrate to expand the clinical potential for translational cardiometabolic medicine.
Collapse
Affiliation(s)
- Abhishek Shastry
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Kimberly Dunham-Snary
- Department of Medicine, Queen's University, Kingston, ON, Canada; Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
44
|
Di Leo V, Bernardino Gomes TM, Vincent AE. Interactions of mitochondrial and skeletal muscle biology in mitochondrial myopathy. Biochem J 2023; 480:1767-1789. [PMID: 37965929 PMCID: PMC10657187 DOI: 10.1042/bcj20220233] [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: 09/06/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023]
Abstract
Mitochondrial dysfunction in skeletal muscle fibres occurs with both healthy aging and a range of neuromuscular diseases. The impact of mitochondrial dysfunction in skeletal muscle and the way muscle fibres adapt to this dysfunction is important to understand disease mechanisms and to develop therapeutic interventions. Furthermore, interactions between mitochondrial dysfunction and skeletal muscle biology, in mitochondrial myopathy, likely have important implications for normal muscle function and physiology. In this review, we will try to give an overview of what is known to date about these interactions including metabolic remodelling, mitochondrial morphology, mitochondrial turnover, cellular processes and muscle cell structure and function. Each of these topics is at a different stage of understanding, with some being well researched and understood, and others in their infancy. Furthermore, some of what we know comes from disease models. Whilst some findings are confirmed in humans, where this is not yet the case, we must be cautious in interpreting findings in the context of human muscle and disease. Here, our goal is to discuss what is known, highlight what is unknown and give a perspective on the future direction of research in this area.
Collapse
Affiliation(s)
- Valeria Di Leo
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, U.K
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, U.K
| | - Tiago M. Bernardino Gomes
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, U.K
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, U.K
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, U.K
| | - Amy E. Vincent
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, U.K
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, U.K
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, U.K
| |
Collapse
|
45
|
Luukkonen PK, Porthan K, Ahlholm N, Rosqvist F, Dufour S, Zhang XM, Lehtimäki TE, Seppänen W, Orho-Melander M, Hodson L, Petersen KF, Shulman GI, Yki-Järvinen H. The PNPLA3 I148M variant increases ketogenesis and decreases hepatic de novo lipogenesis and mitochondrial function in humans. Cell Metab 2023; 35:1887-1896.e5. [PMID: 37909034 DOI: 10.1016/j.cmet.2023.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/26/2023] [Accepted: 10/12/2023] [Indexed: 11/02/2023]
Abstract
The PNPLA3 I148M variant is the major genetic risk factor for all stages of fatty liver disease, but the underlying pathophysiology remains unclear. We studied the effect of this variant on hepatic metabolism in homozygous carriers and non-carriers under multiple physiological conditions with state-of-the-art stable isotope techniques. After an overnight fast, carriers had higher plasma β-hydroxybutyrate concentrations and lower hepatic de novo lipogenesis (DNL) compared to non-carriers. After a mixed meal, fatty acids were channeled toward ketogenesis in carriers, which was associated with an increase in hepatic mitochondrial redox state. During a ketogenic diet, carriers manifested increased rates of intrahepatic lipolysis, increased plasma β-hydroxybutyrate concentrations, and decreased rates of hepatic mitochondrial citrate synthase flux. These studies demonstrate that homozygous PNPLA3 I148M carriers have hepatic mitochondrial dysfunction leading to reduced DNL and channeling of carbons to ketogenesis. These findings have implications for understanding why the PNPLA3 variant predisposes to progressive liver disease.
Collapse
Affiliation(s)
- Panu K Luukkonen
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA; Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Abdominal Center, Helsinki University Hospital, Helsinki, Finland.
| | - Kimmo Porthan
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Noora Ahlholm
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Fredrik Rosqvist
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford & NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK; Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Sylvie Dufour
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA; Yale Diabetes Research Center, Yale School of Medicine, New Haven, CT, USA
| | - Xian-Man Zhang
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA; Yale Diabetes Research Center, Yale School of Medicine, New Haven, CT, USA
| | - Tiina E Lehtimäki
- Department of Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Wenla Seppänen
- Department of Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Marju Orho-Melander
- Department of Clinical Sciences, Diabetes and Endocrinology, University Hospital Malmö, Lund University, Malmö, Sweden
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford & NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - Kitt Falk Petersen
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA; Yale Diabetes Research Center, Yale School of Medicine, New Haven, CT, USA
| | - Gerald I Shulman
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA; Yale Diabetes Research Center, Yale School of Medicine, New Haven, CT, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Hannele Yki-Järvinen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
46
|
Piroli GG, Manuel AM, McCain RS, Smith HH, Ozohanics O, Mellid S, Cox JH, Cotham WE, Walla MD, Cascón A, Ambrus A, Frizzell N. Defective function of α-ketoglutarate dehydrogenase exacerbates mitochondrial ATP deficits during complex I deficiency. Redox Biol 2023; 67:102932. [PMID: 37883842 PMCID: PMC10618796 DOI: 10.1016/j.redox.2023.102932] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
The NDUFS4 knockout (KO) mouse phenotype resembles the human Complex I deficiency Leigh Syndrome. The irreversible succination of protein thiols by fumarate is increased in select regions of the NDUFS4 KO brain affected by neurodegeneration. We report that dihydrolipoyllysine-residue succinyltransferase (DLST), a component of the α-ketoglutarate dehydrogenase complex (KGDHC) of the tricarboxylic acid (TCA) cycle, is succinated in the affected regions of the NDUFS4 KO brain. Succination of DLST reduced KGDHC activity in the brainstem (BS) and olfactory bulb (OB) of KO mice. The defective production of KGDHC derived succinyl-CoA resulted in decreased mitochondrial substrate level phosphorylation (SLP), further aggravating the existing oxidative phosphorylation (OXPHOS) ATP deficit. Protein succinylation, an acylation modification that requires succinyl-CoA, was reduced in the KO mice. Modeling succination of a cysteine in the spatial vicinity of the DLST active site or introduction of succinomimetic mutations recapitulates these metabolic deficits. Our data demonstrate that the biochemical deficit extends beyond impaired Complex I assembly and OXPHOS deficiency, functionally impairing select components of the TCA cycle to drive metabolic perturbations in affected neurons.
Collapse
Affiliation(s)
- Gerardo G Piroli
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - Allison M Manuel
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - Richard S McCain
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - Holland H Smith
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - Oliver Ozohanics
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Sara Mellid
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), 28029, Madrid, Spain
| | - J Hunter Cox
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - William E Cotham
- Mass Spectrometry Center, Department of Chemistry & Biochemistry, University of South Carolina, Columbia, SC, 29205, USA
| | - Michael D Walla
- Mass Spectrometry Center, Department of Chemistry & Biochemistry, University of South Carolina, Columbia, SC, 29205, USA
| | - Alberto Cascón
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), 28029, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Attila Ambrus
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Norma Frizzell
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA.
| |
Collapse
|
47
|
Vellama H, Eskla KL, Eichelmann H, Hüva A, Tennant DA, Thakker A, Roberts J, Jagomäe T, Porosk R, Laisk A, Oja V, Rämma H, Volke V, Vasar E, Luuk H. VHL-deficiency leads to reductive stress in renal cells. Free Radic Biol Med 2023; 208:1-12. [PMID: 37506952 PMCID: PMC10602395 DOI: 10.1016/j.freeradbiomed.2023.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/10/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
Heritable renal cancer syndromes (RCS) are associated with numerous chromosomal alterations including inactivating mutations in von Hippel-Lindau (VHL) gene. Here we identify a novel aspect of the phenotype in VHL-deficient human renal cells. We call it reductive stress as it is characterised by increased NADH/NAD+ ratio that is associated with impaired cellular respiration, impaired CAC activity, upregulation of reductive carboxylation of glutamine and accumulation of lipid droplets in VHL-deficient cells. Reductive stress was mitigated by glucose depletion and supplementation with pyruvate or resazurin, a redox-reactive agent. This study demonstrates for the first time that reductive stress is a part of the phenotype associated with VHL-deficiency in renal cells and indicates that the reversal of reductive stress can augment respiratory activity and CAC activity, suggesting a strategy for altering the metabolic profile of VHL-deficient tumours.
Collapse
Affiliation(s)
- Hans Vellama
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia; Centre of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kattri-Liis Eskla
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia; Centre of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia.
| | - Hillar Eichelmann
- Institute of Biomedicine and Translational Medicine, Department of Pathophysiology, University of Tartu, Tartu, Estonia; Institute of Technology, University of Tartu, Tartu, Estonia
| | - Andria Hüva
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alpesh Thakker
- Institute of Metabolism and Systems Research, 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
| | - Toomas Jagomäe
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia; Centre of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Rando Porosk
- Institute of Biomedicine and Translational Medicine, Department of Biochemistry, University of Tartu, Tartu, Estonia
| | - Agu Laisk
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Vello Oja
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Heikko Rämma
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Vallo Volke
- Institute of Biomedicine and Translational Medicine, Department of Pathophysiology, University of Tartu, Tartu, Estonia
| | - Eero Vasar
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia; Centre of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Hendrik Luuk
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, Tartu, Estonia; Centre of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
48
|
Vardar Acar N, Özgül RK. A big picture of the mitochondria-mediated signals: From mitochondria to organism. Biochem Biophys Res Commun 2023; 678:45-61. [PMID: 37619311 DOI: 10.1016/j.bbrc.2023.08.032] [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: 06/06/2023] [Revised: 08/02/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Mitochondria, well-known for years as the powerhouse and biosynthetic center of the cell, are dynamic signaling organelles beyond their energy production and biosynthesis functions. The metabolic functions of mitochondria, playing an important role in various biological events both in physiological and stress conditions, transform them into important cellular stress sensors. Mitochondria constantly communicate with the rest of the cell and even from other cells to the organism, transmitting stress signals including oxidative and reductive stress or adaptive signals such as mitohormesis. Mitochondrial signal transduction has a vital function in regulating integrity of human genome, organelles, cells, and ultimately organism.
Collapse
Affiliation(s)
- Neşe Vardar Acar
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - R Köksal Özgül
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
49
|
Du J, Zhang C, Liu F, Liu X, Wang D, Zhao D, Shui G, Zhao Y, Yan C. Distinctive metabolic remodeling in TYMP deficiency beyond mitochondrial dysfunction. J Mol Med (Berl) 2023; 101:1237-1253. [PMID: 37603049 DOI: 10.1007/s00109-023-02358-9] [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: 01/02/2023] [Revised: 07/09/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is caused by mutations in the TYMP gene, which encodes thymidine phosphorylase (TP). As a cytosolic metabolic enzyme, TP defects affect biological processes that are thought to not be limited to the abnormal replication of mitochondrial DNA. This study aimed to elucidate the characteristic metabolic alterations and associated homeostatic regulation caused by TYMP deficiency. The pathogenicity of novel TYMP variants was evaluated in terms of clinical features, genetic analysis, and structural instability. We analyzed plasma samples from three patients with MNGIE; three patients with m.3243A > G mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS); and four healthy controls (HC) using both targeted and untargeted metabolomics techniques. Transcriptomics analysis and bioenergetic studies were performed on skin fibroblasts from participants in these three groups. A TYMP overexpression experiment was conducted to rescue the observed changes. Compared with controls, specific alterations in nucleosides, bile acids, and steroid metabolites were identified in the plasma of MNGIE patients. Comparable mitochondrial dysfunction was present in fibroblasts from patients with TYMP deficiency and in those from patients with the m.3243A > G mutation. Distinctively decreased sterol regulatory element binding protein (SREBP) regulated cholesterol metabolism and fatty acid (FA) biosynthesis as well as reduced FA degradation were revealed in fibroblasts with TYMP deficiency. The restoration of thymidine phosphorylase activity rescued the observed changes in MNGIE fibroblasts. Our findings indicated that more widespread metabolic disturbance may be caused by TYMP deficiency in addition to mitochondrial dysfunction, which expands our knowledge of the biochemical outcome of TYMP deficiency. KEY MESSAGES: Distinct metabolic profiles in patients with TYMP deficiency compared to those with m.3243A > G mutation. TYMP deficiency leads to a global disruption of nucleoside metabolism. Cholesterol and fatty acid metabolism are inhibited in individuals with MNGIE. TYMP is functionally related to SREBP-regulated pathways. Potential metabolite biomarkers that could be valuable clinical tools to improve the diagnosis of MNGIE.
Collapse
Affiliation(s)
- Jixiang Du
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chao Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Fuchen Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xihan Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dongdong Wang
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dandan Zhao
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 101408, China
| | - Yuying Zhao
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, 266103, China.
| |
Collapse
|
50
|
Broeks MH, Meijer NWF, Westland D, Bosma M, Gerrits J, German HM, Ciapaite J, van Karnebeek CDM, Wanders RJA, Zwartkruis FJT, Verhoeven-Duif NM, Jans JJM. The malate-aspartate shuttle is important for de novo serine biosynthesis. Cell Rep 2023; 42:113043. [PMID: 37647199 DOI: 10.1016/j.celrep.2023.113043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 05/17/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023] Open
Abstract
The malate-aspartate shuttle (MAS) is a redox shuttle that transports reducing equivalents across the inner mitochondrial membrane while recycling cytosolic NADH to NAD+. We genetically disrupted each MAS component to generate a panel of MAS-deficient HEK293 cell lines in which we performed [U-13C]-glucose tracing. MAS-deficient cells have reduced serine biosynthesis, which strongly correlates with the lactate M+3/pyruvate M+3 ratio (reflective of the cytosolic NAD+/NADH ratio), consistent with the NAD+ dependency of phosphoglycerate dehydrogenase in the serine synthesis pathway. Among the MAS-deficient cells, those lacking malate dehydrogenase 1 (MDH1) show the most severe metabolic disruptions, whereas oxoglutarate-malate carrier (OGC)- and MDH2-deficient cells are less affected. Increasing the NAD+-regenerating capacity using pyruvate supplementation resolves most of the metabolic disturbances. Overall, we show that the MAS is important for de novo serine biosynthesis, implying that serine supplementation could be used as a therapeutic strategy for MAS defects and possibly other redox disorders.
Collapse
Affiliation(s)
- Melissa H Broeks
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands.
| | - Nils W F Meijer
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Denise Westland
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Marjolein Bosma
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Johan Gerrits
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Hannah M German
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Jolita Ciapaite
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Clara D M van Karnebeek
- Emma Center for Personalized Medicine, Departments of Pediatrics and Human Genetics, Amsterdam University Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Departments of Pediatrics and Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Ronald J A Wanders
- Departments of Pediatrics and Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Fried J T Zwartkruis
- dLAB, Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Nanda M Verhoeven-Duif
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Judith J M Jans
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands.
| |
Collapse
|