1
|
Eid F, El Ahmad P, Khoury R, El Masri D, El Zoghby Y, Sahlloul Y, Fadel J, Haddad Z, Mezher A, Ghayad LM, El Sabbagh Y, Gerges L, Lakis M, Sahyoun C, El Khoury G, Stephan JS, Sleiman SF. α-Ketoglutarate Is a Circulatory Exercise Factor That Promotes Learning and Memory Recall and Has Antidepressant Properties. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100477. [PMID: 40248276 PMCID: PMC12005275 DOI: 10.1016/j.bpsgos.2025.100477] [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: 07/11/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 04/19/2025] Open
Abstract
Background Depression poses a significant societal burden, necessitating effective treatment options. Conventional approaches often fall short, highlighting the need for alternatives. Exercise has emerged as a promising nonpharmacological strategy for improving mental health outcomes. Exercise promotes memory recall and alleviates depression by modulating BDNF (brain-derived neurotrophic factor) expression. The effects of exercise on BDNF are influenced by circulatory metabolites known as exercise factors. Methods Associative and spatial memory were evaluated in mice receiving α-ketoglutarate (aKG) and in exercise mice given a glutaminase inhibitor. To prevent and treat depression-like behaviors, male mice underwent daily defeat sessions by a CD1 aggressor for 10 days. Behavior was assessed on day 11 using social interaction and open-field tests. Mice received aKG for 5 days prior to the stress paradigm or as treatment for 14 days following the stress paradigm, after which social behavior was reassessed. BDNF signaling was examined via Western blots. Results aKG was identified as a metabolite released into the bloodstream following exercise in male mice. aKG was shown to mediate the positive effects of exercise on spatial learning and memory formation. aKG was also shown to have prophylactic and antidepressant effects in a chronic social defeat stress model of depression. Conclusions aKG acts as a prophylactic and antidepressant to effectively counteract social avoidance behaviors by modulating BDNF levels in the hippocampus and nucleus accumbens.
Collapse
Affiliation(s)
- Fady Eid
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Perla El Ahmad
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Reine Khoury
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Diala El Masri
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Yara El Zoghby
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Yasmin Sahlloul
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Joanna Fadel
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Zena Haddad
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Amar Mezher
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Litsa Maria Ghayad
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Yorgo El Sabbagh
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Lea Gerges
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Mahmoud Lakis
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Christopher Sahyoun
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Ghinwa El Khoury
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Joseph S. Stephan
- School of Medicine, Lebanese American University, Lebanon, Byblos, Lebanon
| | - Sama F. Sleiman
- Biological Sciences Program, Department of Natural Sciences, Lebanese American University, Lebanon, Byblos, Lebanon
| |
Collapse
|
2
|
Willemse L, Terburgh K, Louw R. The therapeutic potential of a polyunsaturated fatty acid-enriched high-fat diet in Leigh syndrome: Insights from a preclinical model. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167873. [PMID: 40288592 DOI: 10.1016/j.bbadis.2025.167873] [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: 11/27/2024] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Leigh syndrome is often caused by Ndufs4 mutations. The Ndufs4 knockout (KO) mouse model recapitulates key disease features, including systemic inflammation, neurodegeneration, and motor deficits. While dietary interventions such as the ketogenic diet show promise in mitigating mitochondrial dysfunction, conflicting results highlight uncertainties regarding its efficacy. Here, we evaluate the therapeutic potential of a polyunsaturated fatty acid (PUFA)-enriched high-fat diet (HFD) in Ndufs4 KO mice. METHODS Dietary intervention began at postnatal day 23, with mice receiving either a normal diet (ND) or a HFD enriched with PUFAs. Phenotypic evaluation, including locomotor function, clasping behaviour, and survival, continued until natural death. In a second group of animals, biochemical analyses were conducted after three weeks on the diets, using Western blot to evaluate neurometabolic and inflammatory regulators, flow cytometry to quantify serum inflammation markers, and metabolic profiling to identify alterations in neurometabolism and the neurolipidome. RESULTS The HFD significantly extended lifespan and improved clasping behaviour in Ndufs4 KO mice but had no effect on locomotor activity or grip strength decline. While whole-brain mTOR (p70S6K1, 4E-BP1) and SIRT1 (PGC1-α, TNF-α) signalling pathways remained unaffected, the diet significantly reduced serum pro-inflammatory markers TNF and IL-6. Furthermore, the PUFA-enriched HFD partially restored disruptions in TCA cycle, ketone body, branched-chain amino acid, and lipid metabolism, indicating potential metabolic reprogramming. CONCLUSION Dietary interventions, such as a PUFA-enriched HFD, may alleviate systemic inflammation, partially correct metabolic imbalances, and mitigate specific disease phenotypes in Leigh syndrome, warranting further investigation into the underlying mechanisms and broader therapeutic applications.
Collapse
Affiliation(s)
- Luciano Willemse
- Mitochondria Research Group, Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Karin Terburgh
- Mitochondria Research Group, Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Roan Louw
- Mitochondria Research Group, Human Metabolomics, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
3
|
Magro G, Laterza V, Tosto F. Leigh Syndrome: A Comprehensive Review of the Disease and Present and Future Treatments. Biomedicines 2025; 13:733. [PMID: 40149709 PMCID: PMC11940177 DOI: 10.3390/biomedicines13030733] [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: 02/17/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Leigh syndrome (LS) is a severe neurodegenerative condition with an early onset, typically during early childhood or infancy. The disorder exhibits substantial clinical and genetic diversity. From a clinical standpoint, Leigh syndrome showcases a broad range of irregularities, ranging from severe neurological issues to minimal or no discernible abnormalities. The central nervous system is most affected, resulting in psychomotor retardation, seizures, nystagmus, ophthalmoparesis, optic atrophy, ataxia, dystonia, or respiratory failure. Some patients also experience involvement of the peripheral nervous system, such as polyneuropathy or myopathy, as well as non-neurological anomalies, such as diabetes, short stature, hypertrichosis, cardiomyopathy, anemia, renal failure, vomiting, or diarrhea (Leigh-like syndrome). Mutations associated with Leigh syndrome impact genes in both the mitochondrial and nuclear genomes. Presently, LS remains without a cure and shows limited response to various treatments, although certain case reports suggest potential improvement with supplements. Ongoing preclinical studies are actively exploring new treatment approaches. This review comprehensively outlines the genetic underpinnings of LS, its current treatment methods, and preclinical investigations, with a particular focus on treatment.
Collapse
Affiliation(s)
- Giuseppe Magro
- Department of Neuroscience, “Giovanni Paolo II” Hospital, 88100 Lamezia Terme, Italy
| | - Vincenzo Laterza
- Department of Medical and Surgical Sciences, Institute of Neurology, Magna Graecia University, 88100 Catanzaro, Italy
| | - Federico Tosto
- Department of Neuroscience, “Giovanni Paolo II” Hospital, 88100 Lamezia Terme, Italy
| |
Collapse
|
4
|
Mouli K, Liopo AV, McHugh EA, Underwood E, Zhao J, Dash PK, Vo ATT, Malojirao VH, Hegde ML, Tour JM, Derry PJ, Kent TA. Oxidized Carbon Nanoparticles Enhance Cellular Energetics With Application to Injured Brain. Adv Healthc Mater 2025; 14:e2401629. [PMID: 39329414 PMCID: PMC11937864 DOI: 10.1002/adhm.202401629] [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: 05/02/2024] [Revised: 07/26/2024] [Indexed: 09/28/2024]
Abstract
Pro-energetic effects of functionalized, oxidized carbon nanozymes (OCNs) are reported. OCNs, derived from harsh acid oxidation of single-wall carbon nanotubes or activated charcoal are previously shown to possess multiple nanozymatic activities including mimicking superoxide dismutase and catalyzing the oxidation of reduced nicotinamide adenine dinucleotide (NADH) to NAD+. These actions are predicted to generate a glycolytic shift and enhance mitochondrial energetics under impaired conditions. Impaired mitochondrial energy metabolism is increasingly recognized as an important facet of traumatic brain injury (TBI) pathophysiology and decreases the efficiency of electron transport chain (ETC)-coupled adenosine triphosphate (ATP) and NAD+ regeneration. In vitro, OCNs promote a pro-aerobic shift in energy metabolism that persists through ETC inhibition and enhances glycolytic flux, glycolytic ATP production, and cellular generation of lactate, a crucial auxiliary substrate for energy metabolism. To address specific mechanisms of iron injury from hemorrhage, OCNs with the iron chelator, deferoxamine (DEF), covalently-linked were synthesized. DEF-linked OCNs induce a glycolytic shift in-vitro and in-vivo in tissue sections from a rat model of TBI complicated by hemorrhagic contusion. OCNs further reduced hemorrhage volumes 3 days following TBI. These results suggest OCNs are promising as pleiotropic mediators of cell and tissue resilience to injury.
Collapse
Affiliation(s)
- Karthik Mouli
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
| | - Anton V. Liopo
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- Department of ChemistryRice UniversityHoustonTX77005USA
| | - Emily A. McHugh
- Department of ChemistryRice UniversityHoustonTX77005USA
- Smalley‐Curl InstituteRice UniversityHoustonTX77005USA
| | - Erica Underwood
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Jing Zhao
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Pramod K. Dash
- Department of Neurobiology and AnatomyThe University of TX McGovern Medical SchoolHoustonTX77030USA
| | - Anh T. T. Vo
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
| | - Vikas H. Malojirao
- Center for NeuroregenerationDepartment of NeurosurgeryDivision of DNA Repair ResearchHouston Methodist Research InstituteHoustonTX77030USA
| | - Muralidhar L. Hegde
- Center for NeuroregenerationDepartment of NeurosurgeryDivision of DNA Repair ResearchHouston Methodist Research InstituteHoustonTX77030USA
- Department of NeurosciencesWeill Cornell Medical CollegeNew YorkNYUSA
- EnMedSchool of Engineering MedicineTexas A&M UniversityHouston77030USA
| | - James M. Tour
- Department of ChemistryRice UniversityHoustonTX77005USA
- Smalley‐Curl InstituteRice UniversityHoustonTX77005USA
- Welch Institute for Advanced MaterialsRice UniversityHoustonTX77005USA
- The NanoCarbon CenterRice UniversityHoustonTX77005USA
| | - Paul J. Derry
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- EnMedSchool of Engineering MedicineTexas A&M UniversityHouston77030USA
| | - Thomas A. Kent
- Center for Genomics and Precision MedicineDepartment of Translational MedicineInstitute of Biosciences and TechnologyTexas A&M Health Science CenterHoustonTX77030USA
- Department of ChemistryRice UniversityHoustonTX77005USA
- Stanley H. Appel Department of NeurologyHouston Methodist Hospital and Research InstituteHoustonTX77030USA
| |
Collapse
|
5
|
Lin WS. Two tales of therapeutic innovations for Leigh syndrome spectrum. J Neurogenet 2025; 39:4-6. [PMID: 40050739 DOI: 10.1080/01677063.2025.2473087] [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: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 04/12/2025]
Abstract
Leigh syndrome spectrum is the most common form of childhood-onset mitochondrial encephalopathy and is characterized by progressive neurodegeneration. Treatment options for this condition remain limited to date. Nonetheless, two lines of research endeavor in the past decade have shown encouraging results worthy of further investigations. First, therapeutic hypoxia appears to improve neurological outcomes, which is somewhat counterintuitive but supported by preclinical evidence. Furthermore, nicotinic acid or nicotinamide riboside could be an adjunctive therapy that enhances the neuroprotective effect of hypoxia. Second, preclinical studies and preliminary clinical experience suggest that sildenafil is potentially disease-modifying for Leigh syndrome. Sildenafil has already been used to treat pulmonary hypertension, and its repurposing for Leigh syndrome has been endorsed by European Medicines Agency. This perspective aims to raise awareness about these progresses, as well as to call for more clinical studies to ensure safe and effective implementation of these treatment approaches in clinical practice.
Collapse
Affiliation(s)
- Wei-Sheng Lin
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
6
|
Xu Y, Wang H, Li H, Wei C, Zhu Z, Zhao Y, Zhu J, Lei M, Sun Y, Yang Q. Nicotinamide Riboside Supplementation Alleviates Testicular Aging Induced by Disruption of Qprt-Dependent NAD + De Novo Synthesis in Mice. Aging Cell 2025:e70004. [PMID: 39902575 DOI: 10.1111/acel.70004] [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: 11/04/2024] [Revised: 12/21/2024] [Accepted: 01/09/2025] [Indexed: 02/05/2025] Open
Abstract
Recent studies have shown that disruptions in the nicotinamide adenine dinucleotide (NAD+) de novo synthesis pathway accelerate ovarian aging, yet its role in spermatogenesis remains largely unknown. In this study, we investigated the impact of the NAD+ de novo synthesis pathway on spermatogenesis by generating Qprt-deficient mice using CRISPR-Cas9 to target quinolinate phosphoribosyl transferase (Qprt), a key enzyme predominantly expressed in spermatocytes. Our results revealed that the deletion of Qprt did not affect NAD+ levels or spermatogenesis in the testes of 3-month-old mice. However, from 6 months of age onward, Qprt-deficient mice exhibited significantly reduced NAD+ levels in the testes compared to wild-type (WT) controls, along with a notable decrease in germ cell numbers and increased apoptosis. Additionally, these mice demonstrated mitochondrial dysfunction in spermatocytes, impaired progression through prophase I of meiosis, defective double-strand break (DSB) repair, and abnormal meiotic sex chromosome inactivation. Importantly, supplementation with the NAD+ precursor nicotinamide riboside (NR) in Qprt-deficient mice restored NAD+ levels and rescued the spermatogenic defects. These findings underscore the critical role of NAD+ de novo synthesis in maintaining NAD+ homeostasis and highlight its importance in meiotic recombination and meiotic sex chromosome inactivation in spermatogenesis.
Collapse
Affiliation(s)
- Yining Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huan Wang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Li
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chenlu Wei
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenye Zhu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanqing Zhao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiajia Zhu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Lei
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingling Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Goolab S, Terburgh K, du Plessis C, Scholefield J, Louw R. CRISPR-Cas9 mediated knockout of NDUFS4 in human iPSCs: A model for mitochondrial complex I deficiency. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167569. [PMID: 39547516 DOI: 10.1016/j.bbadis.2024.167569] [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: 05/31/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Mitochondrial diseases, often caused by defects in complex I (CI) of the oxidative phosphorylation system, currently lack curative treatments. Human-relevant, high-throughput drug screening platforms are crucial for the discovery of effective therapeutics, with induced pluripotent stem cells (iPSCs) emerging as a valuable technology for this purpose. Here, we present a novel iPSC model of NDUFS4-related CI deficiency that displays a strong metabolic phenotype in the pluripotent state. Human iPSCs were edited using CRISPR-Cas9 to target the NDUFS4 gene, generating isogenic NDUFS4 knockout (KO) cell lines. Sanger sequencing detected heterozygous biallelic deletions, whereas no indel mutations were found in isogenic control cells. Western blotting confirmed the absence of NDUFS4 protein in KO iPSCs and CI enzyme kinetics showed a ~56 % reduction in activity compared to isogenic controls. Comprehensive metabolomic profiling revealed a distinct metabolic phenotype in NDUFS4 KO iPSCs, predominantly associated with an elevated NADH/NAD+ ratio, consistent with alterations observed in other models of mitochondrial dysfunction. Additionally, β-lapachone, a recognized NAD+ modulator, alleviated reductive stress in KO iPSCs by modifying the redox state in both the cytosol and mitochondria. Although undifferentiated iPSCs cannot fully replicate the complex cellular dynamics of the disease seen in vivo, these findings highlight the utility of iPSCs in providing a relevant metabolic milieu that can facilitate early-stage, high-throughput exploration of therapeutic strategies for mitochondrial dysfunction.
Collapse
Affiliation(s)
- Shivani Goolab
- Bioengineering and Integrated Genomics Group, Future Productions: Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Karin Terburgh
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Charl du Plessis
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Janine Scholefield
- Bioengineering and Integrated Genomics Group, Future Productions: Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa; Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Division of Human Genetics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
8
|
Zhang X, Zhang B, Tao Z, Liang J. Mitochondrial disease and epilepsy in children. Front Neurol 2025; 15:1499876. [PMID: 39850733 PMCID: PMC11754068 DOI: 10.3389/fneur.2024.1499876] [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: 09/22/2024] [Accepted: 12/26/2024] [Indexed: 01/25/2025] Open
Abstract
Mitochondria is the cell's powerhouse. Mitochondrial disease refers to a group of clinically heterogeneous disorders caused by dysfunction in the mitochondrial respiratory chain, often due to mutations in mitochondrial DNA (mtDNA) or nuclear DNA (nDNA) that encodes mitochondrial proteins. This dysfunction can lead to a variety of clinical phenotypes, particularly affecting organs with high energy demands, such as the brain and muscles. Epilepsy is a prevalent neurological disorder in children and is also a frequent manifestation of mitochondrial disease. The exact mechanisms underlying epilepsy in mitochondrial disease remain unclear and are thought to involve multiple contributing factors. This review explores common mitochondrial diseases associated with epilepsy, focusing on their prevalence, seizure types, EEG features, therapeutic strategies, and outcomes. It also summarizes the relationship between the molecular genetics of mitochondrial respiratory chain components and the development of epilepsy.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Pediatric Neurology, Children's Medical Center, First Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Pediatric Neurology, Changchun, China
- Neuromedical Center, First Hospital of Jilin University, Changchun, China
| | - Bo Zhang
- Department of Pediatric Neurology, Children's Medical Center, First Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Pediatric Neurology, Changchun, China
- Neuromedical Center, First Hospital of Jilin University, Changchun, China
| | - Zhiming Tao
- Department of Pediatric Neurology, Children's Medical Center, First Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Pediatric Neurology, Changchun, China
- Neuromedical Center, First Hospital of Jilin University, Changchun, China
| | - Jianmin Liang
- Department of Pediatric Neurology, Children's Medical Center, First Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Pediatric Neurology, Changchun, China
- Neuromedical Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
Di Michele M, Attina A, Roux PF, Tabet I, Laguesse S, Florido J, Houdeville M, Choquet A, Encislai B, Arena G, De Blasio C, Wendling O, Frenois FX, Papon L, Stuani L, Fuentes M, Jahannault Talignani C, Rousseau M, Guégan J, Buscail Y, Dupré P, Michaud HA, Rodier G, Bellvert F, Kulyk H, Ferraro Peyret C, Mathieu H, Close P, Rapino F, Chaveroux C, Pirot N, Rubio L, Torro A, Sorg T, Ango F, Hirtz C, Compan V, Lebigot E, Legati A, Ghezzi D, Nguyen L, David A, Sardet C, Lacroix M, Le Cam L. E4F1 coordinates pyruvate metabolism and the activity of the elongator complex to ensure translation fidelity during brain development. Nat Commun 2025; 16:67. [PMID: 39747033 PMCID: PMC11696611 DOI: 10.1038/s41467-024-55444-y] [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: 07/13/2022] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Pyruvate metabolism defects lead to severe neuropathies such as the Leigh syndrome (LS) but the molecular mechanisms underlying neuronal cell death remain poorly understood. Here, we unravel a connection between pyruvate metabolism and the regulation of the epitranscriptome that plays an essential role during brain development. Using genetically engineered mouse model and primary neuronal cells, we identify the transcription factor E4F1 as a key coordinator of AcetylCoenzyme A (AcCoA) production by the pyruvate dehydrogenase complex (PDC) and its utilization as an essential co-factor by the Elongator complex to acetylate tRNAs at the wobble position uridine 34 (U34). E4F1-mediated direct transcriptional regulation of Dlat and Elp3, two genes encoding key subunits of the PDC and of the Elongator complex, respectively, ensures proper translation fidelity and cell survival in the central nervous system (CNS) during mouse embryonic development. Furthermore, analysis of PDH-deficient cells highlight a crosstalk linking the PDC to ELP3 expression that is perturbed in LS patients.
Collapse
Affiliation(s)
- Michela Di Michele
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France.
- Equipe labélisée Ligue Contre le Cancer, Paris, France.
- Institut des Biomolécules Max Mousseron (IBMM), UMR-5247, Univ. Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Aurore Attina
- IRMB-PPC, Univ. Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Pierre-François Roux
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Imène Tabet
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Sophie Laguesse
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | - Javier Florido
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Morane Houdeville
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Armelle Choquet
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Betty Encislai
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Giuseppe Arena
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Carlo De Blasio
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Olivia Wendling
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | | | - Laura Papon
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Lucille Stuani
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Maryse Fuentes
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Céline Jahannault Talignani
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Mélanie Rousseau
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Justine Guégan
- Data Analysis Core Platform, Institut du Cerveau - Paris Brain Institute - ICM, Sorbonne Université, INSERM, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Yoan Buscail
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- BioCampus, RHEM, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pierrick Dupré
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Henri-Alexandre Michaud
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- SIMCaT plateform, Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Geneviève Rodier
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Floriant Bellvert
- MetaToul-MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
- Toulouse Biotechnologie Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Hanna Kulyk
- MetaToul-MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
- Toulouse Biotechnologie Institute (TBI), Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Carole Ferraro Peyret
- Univ. Lyon, Claude Bernard University, LBTI UMR CNRS 5305, Faculty of Pharmacy, Lyon, France
- Hospices Civils de Lyon, AURAGEN, Edouard Herriot Hospital, Lyon, France
| | - Hugo Mathieu
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Pierre Close
- Laboratory of Cancer Signaling, GIGA-Institute, University of Liège, 4000, Liège, Belgium
- WELBIO department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Francesca Rapino
- Laboratory of Cancer Signaling, GIGA-Institute, University of Liège, 4000, Liège, Belgium
| | - Cédric Chaveroux
- Univ. Lyon, Claude Bernard University, LBTI UMR CNRS 5305, Faculty of Pharmacy, Lyon, France
| | - Nelly Pirot
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- BioCampus, RHEM, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Lucie Rubio
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Adeline Torro
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Tania Sorg
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | - Fabrice Ango
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Christophe Hirtz
- IRMB-PPC, Univ. Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Vincent Compan
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Elise Lebigot
- Biochemistry Department, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre, France
| | - Andrea Legati
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniele Ghezzi
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Laurent Nguyen
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
- WELBIO department, WEL Research Institute, avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Alexandre David
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
- IRMB-PPC, Univ. Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Claude Sardet
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Matthieu Lacroix
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France.
- Equipe labélisée Ligue Contre le Cancer, Paris, France.
| | - Laurent Le Cam
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France.
- Equipe labélisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
10
|
Ishima T, Kimura N, Kobayashi M, Watanabe C, Jimbo EF, Kobayashi R, Horii T, Hatada I, Murayama K, Ohtake A, Nagai R, Osaka H, Aizawa K. NADH Reductive Stress and Its Correlation with Disease Severity in Leigh Syndrome: A Pilot Study Using Patient Fibroblasts and a Mouse Model. Biomolecules 2024; 15:38. [PMID: 39858433 PMCID: PMC11764390 DOI: 10.3390/biom15010038] [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: 11/20/2024] [Revised: 12/25/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a critical cofactor in mitochondrial energy production. The NADH/NAD+ ratio, reflecting the balance between NADH (reduced) and NAD+ (oxidized), is a key marker for the severity of mitochondrial diseases. We recently developed a streamlined LC-MS/MS method for the precise measurement of NADH and NAD+. Utilizing this technique, we quantified NADH and NAD+ levels in fibroblasts derived from pediatric patients and in a Leigh syndrome mouse model in which mitochondrial respiratory chain complex I subunit Ndufs4 is knocked out (KO). In patient-derived fibroblasts, NAD+ levels did not differ significantly from those of healthy controls (p = 0.79); however, NADH levels were significantly elevated (p = 0.04), indicating increased NADH reductive stress. This increase, observed despite comparable total NAD(H) levels between the groups, was attributed to elevated NADH levels. Similarly, in the mouse model, NADH levels were significantly increased in the KO group (p = 0.002), further suggesting that NADH elevation drives reductive stress. This precise method for NADH measurement is expected to outperform conventional assays, such as those for lactate, providing a simpler and more reliable means of assessing disease progression.
Collapse
Affiliation(s)
- Tamaki Ishima
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Natsuka Kimura
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Mizuki Kobayashi
- Department of Pediatrics, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Chika Watanabe
- Department of Pediatrics, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Eriko F. Jimbo
- Department of Pediatrics, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Ryosuke Kobayashi
- Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | - Takuro Horii
- Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | - Izuho Hatada
- Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Maebashi 371-8511, Japan
| | - Kei Murayama
- Center for Medical Genetics, Chiba Children’s Hospital, Chiba 266-0007, Japan
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8431, Japan
| | - Akira Ohtake
- Department of Clinical Genomics, Faculty of Medicine, Saitama Medical University, Moroyama 350-0495, Japan
- Department of Pediatrics, Faculty of Medicine, Saitama Medical University, Moroyama 350-0495, Japan
| | - Ryozo Nagai
- Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Kenichi Aizawa
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
- 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
|
11
|
Huang G, Li Z, Liu X, Guan M, Zhou S, Zhong X, Zheng T, Xin D, Gu X, Mu D, Guo Y, Zhang L, Zhang L, Lu QR, He X. DOR activation in mature oligodendrocytes regulates α-ketoglutarate metabolism leading to enhanced remyelination in aged mice. Nat Neurosci 2024; 27:2073-2085. [PMID: 39266660 DOI: 10.1038/s41593-024-01754-9] [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: 07/20/2023] [Accepted: 08/07/2024] [Indexed: 09/14/2024]
Abstract
The decreased ability of mature oligodendrocytes to produce myelin negatively affects remyelination in demyelinating diseases and aging, but the underlying mechanisms are incompletely understood. In the present study, we identify a mature oligodendrocyte-enriched transcriptional coregulator diabetes- and obesity-related gene (DOR)/tumor protein p53-inducible nuclear protein 2 (TP53INP2), downregulated in demyelinated lesions of donors with multiple sclerosis and in aged oligodendrocyte-lineage cells. Dor ablation in mice of both sexes results in defective myelinogenesis and remyelination. Genomic occupancy in oligodendrocytes and transcriptome profiling of the optic nerves of wild-type and Dor conditional knockout mice reveal that DOR and SOX10 co-occupy enhancers of critical myelinogenesis-associated genes including Prr18, encoding an oligodendrocyte-enriched, proline-rich factor. We show that DOR targets regulatory elements of genes responsible for α-ketoglutarate biosynthesis in mature oligodendrocytes and is essential for α-ketoglutarate production and lipid biosynthesis. Supplementation with α-ketoglutarate restores oligodendrocyte-maturation defects in Dor-deficient adult mice and improves remyelination after lysolecithin-induced demyelination and cognitive function in 17-month-old wild-type mice. Our data suggest that activation of α-ketoglutarate metabolism in mature oligodendrocytes can promote myelin production during demyelination and aging.
Collapse
Affiliation(s)
- Guojiao Huang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhidan Li
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xuezhao Liu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Menglong Guan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaowen Zhong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tao Zheng
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dazhuan Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Dezhi Mu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yingkun Guo
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lin Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liguo Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xuelian He
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Nakai R, Varnum S, Field RL, Shi H, Giwa R, Jia W, Krysa SJ, Cohen EF, Borcherding N, Saneto RP, Tsai RC, Suganuma M, Ohta H, Yokota T, Brestoff JR. Mitochondria transfer-based therapies reduce the morbidity and mortality of Leigh syndrome. Nat Metab 2024; 6:1886-1896. [PMID: 39223312 DOI: 10.1038/s42255-024-01125-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria transfer is a recently described phenomenon in which donor cells deliver mitochondria to acceptor cells1-3. One possible consequence of mitochondria transfer is energetic support of neighbouring cells; for example, exogenous healthy mitochondria can rescue cell-intrinsic defects in mitochondrial metabolism in cultured ρ0 cells or Ndufs4-/- peritoneal macrophages4-7. Exposing haematopoietic stem cells to purified mitochondria before autologous haematopoietic stem cell transplantation allowed for treatment of anaemia in patients with large-scale mitochondrial DNA mutations8,9, and mitochondria transplantation was shown to minimize ischaemic damage to the heart10-12, brain13-15 and limbs16. However, the therapeutic potential of using mitochondria transfer-based therapies to treat inherited mitochondrial diseases is unclear. Here we demonstrate improved morbidity and mortality of the Ndufs4-/- mouse model of Leigh syndrome (LS) in multiple treatment paradigms associated with mitochondria transfer. Transplantation of bone marrow from wild-type mice, which is associated with release of haematopoietic cell-derived extracellular mitochondria into circulation and transfer of mitochondria to host cells in multiple organs, ameliorates LS in mice. Furthermore, administering isolated mitochondria from wild-type mice extends lifespan, improves neurological function and increases energy expenditure of Ndufs4-/- mice, whereas mitochondria from Ndufs4-/- mice did not improve neurological function. Finally, we demonstrate that cross-species administration of human mitochondria to Ndufs4-/- mice also improves LS. These data suggest that mitochondria transfer-related approaches can be harnessed to treat mitochondrial diseases, such as LS.
Collapse
Affiliation(s)
- Ritsuko Nakai
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
- Department of Hematology, Sakai City Medical Center, Sakai, Japan
| | - Stella Varnum
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachael L Field
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Henyun Shi
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Rocky Giwa
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wentong Jia
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Samantha J Krysa
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Eva F Cohen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Russell P Saneto
- Neuroscience Institute, Center for Integrated Brain Research, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | | | | | | | - Takafumi Yokota
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan.
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan.
| | - Jonathan R Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
13
|
Walker MA, Tian R. NAD metabolism and heart failure: Mechanisms and therapeutic potentials. J Mol Cell Cardiol 2024; 195:45-54. [PMID: 39096536 PMCID: PMC11390314 DOI: 10.1016/j.yjmcc.2024.07.008] [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: 04/20/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/05/2024]
Abstract
Nicotinamide adenine dinucleotide provides the critical redox pair, NAD+ and NADH, for cellular energy metabolism. In addition, NAD+ is the precursor for de novo NADP+ synthesis as well as the co-substrates for CD38, poly(ADP-ribose) polymerase and sirtuins, thus, playing a central role in the regulation of oxidative stress and cell signaling. Declines of the NAD+ level and altered NAD+/NADH redox states have been observed in cardiometabolic diseases of various etiologies. NAD based therapies have emerged as a promising strategy to treat cardiovascular disease. Strategies that reduce NAD+ consumption or promote NAD+ production have repleted intracellular NAD+ or normalized NAD+/NADH redox in preclinical studies. These interventions have shown cardioprotective effects in multiple models suggesting a great promise of the NAD+ elevating therapy. Mechanisms for the benefit of boosting NAD+ level, however, remain incompletely understood. Moreover, despite the robust pre-clinical studies there are still challenges to translate the therapy to clinic. Here, we review the most up to date literature on mechanisms underlying the NAD+ elevating interventions and discuss the progress of human studies. We also aim to provide a better understanding of how NAD metabolism is changed in failing hearts with a particular emphasis on types of strategies employed and methods to target these pathways. Finally, we conclude with a comprehensive assessment of the challenges in developing NAD-based therapies for heart diseases, and to provide a perspective on the future of the targeting strategies.
Collapse
Affiliation(s)
- Matthew A Walker
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
14
|
Zhu R, Kang Y, Li Q, Peng K, Shi X, Yin Z, Xuan Y. Alpha-tocopherol inhibits ferroptosis and promotes neural function recovery in rats with spinal cord injury via downregulating Alox15. Biomed Pharmacother 2024; 175:116734. [PMID: 38754264 DOI: 10.1016/j.biopha.2024.116734] [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: 09/05/2023] [Revised: 04/27/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
Spinal cord injury (SCI) is a type of central nervous system (CNS) injury in which ferroptosis is becoming a promising target for treatment. Alpha-tocopherol (Vitamin E, Vit E) is a compound with anti-ferroptosis activity. The mechanism of alpha-tocopherol in regulating ferroptosis after SCI has not been deeply studied. In this study, rats with SCI were treated by Alpha-tocopherol based on bioinformatic analysis and molecular docking prediction. Behavioral tests and histological findings showed that Alpha-tocopherol promoted neural function recovery and tissue repairment in rats with SCI. Subsequently, regulatory effects of Alpha-tocopherol on Alox15 and ferroptosis were detected and then localized by immunofluorescence. In vitro, alpha-tocopherol improved the ROS accumulation, iron overload, lipid peroxidation and mitochondrial dysfunction. The effects of Alpha-tocopherol on the expression of Alox15, Ptgs2 and 4Hne were validated in vitro. Finally, the inhibitory effects of Alpha-tocopherol on Alox15 and ferroptosis were weakened by the mutation of 87th residue of Alox15. In summary, alpha-tocopherol could alleviate SCI-induced ferroptosis by downregulating Alox15 to promote neural function recovery in rats with SCI. Findings in this study could help further our understanding on SCI-induced ferroptosis and provide a novel insight for treating SCI.
Collapse
Affiliation(s)
- Rui Zhu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, 218 Jixi Road, Hefei 230022, China; Department of Orthopedics, Hefei Orthopedics Hospital, 58 Chaohu Northern Road, Hefei 238001, China
| | - Yu Kang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, 218 Jixi Road, Hefei 230022, China
| | - Qiangwei Li
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Kai Peng
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, 218 Jixi Road, Hefei 230022, China; The Key Laboratory of Microbiology and Parasitology of Anhui Province, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Xuanming Shi
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, China.
| | - Zongsheng Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, 218 Jixi Road, Hefei 230022, China.
| | - Yong Xuan
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, 218 Jixi Road, Hefei 230022, China; Department of Orthopedics, The Second People's Hospital of Hefei, 246 Heping Road, Hefei 230011, China.
| |
Collapse
|
15
|
Saleh TA, Whitson JA, Keiser P, Prasad P, Jenkins BC, Sodeinde T, Mann C, Rabinovitch PS, McReynolds MR, Sweetwyne MT. Metabolite accumulation from oral NMN supplementation drives aging-specific kidney inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588624. [PMID: 38645109 PMCID: PMC11030441 DOI: 10.1101/2024.04.09.588624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The mitochondrial-rich renal tubule cells are key regulators of blood homeostasis via excretion and reabsorption of metabolic waste. With age, tubules are subject to increasing mitochondrial dysfunction and declining nicotinamide adenine dinucleotide (NAD+) levels, both hampering ATP production efficiency. We tested two mitochondrial interventions in young (6-mo) and aged (26-mo) adult male mice: elamipretide (ELAM), a tetrapeptide in clinical trials that improves mitochondrial structure and function, and nicotinamide mononucleotide (NMN), an NAD+ intermediate and commercially available oral supplement. Kidneys were analyzed from young and aged mice after eight weeks of treatment with ELAM (3 mg/kg/day), NMN (300 mg/kg/day), or from aged mice treated with the two interventions combined (ELAM+NMN). We hypothesized that combining pharmacologic treatments to ameliorate mitochondrial dysfunction and boost NAD+ levels, would more effectively reduce kidney aging than either intervention alone. Unexpectedly, in aged kidneys, NMN increased expression of genetic markers of inflammation (IL-1-beta; and Ccl2) and tubule injury (Kim-1). Metabolomics of endpoint sera showed that NMN-treated aged mice had higher circulating levels of uremic toxins than either aged controls or young NMN-treated mice. ELAM+NMN-treated aged mice accumulated uremic toxins like NMN-only aged mice, but reduced IL-1-beta; and Ccl2 kidney mRNA. This suggests that pre-existing mitochondrial dysfunction in aged kidney underlies susceptibility to inflammatory signaling with NMN supplementation in aged, but not young, mice. These findings demonstrate age and tissue dependent effects on downstream metabolic accumulation from NMN and highlight the need for targeted analysis of aged kidneys to assess the safety of anti-aging supplements in older populations.
Collapse
|
16
|
Sonsalla G, Malpartida AB, Riedemann T, Gusic M, Rusha E, Bulli G, Najas S, Janjic A, Hersbach BA, Smialowski P, Drukker M, Enard W, Prehn JHM, Prokisch H, Götz M, Masserdotti G. Direct neuronal reprogramming of NDUFS4 patient cells identifies the unfolded protein response as a novel general reprogramming hurdle. Neuron 2024; 112:1117-1132.e9. [PMID: 38266647 PMCID: PMC10994141 DOI: 10.1016/j.neuron.2023.12.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/12/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024]
Abstract
Mitochondria account for essential cellular pathways, from ATP production to nucleotide metabolism, and their deficits lead to neurological disorders and contribute to the onset of age-related diseases. Direct neuronal reprogramming aims at replacing neurons lost in such conditions, but very little is known about the impact of mitochondrial dysfunction on the direct reprogramming of human cells. Here, we explore the effects of mitochondrial dysfunction on the neuronal reprogramming of induced pluripotent stem cell (iPSC)-derived astrocytes carrying mutations in the NDUFS4 gene, important for Complex I and associated with Leigh syndrome. This led to the identification of the unfolded protein response as a major hurdle in the direct neuronal conversion of not only astrocytes and fibroblasts from patients but also control human astrocytes and fibroblasts. Its transient inhibition potently improves reprogramming by influencing the mitochondria-endoplasmic-reticulum-stress-mediated pathways. Taken together, disease modeling using patient cells unraveled novel general hurdles and ways to overcome these in human astrocyte-to-neuron reprogramming.
Collapse
Affiliation(s)
- Giovanna Sonsalla
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Graduate School of Systemic Neurosciences, BMC, LMU Munich, Planegg-Martinsried 82152 Germany
| | - Ana Belen Malpartida
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; International Max Planck Research School (IMPRS) for Molecular Life Sciences, Planegg-Martinsried 82152, Germany
| | - Therese Riedemann
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Mirjana Gusic
- Institute of Neurogenomics, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Ejona Rusha
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany
| | - Giorgia Bulli
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Graduate School of Systemic Neurosciences, BMC, LMU Munich, Planegg-Martinsried 82152 Germany
| | - Sonia Najas
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Aleks Janjic
- Anthropology and Human Genomics, Faculty of Biology, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Bob A Hersbach
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Graduate School of Systemic Neurosciences, BMC, LMU Munich, Planegg-Martinsried 82152 Germany
| | - Pawel Smialowski
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Biomedical Center Munich, Bioinformatic Core Facility, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Micha Drukker
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Gorlaeus Building, 2333 CC RA, Leiden, the Netherlands
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, LMU Munich, Planegg-Martinsried 82152, Germany
| | - Jochen H M Prehn
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Holger Prokisch
- Institute of Neurogenomics, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764 Neuherberg, Germany; Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Magdalena Götz
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Excellence Cluster of Systems Neurology (SYNERGY), Munich, Germany.
| | - Giacomo Masserdotti
- Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany.
| |
Collapse
|
17
|
Rahman SU, Qadeer A, Wu Z. Role and Potential Mechanisms of Nicotinamide Mononucleotide in Aging. Aging Dis 2024; 15:565-583. [PMID: 37548938 PMCID: PMC10917541 DOI: 10.14336/ad.2023.0519-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/19/2023] [Indexed: 08/08/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) has recently attracted much attention due to its role in aging and lifespan extension. NAD+ directly and indirectly affects many cellular processes, including metabolic pathways, DNA repair, and immune cell activities. These mechanisms are critical for maintaining cellular homeostasis. However, the decline in NAD+ levels with aging impairs tissue function, which has been associated with several age-related diseases. In fact, the aging population has been steadily increasing worldwide, and it is important to restore NAD+ levels and reverse or delay these age-related disorders. Therefore, there is an increasing demand for healthy products that can mitigate aging, extend lifespan, and halt age-related consequences. In this case, several studies in humans and animals have targeted NAD+ metabolism with NAD+ intermediates. Among them, nicotinamide mononucleotide (NMN), a precursor in the biosynthesis of NAD+, has recently received much attention from the scientific community for its anti-aging properties. In model organisms, ingestion of NMN has been shown to improve age-related diseases and probably delay death. Here, we review aspects of NMN biosynthesis and the mechanism of its absorption, as well as potential anti-aging mechanisms of NMN, including recent preclinical and clinical tests, adverse effects, limitations, and perceived challenges.
Collapse
Affiliation(s)
- Sajid Ur Rahman
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Abdul Qadeer
- Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Ziyun Wu
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
18
|
Thompson LP, Song H, Hartnett J. Nicotinamide Riboside, an NAD + Precursor, Protects Against Cardiac Mitochondrial Dysfunction in Fetal Guinea Pigs Exposed to Gestational Hypoxia. Reprod Sci 2024; 31:975-986. [PMID: 37957471 PMCID: PMC10959782 DOI: 10.1007/s43032-023-01387-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023]
Abstract
Gestational hypoxia inhibits mitochondrial function in the fetal heart and placenta contributing to fetal growth restriction and organ dysfunction. NAD + deficiency may contribute to a metabolic deficit by inhibiting oxidative phosphorylation and ATP synthesis. We tested the effects of nicotinamide riboside (NR), an NAD + precursor, as a treatment for reversing known mitochondrial dysfunction in hypoxic fetal hearts. Pregnant guinea pigs were housed in room air (normoxia) or placed in a hypoxic chamber (10.5%O2) for the last 14 days of gestation (term = 65 days) and administered either water or NR (1.6 mg/ml) in the drinking bottle. Fetuses were excised at term, and NAD + levels of maternal liver, placenta, and fetal heart ventricles were measured. Indices of mitochondrial function (complex IV activity, sirtuin 3 activity, protein acetylation) and ATP synthesis were measured in fetal heart ventricles of NR-treated/untreated normoxic and hypoxic animals. Hypoxia reduced fetal body weight in both sexes (p = 0.01), which was prevented by NR. Hypoxia had no effect on maternal liver NAD + levels but decreased (p = 0.04) placenta NAD + levels, the latter normalized with NR treatment. Hypoxia had no effect on fetal heart NAD + but decreased (p < 0.05) mitochondrial complex IV and sirtuin 3 activities, ATP content, and increased mitochondrial acetylation, which were all normalized with maternal NR. Hypoxia increased (p < 0.05) mitochondrial acetylation in female fetal hearts but had no effect on other mitochondrial indices. We conclude that maternal NR is an effective treatment for normalizing mitochondrial dysfunction and ATP synthesis in the hypoxic fetal heart.
Collapse
Affiliation(s)
- Loren P Thompson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| | - Hong Song
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Jamie Hartnett
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| |
Collapse
|
19
|
Kopecky BJ, Lavine KJ. Cardiac macrophage metabolism in health and disease. Trends Endocrinol Metab 2024; 35:249-262. [PMID: 37993313 PMCID: PMC10949041 DOI: 10.1016/j.tem.2023.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
Cardiac macrophages are essential mediators of cardiac development, tissue homeostasis, and response to injury. Cell-intrinsic shifts in metabolism and availability of metabolites regulate macrophage function. The human and mouse heart contain a heterogeneous compilation of cardiac macrophages that are derived from at least two distinct lineages. In this review, we detail the unique functional roles and metabolic profiles of tissue-resident and monocyte-derived cardiac macrophages during embryonic development and adult tissue homeostasis and in response to pathologic and physiologic stressors. We discuss the metabolic preferences of each macrophage lineage and how metabolism influences monocyte fate specification. Finally, we highlight the contribution of cardiac macrophages and derived metabolites on cell-cell communication, metabolic health, and disease pathogenesis.
Collapse
Affiliation(s)
- Benjamin J Kopecky
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kory J Lavine
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
20
|
Zhang Q, Li Z, Li Q, Trammell SA, Schmidt MS, Pires KM, Cai J, Zhang Y, Kenny H, Boudina S, Brenner C, Abel ED. Control of NAD + homeostasis by autophagic flux modulates mitochondrial and cardiac function. EMBO J 2024; 43:362-390. [PMID: 38212381 PMCID: PMC10897141 DOI: 10.1038/s44318-023-00009-w] [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: 01/20/2023] [Revised: 10/31/2023] [Accepted: 11/08/2023] [Indexed: 01/13/2024] Open
Abstract
Impaired autophagy is known to cause mitochondrial dysfunction and heart failure, in part due to altered mitophagy and protein quality control. However, whether additional mechanisms are involved in the development of mitochondrial dysfunction and heart failure in the setting of deficient autophagic flux remains poorly explored. Here, we show that impaired autophagic flux reduces nicotinamide adenine dinucleotide (NAD+) availability in cardiomyocytes. NAD+ deficiency upon autophagic impairment is attributable to the induction of nicotinamide N-methyltransferase (NNMT), which methylates the NAD+ precursor nicotinamide (NAM) to generate N-methyl-nicotinamide (MeNAM). The administration of nicotinamide mononucleotide (NMN) or inhibition of NNMT activity in autophagy-deficient hearts and cardiomyocytes restores NAD+ levels and ameliorates cardiac and mitochondrial dysfunction. Mechanistically, autophagic inhibition causes the accumulation of SQSTM1, which activates NF-κB signaling and promotes NNMT transcription. In summary, we describe a novel mechanism illustrating how autophagic flux maintains mitochondrial and cardiac function by mediating SQSTM1-NF-κB-NNMT signaling and controlling the cellular levels of NAD+.
Collapse
Affiliation(s)
- Quanjiang Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Zhonggang Li
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Human Genetics, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Qiuxia Li
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Samuel Aj Trammell
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Mark S Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Karla Maria Pires
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jinjin Cai
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Yuan Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Helena Kenny
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sihem Boudina
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, 84112, USA
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Diabetes & Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - E Dale Abel
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
21
|
Yagi M, Do Y, Hirai H, Miki K, Toshima T, Fukahori Y, Setoyama D, Abe C, Nabeshima YI, Kang D, Uchiumi T. Improving lysosomal ferroptosis with NMN administration protects against heart failure. Life Sci Alliance 2023; 6:e202302116. [PMID: 37793777 PMCID: PMC10551641 DOI: 10.26508/lsa.202302116] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023] Open
Abstract
Myocardial mitochondria are primary sites of myocardial energy metabolism. Mitochondrial disorders are associated with various cardiac diseases. We previously showed that mice with cardiomyocyte-specific knockout of the mitochondrial translation factor p32 developed heart failure from dilated cardiomyopathy. Mitochondrial translation defects cause not only mitochondrial dysfunction but also decreased nicotinamide adenine dinucleotide (NAD+) levels, leading to impaired lysosomal acidification and autophagy. In this study, we investigated whether nicotinamide mononucleotide (NMN) administration, which compensates for decreased NAD+ levels, improves heart failure because of mitochondrial dysfunction. NMN administration reduced damaged lysosomes and improved autophagy, thereby reducing heart failure and extending the lifespan in p32cKO mice. We found that lysosomal damage due to mitochondrial dysfunction induced ferroptosis, involving the accumulation of iron in lysosomes and lipid peroxide. The ameliorative effects of NMN supplementation were found to strongly affect lysosomal function rather than mitochondrial function, particularly lysosome-mediated ferroptosis. NMN supplementation can improve lysosomal, rather than mitochondrial, function and prevent chronic heart failure.
Collapse
Affiliation(s)
- Mikako Yagi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yura Do
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Haruka Hirai
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenji Miki
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Toshima
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yukina Fukahori
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Chiaki Abe
- Department of Aging Science and Medicine, Graduate School of Medicine Kyoto University Medical Innovation Center, Kyoto, Japan
| | - Yo-Ichi Nabeshima
- Department of Aging Science and Medicine, Graduate School of Medicine Kyoto University Medical Innovation Center, Kyoto, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Willemen HLDM, Santos Ribeiro PS, Broeks M, Meijer N, Versteeg S, Tiggeler A, de Boer TP, Małecki JM, Falnes PØ, Jans J, Eijkelkamp N. Inflammation-induced mitochondrial and metabolic disturbances in sensory neurons control the switch from acute to chronic pain. Cell Rep Med 2023; 4:101265. [PMID: 37944527 PMCID: PMC10694662 DOI: 10.1016/j.xcrm.2023.101265] [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: 11/16/2022] [Revised: 07/24/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
Pain often persists in patients with an inflammatory disease, even when inflammation has subsided. The molecular mechanisms leading to this failure in pain resolution and the transition to chronic pain are poorly understood. Mitochondrial dysfunction in sensory neurons links to chronic pain, but its role in resolution of inflammatory pain is unclear. Transient inflammation causes neuronal plasticity, called hyperalgesic priming, which impairs resolution of pain induced by a subsequent inflammatory stimulus. We identify that hyperalgesic priming in mice increases the expression of a mitochondrial protein (ATPSc-KMT) and causes mitochondrial and metabolic disturbances in sensory neurons. Inhibition of mitochondrial respiration, knockdown of ATPSCKMT expression, or supplementation of the affected metabolite is sufficient to restore resolution of inflammatory pain and prevents chronic pain development. Thus, inflammation-induced mitochondrial-dependent disturbances in sensory neurons predispose to a failure in resolution of inflammatory pain and development of chronic pain.
Collapse
Affiliation(s)
- Hanneke L D M Willemen
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Patrícia Silva Santos Ribeiro
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Melissa Broeks
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Nils Meijer
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Sabine Versteeg
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Annefien Tiggeler
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 Utrecht, the Netherlands
| | - Jędrzej M Małecki
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; CRES-O - Centre for Embryology and Healthy Development, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Pål Ø Falnes
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; CRES-O - Centre for Embryology and Healthy Development, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Judith Jans
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Niels Eijkelkamp
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands.
| |
Collapse
|
23
|
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
|
24
|
Francisco J, Del Re DP. Inflammation in Myocardial Ischemia/Reperfusion Injury: Underlying Mechanisms and Therapeutic Potential. Antioxidants (Basel) 2023; 12:1944. [PMID: 38001797 PMCID: PMC10669026 DOI: 10.3390/antiox12111944] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Acute myocardial infarction (MI) occurs when blood flow to the myocardium is restricted, leading to cardiac damage and massive loss of viable cardiomyocytes. Timely restoration of coronary flow is considered the gold standard treatment for MI patients and limits infarct size; however, this intervention, known as reperfusion, initiates a complex pathological process that somewhat paradoxically also contributes to cardiac injury. Despite being a sterile environment, ischemia/reperfusion (I/R) injury triggers inflammation, which contributes to infarct expansion and subsequent cardiac remodeling and wound healing. The immune response is comprised of subsets of both myeloid and lymphoid-derived cells that act in concert to modulate the pathogenesis and resolution of I/R injury. Multiple mechanisms, including altered metabolic status, regulate immune cell activation and function in the setting of acute MI, yet our understanding remains incomplete. While numerous studies demonstrated cardiac benefit following strategies that target inflammation in preclinical models, therapeutic attempts to mitigate I/R injury in patients were less successful. Therefore, further investigation leveraging emerging technologies is needed to better characterize this intricate inflammatory response and elucidate its influence on cardiac injury and the progression to heart failure.
Collapse
Affiliation(s)
| | - Dominic P. Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
25
|
Thorp EB. Cardiac macrophages and emerging roles for their metabolism after myocardial infarction. J Clin Invest 2023; 133:e171953. [PMID: 37712418 PMCID: PMC10503791 DOI: 10.1172/jci171953] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Interest in cardioimmunology has reached new heights as the experimental cardiology field works to tap the unrealized potential of immunotherapy for clinical care. Within this space is the cardiac macrophage, a key modulator of cardiac function in health and disease. After a myocardial infarction, myeloid macrophages both protect and harm the heart. To varying degrees, such outcomes are a function of myeloid ontogeny and heterogeneity, as well as functional cellular plasticity. Diversity is further shaped by the extracellular milieu, which fluctuates considerably after coronary occlusion. Ischemic limitation of nutrients constrains the metabolic potential of immune cells, and accumulating evidence supports a paradigm whereby macrophage metabolism is coupled to divergent inflammatory consequences, although experimental evidence for this in the heart is just emerging. Herein we examine the heterogeneous cardiac macrophage response following ischemic injury, with a focus on integrating putative contributions of immunometabolism and implications for therapeutically relevant cardiac injury versus cardiac repair.
Collapse
|
26
|
Bornstein R, Mulholland MT, Sedensky M, Morgan P, Johnson SC. Glutamine metabolism in diseases associated with mitochondrial dysfunction. Mol Cell Neurosci 2023; 126:103887. [PMID: 37586651 PMCID: PMC10773532 DOI: 10.1016/j.mcn.2023.103887] [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: 05/19/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023] Open
Abstract
Mitochondrial dysfunction can arise from genetic defects or environmental exposures and impact a wide range of biological processes. Among these are metabolic pathways involved in glutamine catabolism, anabolism, and glutamine-glutamate cycling. In recent years, altered glutamine metabolism has been found to play important roles in the pathologic consequences of mitochondrial dysfunction. Glutamine is a pleiotropic molecule, not only providing an alternate carbon source to glucose in certain conditions, but also playing unique roles in cellular communication in neurons and astrocytes. Glutamine consumption and catabolic flux can be significantly altered in settings of genetic mitochondrial defects or exposure to mitochondrial toxins, and alterations to glutamine metabolism appears to play a particularly significant role in neurodegenerative diseases. These include primary mitochondrial diseases like Leigh syndrome (subacute necrotizing encephalopathy) and MELAS (mitochondrial myopathy with encephalopathy, lactic acidosis, and stroke-like episodes), as well as complex age-related neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Pharmacologic interventions targeting glutamine metabolizing and catabolizing pathways appear to provide some benefits in cell and animal models of these diseases, indicating glutamine metabolism may be a clinically relevant target. In this review, we discuss glutamine metabolism, mitochondrial disease, the impact of mitochondrial dysfunction on glutamine metabolic processes, glutamine in neurodegeneration, and candidate targets for therapeutic intervention.
Collapse
Affiliation(s)
- Rebecca Bornstein
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, USA
| | - Michael T Mulholland
- Department of Applied Sciences, Translational Bioscience, Northumbria University, Newcastle, UK
| | - Margaret Sedensky
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, USA
| | - Phil Morgan
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, USA
| | - Simon C Johnson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA; Department of Neurology, University of Washington, Seattle, USA; Department of Applied Sciences, Translational Bioscience, Northumbria University, Newcastle, UK.
| |
Collapse
|
27
|
Yang S, Park JH, Lu HC. Axonal energy metabolism, and the effects in aging and neurodegenerative diseases. Mol Neurodegener 2023; 18:49. [PMID: 37475056 PMCID: PMC10357692 DOI: 10.1186/s13024-023-00634-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/08/2023] [Indexed: 07/22/2023] Open
Abstract
Human studies consistently identify bioenergetic maladaptations in brains upon aging and neurodegenerative disorders of aging (NDAs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and Amyotrophic lateral sclerosis. Glucose is the major brain fuel and glucose hypometabolism has been observed in brain regions vulnerable to aging and NDAs. Many neurodegenerative susceptible regions are in the topological central hub of the brain connectome, linked by densely interconnected long-range axons. Axons, key components of the connectome, have high metabolic needs to support neurotransmission and other essential activities. Long-range axons are particularly vulnerable to injury, neurotoxin exposure, protein stress, lysosomal dysfunction, etc. Axonopathy is often an early sign of neurodegeneration. Recent studies ascribe axonal maintenance failures to local bioenergetic dysregulation. With this review, we aim to stimulate research in exploring metabolically oriented neuroprotection strategies to enhance or normalize bioenergetics in NDA models. Here we start by summarizing evidence from human patients and animal models to reveal the correlation between glucose hypometabolism and connectomic disintegration upon aging/NDAs. To encourage mechanistic investigations on how axonal bioenergetic dysregulation occurs during aging/NDAs, we first review the current literature on axonal bioenergetics in distinct axonal subdomains: axon initial segments, myelinated axonal segments, and axonal arbors harboring pre-synaptic boutons. In each subdomain, we focus on the organization, activity-dependent regulation of the bioenergetic system, and external glial support. Second, we review the mechanisms regulating axonal nicotinamide adenine dinucleotide (NAD+) homeostasis, an essential molecule for energy metabolism processes, including NAD+ biosynthetic, recycling, and consuming pathways. Third, we highlight the innate metabolic vulnerability of the brain connectome and discuss its perturbation during aging and NDAs. As axonal bioenergetic deficits are developing into NDAs, especially in asymptomatic phase, they are likely exaggerated further by impaired NAD+ homeostasis, the high energetic cost of neural network hyperactivity, and glial pathology. Future research in interrogating the causal relationship between metabolic vulnerability, axonopathy, amyloid/tau pathology, and cognitive decline will provide fundamental knowledge for developing therapeutic interventions.
Collapse
Affiliation(s)
- Sen Yang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jung Hyun Park
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
28
|
Meng L, Wu G. Recent advances in small molecules for improving mitochondrial disorders. RSC Adv 2023; 13:20476-20485. [PMID: 37435377 PMCID: PMC10331567 DOI: 10.1039/d3ra03313a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/03/2023] [Indexed: 07/13/2023] Open
Abstract
Mitochondrial disorders are observed in various human diseases, including rare genetic disorders and complex acquired pathologies. Recent advances in molecular biological techniques have dramatically expanded the understanding of multiple pathomechanisms involving mitochondrial disorders. However, the therapeutic methods for mitochondrial disorders are limited. For this reason, there is increasing interest in identifying safe and effective strategies to mitigate mitochondrial impairments. Small-molecule therapies hold promise for improving mitochondrial performance. This review focuses on the latest advances in developing bioactive compounds for treating mitochondrial disease, aiming to provide a broader perspective of fundamental studies that have been carried out to evaluate the effects of small molecules in regulating mitochondrial function. Novel-designed small molecules ameliorating mitochondrial functions are urgent for further research.
Collapse
Affiliation(s)
- Liying Meng
- Department of Central Laboratory and Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University Qingdao China
| | - Guanzhao Wu
- Department of Central Laboratory and Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University Qingdao China
| |
Collapse
|
29
|
Ju WK, Perkins GA, Kim KY, Bastola T, Choi WY, Choi SH. Glaucomatous optic neuropathy: Mitochondrial dynamics, dysfunction and protection in retinal ganglion cells. Prog Retin Eye Res 2023; 95:101136. [PMID: 36400670 DOI: 10.1016/j.preteyeres.2022.101136] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/04/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022]
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide and is characterized by a slow, progressive, and multifactorial degeneration of retinal ganglion cells (RGCs) and their axons, resulting in vision loss. Despite its high prevalence in individuals 60 years of age and older, the causing factors contributing to glaucoma progression are currently not well characterized. Intraocular pressure (IOP) is the only proven treatable risk factor. However, lowering IOP is insufficient for preventing disease progression. One of the significant interests in glaucoma pathogenesis is understanding the structural and functional impairment of mitochondria in RGCs and their axons and synapses. Glaucomatous risk factors such as IOP elevation, aging, genetic variation, neuroinflammation, neurotrophic factor deprivation, and vascular dysregulation, are potential inducers for mitochondrial dysfunction in glaucoma. Because oxidative phosphorylation stress-mediated mitochondrial dysfunction is associated with structural and functional impairment of mitochondria in glaucomatous RGCs, understanding the underlying mechanisms and relationship between structural and functional alterations in mitochondria would be beneficial to developing mitochondria-related neuroprotection in RGCs and their axons and synapses against glaucomatous neurodegeneration. Here, we review the current studies focusing on mitochondrial dynamics-based structural and functional alterations in the mitochondria of glaucomatous RGCs and therapeutic strategies to protect RGCs against glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Won-Kyu Ju
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tonking Bastola
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA
| | - Woo-Young Choi
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA; Department of Plastic Surgery, College of Medicine, Chosun University, Gwang-ju, South Korea
| | - Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
30
|
DeBerge M, Chaudhary R, Schroth S, Thorp EB. Immunometabolism at the Heart of Cardiovascular Disease. JACC Basic Transl Sci 2023; 8:884-904. [PMID: 37547069 PMCID: PMC10401297 DOI: 10.1016/j.jacbts.2022.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 08/08/2023]
Abstract
Immune cell function among the myocardium, now more than ever, is appreciated to regulate cardiac function and pathophysiology. This is the case for both innate immunity, which includes neutrophils, monocytes, dendritic cells, and macrophages, as well as adaptive immunity, which includes T cells and B cells. This function is fueled by cell-intrinsic shifts in metabolism, such as glycolysis and oxidative phosphorylation, as well as metabolite availability, which originates from the surrounding extracellular milieu and varies during ischemia and metabolic syndrome. Immune cell crosstalk with cardiac parenchymal cells, such as cardiomyocytes and fibroblasts, is also regulated by complex cellular metabolic circuits. Although our understanding of immunometabolism has advanced rapidly over the past decade, in part through valuable insights made in cultured cells, there remains much to learn about contributions of in vivo immunometabolism and directly within the myocardium. Insight into such fundamental cell and molecular mechanisms holds potential to inform interventions that shift the balance of immunometabolism from maladaptive to cardioprotective and potentially even regenerative. Herein, we review our current working understanding of immunometabolism, specifically in the settings of sterile ischemic cardiac injury or cardiometabolic disease, both of which contribute to the onset of heart failure. We also discuss current gaps in knowledge in this context and therapeutic implications.
Collapse
Affiliation(s)
| | | | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
31
|
Olkhova EA, Smith LA, Bradshaw C, Gorman GS, Erskine D, Ng YS. Neurological Phenotypes in Mouse Models of Mitochondrial Disease and Relevance to Human Neuropathology. Int J Mol Sci 2023; 24:ijms24119698. [PMID: 37298649 DOI: 10.3390/ijms24119698] [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: 05/13/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Mitochondrial diseases represent the most common inherited neurometabolic disorders, for which no effective therapy currently exists for most patients. The unmet clinical need requires a more comprehensive understanding of the disease mechanisms and the development of reliable and robust in vivo models that accurately recapitulate human disease. This review aims to summarise and discuss various mouse models harbouring transgenic impairments in genes that regulate mitochondrial function, specifically their neurological phenotype and neuropathological features. Ataxia secondary to cerebellar impairment is one of the most prevalent neurological features of mouse models of mitochondrial dysfunction, consistent with the observation that progressive cerebellar ataxia is a common neurological manifestation in patients with mitochondrial disease. The loss of Purkinje neurons is a shared neuropathological finding in human post-mortem tissues and numerous mouse models. However, none of the existing mouse models recapitulate other devastating neurological phenotypes, such as refractory focal seizures and stroke-like episodes seen in patients. Additionally, we discuss the roles of reactive astrogliosis and microglial reactivity, which may be driving the neuropathology in some of the mouse models of mitochondrial dysfunction, as well as mechanisms through which cellular death may occur, beyond apoptosis, in neurons undergoing mitochondrial bioenergy crisis.
Collapse
Affiliation(s)
- Elizaveta A Olkhova
- 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
| | - Laura A Smith
- 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
| | - Carla Bradshaw
- 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
| | - 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
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Daniel Erskine
- 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 Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Yi Shiau Ng
- 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
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
32
|
Bitto A, Grillo AS, Ito TK, Stanaway IB, Nguyen BMG, Ying K, Tung H, Smith K, Tran N, Velikanje G, Urfer SR, Snyder JM, Barton J, Sharma A, Kayser EB, Wang L, Smith DL, Thompson JW, DuBois L, DePaolo W, Kaeberlein M. Acarbose suppresses symptoms of mitochondrial disease in a mouse model of Leigh syndrome. Nat Metab 2023; 5:955-967. [PMID: 37365290 DOI: 10.1038/s42255-023-00815-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/04/2023] [Indexed: 06/28/2023]
Abstract
Mitochondrial diseases represent a spectrum of disorders caused by impaired mitochondrial function, ranging in severity from mortality during infancy to progressive adult-onset disease. Mitochondrial dysfunction is also recognized as a molecular hallmark of the biological ageing process. Rapamycin, a drug that increases lifespan and health during normative ageing, also increases survival and reduces neurological symptoms in a mouse model of the severe mitochondrial disease Leigh syndrome. The Ndufs4 knockout (Ndufs4-/-) mouse lacks the complex I subunit NDUFS4 and shows rapid onset and progression of neurodegeneration mimicking patients with Leigh syndrome. Here we show that another drug that extends lifespan and delays normative ageing in mice, acarbose, also suppresses symptoms of disease and improves survival of Ndufs4-/- mice. Unlike rapamycin, acarbose rescues disease phenotypes independently of inhibition of the mechanistic target of rapamycin. Furthermore, rapamycin and acarbose have additive effects in delaying neurological symptoms and increasing maximum lifespan in Ndufs4-/- mice. We find that acarbose remodels the intestinal microbiome and alters the production of short-chain fatty acids. Supplementation with tributyrin, a source of butyric acid, recapitulates some effects of acarbose on lifespan and disease progression, while depletion of the endogenous microbiome in Ndufs4-/- mice appears to fully recapitulate the effects of acarbose on healthspan and lifespan in these animals. To our knowledge, this study provides the first evidence that alteration of the gut microbiome plays a significant role in severe mitochondrial disease and provides further support for the model that biological ageing and severe mitochondrial disorders share underlying common mechanisms.
Collapse
Affiliation(s)
- Alessandro Bitto
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Anthony S Grillo
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Takashi K Ito
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Ian B Stanaway
- Division of Nephrology, School of Medicine, University of Washington, Seattle, WA, USA
- Harborview Medical Center, Kidney Research Institute, Seattle, WA, USA
| | - Bao M G Nguyen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kejun Ying
- T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | | | - Ngoc Tran
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Gunnar Velikanje
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Silvan R Urfer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Jessica M Snyder
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Jacob Barton
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Ayush Sharma
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Daniel L Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Will Thompson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Laura DuBois
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - William DePaolo
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
33
|
van Kraaij SJW, Pereira DR, Smal B, Summo L, Konkel A, Lossie J, Busjahn A, Grammatopoulos TN, Klaassen E, Fischer R, Schunck WH, Gal P, Moerland M. Identification of peripheral vascular function measures and circulating biomarkers of mitochondrial function in patients with mitochondrial disease. Clin Transl Sci 2023. [PMID: 37177864 DOI: 10.1111/cts.13530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
The development of pharmacological therapies for mitochondrial diseases is hampered by the lack of tissue-level and circulating biomarkers reflecting effects of compounds on endothelial and mitochondrial function. This phase 0 study aimed to identify biomarkers differentiating between patients with mitochondrial disease and healthy volunteers (HVs). In this cross-sectional case-control study, eight participants with mitochondrial disease and eight HVs matched on age, sex, and body mass index underwent study assessments consisting of blood collection for evaluation of plasma and serum biomarkers, mitochondrial function in peripheral blood mononuclear cells (PBMCs), and an array of imaging methods for assessment of (micro)circulation. Plasma biomarkers GDF-15, IL-6, NT-proBNP, and cTNI were significantly elevated in patients compared to HVs, as were several clinical chemistry and hematology markers. No differences between groups were found for mitochondrial membrane potential, mitochondrial reactive oxygen production, oxygen consumption rate, or extracellular acidification rate in PBMCs. Imaging revealed significantly higher nicotinamide-adenine-dinucleotide-hydrogen (NADH) content in skin as well as reduced passive leg movement-induced hyperemia in patients. This study confirmed results of earlier studies regarding plasma biomarkers in mitochondrial disease and identified several imaging techniques that could detect functional differences at the tissue level between participants with mitochondrial disease and HVs. However, assays of mitochondrial function in PBMCs did not show differences between participants with mitochondrial disease and HVs, possibly reflecting compensatory mechanisms and heterogeneity in mutational load. In future clinical trials, using a mix of imaging and blood-based biomarkers may be advisable, as well as combining these with an in vivo challenge to disturb homeostasis.
Collapse
Affiliation(s)
- Sebastiaan J W van Kraaij
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Bastiaan Smal
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | | | | | | | | | | | | | - Wolf-Hagen Schunck
- OMEICOS Therapeutics GmbH, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Pim Gal
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Medical Centre, Leiden, The Netherlands
| | - Matthijs Moerland
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
34
|
Chen B, Daneshgar N, Lee HC, Song LS, Dai DF. Mitochondrial Oxidative Stress Mediates Bradyarrhythmia in Leigh Syndrome Mitochondrial Disease Mice. Antioxidants (Basel) 2023; 12:antiox12051001. [PMID: 37237867 PMCID: PMC10215409 DOI: 10.3390/antiox12051001] [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: 02/27/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Mitochondrial oxidative stress has been implicated in aging and several cardiovascular diseases, including heart failure and cardiomyopathy, ventricular tachycardia, and atrial fibrillation. The role of mitochondrial oxidative stress in bradyarrhythmia is less clear. Mice with a germline deletion of Ndufs4 subunit respiratory complex I develop severe mitochondrial encephalomyopathy resembling Leigh Syndrome (LS). Several types of cardiac bradyarrhythmia are present in LS mice, including a frequent sinus node dysfunction and episodic atrioventricular (AV) block. Treatment with the mitochondrial antioxidant Mitotempo or mitochondrial protective peptide SS31 significantly ameliorated the bradyarrhythmia and extended the lifespan of LS mice. Using an ex vivo Langendorff perfused heart with live confocal imaging of mitochondrial and total cellular reactive oxygen species (ROS), we showed increased ROS in the LS heart, which was potentiated by ischemia-reperfusion. A simultaneous ECG recording showed a sinus node dysfunction and AV block concurrent with the severity of the oxidative stress. Treatment with Mitotempo abolished ROS and restored the sinus rhythm. Our study reveals robust evidence of the direct mechanistic roles of mitochondrial and total ROS in bradyarrhythmia in the setting of LS mitochondrial cardiomyopathy. Our study also supports the potential clinical application of mitochondrial-targeted antioxidants or SS31 for the treatment of LS patients.
Collapse
Affiliation(s)
- Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Nastaran Daneshgar
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Hsiang-Chun Lee
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Dao-Fu Dai
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
35
|
Karaa A, Klopstock T. Clinical trials in mitochondrial diseases. HANDBOOK OF CLINICAL NEUROLOGY 2023; 194:229-250. [PMID: 36813315 DOI: 10.1016/b978-0-12-821751-1.00002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Primary mitochondrial diseases are some of the most common and complex inherited inborn errors of metabolism. Their molecular and phenotypic diversity has led to difficulties in finding disease-modifying therapies and clinical trial efforts have been slow due to multiple significant challenges. Lack of robust natural history data, difficulties in finding specific biomarkers, absence of well-validated outcome measures, and small patient numbers have made clinical trial design and conduct difficult. Encouragingly, new interest in treating mitochondrial dysfunction in common diseases and regulatory incentives to develop therapies for rare conditions have led to significant interest and efforts to develop drugs for primary mitochondrial diseases. Here, we review past and present clinical trials and future strategies of drug development in primary mitochondrial diseases.
Collapse
Affiliation(s)
- Amel Karaa
- Mitochondrial Disease Program, Division of Medical Genetics and Metabolism, Massachusetts General Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institute, University Hospital, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Network for mitochondrial disorders (mitoNET), Munich, Germany
| |
Collapse
|
36
|
Kelly CJ, Couch RK, Ha VT, Bodart CM, Wu J, Huff S, Herrel NT, Kim HD, Zimmermann AO, Shattuck J, Pan YC, Kaeberlein M, Grillo AS. Iron status influences mitochondrial disease progression in Complex I-deficient mice. eLife 2023; 12:e75825. [PMID: 36799301 PMCID: PMC10030112 DOI: 10.7554/elife.75825] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Mitochondrial dysfunction caused by aberrant Complex I assembly and reduced activity of the electron transport chain is pathogenic in many genetic and age-related diseases. Mice missing the Complex I subunit NADH dehydrogenase [ubiquinone] iron-sulfur protein 4 (NDUFS4) are a leading mammalian model of severe mitochondrial disease that exhibit many characteristic symptoms of Leigh Syndrome including oxidative stress, neuroinflammation, brain lesions, and premature death. NDUFS4 knockout mice have decreased expression of nearly every Complex I subunit. As Complex I normally contains at least 8 iron-sulfur clusters and more than 25 iron atoms, we asked whether a deficiency of Complex I may lead to iron perturbations, thereby accelerating disease progression. Consistent with this, iron supplementation accelerates symptoms of brain degeneration in these mice, while iron restriction delays the onset of these symptoms, reduces neuroinflammation, and increases survival. NDUFS4 knockout mice display signs of iron overload in the liver including increased expression of hepcidin and show changes in iron-responsive element-regulated proteins consistent with increased cellular iron that were prevented by iron restriction. These results suggest that perturbed iron homeostasis may contribute to pathology in Leigh Syndrome and possibly other mitochondrial disorders.
Collapse
Affiliation(s)
- CJ Kelly
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Reid K Couch
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Vivian T Ha
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Camille M Bodart
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Judy Wu
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Sydney Huff
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Nicole T Herrel
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Hyunsung D Kim
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Azaad O Zimmermann
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Jessica Shattuck
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Yu-Chen Pan
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Matt Kaeberlein
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| | - Anthony S Grillo
- Department of Laboratory Medicine & Pathology, University of WashingtonSeattleUnited States
| |
Collapse
|
37
|
Morel JD, Sleiman MB, Li TY, von Alvensleben G, Bachmann AM, Hofer D, Broeckx E, Ma JY, Carreira V, Chen T, Azhar N, Gonzalez-Villalobos RA, Breyer M, Reilly D, Mullican S, Auwerx J. Mitochondrial and NAD+ metabolism predict recovery from acute kidney injury in a diverse mouse population. JCI Insight 2023; 8:164626. [PMID: 36752209 PMCID: PMC9977436 DOI: 10.1172/jci.insight.164626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/29/2022] [Indexed: 02/09/2023] Open
Abstract
Acute kidney failure and chronic kidney disease are global health issues steadily rising in incidence and prevalence. Animal models on a single genetic background have so far failed to recapitulate the clinical presentation of human nephropathies. Here, we used a simple model of folic acid-induced kidney injury in 7 highly diverse mouse strains. We measured plasma and urine parameters, as well as renal histopathology and mRNA expression data, at 1, 2, and 6 weeks after injury, covering the early recovery and long-term remission. We observed an extensive strain-specific response ranging from complete resistance of the CAST/EiJ to high sensitivity of the C57BL/6J, DBA/2J, and PWK/PhJ strains. In susceptible strains, the severe early kidney injury was accompanied by the induction of mitochondrial stress response (MSR) genes and the attenuation of NAD+ synthesis pathways. This is associated with delayed healing and a prolonged inflammatory and adaptive immune response 6 weeks after insult, heralding a transition to chronic kidney disease. Through a thorough comparison of the transcriptomic response in mouse and human disease, we show that critical metabolic gene alterations were shared across species, and we highlight the PWK/PhJ strain as an emergent model of transition from acute kidney injury to chronic disease.
Collapse
Affiliation(s)
- Jean-David Morel
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Terytty Yang Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Giacomo von Alvensleben
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alexis M. Bachmann
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Dina Hofer
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ellen Broeckx
- Janssen Research and Development LLC, Raritan, New Jersey, USA
| | - Jing Ying Ma
- Janssen Research and Development LLC, Raritan, New Jersey, USA
| | | | - Tao Chen
- Janssen Research and Development LLC, Raritan, New Jersey, USA
| | - Nabil Azhar
- Janssen Research and Development LLC, Raritan, New Jersey, USA
| | | | - Matthew Breyer
- Janssen Research and Development LLC, Raritan, New Jersey, USA
| | - Dermot Reilly
- Janssen Research and Development LLC, Raritan, New Jersey, USA
| | | | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
38
|
Jobst M, Kiss E, Gerner C, Marko D, Del Favero G. Activation of autophagy triggers mitochondrial loss and changes acetylation profile relevant for mechanotransduction in bladder cancer cells. Arch Toxicol 2023; 97:217-233. [PMID: 36214828 PMCID: PMC9816236 DOI: 10.1007/s00204-022-03375-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/05/2022] [Indexed: 01/19/2023]
Abstract
Bladder cells are constantly exposed to multiple xenobiotics and bioactive metabolites. In addition to this challenging chemical environment, they are also exposed to shear stress originating from urine and interstitial fluids. Hence, physiological function of bladder cells relies on a high biochemical and biomechanical adaptive competence, which, in turn, is largely supported via autophagy-related mechanisms. As a negative side of this plasticity, bladder cancer cells are known to adapt readily to chemotherapeutic programs. At the molecular level, autophagy was described to support resistance against pharmacological treatments and to contribute to the maintenance of cell structure and metabolic competence. In this study, we enhanced autophagy with rapamycin (1-100 nM) and assessed its effects on the motility of bladder cells, as well as the capability to respond to shear stress. We observed that rapamycin reduced cell migration and the mechanical-induced translocation potential of Krüppel-like transcription factor 2 (KLF2). These effects were accompanied by a rearrangement of cytoskeletal elements and mitochondrial loss. In parallel, intracellular acetylation levels were decreased. Mechanistically, inhibition of the NAD + -dependent deacetylase sirtuin-1 (SIRT1) with nicotinamide (NAM; 0.1-5 mM) restored acetylation levels hampered by rapamycin and cell motility. Taken together, we described the effects of rapamycin on cytoskeletal elements crucial for mechanotransduction and the dependency of these changes on the mitochondrial turnover caused by autophagy activation. Additionally, we could show that targeted metabolic intervention could revert the outcome of autophagy activation, reinforcing the idea that bladder cells can easily adapt to multiple xenobiotics and circumvent in this way the effects of single chemicals.
Collapse
Affiliation(s)
- Maximilian Jobst
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090 Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal, Imaging, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090 Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090 Vienna, Austria ,Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090 Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090 Vienna, Austria ,Core Facility Multimodal, Imaging, Faculty of Chemistry, University of Vienna, Währingerstr. 38-40, 1090 Vienna, Austria
| |
Collapse
|
39
|
Warwick AM, Bomze HM, Wang L, Klingeborn M, Hao Y, Stinnett SS, Gospe III SM. Continuous Hypoxia Reduces Retinal Ganglion Cell Degeneration in a Mouse Model of Mitochondrial Optic Neuropathy. Invest Ophthalmol Vis Sci 2022; 63:21. [PMID: 36538003 PMCID: PMC9769749 DOI: 10.1167/iovs.63.13.21] [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] [Indexed: 12/24/2022] Open
Abstract
Purpose To test whether continuous hypoxia is neuroprotective to retinal ganglion cells (RGCs) in a mouse model of mitochondrial optic neuropathy. Methods RGC degeneration was assessed in genetically modified mice in which the floxed gene for the complex I subunit NDUFS4 is deleted from RGCs using Vlgut2-driven Cre recombinase. Beginning at postnatal day 25 (P25), Vglut2-Cre;ndufs4loxP/loxP mice and control littermates were housed under hypoxia (11% oxygen) or kept under normoxia (21% oxygen). Survival of RGC somas and axons was assessed at P60 and P90 via histological analysis of retinal flatmounts and optic nerve cross-sections, respectively. Retinal tissue was also assessed for gliosis and neuroinflammation using western blot and immunofluorescence. Results Consistent with our previous characterization of this model, at least one-third of RGCs had degenerated by P60 in Vglut2-Cre;ndufs4loxP/loxP mice remaining under normoxia. However, continuous hypoxia resulted in complete rescue of RGC somas and axons at this time point, with normal axonal myelination observed on electron microscopy. Though only partial, hypoxia-mediated rescue of complex I-deficient RGC somas and axons remained significant at P90. Hypoxia prevented reactive gliosis at P60, but the retinal accumulation of Iba1+ mononuclear phagocytic cells was not substantially reduced. Conclusions Continuous hypoxia achieved dramatic rescue of early RGC degeneration in mice with severe mitochondrial dysfunction. Although complete rescue was not durable to P90, our observations suggest that investigating the mechanisms underlying hypoxia-mediated neuroprotection of RGCs may identify useful therapeutic strategies for optic neuropathies resulting from less profound mitochondrial impairment, such as Leber hereditary optic neuropathy.
Collapse
Affiliation(s)
- Alexander M. Warwick
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Howard M. Bomze
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States,Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Luyu Wang
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Mikael Klingeborn
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Ying Hao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Sandra S. Stinnett
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Sidney M. Gospe III
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
40
|
Elucidation of the Role of FAM210B in Mitochondrial Metabolism and Erythropoiesis. Mol Cell Biol 2022; 42:e0014322. [PMID: 36374104 PMCID: PMC9753634 DOI: 10.1128/mcb.00143-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Mitochondria play essential and specific roles during erythroid differentiation. Recently, FAM210B, encoding a mitochondrial inner membrane protein, has been identified as a novel target of GATA-1, as well as an erythropoietin-inducible gene. While FAM210B protein is involved in regulate mitochondrial metabolism and heme biosynthesis, its detailed function remains unknown. Here, we generated both knockout and knockdown of endogenous FAM210B in human induced pluripotent stem-derived erythroid progenitor (HiDEP) cells using CRISPR/Cas9 methodology. Intriguingly, erythroid differentiation was more pronounced in the FAM210B-depleted cells, and this resulted in increased frequency of orthochromatic erythroblasts and decreased frequencies of basophilic/polychromatic erythroblasts. Comprehensive metabolite analysis and functional analysis indicated that oxygen consumption rates and the NAD (NAD+)/NADH ratio were significantly decreased, while lactate production was significantly increased in FAM210B deletion HiDEP cells, indicating involvement of FAM210B in mitochondrial energy metabolism in erythroblasts. Finally, we purified FAM210B-interacting protein from K562 cells that stably expressed His/biotin-tagged FAM210B. Mass spectrometry analysis of the His/biotin-purified material indicated interactions with multiple subunits of mitochondrial ATP synthases, such as subunit alpha (ATP5A) and beta (ATP5B). Our results suggested that FAM210B contributes prominently to erythroid differentiation by regulating mitochondrial energy metabolism. Our results provide insights into the pathophysiology of dysregulated hematopoiesis.
Collapse
|
41
|
Bennett CF, Ronayne CT, Puigserver P. Targeting adaptive cellular responses to mitochondrial bioenergetic deficiencies in human disease. FEBS J 2022; 289:6969-6993. [PMID: 34510753 PMCID: PMC8917243 DOI: 10.1111/febs.16195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/25/2021] [Accepted: 09/10/2021] [Indexed: 01/13/2023]
Abstract
Mitochondrial dysfunction is increasingly appreciated as a central contributor to human disease. Oxidative metabolism at the mitochondrial respiratory chain produces ATP and is intricately tied to redox homeostasis and biosynthetic pathways. Metabolic stress arising from genetic mutations in mitochondrial genes and environmental factors such as malnutrition or overnutrition is perceived by the cell and leads to adaptive and maladaptive responses that can underlie pathology. Here, we will outline cellular sensors that react to alterations in energy production, organellar redox, and metabolites stemming from mitochondrial disease (MD) mutations. MD is a heterogeneous group of disorders primarily defined by defects in mitochondrial oxidative phosphorylation from nuclear or mitochondrial-encoded gene mutations. Preclinical therapies that improve fitness of MD mouse models have been recently identified. Targeting metabolic/energetic deficiencies, maladaptive signaling processes, and hyper-oxygenation of tissues are all strategies aside from direct genetic approaches that hold therapeutic promise. A further mechanistic understanding of these curative processes as well as the identification of novel targets will significantly impact mitochondrial biology and disease research.
Collapse
Affiliation(s)
- Christopher F Bennett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Conor T Ronayne
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Martyniuk V, Khoma V, Matskiv T, Baranovsky V, Orlova-Hudim K, Gylytė B, Symchak R, Matciuk O, Gnatyshyna L, Manusadžianas L, Stoliar O. Indication of the impact of environmental stress on the responses of the bivalve mollusk Unio tumidus to ibuprofen and microplastics based on biomarkers of reductive stress and apoptosis. Comp Biochem Physiol C Toxicol Pharmacol 2022; 261:109425. [PMID: 35914710 DOI: 10.1016/j.cbpc.2022.109425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/19/2022]
Abstract
The vulnerability of bivalve mollusks to micropollutants is estimated mainly in single model exposures. However, chronic environmental stress and complex exposures can modulate their responses. To evaluate the impact of population-dependent adaptations on the ability to react to common micropollutants, we compared freshwater bivalves Unio tumidus from two distinct populations, pure (Pr) and contaminated (Ct), in their exposures to microplastics (MP, 1 mg L-1, size 0.1-0.5 mm), pharmaceutical ibuprofen (IBU, 0.8 μg L-1), or their combination (Mix) for 14 days. Control groups from both sites showed remarkable differences, with lower levels of total antioxidant capacity (TAC), metallothionein protein (MTSH), NADH and NAD+, cytochrome P450-related EROD, glutathione-S transferase (GST), and citrate synthase (CS) but higher levels of GSH, GSSG, caspase-3 and cathepsin D (CTD) in the Ct-control group. These data indicate a chronic stress impact in the Ct population. Under exposures, we found an almost common strategy in both populations for NAD+/NADH and MTSH suppression and CTD induction. Additionally, Mix exposure caused an increase in CS, and IBU did not change GSH in both populations. However, the expected response to IBU - the suppression of caspase-3 - was indicated only in PrIBU- and PrMix-mollusks. CTD efflux increased dramatically only in PrMP- and PrMix- groups, and suppression of EROD and GST was detected in the PrMix-group. According to discriminant analysis, exposed Pr-groups were highly differentiated from control, whereas Ct-control and exposed groups had common localization demonstrating high resistance to environmental stress. Thus, the same exposures resulted in different adverse outcome pathways depending on the population.
Collapse
Affiliation(s)
- Viktoria Martyniuk
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Vira Khoma
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine.
| | - Tetiana Matskiv
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine; I.Ya. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine.
| | - Vitaliy Baranovsky
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine.
| | | | | | - Ruslan Symchak
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine
| | - Oksana Matciuk
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine.
| | - Lesya Gnatyshyna
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine; I.Ya. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine.
| | | | - Oksana Stoliar
- Ternopil Volodymyr Hnatiuk National Pedagogical University, Ternopil, Ukraine.
| |
Collapse
|
43
|
Belal S, Goudenège D, Bocca C, Dumont F, Chao De La Barca JM, Desquiret-Dumas V, Gueguen N, Geffroy G, Benyahia R, Kane S, Khiati S, Bris C, Aranyi T, Stockholm D, Inisan A, Renaud A, Barth M, Simard G, Reynier P, Letournel F, Lenaers G, Bonneau D, Chevrollier A, Procaccio V. Glutamate-Induced Deregulation of Krebs Cycle in Mitochondrial Encephalopathy Lactic Acidosis Syndrome Stroke-Like Episodes (MELAS) Syndrome Is Alleviated by Ketone Body Exposure. Biomedicines 2022; 10:biomedicines10071665. [PMID: 35884972 PMCID: PMC9312837 DOI: 10.3390/biomedicines10071665] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/19/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: The development of mitochondrial medicine has been severely impeded by a lack of effective therapies. (2) Methods: To better understand Mitochondrial Encephalopathy Lactic Acidosis Syndrome Stroke-like episodes (MELAS) syndrome, neuronal cybrid cells carrying different mutation loads of the m.3243A > G mitochondrial DNA variant were analysed using a multi-omic approach. (3) Results: Specific metabolomic signatures revealed that the glutamate pathway was significantly increased in MELAS cells with a direct correlation between glutamate concentration and the m.3243A > G heteroplasmy level. Transcriptomic analysis in mutant cells further revealed alterations in specific gene clusters, including those of the glutamate, gamma-aminobutyric acid pathways, and tricarboxylic acid (TCA) cycle. These results were supported by post-mortem brain tissue analysis from a MELAS patient, confirming the glutamate dysregulation. Exposure of MELAS cells to ketone bodies significantly reduced the glutamate level and improved mitochondrial functions, reducing the accumulation of several intermediate metabolites of the TCA cycle and alleviating the NADH-redox imbalance. (4) Conclusions: Thus, a multi-omic integrated approach to MELAS cells revealed glutamate as a promising disease biomarker, while also indicating that a ketogenic diet should be tested in MELAS patients.
Collapse
Affiliation(s)
- Sophie Belal
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - David Goudenège
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Cinzia Bocca
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Florent Dumont
- Signalling and Cardiovascular Pathophysiology, INSERM UMR-S 1180, University of Paris-Saclay, 92296 Châtenay-Malabry, France;
| | - Juan Manuel Chao De La Barca
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Valérie Desquiret-Dumas
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Naïg Gueguen
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Guillaume Geffroy
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Rayane Benyahia
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Selma Kane
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Salim Khiati
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Céline Bris
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Tamas Aranyi
- Institute of Enzymology, Research Center for Natural Sciences, H-1519 Budapest, Hungary;
- Department of Molecular Biology, Semmelweis University of Medicine, H-1519 Budapest, Hungary
| | - Daniel Stockholm
- Ecole Pratique des Hautes Etudes, PSL Research University, 75014 Paris, France;
- Centre de Recherche Saint-Antoine, UMRS-938, INSERM, Sorbonne Université, F-75012 Paris, France
| | - Aurore Inisan
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Aurélie Renaud
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Magalie Barth
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Gilles Simard
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Pascal Reynier
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Franck Letournel
- Department of Neurobiology-Neuropathology, Angers Hospital, 49933 Angers, France;
- UMR INSERM 1066-CNRS 6021, MINT Laboratory, 49933 Angers, France
| | - Guy Lenaers
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Service de Neurologie, CHU d'Angers, 49933 Angers, France
| | - Dominique Bonneau
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Arnaud Chevrollier
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Vincent Procaccio
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
- Correspondence:
| |
Collapse
|
44
|
Yoon J, Daneshgar N, Chu Y, Chen B, Hefti M, Vikram A, Irani K, Song L, Brenner C, Abel ED, London B, Dai D. Metabolic rescue ameliorates mitochondrial encephalo-cardiomyopathy in murine and human iPSC models of Leigh syndrome. Clin Transl Med 2022; 12:e954. [PMID: 35872650 PMCID: PMC9309541 DOI: 10.1002/ctm2.954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Mice with deletion of complex I subunit Ndufs4 develop mitochondrial encephalomyopathy resembling Leigh syndrome (LS). The metabolic derangement and underlying mechanisms of cardio-encephalomyopathy in LS remains incompletely understood. METHODS We performed echocardiography, electrophysiology, confocal microscopy, metabolic and molecular/morphometric analysis of the mice lacking Ndufs4. HEK293 cells, human iPS cells-derived cardiomyocytes and neurons were used to determine the mechanistic role of mitochondrial complex I deficiency. RESULTS LS mice develop severe cardiac bradyarrhythmia and diastolic dysfunction. Human-induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) with Ndufs4 deletion recapitulate LS cardiomyopathy. Mechanistically, we demonstrate a direct link between complex I deficiency, decreased intracellular (nicotinamide adenine dinucleotide) NAD+ /NADH and bradyarrhythmia, mediated by hyperacetylation of the cardiac sodium channel NaV 1.5, particularly at K1479 site. Neuronal apoptosis in the cerebellar and midbrain regions in LS mice was associated with hyperacetylation of p53 and activation of microglia. Targeted metabolomics revealed increases in several amino acids and citric acid cycle intermediates, likely due to impairment of NAD+ -dependent dehydrogenases, and a substantial decrease in reduced Glutathione (GSH). Metabolic rescue by nicotinamide riboside (NR) supplementation increased intracellular NAD+ / NADH, restored metabolic derangement, reversed protein hyperacetylation through NAD+ -dependent Sirtuin deacetylase, and ameliorated cardiomyopathic phenotypes, concomitant with improvement of NaV 1.5 current and SERCA2a function measured by Ca2+ -transients. NR also attenuated neuronal apoptosis and microglial activation in the LS brain and human iPS-derived neurons with Ndufs4 deletion. CONCLUSIONS Our study reveals direct mechanistic explanations of the observed cardiac bradyarrhythmia, diastolic dysfunction and neuronal apoptosis in mouse and human induced pluripotent stem cells (iPSC) models of LS.
Collapse
Affiliation(s)
- Jin‐Young Yoon
- Division of Cardiovascular Medicine, Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Nastaran Daneshgar
- Department of Pathology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Yi Chu
- Department of Pathology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Marco Hefti
- Department of Pathology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Ajit Vikram
- Division of Cardiovascular Medicine, Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Kaikobad Irani
- Division of Cardiovascular Medicine, Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Long‐Sheng Song
- Division of Cardiovascular Medicine, Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Charles Brenner
- Department of Diabetes & Cancer MetabolismCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - E. Dale Abel
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Department of Internal MedicineUniversity of Iowa Carver College of Medicine, University of IowaIowa CityIowaUSA
| | - Barry London
- Division of Cardiovascular Medicine, Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Dao‐Fu Dai
- Department of Pathology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
45
|
Mitochondrial DNA Is a Vital Driving Force in Ischemia-Reperfusion Injury in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6235747. [PMID: 35620580 PMCID: PMC9129988 DOI: 10.1155/2022/6235747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022]
Abstract
According to the latest Global Burden of Disease Study, cardiovascular disease (CVD) is the leading cause of death, and ischemic heart disease and stroke are the cause of death in approximately half of CVD patients. In CVD, mitochondrial dysfunction following ischemia-reperfusion (I/R) injury results in heart failure. The proper functioning of oxidative phosphorylation (OXPHOS) and the mitochondrial life cycle in cardiac mitochondria are closely related to mitochondrial DNA (mtDNA). Following myocardial I/R injury, mitochondria activate multiple repair and clearance mechanisms to repair damaged mtDNA. When these repair mechanisms are insufficient to restore the structure and function of mtDNA, irreversible mtDNA damage occurs, leading to mtDNA mutations. Since mtDNA mutations aggravate OXPHOS dysfunction and affect mitophagy, mtDNA mutation accumulation leads to leakage of mtDNA and proteins outside the mitochondria, inducing an innate immune response, aggravating cardiovascular injury, and leading to the need for external interventions to stop or slow the disease course. On the other hand, mtDNA released into the circulation after cardiac injury can serve as a biomarker for CVD diagnosis and prognosis. This article reviews the pathogenic basis and related research findings of mtDNA oxidative damage and mtDNA leak-triggered innate immune response associated with I/R injury in CVD and summarizes therapeutic options that target mtDNA.
Collapse
|
46
|
Yuan Y, Liang B, Liu XL, Liu WJ, Huang BH, Yang SB, Gao YZ, Meng JS, Li MJ, Ye T, Wang CZ, Hu XK, Xing DM. Targeting NAD+: is it a common strategy to delay heart aging? Cell Death Dis 2022; 8:230. [PMID: 35474295 PMCID: PMC9042931 DOI: 10.1038/s41420-022-01031-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/17/2022]
Abstract
Heart aging is the main susceptible factor to coronary heart disease and significantly increases the risk of heart failure, especially when the aging heart is suffering from ischemia-reperfusion injury. Numerous studies with NAD+ supplementations have suggested its use in anti-aging treatment. However, systematic reviews regarding the overall role of NAD+ in cardiac aging are scarce. The relationship between NAD+ signaling and heart aging has yet to be clarified. This review comprehensively summarizes the current studies on the role of NAD+ signaling in delaying heart aging from the following aspects: the influence of NAD+ supplementations on the aging heart; the relationship and cross-talks between NAD+ signaling and other cardiac aging-related signaling pathways; Importantly, the therapeutic potential of targeting NAD+ in delaying heart aging will be discussed. In brief, NAD+ plays a vital role in delaying heart aging. However, the abnormalities such as altered glucose and lipid metabolism, oxidative stress, and calcium overload could also interfere with NAD+ function in the heart. Therefore, the specific physiopathology of the aging heart should be considered before applying NAD+ supplementations. We believe that this article will help augment our understanding of heart aging mechanisms. In the meantime, it provides invaluable insights into possible therapeutic strategies for preventing age-related heart diseases in clinical settings.
Collapse
Affiliation(s)
- Yang Yuan
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Bing Liang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Xin-Lin Liu
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Wen-Jing Liu
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Bing-Huan Huang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Shan-Bo Yang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Yuan-Zhen Gao
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Jing-Sen Meng
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Meng-Jiao Li
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Ting Ye
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Chuan-Zhi Wang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Xiao-Kun Hu
- Interventional Medicine Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dong-Ming Xing
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China. .,School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
47
|
van de Wal MAE, Adjobo-Hermans MJW, Keijer J, Schirris TJJ, Homberg JR, Wieckowski MR, Grefte S, van Schothorst EM, van Karnebeek C, Quintana A, Koopman WJH. Ndufs4 knockout mouse models of Leigh syndrome: pathophysiology and intervention. Brain 2022. [PMID: 34849584 DOI: 10.1093/brain/awab426%jbrain] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Mitochondria are small cellular constituents that generate cellular energy (ATP) by oxidative phosphorylation (OXPHOS). Dysfunction of these organelles is linked to a heterogeneous group of multisystemic disorders, including diabetes, cancer, ageing-related pathologies and rare mitochondrial diseases. With respect to the latter, mutations in subunit-encoding genes and assembly factors of the first OXPHOS complex (complex I) induce isolated complex I deficiency and Leigh syndrome. This syndrome is an early-onset, often fatal, encephalopathy with a variable clinical presentation and poor prognosis due to the lack of effective intervention strategies. Mutations in the nuclear DNA-encoded NDUFS4 gene, encoding the NADH:ubiquinone oxidoreductase subunit S4 (NDUFS4) of complex I, induce 'mitochondrial complex I deficiency, nuclear type 1' (MC1DN1) and Leigh syndrome in paediatric patients. A variety of (tissue-specific) Ndufs4 knockout mouse models were developed to study the Leigh syndrome pathomechanism and intervention testing. Here, we review and discuss the role of complex I and NDUFS4 mutations in human mitochondrial disease, and review how the analysis of Ndufs4 knockout mouse models has generated new insights into the MC1ND1/Leigh syndrome pathomechanism and its therapeutic targeting.
Collapse
Affiliation(s)
- Melissa A E van de Wal
- Department of Pediatrics, Amalia Children's Hospital, RIMLS, RCMM, Radboudumc, Nijmegen, The Netherlands
| | | | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Tom J J Schirris
- Department of Pharmacology and Toxicology, RIMLS, RCMM, Radboudumc, Nijmegen, The Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, The Netherlands
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Sander Grefte
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | | | - Clara van Karnebeek
- Department of Pediatrics, Amalia Children's Hospital, RIMLS, RCMM, Radboudumc, Nijmegen, The Netherlands
- Department of Pediatrics, Emma Personalized Medicine Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Human Genetics, Emma Personalized Medicine Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Albert Quintana
- Mitochondrial Neuropathology Laboratory, Institut de Neurociències and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Werner J H Koopman
- Department of Pediatrics, Amalia Children's Hospital, RIMLS, RCMM, Radboudumc, Nijmegen, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
48
|
Gautam M, Gunay A, Chandel NS, Ozdinler PH. Mitochondrial dysregulation occurs early in ALS motor cortex with TDP-43 pathology and suggests maintaining NAD + balance as a therapeutic strategy. Sci Rep 2022; 12:4287. [PMID: 35277554 PMCID: PMC8917163 DOI: 10.1038/s41598-022-08068-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial defects result in dysregulation of metabolomics and energy homeostasis that are detected in upper motor neurons (UMNs) with TDP-43 pathology, a pathology that is predominantly present in both familial and sporadic cases of amyotrophic lateral sclerosis (ALS). While same mitochondrial problems are present in the UMNs of ALS patients with TDP-43 pathology and UMNs of TDP-43 mouse models, and since pathologies are shared at a cellular level, regardless of species, we first analyzed the metabolite profile of both healthy and diseased motor cortex to investigate whether metabolomic changes occur with respect to TDP-43 pathology. High-performance liquid chromatography, high-resolution mass spectrometry and tandem mass spectrometry (HPLC-MS/MS) for metabolite profiling began to suggest that reduced levels of NAD+ is one of the underlying causes of metabolomic problems. Since nicotinamide mononucleotide (NMN) was reported to restore NAD+ levels, we next investigated whether NMN treatment would improve the health of diseased corticospinal motor neurons (CSMN, a.k.a. UMN in mice). prpTDP-43A315T-UeGFP mice, the CSMN reporter line with TDP-43 pathology, allowed cell-type specific responses of CSMN to NMN treatment to be assessed in vitro. Our results show that metabolomic defects occur early in ALS motor cortex and establishing NAD+ balance could offer therapeutic benefit to UMNs with TDP-43 pathology.
Collapse
Affiliation(s)
- Mukesh Gautam
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Aksu Gunay
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Navdeep S Chandel
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - P Hande Ozdinler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA. .,Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60611, USA. .,Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA. .,Feinberg School of Medicine, Les Turner ALS Center at Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
49
|
McClements L, Richards C, Patel N, Chen H, Sesperez K, Bubb KJ, Karlstaedt A, Aksentijevic D. Impact of reduced uterine perfusion pressure model of preeclampsia on metabolism of placenta, maternal and fetal hearts. Sci Rep 2022; 12:1111. [PMID: 35064159 PMCID: PMC8782944 DOI: 10.1038/s41598-022-05120-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/31/2021] [Indexed: 12/23/2022] Open
Abstract
Preeclampsia is a cardiovascular pregnancy complication characterised by new onset hypertension and organ damage or intrauterine growth restriction. It is one of the leading causes of maternal and fetal mortality in pregnancy globally. Short of pre-term delivery of the fetus and placenta, treatment options are limited. Consequently, preeclampsia leads to increased cardiovascular disease risk in both mothers and offspring later in life. Here we aim to examine the impact of the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia on the maternal cardiovascular system, placental and fetal heart metabolism. The surgical RUPP model was induced in pregnant rats by applying silver clips around the aorta and uterine arteries on gestational day 14, resulting in ~ 40% uterine blood flow reduction. The experiment was terminated on gestational day 19 and metabolomic profile of placentae, maternal and fetal hearts analysed using high-resolution 1H NMR spectroscopy. Impairment of uterine perfusion in RUPP rats caused placental and cardiac hypoxia and a series of metabolic adaptations: altered energetics, carbohydrate, lipid and amino acid metabolism of placentae and maternal hearts. Comparatively, the fetal metabolic phenotype was mildly affected. Nevertheless, long-term effects of these changes in both mothers and the offspring should be investigated further in the future.
Collapse
Affiliation(s)
- Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Claire Richards
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Nikayla Patel
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Hao Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Kimberly Sesperez
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Kristen J Bubb
- Biomedical Discovery Institute, Monash University, Melbourne, Australia
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA, 127 San Vincente Blvd, 90048
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
50
|
Stokes JC, Bornstein RL, James K, Park KY, Spencer KA, Vo K, Snell JC, Johnson BM, Morgan PG, Sedensky MM, Baertsch NA, Johnson SC. Leukocytes mediate disease pathogenesis in the Ndufs4(KO) mouse model of Leigh syndrome. JCI Insight 2022; 7:156522. [PMID: 35050903 PMCID: PMC8983133 DOI: 10.1172/jci.insight.156522] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/19/2022] [Indexed: 11/23/2022] Open
Abstract
Symmetric, progressive, necrotizing lesions in the brainstem are a defining feature of Leigh syndrome (LS). A mechanistic understanding of the pathogenesis of these lesions has been elusive. Here, we report that leukocyte proliferation is causally involved in the pathogenesis of LS. Depleting leukocytes with a colony-stimulating factor 1 receptor inhibitor disrupted disease progression, including suppression of CNS lesion formation and a substantial extension of survival. Leukocyte depletion rescued diverse symptoms, including seizures, respiratory center function, hyperlactemia, and neurologic sequelae. These data reveal a mechanistic explanation for the beneficial effects of mTOR inhibition. More importantly, these findings dramatically alter our understanding of the pathogenesis of LS, demonstrating that immune involvement is causal in disease. This work has important implications for the mechanisms of mitochondrial disease and may lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Julia C Stokes
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Rebecca L Bornstein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, United States of America
| | - Katerina James
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Kyung Yeon Park
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Kira A Spencer
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Katie Vo
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - John C Snell
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Brittany M Johnson
- Department of Neurology, University of Washington, Seattle, United States of America
| | - Philip G Morgan
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Margaret M Sedensky
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Nathan A Baertsch
- Department of Pediatrics, University of Washington, Seattle, United States of America
| | - Simon C Johnson
- Department of Neurology, University of Washington, Seattle, United States of America
| |
Collapse
|