1
|
Wen H, Deng H, Li B, Chen J, Zhu J, Zhang X, Yoshida S, Zhou Y. Mitochondrial diseases: from molecular mechanisms to therapeutic advances. Signal Transduct Target Ther 2025; 10:9. [PMID: 39788934 PMCID: PMC11724432 DOI: 10.1038/s41392-024-02044-3] [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: 07/02/2024] [Revised: 09/28/2024] [Accepted: 10/31/2024] [Indexed: 01/12/2025] Open
Abstract
Mitochondria are essential for cellular function and viability, serving as central hubs of metabolism and signaling. They possess various metabolic and quality control mechanisms crucial for maintaining normal cellular activities. Mitochondrial genetic disorders can arise from a wide range of mutations in either mitochondrial or nuclear DNA, which encode mitochondrial proteins or other contents. These genetic defects can lead to a breakdown of mitochondrial function and metabolism, such as the collapse of oxidative phosphorylation, one of the mitochondria's most critical functions. Mitochondrial diseases, a common group of genetic disorders, are characterized by significant phenotypic and genetic heterogeneity. Clinical symptoms can manifest in various systems and organs throughout the body, with differing degrees and forms of severity. The complexity of the relationship between mitochondria and mitochondrial diseases results in an inadequate understanding of the genotype-phenotype correlation of these diseases, historically making diagnosis and treatment challenging and often leading to unsatisfactory clinical outcomes. However, recent advancements in research and technology have significantly improved our understanding and management of these conditions. Clinical translations of mitochondria-related therapies are actively progressing. This review focuses on the physiological mechanisms of mitochondria, the pathogenesis of mitochondrial diseases, and potential diagnostic and therapeutic applications. Additionally, this review discusses future perspectives on mitochondrial genetic diseases.
Collapse
Affiliation(s)
- Haipeng Wen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Hui Deng
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junyu Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junye Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Xian Zhang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
2
|
Cao B, Lu H, Liu P, Zhang Y, Wang C. Serum metabolomics signature of maternally inherited diabetes and deafness by gas chromatography-time of flight mass spectrometry. J Diabetes Investig 2025; 16:146-153. [PMID: 39480690 DOI: 10.1111/jdi.14334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/16/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
AIMS/INTRODUCTION The aim of this study was to identify a metabolic signature of MIDD as compared to healthy controls and other types of diabetes. METHODS We performed a comprehensive serum metabolomic analysis using gas chromatography-time of flight mass spectrometry (GC-TOFMS) in participants diagnosed with MIDD (n = 14), latent autoimmune diabetes in adults (LADA) (n = 14), type 2 diabetes mellitus (n = 14), and healthy controls (n = 14). Each group was matched for gender and age. RESULTS There were significant metabolic differences among MIDD and other diabetic and control groups. Compared with control, MIDD patients had high levels of carbohydrates (glucose, galactose, mannose, sorbose, and maltose), fatty acids (2-Hydroxybutyric acid, eicosapentaenoic acid, and octadecanoic acid), and other metabolites (alanine, threonic acid, cholesterol, lactic acid, and gluconic acid), but low level of threonine. Compared with LADA, MIDD patients had high levels of threonic acid and some amino acids (alanine, tryptophan, histidine, proline, glutamine, and creatine) but low levels of serine. Compared with type 2 diabetes mellitus, MIDD patients had high levels of citrulline, creatine, 3-Amino-2-piperidone, but low levels of ornithine, fatty acids (arachidonic acid and octadecanoic acid), and intermediates of the tricarboxylic acid cycle (malic acid and succinic acid). CONCLUSIONS Our study identified a specific metabolic profile related to glycolysis and the tricarboxylic acid cycle in MIDD that differs from healthy controls and other types of diabetes. This unique metabolic signature provides new perspectives for understanding the pathophysiology and underlying mechanisms of MIDD.
Collapse
Affiliation(s)
- Baige Cao
- Department of Endocrinology & Metabolism, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huijuan Lu
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Liu
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinan Zhang
- The Metabolic Disease Biobank, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Congrong Wang
- Department of Endocrinology & Metabolism, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
3
|
Yamashita S. Late-onset primary muscle diseases mimicking sarcopenia. Geriatr Gerontol Int 2024; 24:1099-1110. [PMID: 39402847 DOI: 10.1111/ggi.15000] [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: 06/06/2024] [Revised: 09/15/2024] [Accepted: 09/27/2024] [Indexed: 11/08/2024]
Abstract
Sarcopenia is an age-related loss of skeletal muscle mass, strength, and function that causes various health problems. In contrast, late-onset primary myopathies, which occur in the older population, are caused by a variety of factors, including genetic mutations, autoimmune processes, and metabolic abnormalities. Although sarcopenia and primary myopathy are two distinct disease processes, their symptoms can overlap, making differentiation challenging. The diagnostic criteria for sarcopenia have evolved over time, and various criteria have been proposed by expert groups. Late-onset primary muscle diseases such as inclusion body myositis, sporadic late-onset nemaline myopathy, muscular dystrophies, distal myopathies, myofibrillar myopathies, metabolic myopathies, and mitochondrial myopathies share common pathogenic mechanisms with sarcopenia, further complicating the diagnostic process. Appropriate clinical evaluation, including detailed history-taking, physical examination, and diagnostic testing, is essential for accurate diagnosis and management. Treatment approaches, including exercise, nutritional support, and disease-specific therapies, must be tailored to the characteristics of each disease. Despite these differences, sarcopenia and primary myopathies require careful consideration in the clinical setting for proper diagnosis and management. This review outlines the evolution of diagnostic criteria and diagnostic items for sarcopenia, late-onset primary myopathies that should be differentiated from sarcopenia, common pathomechanisms, and diagnostic algorithms to properly differentiate primary myopathies. Geriatr Gerontol Int 2024; 24: 1099-1110.
Collapse
Affiliation(s)
- Satoshi Yamashita
- Department of Neurology, International University of Health and Welfare Narita Hospital, Narita, Japan
| |
Collapse
|
4
|
Lingampelly SS, Naviaux JC, Heuer LS, Monk JM, Li K, Wang L, Haapanen L, Kelland CA, Van de Water J, Naviaux RK. Metabolic network analysis of pre-ASD newborns and 5-year-old children with autism spectrum disorder. Commun Biol 2024; 7:536. [PMID: 38729981 PMCID: PMC11549098 DOI: 10.1038/s42003-024-06102-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: 10/19/2023] [Accepted: 03/22/2024] [Indexed: 05/12/2024] Open
Abstract
Classical metabolomic and new metabolic network methods were used to study the developmental features of autism spectrum disorder (ASD) in newborns (n = 205) and 5-year-old children (n = 53). Eighty percent of the metabolic impact in ASD was caused by 14 shared biochemical pathways that led to decreased anti-inflammatory and antioxidant defenses, and to increased physiologic stress molecules like lactate, glycerol, cholesterol, and ceramides. CIRCOS plots and a new metabolic network parameter,V ° net, revealed differences in both the kind and degree of network connectivity. Of 50 biochemical pathways and 450 polar and lipid metabolites examined, the developmental regulation of the purine network was most changed. Purine network hub analysis revealed a 17-fold reversal in typically developing children. This purine network reversal did not occur in ASD. These results revealed previously unknown metabolic phenotypes, identified new developmental states of the metabolic correlation network, and underscored the role of mitochondrial functional changes, purine metabolism, and purinergic signaling in autism spectrum disorder.
Collapse
Grants
- UL1 TR001442 NCATS NIH HHS
- 7274 Autism Speaks (Autism Speaks Inc.)
- This work was funded in part by philanthropic gifts to the Naviaux Lab from the UCSD Christini Fund, the Lennox Foundation, the William Wright Family Foundation, Malone Family Foundation, the Brain Foundation, the Westreich Foundation, the Aloe family, the Harb family, Marc Spilo and all the others who contributed to the Aloe family autism research fund, the N of One Autism Research Foundation, the UCSD Mitochondrial Disease Research Fund, the JMS Fund, Linda Clark, Jeanne Conrad, David Cannistraro, the Kirby and Katie Mano Family, Simon and Evelyn Foo, Wing-kun Tam, Gita and Anurag Gupta, the Brent Kaufman Family, and the Daniel and Kelly White Family, and grassroots support from over 2000 individuals from around the world who have each provided gifts in the past year to support Naviaux Lab research. The REDCap software system used in this study was provided by the UCSD Clinical and Translational Research Center and supported by Award Number UL1TR001442 from the National Center for Research Resources. Financial supporters for this study had no role in study design, data collection, analysis, interpretation, writing, or publication of this work.
Collapse
Affiliation(s)
- Sai Sachin Lingampelly
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
| | - Jane C Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Neuroscience, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
| | - Luke S Heuer
- The UC Davis MIND Institute, University of California, Davis, Davis, CA, 95616, USA
| | - Jonathan M Monk
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Macao Polytechnic University, Macau, China
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA
| | - Lori Haapanen
- The UC Davis MIND Institute, University of California, Davis, Davis, CA, 95616, USA
| | - Chelsea A Kelland
- The UC Davis MIND Institute, University of California, Davis, Davis, CA, 95616, USA
| | - Judy Van de Water
- The UC Davis MIND Institute, University of California, Davis, Davis, CA, 95616, USA
- Department of Rheumatology and Allergy, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA.
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA.
- Department of Pediatrics, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA.
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, 92103-8467, USA.
| |
Collapse
|
5
|
Gervasoni J, Primiano A, Cicchinelli M, Santucci L, Servidei S, Urbani A, Primiano G, Iavarone F. Mitochondrial Biomarkers in the Omics Era: A Clinical-Pathophysiological Perspective. Int J Mol Sci 2024; 25:4855. [PMID: 38732076 PMCID: PMC11084339 DOI: 10.3390/ijms25094855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Mitochondrial diseases (MDs) affect 4300 individuals, with different ages of presentation and manifestation in any organ. How defects in mitochondria can cause such a diverse range of human diseases remains poorly understood. In recent years, several published research articles regarding the metabolic and protein profiles of these neurogenetic disorders have helped shed light on the pathogenetic mechanisms. By investigating different pathways in MDs, often with the aim of identifying disease biomarkers, it is possible to identify molecular processes underlying the disease. In this perspective, omics technologies such as proteomics and metabolomics considered in this review, can support unresolved mitochondrial questions, helping to improve outcomes for patients.
Collapse
Affiliation(s)
- Jacopo Gervasoni
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
| | - Aniello Primiano
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
| | - Michela Cicchinelli
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy;
| | - Lavinia Santucci
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
| | - Serenella Servidei
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Andrea Urbani
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy;
| | - Guido Primiano
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Federica Iavarone
- Fondazione Policlinico Universitario ‘Agostino Gemelli’ IRCCS, 00168 Rome, Italy; (J.G.); (A.P.); (L.S.); (S.S.); (G.P.)
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy;
| |
Collapse
|
6
|
Guglielmi V, Cheli M, Tonin P, Vattemi G. Sporadic Inclusion Body Myositis at the Crossroads between Muscle Degeneration, Inflammation, and Aging. Int J Mol Sci 2024; 25:2742. [PMID: 38473988 PMCID: PMC10932328 DOI: 10.3390/ijms25052742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Sporadic inclusion body myositis (sIBM) is the most common muscle disease of older people and is clinically characterized by slowly progressive asymmetrical muscle weakness, predominantly affecting the quadriceps, deep finger flexors, and foot extensors. At present, there are no enduring treatments for this relentless disease that eventually leads to severe disability and wheelchair dependency. Although sIBM is considered a rare muscle disorder, its prevalence is certainly higher as the disease is often undiagnosed or misdiagnosed. The histopathological phenotype of sIBM muscle biopsy includes muscle fiber degeneration and endomysial lymphocytic infiltrates that mainly consist of cytotoxic CD8+ T cells surrounding nonnecrotic muscle fibers expressing MHCI. Muscle fiber degeneration is characterized by vacuolization and the accumulation of congophilic misfolded multi-protein aggregates, mainly in their non-vacuolated cytoplasm. Many players have been identified in sIBM pathogenesis, including environmental factors, autoimmunity, abnormalities of protein transcription and processing, the accumulation of several toxic proteins, the impairment of autophagy and the ubiquitin-proteasome system, oxidative and nitrative stress, endoplasmic reticulum stress, myonuclear degeneration, and mitochondrial dysfunction. Aging has also been proposed as a contributor to the disease. However, the interplay between these processes and the primary event that leads to the coexistence of autoimmune and degenerative changes is still under debate. Here, we outline our current understanding of disease pathogenesis, focusing on degenerative mechanisms, and discuss the possible involvement of aging.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Cellular and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marta Cheli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| | - Paola Tonin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| | - Gaetano Vattemi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| |
Collapse
|
7
|
Nagy-Grócz G, Spekker E, Vécsei L. Kynurenines, Neuronal Excitotoxicity, and Mitochondrial Oxidative Stress: Role of the Intestinal Flora. Int J Mol Sci 2024; 25:1698. [PMID: 38338981 PMCID: PMC10855176 DOI: 10.3390/ijms25031698] [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: 01/04/2024] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
The intestinal flora has been the focus of numerous investigations recently, with inquiries not just into the gastrointestinal aspects but also the pathomechanism of other diseases such as nervous system disorders and mitochondrial diseases. Mitochondrial disorders are the most common type of inheritable metabolic illness caused by mutations of mitochondrial and nuclear DNA. Despite the intensive research, its diagnosis is usually difficult, and unfortunately, treating it challenges physicians. Metabolites of the kynurenine pathway are linked to many disorders, such as depression, schizophrenia, migraine, and also diseases associated with impaired mitochondrial function. The kynurenine pathway includes many substances, for instance kynurenic acid and quinolinic acid. In this review, we would like to show a possible link between the metabolites of the kynurenine pathway and mitochondrial stress in the context of intestinal flora. Furthermore, we summarize the possible markers of and future therapeutic options for the kynurenine pathway in excitotoxicity and mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Gábor Nagy-Grócz
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary;
- Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| | | | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary;
- HUN-REN-SZTE Neuroscience Research Group, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
8
|
Suciu I, Delp J, Gutbier S, Suess J, Henschke L, Celardo I, Mayer TU, Amelio I, Leist M. Definition of the Neurotoxicity-Associated Metabolic Signature Triggered by Berberine and Other Respiratory Chain Inhibitors. Antioxidants (Basel) 2023; 13:49. [PMID: 38247474 PMCID: PMC10812665 DOI: 10.3390/antiox13010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
To characterize the hits from a phenotypic neurotoxicity screen, we obtained transcriptomics data for valinomycin, diethylstilbestrol, colchicine, rotenone, 1-methyl-4-phenylpyridinium (MPP), carbaryl and berberine (Ber). For all compounds, the concentration triggering neurite degeneration correlated with the onset of gene expression changes. The mechanistically diverse toxicants caused similar patterns of gene regulation: the responses were dominated by cell de-differentiation and a triggering of canonical stress response pathways driven by ATF4 and NRF2. To obtain more detailed and specific information on the modes-of-action, the effects on energy metabolism (respiration and glycolysis) were measured. Ber, rotenone and MPP inhibited the mitochondrial respiratory chain and they shared complex I as the target. This group of toxicants was further evaluated by metabolomics under experimental conditions that did not deplete ATP. Ber (204 changed metabolites) showed similar effects as MPP and rotenone. The overall metabolic situation was characterized by oxidative stress, an over-abundance of NADH (>1000% increase) and a re-routing of metabolism in order to dispose of the nitrogen resulting from increased amino acid turnover. This unique overall pattern led to the accumulation of metabolites known as biomarkers of neurodegeneration (saccharopine, aminoadipate and branched-chain ketoacids). These findings suggest that neurotoxicity of mitochondrial inhibitors may result from an ensemble of metabolic changes rather than from a simple ATP depletion. The combi-omics approach used here provided richer and more specific MoA data than the more common transcriptomics analysis alone. As Ber, a human drug and food supplement, mimicked closely the mode-of-action of known neurotoxicants, its potential hazard requires further investigation.
Collapse
Affiliation(s)
- Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Graduate School of Chemical Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Johannes Delp
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Simon Gutbier
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Julian Suess
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Lars Henschke
- Graduate School of Chemical Biology, University of Konstanz, 78464 Konstanz, Germany
- Department of Molecular Genetics, University of Konstanz, 78464 Konstanz, Germany
| | - Ivana Celardo
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Thomas U. Mayer
- Department of Molecular Genetics, University of Konstanz, 78464 Konstanz, Germany
| | - Ivano Amelio
- Division for Systems Toxicology, Department of Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| |
Collapse
|
9
|
Pan LA, Naviaux JC, Wang L, Li K, Monk JM, Lingampelly SS, Segreti AM, Bloom K, Vockley J, Tarnopolsky MA, Finegold DN, Peters DG, Naviaux RK. Metabolic features of treatment-refractory major depressive disorder with suicidal ideation. Transl Psychiatry 2023; 13:393. [PMID: 38097555 PMCID: PMC10721812 DOI: 10.1038/s41398-023-02696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/18/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
Peripheral blood metabolomics was used to gain chemical insight into the biology of treatment-refractory Major Depressive Disorder with suicidal ideation, and to identify individualized differences for personalized care. The study cohort consisted of 99 patients with treatment-refractory major depressive disorder and suicidal ideation (trMDD-SI n = 52 females and 47 males) and 94 age- and sex-matched healthy controls (n = 48 females and 46 males). The median age was 29 years (IQR 22-42). Targeted, broad-spectrum metabolomics measured 448 metabolites. Fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15) were measured as biomarkers of mitochondrial dysfunction. The diagnostic accuracy of plasma metabolomics was over 90% (95%CI: 0.80-1.0) by area under the receiver operator characteristic (AUROC) curve analysis. Over 55% of the metabolic impact in males and 75% in females came from abnormalities in lipids. Modified purines and pyrimidines from tRNA, rRNA, and mRNA turnover were increased in the trMDD-SI group. FGF21 was increased in both males and females. Increased lactate, glutamate, and saccharopine, and decreased cystine provided evidence of reductive stress. Seventy-five percent of the metabolomic abnormalities found were individualized. Personalized deficiencies in CoQ10, flavin adenine dinucleotide (FAD), citrulline, lutein, carnitine, or folate were found. Pathways regulated by mitochondrial function dominated the metabolic signature. Peripheral blood metabolomics identified mitochondrial dysfunction and reductive stress as common denominators in suicidal ideation associated with treatment-refractory major depressive disorder. Individualized metabolic differences were found that may help with personalized management.
Collapse
Affiliation(s)
- Lisa A Pan
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Panomics Mental Health Initiative, Pittsburgh, PA, USA.
- New Hope Molecular, LLC, Pittsburgh, PA, USA.
- New Hope Molecular, LLC, 750 Washington Rd, Suite 19, Pittsburgh, PA, 15228, USA.
| | - Jane C Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Jonathan M Monk
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Sai Sachin Lingampelly
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA
| | - Anna Maria Segreti
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kaitlyn Bloom
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jerry Vockley
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - David N Finegold
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Panomics Mental Health Initiative, Pittsburgh, PA, USA
- New Hope Molecular, LLC, Pittsburgh, PA, USA
| | - David G Peters
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- School of Public Health, Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Panomics Mental Health Initiative, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Pediatrics, University of California, San Diego School of Medicine, San Diego, CA, USA.
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, USA.
| |
Collapse
|
10
|
Peng X, Ma LX, Xiao C, Zhang ZZ, Zhu M, Hong DJ, Zhan YA. Weaning difficulty after severe pneumonia in adult-onset mitochondrial myopathy with A3243G mutation in the mitochondrial tRNA gene: A case report. Heliyon 2023; 9:e23300. [PMID: 38149187 PMCID: PMC10750057 DOI: 10.1016/j.heliyon.2023.e23300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Background Mitochondrial myopathy is a group of diseases caused by abnormal mitochondrial structure or function. The mitochondrial myopathy impacts muscles of the whole body and exhibits variable symptoms. Respiratory muscle deficits deteriorate pulmonary function in patients with severe pneumonia. Case presentation We report the case of a male patient with severe pneumonia-induced respiratory failure. He was abnormally dependent invasive ventilator-assisted ventilation after his condition had improved. Then we found abnormal ventilator waveform and a decline in muscle strength of him. Mitochondrial myopathy was ultimately confirmed by muscle pathological biopsy and body fluid genetic testing. Vitamin B complex, coenzyme Q10, Neprinol AFD, l-arginine, and MITO-TONIC were used to improve mitochondrial function and muscle metabolism. After treatment, discomfort associated with chest tightness, fatigue, cough, and sputum disappeared, and the patient was discharged. Conclusion This case presented an uncommon cause of difficult weaning and extubation-acute onset of mitochondrial myopathy. Muscle biopsy and genetic testing of body fluid are essential for diagnosing mitochondrial myopathy. The A3243G mutation in the MT-TL1 gene of mitochondrial DNA contributes to pathogenesis of this case.
Collapse
Affiliation(s)
- Xiong Peng
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Li-xiu Ma
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ce Xiao
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhi-zhe Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Min Zhu
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Dao-jun Hong
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi-an Zhan
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
11
|
Du J, Zhang C, Liu F, Liu X, Wang D, Zhao D, Shui G, Zhao Y, Yan C. Distinctive metabolic remodeling in TYMP deficiency beyond mitochondrial dysfunction. J Mol Med (Berl) 2023; 101:1237-1253. [PMID: 37603049 DOI: 10.1007/s00109-023-02358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 07/09/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is caused by mutations in the TYMP gene, which encodes thymidine phosphorylase (TP). As a cytosolic metabolic enzyme, TP defects affect biological processes that are thought to not be limited to the abnormal replication of mitochondrial DNA. This study aimed to elucidate the characteristic metabolic alterations and associated homeostatic regulation caused by TYMP deficiency. The pathogenicity of novel TYMP variants was evaluated in terms of clinical features, genetic analysis, and structural instability. We analyzed plasma samples from three patients with MNGIE; three patients with m.3243A > G mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS); and four healthy controls (HC) using both targeted and untargeted metabolomics techniques. Transcriptomics analysis and bioenergetic studies were performed on skin fibroblasts from participants in these three groups. A TYMP overexpression experiment was conducted to rescue the observed changes. Compared with controls, specific alterations in nucleosides, bile acids, and steroid metabolites were identified in the plasma of MNGIE patients. Comparable mitochondrial dysfunction was present in fibroblasts from patients with TYMP deficiency and in those from patients with the m.3243A > G mutation. Distinctively decreased sterol regulatory element binding protein (SREBP) regulated cholesterol metabolism and fatty acid (FA) biosynthesis as well as reduced FA degradation were revealed in fibroblasts with TYMP deficiency. The restoration of thymidine phosphorylase activity rescued the observed changes in MNGIE fibroblasts. Our findings indicated that more widespread metabolic disturbance may be caused by TYMP deficiency in addition to mitochondrial dysfunction, which expands our knowledge of the biochemical outcome of TYMP deficiency. KEY MESSAGES: Distinct metabolic profiles in patients with TYMP deficiency compared to those with m.3243A > G mutation. TYMP deficiency leads to a global disruption of nucleoside metabolism. Cholesterol and fatty acid metabolism are inhibited in individuals with MNGIE. TYMP is functionally related to SREBP-regulated pathways. Potential metabolite biomarkers that could be valuable clinical tools to improve the diagnosis of MNGIE.
Collapse
Affiliation(s)
- Jixiang Du
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chao Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Fuchen Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xihan Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dongdong Wang
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dandan Zhao
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 101408, China
| | - Yuying Zhao
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, 266103, China.
| |
Collapse
|
12
|
Cantó-Santos J, Valls-Roca L, Tobías E, Oliva C, García-García FJ, Guitart-Mampel M, Andújar-Sánchez F, Esteve-Codina A, Martín-Mur B, Padrosa J, Aránega R, Moreno-Lozano PJ, Milisenda JC, Artuch R, Grau-Junyent JM, Garrabou G. Integrated Multi-Omics Analysis for Inferring Molecular Players in Inclusion Body Myositis. Antioxidants (Basel) 2023; 12:1639. [PMID: 37627634 PMCID: PMC10452026 DOI: 10.3390/antiox12081639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Inclusion body myositis (IBM) is an acquired inflammatory myopathy affecting proximal and distal muscles that leads to weakness in patients over 50. It is diagnosed based on clinical and histological findings in muscle related to inflammation, degeneration, and mitochondria. In relation to IBM, a shortage of validated disease models and a lack of biomarkers and effective treatments constitute an unmet medical need. To overcome these hurdles, we performed an omics analysis of multiple samples from IBM patients (saliva, fibroblasts, urine, plasma, and muscle) to gain insight into the pathophysiology of IBM. Degeneration was evident due to the presence of amyloid β peptide 1-42 (Aβ1-42) in the saliva of the analyzed IBM patients. The presence of metabolic disarrangements in IBM was indicated by an imbalanced organic acid profile in fibroblasts and urine. Specifically, abnormal levels of L-pyroglutamic and orotic acid were supported by the abnormal expression of related metabolites in plasma and urine (glutathione and pyrimidines) and the aberrant expression of upstream gene regulators (L2HGDH, IDH2, OPLAH, and ASL) in muscle. Combined levels of L-pyroglutamic and orotic acid displayed an outstanding biomarker signature in urine with 100% sensitivity and specificity. The confirmation of systemic metabolic disarrangements in IBM and the identification of novel biomarkers reported herein unveil novel insights that require validation in larger cohorts.
Collapse
Affiliation(s)
- Judith Cantó-Santos
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Laura Valls-Roca
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Ester Tobías
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Clara Oliva
- Department of Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain; (C.O.); (R.A.)
| | - Francesc Josep García-García
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Mariona Guitart-Mampel
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Félix Andújar-Sánchez
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (A.E.-C.); (B.M.-M.)
- Department of Medicine and Health Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Beatriz Martín-Mur
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (A.E.-C.); (B.M.-M.)
| | - Joan Padrosa
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Raquel Aránega
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Pedro J. Moreno-Lozano
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - José César Milisenda
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Rafael Artuch
- Department of Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain; (C.O.); (R.A.)
| | - Josep M. Grau-Junyent
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| | - Glòria Garrabou
- Inherited Metabolic Disorders and Muscular Diseases Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (J.C.-S.); (L.V.-R.); (E.T.); (F.J.G.-G.); (M.G.-M.); (F.A.-S.); (J.P.); (R.A.); (P.J.M.-L.)
- Department of Internal Medicine, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- CIBERER—Spanish Biomedical Research Centre in Rare Diseases, 28029 Madrid, Spain
| |
Collapse
|
13
|
Singh P, Gollapalli K, Mangiola S, Schranner D, Yusuf MA, Chamoli M, Shi SL, Bastos BL, Nair T, Riermeier A, Vayndorf EM, Wu JZ, Nilakhe A, Nguyen CQ, Muir M, Kiflezghi MG, Foulger A, Junker A, Devine J, Sharan K, Chinta SJ, Rajput S, Rane A, Baumert P, Schönfelder M, Iavarone F, Lorenzo GD, Kumari S, Gupta A, Sarkar R, Khyriem C, Chawla AS, Sharma A, Sarper N, Chattopadhyay N, Biswal BK, Settembre C, Nagarajan P, Targoff KL, Picard M, Gupta S, Velagapudi V, Papenfuss AT, Kaya A, Ferreira MG, Kennedy BK, Andersen JK, Lithgow GJ, Ali AM, Mukhopadhyay A, Palotie A, Kastenmüller G, Kaeberlein M, Wackerhage H, Pal B, Yadav VK. Taurine deficiency as a driver of aging. Science 2023; 380:eabn9257. [PMID: 37289866 PMCID: PMC10630957 DOI: 10.1126/science.abn9257] [Citation(s) in RCA: 176] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 04/14/2023] [Indexed: 06/10/2023]
Abstract
Aging is associated with changes in circulating levels of various molecules, some of which remain undefined. We find that concentrations of circulating taurine decline with aging in mice, monkeys, and humans. A reversal of this decline through taurine supplementation increased the health span (the period of healthy living) and life span in mice and health span in monkeys. Mechanistically, taurine reduced cellular senescence, protected against telomerase deficiency, suppressed mitochondrial dysfunction, decreased DNA damage, and attenuated inflammaging. In humans, lower taurine concentrations correlated with several age-related diseases and taurine concentrations increased after acute endurance exercise. Thus, taurine deficiency may be a driver of aging because its reversal increases health span in worms, rodents, and primates and life span in worms and rodents. Clinical trials in humans seem warranted to test whether taurine deficiency might drive aging in humans.
Collapse
Affiliation(s)
- Parminder Singh
- Metabolic Research Laboratories, National Institute of Immunology; New Delhi, India
| | - Kishore Gollapalli
- Vagelos College of Physicians and Surgeons, Columbia University; New York, USA
| | - Stefano Mangiola
- Department of Medical Biology, University of Melbourne; Melbourne, Australia
- School of Cancer Medicine, La Trobe University; Bundoora, Australia
- Olivia Newton-John Cancer Research Institute; Heidelberg, Australia
| | - Daniela Schranner
- Exercise Biology Group, Technical University of Munich; Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München; Neuherberg, Germany
| | - Mohd Aslam Yusuf
- Department of Bioengineering, Integral University; Lucknow, India
| | - Manish Chamoli
- Buck Institute of Age Research, 8001 Redwood Blvd; California, USA
| | - Sting L. Shi
- Vagelos College of Physicians and Surgeons, Columbia University; New York, USA
| | - Bruno Lopes Bastos
- Institute for Research on Cancer and Aging of Nice (IRCAN); Nice, France
| | - Tripti Nair
- Molecular Aging Laboratory, National Institute of Immunology; New Delhi, India
| | - Annett Riermeier
- Exercise Biology Group, Technical University of Munich; Munich, Germany
| | - Elena M. Vayndorf
- Department of Laboratory Medicine and Pathology, University of Washington; WA, USA
| | - Judy Z. Wu
- Department of Laboratory Medicine and Pathology, University of Washington; WA, USA
| | - Aishwarya Nilakhe
- Metabolic Research Laboratories, National Institute of Immunology; New Delhi, India
| | - Christina Q. Nguyen
- Department of Laboratory Medicine and Pathology, University of Washington; WA, USA
| | - Michael Muir
- Department of Laboratory Medicine and Pathology, University of Washington; WA, USA
| | - Michael G. Kiflezghi
- Department of Laboratory Medicine and Pathology, University of Washington; WA, USA
| | - Anna Foulger
- Buck Institute of Age Research, 8001 Redwood Blvd; California, USA
| | - Alex Junker
- Department of Neurology, Columbia University; New York, USA
| | - Jack Devine
- Department of Neurology, Columbia University; New York, USA
| | - Kunal Sharan
- Mouse Genetics Project, Wellcome Sanger Institute; Cambridge, UK
| | | | - Swati Rajput
- Division of Endocrinology, CSIR-Central Drug Research Institute; Lucknow, India
| | - Anand Rane
- Buck Institute of Age Research, 8001 Redwood Blvd; California, USA
| | - Philipp Baumert
- Exercise Biology Group, Technical University of Munich; Munich, Germany
| | | | | | | | - Swati Kumari
- Metabolic Research Laboratories, National Institute of Immunology; New Delhi, India
| | - Alka Gupta
- Metabolic Research Laboratories, National Institute of Immunology; New Delhi, India
| | - Rajesh Sarkar
- Metabolic Research Laboratories, National Institute of Immunology; New Delhi, India
| | - Costerwell Khyriem
- Harry Perkins Institute of Medical Research; Perth, Australia
- Curtin Medical School, Curtin University; Perth, Australia
| | - Amanpreet S. Chawla
- Immunobiology Laboratory, National Institute of Immunology; New Delhi, India
- MRC-Protein Phosphorylation and Ubiquitination Unit, University of Dundee; Dundee, UK
| | - Ankur Sharma
- Harry Perkins Institute of Medical Research; Perth, Australia
- Curtin Medical School, Curtin University; Perth, Australia
| | - Nazan Sarper
- Pediatrics and Pediatric Hematology, Kocaeli University Hospital; Kocaeli, Turkey
| | | | - Bichitra K. Biswal
- Metabolic Research Laboratories, National Institute of Immunology; New Delhi, India
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM); Pozzuoli, Italy
- Department of Clinical Medicine and Surgery, Federico II University; Naples, Italy
| | - Perumal Nagarajan
- Primate Research Facility, National Institute of Immunology; New Delhi, India
- Small Animal Research Facility, National Institute of Immunology; New Delhi, India
| | - Kimara L. Targoff
- Division of Cardiology, Department of Pediatrics, Columbia University; New York, USA
| | - Martin Picard
- Department of Neurology, Columbia University; New York, USA
| | - Sarika Gupta
- Metabolic Research Laboratories, National Institute of Immunology; New Delhi, India
| | - Vidya Velagapudi
- Institute for Molecular Medicine Finland FIMM, University of Helsinki; Helsinki, Finland
| | | | - Alaattin Kaya
- Department of Biology, Virginia Commonwealth University; Virginia, USA
| | | | - Brian K. Kennedy
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore; Singapore, Singapore
- Centre for Healthy Longevity, National University Health System; Singapore, Singapore
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore; Singapore, Singapore
| | | | | | - Abdullah Mahmood Ali
- Department of Medicine, Columbia University Irving Medical Center; New York, USA
| | - Arnab Mukhopadhyay
- Molecular Aging Laboratory, National Institute of Immunology; New Delhi, India
| | - Aarno Palotie
- Institute for Molecular Medicine Finland FIMM, University of Helsinki; Helsinki, Finland
- Broad Institute of Harvard and MIT; Cambridge, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital; Boston, USA
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München; Neuherberg, Germany
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington; WA, USA
| | | | - Bhupinder Pal
- Department of Medical Biology, University of Melbourne; Melbourne, Australia
- School of Cancer Medicine, La Trobe University; Bundoora, Australia
| | - Vijay K. Yadav
- Metabolic Research Laboratories, National Institute of Immunology; New Delhi, India
- Vagelos College of Physicians and Surgeons, Columbia University; New York, USA
- Mouse Genetics Project, Wellcome Sanger Institute; Cambridge, UK
- Department of Genetics and Development, Columbia University; New York, USA
| |
Collapse
|
14
|
Hoogstraten CA, Lyon JJ, Smeitink JAM, Russel FGM, Schirris TJJ. Time to Change: A Systems Pharmacology Approach to Disentangle Mechanisms of Drug-Induced Mitochondrial Toxicity. Pharmacol Rev 2023; 75:463-486. [PMID: 36627212 DOI: 10.1124/pharmrev.122.000568] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 01/11/2023] Open
Abstract
An increasing number of commonly prescribed drugs are known to interfere with mitochondrial function, which is associated with almost half of all Food and Drug Administration black box warnings, a variety of drug withdrawals, and attrition of drug candidates. This can mainly be attributed to a historic lack of sensitive and specific assays to identify the mechanisms underlying mitochondrial toxicity during drug development. In the last decade, a better understanding of drug-induced mitochondrial dysfunction has been achieved by network-based and structure-based systems pharmacological approaches. Here, we propose the implementation of a tiered systems pharmacology approach to detect adverse mitochondrial drug effects during preclinical drug development, which is based on a toolset developed to study inherited mitochondrial disease. This includes phenotypic characterization, profiling of key metabolic alterations, mechanistic studies, and functional in vitro and in vivo studies. Combined with binding pocket similarity comparisons and bottom-up as well as top-down metabolic network modeling, this tiered approach enables identification of mechanisms underlying drug-induced mitochondrial dysfunction. After validation of these off-target mechanisms, drug candidates can be adjusted to minimize mitochondrial activity. Implementing such a tiered systems pharmacology approach could lead to a more efficient drug development trajectory due to lower drug attrition rates and ultimately contribute to the development of safer drugs. SIGNIFICANCE STATEMENT: Many commonly prescribed drugs adversely affect mitochondrial function, which can be detected using phenotypic assays. However, these methods provide only limited insight into the underlying mechanisms. In recent years, a better understanding of drug-induced mitochondrial dysfunction has been achieved by network-based and structure-based system pharmacological approaches. Their implementation in preclinical drug development could reduce the number of drug failures, contributing to safer drug design.
Collapse
Affiliation(s)
- Charlotte A Hoogstraten
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| | - Jonathan J Lyon
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| | - Jan A M Smeitink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| | - Tom J J Schirris
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (C.A.H., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (C.A.H., J.A.M.S., F.G.M.R., T.J.J.S.), and Department of Pediatrics (J.A.M.S.), Radboud University Medical Center, Nijmegen, The Netherlands; GlaxoSmithKline, Safety Assessment, Ware, Hertfordshire, United Kingdom (J.J.L.); and Khondrion BV, Nijmegen, The Netherlands (J.A.M.S.)
| |
Collapse
|
15
|
Cantó-Santos J, Valls-Roca L, Tobías E, García-García FJ, Guitart-Mampel M, Esteve-Codina A, Martín-Mur B, Casado M, Artuch R, Solsona-Vilarrasa E, Fernandez-Checa JC, García-Ruiz C, Rentero C, Enrich C, Moreno-Lozano PJ, Milisenda JC, Cardellach F, Grau-Junyent JM, Garrabou G. Unravelling inclusion body myositis using a patient-derived fibroblast model. J Cachexia Sarcopenia Muscle 2023; 14:964-977. [PMID: 36860172 PMCID: PMC10067507 DOI: 10.1002/jcsm.13178] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 11/24/2022] [Accepted: 01/02/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Inclusion body myositis (IBM) is an inflammatory myopathy clinically characterized by proximal and distal muscle weakness, with inflammatory infiltrates, rimmed vacuoles and mitochondrial changes in muscle histopathology. There is scarce knowledge on IBM aetiology, and non-established biomarkers or effective treatments are available, partly due to the lack of validated disease models. METHODS We have performed transcriptomics and functional validation of IBM muscle pathological hallmarks in fibroblasts from IBM patients (n = 14) and healthy controls (n = 12), paired by age and sex. The results comprise an mRNA-seq, together with functional inflammatory, autophagy, mitochondrial and metabolic changes between patients and controls. RESULTS Gene expression profile of IBM vs control fibroblasts revealed 778 differentially expressed genes (P-value adj < 0.05) related to inflammation, mitochondria, cell cycle regulation and metabolism. Functionally, an increased inflammatory profile was observed in IBM fibroblasts with higher supernatant cytokine secretion (three-fold increase). Autophagy was reduced considering basal protein mediators (18.4% reduced), time-course autophagosome formation (LC3BII 39% reduced, P-value < 0.05), and autophagosome microscopic evaluation. Mitochondria displayed reduced genetic content (by 33.9%, P-value < 0.05) and function (30.2%-decrease in respiration, 45.6%-decline in enzymatic activity (P-value < 0.001), 14.3%-higher oxidative stress, 135.2%-increased antioxidant defence (P-value < 0.05), 11.6%-reduced mitochondrial membrane potential (P-value < 0.05) and 42.8%-reduced mitochondrial elongation (P-value < 0.05)). In accordance, at the metabolite level, organic acid showed a 1.8-fold change increase, with conserved amino acid profile. Correlating to disease evolution, oxidative stress and inflammation emerge as potential markers of prognosis. CONCLUSIONS These findings confirm the presence of molecular disturbances in peripheral tissues from IBM patients and prompt patients' derived fibroblasts as a promising disease model, which may eventually be exported to other neuromuscular disorders. We additionally identify new molecular players in IBM associated with disease progression, setting the path to deepen in disease aetiology, in the identification of novel biomarkers or in the standardization of biomimetic platforms to assay new therapeutic strategies for preclinical studies.
Collapse
Affiliation(s)
- Judith Cantó-Santos
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Laura Valls-Roca
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Ester Tobías
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Francesc Josep García-García
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Mariona Guitart-Mampel
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Beatriz Martín-Mur
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mercedes Casado
- CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain.,Department of Clinical Biochemistry, Institut de Recerca Sant Joan de Déu; Esplugues de Llobregat, Barcelona, Spain
| | - Rafael Artuch
- CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain.,Department of Clinical Biochemistry, Institut de Recerca Sant Joan de Déu; Esplugues de Llobregat, Barcelona, Spain
| | - Estel Solsona-Vilarrasa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Liver Unit-HCB-IDIBAPS, Barcelona, Spain.,CIBEREHD-Spanish Biomedical Research Centre in Hepatic and Digestive Diseases, Madrid, Spain
| | - José Carlos Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Liver Unit-HCB-IDIBAPS, Barcelona, Spain.,CIBEREHD-Spanish Biomedical Research Centre in Hepatic and Digestive Diseases, Madrid, Spain
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Liver Unit-HCB-IDIBAPS, Barcelona, Spain.,CIBEREHD-Spanish Biomedical Research Centre in Hepatic and Digestive Diseases, Madrid, Spain
| | - Carles Rentero
- Department of Biomedicine, Cell Biology Unit, CELLEX-IDIBAPS, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Carlos Enrich
- Department of Biomedicine, Cell Biology Unit, CELLEX-IDIBAPS, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Pedro J Moreno-Lozano
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - José César Milisenda
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Francesc Cardellach
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Josep M Grau-Junyent
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| | - Glòria Garrabou
- Muscle Research and Mitochondrial Function Lab, Centre de Recerca Biomèdica CELLEX - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Department of Internal Medicine, Hospital Clinic of Barcelona, Barcelona, Spain.,CIBERER-Spanish Biomedical Research Centre in Rare Diseases, Madrid, Spain
| |
Collapse
|
16
|
Blood biomarkers of mitochondrial disease-One for all or all for one? HANDBOOK OF CLINICAL NEUROLOGY 2023; 194:251-257. [PMID: 36813317 DOI: 10.1016/b978-0-12-821751-1.00006-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The mitochondrial disease group consists of different disorders with unprecedented variability of clinical manifestations and tissue-specific symptoms. Their tissue-specific stress responses vary depending on the patients' age and type of dysfunction. These responses include secretion of metabolically active signal molecules to systemic circulation. Such signals-metabolites or metabokines-can be also utilized as biomarkers. During the past 10 years, metabolite and metabokine biomarkers have been described for mitochondrial disease diagnosis and follow-up, to complement the conventional blood biomarkers lactate, pyruvate and alanine. These new tools include metabokines FGF21 and GDF15; cofactors (NAD-forms); sets of metabolites (multibiomarkers) and the full metabolome. FGF21 and GDF15 are messengers of mitochondrial integrated stress response that together outperform the conventional biomarkers in specificity and sensitivity for muscle-manifesting mitochondrial diseases. Metabolite or metabolomic imbalance (e.g., NAD+ deficiency) is a secondary consequence to the primary cause in some diseases, but relevant as a biomarker and a potential indicator of therapy targets. For therapy trials, the optimal biomarker set needs to be tailored to match the disease of interest. The new biomarkers have increased the value of blood samples in mitochondrial disease diagnosis and follow-up, enabling prioritization of patients to different diagnostic paths and having crucial roles in follow-up of therapy effect.
Collapse
|
17
|
Oxidative stress, mitochondrial dysfunction, and respiratory chain enzyme defects in inflammatory myopathies. Autoimmun Rev 2023; 22:103308. [PMID: 36822387 DOI: 10.1016/j.autrev.2023.103308] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023]
Abstract
We investigated the relationship between oxidative stress and inflammatory myopathies. We searched in the current literature the role of mitochondria and respiratory chain defects as sources of oxidative stress and reactive oxygen species production that led to muscle weakness and fatigue. Different molecules and pathways contribute to redox milieu, reactive oxygen species generation, accumulation of misfolded and carbonylated proteins that lose their ability to fulfil cellular activities. Small peptides and physical techniques proved, in mice models, to reduce oxidative stress. We focused on inclusion body myositis, as a major expression of myopathy related to oxidative stress, where mitochondrial abnormalities are causative agents as well. We described the effect of physical exercise in inclusion body myositis that showed to increase strength and to reduce beta amyloid accumulation with subsequent improvement of the mitochondrial functions. We illustrated the influence of epigenetic control on the immune system by non-coding genetic material in the interaction between oxidative stress and inflammatory myopathies.
Collapse
|
18
|
Paredes-Fuentes AJ, Oliva C, Urreizti R, Yubero D, Artuch R. Laboratory testing for mitochondrial diseases: biomarkers for diagnosis and follow-up. Crit Rev Clin Lab Sci 2023; 60:270-289. [PMID: 36694353 DOI: 10.1080/10408363.2023.2166013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The currently available biomarkers generally lack the specificity and sensitivity needed for the diagnosis and follow-up of patients with mitochondrial diseases (MDs). In this group of rare genetic disorders (mutations in approximately 350 genes associated with MDs), all clinical presentations, ages of disease onset and inheritance types are possible. Blood, urine, and cerebrospinal fluid surrogates are well-established biomarkers that are used in clinical practice to assess MD. One of the main challenges is validating specific and sensitive biomarkers for the diagnosis of disease and prediction of disease progression. Profiling of lactate, amino acids, organic acids, and acylcarnitine species is routinely conducted to assess MD patients. New biomarkers, including some proteins and circulating cell-free mitochondrial DNA, with increased diagnostic specificity have been identified in the last decade and have been proposed as potentially useful in the assessment of clinical outcomes. Despite these advances, even these new biomarkers are not sufficiently specific and sensitive to assess MD progression, and new biomarkers that indicate MD progression are urgently needed to monitor the success of novel therapeutic strategies. In this report, we review the mitochondrial biomarkers that are currently analyzed in clinical laboratories, new biomarkers, an overview of the most common laboratory diagnostic techniques, and future directions regarding targeted versus untargeted metabolomic and genomic approaches in the clinical laboratory setting. Brief descriptions of the current methodologies are also provided.
Collapse
Affiliation(s)
- Abraham J Paredes-Fuentes
- Division of Inborn Errors of Metabolism-IBC, Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Clara Oliva
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Roser Urreizti
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Delia Yubero
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,Department of Genetic and Molecular Medicine-IPER, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Rafael Artuch
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Li Q, Hoppe T. Role of amino acid metabolism in mitochondrial homeostasis. Front Cell Dev Biol 2023; 11:1127618. [PMID: 36923249 PMCID: PMC10008872 DOI: 10.3389/fcell.2023.1127618] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/16/2023] [Indexed: 03/03/2023] Open
Abstract
Mitochondria are central hubs for energy production, metabolism and cellular signal transduction in eukaryotic cells. Maintenance of mitochondrial homeostasis is important for cellular function and survival. In particular, cellular metabolic state is in constant communication with mitochondrial homeostasis. One of the most important metabolic processes that provide energy in the cell is amino acid metabolism. Almost all of the 20 amino acids that serve as the building blocks of proteins are produced or degraded in the mitochondria. The synthesis of the amino acids aspartate and arginine depends on the activity of the respiratory chain, which is essential for cell proliferation. The degradation of branched-chain amino acids mainly occurs in the mitochondrial matrix, contributing to energy metabolism, mitochondrial biogenesis, as well as protein quality control in both mitochondria and cytosol. Dietary supplementation or restriction of amino acids in worms, flies and mice modulates lifespan and health, which has been associated with changes in mitochondrial biogenesis, antioxidant response, as well as the activity of tricarboxylic acid cycle and respiratory chain. Consequently, impaired amino acid metabolism has been associated with both primary mitochondrial diseases and diseases with mitochondrial dysfunction such as cancer. Here, we present recent observations on the crosstalk between amino acid metabolism and mitochondrial homeostasis, summarise the underlying molecular mechanisms to date, and discuss their role in cellular functions and organismal physiology.
Collapse
Affiliation(s)
- Qiaochu Li
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Thorsten Hoppe
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
20
|
Murakami A, Noda S, Kazuta T, Hirano S, Kimura S, Nakanishi H, Matsuo K, Tsujikawa K, Iida M, Koike H, Sakamoto K, Hara Y, Kuru S, Kadomatsu K, Shimamura T, Ogi T, Katsuno M. Metabolome and transcriptome analysis on muscle of sporadic inclusion body myositis. Ann Clin Transl Neurol 2022; 9:1602-1615. [PMID: 36107781 PMCID: PMC9539386 DOI: 10.1002/acn3.51657] [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: 07/12/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/06/2022] Open
Abstract
Objective Methods Results Interpretation
Collapse
Affiliation(s)
- Ayuka Murakami
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
- Department of Neurology National Hospital Organization Suzuka Hospital Suzuka Japan
| | - Seiya Noda
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
- Department of Neurology National Hospital Organization Suzuka Hospital Suzuka Japan
| | - Tomoyuki Kazuta
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
- Department of Neurology National Hospital Organization Suzuka Hospital Suzuka Japan
| | - Satoko Hirano
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
- Department of Neurology National Hospital Organization Suzuka Hospital Suzuka Japan
| | - Seigo Kimura
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
- Department of Neurology National Hospital Organization Suzuka Hospital Suzuka Japan
| | | | - Koji Matsuo
- Department of Neurology Kariya Toyota General Hospital Kariya Japan
| | - Koyo Tsujikawa
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Madoka Iida
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Haruki Koike
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Kazuma Sakamoto
- Department of Biochemistry Nagoya University Graduate School of Medicine Nagoya Japan
- Institute for Glyco‐Core Research (iGCORE), Nagoya University Nagoya Japan
| | - Yuichiro Hara
- Department of Genetics Research Institute of Environmental Medicine (RLeM), Nagoya University Nagoya Japan
- Department of Human Genetics and Molecular Biology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Satoshi Kuru
- Department of Neurology National Hospital Organization Suzuka Hospital Suzuka Japan
| | - Kenji Kadomatsu
- Department of Biochemistry Nagoya University Graduate School of Medicine Nagoya Japan
- Institute for Glyco‐Core Research (iGCORE), Nagoya University Nagoya Japan
| | - Teppei Shimamura
- Division of Systems Biology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Tomoo Ogi
- Department of Genetics Research Institute of Environmental Medicine (RLeM), Nagoya University Nagoya Japan
- Department of Human Genetics and Molecular Biology Nagoya University Graduate School of Medicine Nagoya Japan
| | - Masahisa Katsuno
- Department of Neurology Nagoya University Graduate School of Medicine Nagoya Japan
- Department of Clinical Research Education Nagoya University Graduate School of Medicine Nagoya Japan
| |
Collapse
|
21
|
Metrics of progression and prognosis in untreated adults with thymidine kinase 2 deficiency: An observational study. Neuromuscul Disord 2022; 32:728-735. [PMID: 35907766 DOI: 10.1016/j.nmd.2022.07.399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/23/2022] [Accepted: 07/13/2022] [Indexed: 11/21/2022]
Abstract
This historical cohort study evaluated clinical characteristics of progression and prognosis in adults with thymidine kinase 2 deficiency (TK2d). Records were available for 17 untreated adults with TK2d (mean age of onset, 32 years), including longitudinal data from 6 patients (mean follow-up duration, 26.5 months). Pearson's correlation assessed associations between standard motor and respiratory assessments, clinical characteristics, and laboratory values. Longitudinal data were assessed by linear regression mixed models. Respiratory involvement progressed at an annual rate of 8.16% decrement in forced vital capacity (FVC). Most patients under noninvasive ventilation (NIV) remained ambulant (12/14, 86%), reduced FVC was not associated with concomitant decline in 6-minute walk test (6MWT), and 6MWT results were not correlated with FVC. Disease severity, assessed by age at NIV onset, correlated most strongly at diagnosis with: creatinine levels (r = 0.8036; P = 0.0009), followed by FVC (r = 0.7265; P = 0.0033), mtDNA levels in muscle (r = 0.7933; P = 0.0188), and age at disease onset (r = 0.7128; P = 0.0042). This population of adults with TK2d demonstrates rapid deterioration of respiratory muscles, which progresses independently of motor impairment. The results support FVC at diagnosis, mtDNA levels in muscle, and age at disease onset as prognostic indicators. Creatinine levels may also be potentially prognostic, as previously reported in other neuromuscular disorders.
Collapse
|
22
|
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
|
23
|
Romero-Morales AI, Robertson GL, Rastogi A, Rasmussen ML, Temuri H, McElroy GS, Chakrabarty RP, Hsu L, Almonacid PM, Millis BA, Chandel NS, Cartailler JP, Gama V. Human iPSC-derived cerebral organoids model features of Leigh syndrome and reveal abnormal corticogenesis. Development 2022; 149:275911. [PMID: 35792828 PMCID: PMC9357378 DOI: 10.1242/dev.199914] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 05/18/2022] [Indexed: 01/12/2023]
Abstract
Leigh syndrome (LS) is a rare, inherited neurometabolic disorder that presents with bilateral brain lesions caused by defects in the mitochondrial respiratory chain and associated nuclear-encoded proteins. We generated human induced pluripotent stem cells (iPSCs) from three LS patient-derived fibroblast lines. Using whole-exome and mitochondrial sequencing, we identified unreported mutations in pyruvate dehydrogenase (GM0372, PDH; GM13411, MT-ATP6/PDH) and dihydrolipoyl dehydrogenase (GM01503, DLD). These LS patient-derived iPSC lines were viable and capable of differentiating into progenitor populations, but we identified several abnormalities in three-dimensional differentiation models of brain development. LS patient-derived cerebral organoids showed defects in neural epithelial bud generation, size and cortical architecture at 100 days. The double mutant MT-ATP6/PDH line produced organoid neural precursor cells with abnormal mitochondrial morphology, characterized by fragmentation and disorganization, and showed an increased generation of astrocytes. These studies aim to provide a comprehensive phenotypic characterization of available patient-derived cell lines that can be used to study Leigh syndrome.
Collapse
Affiliation(s)
| | - Gabriella L. Robertson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Anuj Rastogi
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Megan L. Rasmussen
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Hoor Temuri
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Gregory Scott McElroy
- Feinberg School of Medicine, Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ram Prosad Chakrabarty
- Feinberg School of Medicine, Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lawrence Hsu
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology,Vanderbilt University,Nashville, TN 37232, USA
| | | | - Bryan A. Millis
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA,Vanderbilt Biophotonics Center,Vanderbilt University, Nashville, TN 37232, USA
| | - Navdeep S. Chandel
- Feinberg School of Medicine, Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL 60611, USA,Feinberg School of Medicine, Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL 60611, USA
| | - Jean-Philippe Cartailler
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology,Vanderbilt University,Nashville, TN 37232, USA
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA,Creative Data Solutions, Vanderbilt Center for Stem Cell Biology,Vanderbilt University,Nashville, TN 37232, USA,Vanderbilt Brain Institute,Vanderbilt University,Nashville, TN 37232, USA,Author for correspondence ()
| |
Collapse
|
24
|
Järvilehto J, Harjuhaahto S, Palu E, Auranen M, Kvist J, Zetterberg H, Koskivuori J, Lehtonen M, Saukkonen AM, Jokela M, Ylikallio E, Tyynismaa H. Serum Creatine, Not Neurofilament Light, Is Elevated in CHCHD10-Linked Spinal Muscular Atrophy. Front Neurol 2022; 13:793937. [PMID: 35250809 PMCID: PMC8891230 DOI: 10.3389/fneur.2022.793937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To characterize serum biomarkers in mitochondrial CHCHD10-linked spinal muscular atrophy Jokela (SMAJ) type for disease monitoring and for the understanding of pathogenic mechanisms. Methods We collected serum samples from a cohort of 49 patients with SMAJ, all carriers of the heterozygous c.197G>T p.G66V variant in CHCHD10. As controls, we used age- and sex-matched serum samples obtained from Helsinki Biobank. Creatine kinase and creatinine were measured by standard methods. Neurofilament light (NfL) and glial fibrillary acidic protein (GFAP) were measured with single molecule array (Simoa), fibroblast growth factor 21 (FGF-21), and growth differentiation factor 15 (GDF-15) with an enzyme-linked immunosorbent assay. For non-targeted plasma metabolite profiling, samples were analyzed with liquid chromatography high-resolution mass spectrometry. Disease severity was evaluated retrospectively by calculating a symptom-based score. Results Axon degeneration marker, NfL, was unexpectedly not altered in the serum of patients with SMAJ, whereas astrocytic activation marker, GFAP, was slightly decreased. Creatine kinase was elevated in most patients, particularly men. We identified six metabolites that were significantly altered in serum of patients with SMAJ in comparison to controls: increased creatine and pyruvate, and decreased creatinine, taurine, N-acetyl-carnosine, and succinate. Creatine correlated with disease severity. Altered pyruvate and succinate indicated a metabolic response to mitochondrial dysfunction; however, lactate or mitochondrial myopathy markers FGF-21 or GDF-15 was not changed. Conclusions Biomarkers of muscle mass and damage are altered in SMAJ serum, indicating a role for skeletal muscle in disease pathogenesis in addition to neurogenic damage. Despite the minimal mitochondrial pathology in skeletal muscle, signs of a metabolic shift can be detected.
Collapse
Affiliation(s)
- Julius Järvilehto
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sandra Harjuhaahto
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Edouard Palu
- Clinical Neurosciences, Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Mari Auranen
- Clinical Neurosciences, Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Jouni Kvist
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | | | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - Manu Jokela
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
- Department of Neurology, Neuromuscular Research Center, Tampere University Hospital and Tampere University, Tampere, Finland
| | - Emil Ylikallio
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Clinical Neurosciences, Neurology, Helsinki University Hospital, Helsinki, Finland
- *Correspondence: Emil Ylikallio
| | - Henna Tyynismaa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Henna Tyynismaa
| |
Collapse
|
25
|
Denmark D, Ruhoy I, Wittmann B, Ashki H, Koran LM. Altered Plasma Mitochondrial Metabolites in Persistently Symptomatic Individuals after a GBCA-Assisted MRI. TOXICS 2022; 10:56. [PMID: 35202243 PMCID: PMC8879776 DOI: 10.3390/toxics10020056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/24/2022] [Indexed: 12/22/2022]
Abstract
Despite the impressive safety of gadolinium (Gd)-based contrast agents (GBCAs), a small number of patients report the onset of new, severe, ongoing symptoms after even a single exposure-a syndrome termed Gadolinium Deposition Disease (GDD). Mitochondrial dysfunction and oxidative stress have been repeatedly implicated by animal and in vitro studies as mechanisms of Gd/GBCA-related toxicity, and as pathogenic in other diseases with similarities in presentation. Here, we aimed to molecularly characterize and explore potential metabolic associations with GDD symptoms. Detailed clinical phenotypes were systematically obtained for a small cohort of individuals (n = 15) with persistent symptoms attributed to a GBCA-enhanced MRI and consistent with provisional diagnostic criteria for GDD. Global untargeted mass spectroscopy-based metabolomics analyses were performed on plasma samples and examined for relevance with both single marker and pathways approaches. In addition to GDD criteria, frequently reported symptoms resembled those of patients with known mitochondrial-related diseases. Plasma differences compared to a healthy, asymptomatic reference cohort were suggested for 45 of 813 biochemicals. A notable proportion of these are associated with mitochondrial function and related disorders, including nucleotide and energy superpathways, which were over-represented. Although early evidence, coincident clinical and biochemical indications of potential mitochondrial involvement in GDD are remarkable in light of preclinical models showing adverse Gd/GBCA effects on multiple aspects of mitochondrial function. Further research on the potential contributory role of these markers and pathways in persistent symptoms attributed to GBCA exposure is recommended.
Collapse
Affiliation(s)
- DeAunne Denmark
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3710 SW US Veterans Hospital Road, Mail Code R&D40, Portland, OR 97239, USA;
| | - Ilene Ruhoy
- Mount Sinai South Nassau Chiari-EDS Center, 1420 Broadway, Hewlett, NY 11557, USA;
| | - Bryan Wittmann
- Owlstone Medical, 600 Park Offices Drive, Suite 140, Research Triangle Park, NC 27709, USA;
| | - Haleh Ashki
- Prime Genomics, Inc., 319 Bernardo Avenue, Mountain View, CA 94041, USA;
| | - Lorrin M. Koran
- Department of Psychiatry and Behavioral Sciences, OCD Clinic, Stanford University Medical Center, 401 Quarry Road, Stanford, CA 94305, USA
| |
Collapse
|
26
|
Chung H, Jo Y, Ryu D, Jeong C, Choe S, Lee J. Artificial-intelligence-driven discovery of prognostic biomarker for sarcopenia. J Cachexia Sarcopenia Muscle 2021; 12:2220-2230. [PMID: 34704369 PMCID: PMC8718042 DOI: 10.1002/jcsm.12840] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/02/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Sarcopenia is defined as muscle wasting, characterized by a progressive loss of muscle mass and function due to ageing. Diagnosis of sarcopenia typically involves both muscle imaging and the physical performance of people exhibiting signs of muscle weakness. Despite its worldwide prevalence, a molecular method for accurately diagnosing sarcopenia has not been established. METHODS We develop an artificial intelligence (AI) diagnosis model of sarcopenia using a published transcriptome dataset comprising patients from multiple ethnicities. For the AI model for sarcopenia diagnosis, we use a transcriptome database comprising 17 339 genes from 118 subjects. Among the 17 339 genes, we select 27 features as the model inputs. For feature selection, we use a random forest, extreme gradient boosting and adaptive boosting. Using the top 27 features, we propose a four-layer deep neural network, named DSnet-v1, for sarcopenia diagnosis. RESULTS Among isolated testing datasets, DSnet-v1 provides high sensitivity (100%), specificity (94.12%), accuracy (95.83%), balanced accuracy (97.06%) and area under receiver operating characteristics (0.99). To extend the number of patient data, we develop a web application (http://sarcopeniaAI.ml/), where the model can be accessed unrestrictedly to diagnose sarcopenia if the transcriptome is available. A focused analysis of the top 27 genes for their differential or co-expression with other genes implied the potential existence of race-specific factors for sarcopenia, suggesting the possibility of identifying causal factors of sarcopenia when a more extended dataset is provided. CONCLUSIONS Our new AI model, DSnet-v1, accurately diagnoses sarcopenia and is currently available publicly to assist healthcare providers in diagnosing and treating sarcopenia.
Collapse
Affiliation(s)
- Heewon Chung
- Department of Biomedical Engineering, College of Electronics and InformationKyung Hee UniversityYongin‐siGyeonggi‐doRepublic of Korea
| | - Yunju Jo
- Department of Molecular Cell BiologySungkyunkwan University School of MedicineSuwonRepublic of Korea
- Sarcopenia Total Solution CenterWonkwang University School of MedicineIksanRepublic of Korea
| | - Dongryeol Ryu
- Department of Molecular Cell BiologySungkyunkwan University School of MedicineSuwonRepublic of Korea
- Sarcopenia Total Solution CenterWonkwang University School of MedicineIksanRepublic of Korea
| | - Changwon Jeong
- Sarcopenia Total Solution CenterWonkwang University School of MedicineIksanRepublic of Korea
- Medical Convergence Research CenterWonkwang UniversityIksanRepublic of Korea
| | - Seong‐Kyu Choe
- Sarcopenia Total Solution CenterWonkwang University School of MedicineIksanRepublic of Korea
- Department of Microbiology, and Institute of Wonkwang Medical ScienceWonkwang University School of MedicineIksanJeonbukRepublic of Korea
| | - Jinseok Lee
- Department of Biomedical Engineering, College of Electronics and InformationKyung Hee UniversityYongin‐siGyeonggi‐doRepublic of Korea
| |
Collapse
|
27
|
Rewiring cell signalling pathways in pathogenic mtDNA mutations. Trends Cell Biol 2021; 32:391-405. [PMID: 34836781 DOI: 10.1016/j.tcb.2021.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022]
Abstract
Mitochondria generate the energy to sustain cell viability and serve as a hub for cell signalling. Their own genome (mtDNA) encodes genes critical for oxidative phosphorylation. Mutations of mtDNA cause major disease and disability with a wide range of presentations and severity. We review here an emerging body of data suggesting that changes in cell metabolism and signalling pathways in response to the presence of mtDNA mutations play a key role in shaping disease presentation and progression. Understanding the impact of mtDNA mutations on cellular energy homeostasis and signalling pathways seems fundamental to identify novel therapeutic interventions with the potential to improve the prognosis for patients with primary mitochondrial disease.
Collapse
|
28
|
Zárybnický T, Heikkinen A, Kangas SM, Karikoski M, Martínez-Nieto GA, Salo MH, Uusimaa J, Vuolteenaho R, Hinttala R, Sipilä P, Kuure S. Modeling Rare Human Disorders in Mice: The Finnish Disease Heritage. Cells 2021; 10:cells10113158. [PMID: 34831381 PMCID: PMC8621025 DOI: 10.3390/cells10113158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/31/2022] Open
Abstract
The modification of genes in animal models has evidently and comprehensively improved our knowledge on proteins and signaling pathways in human physiology and pathology. In this review, we discuss almost 40 monogenic rare diseases that are enriched in the Finnish population and defined as the Finnish disease heritage (FDH). We will highlight how gene-modified mouse models have greatly facilitated the understanding of the pathological manifestations of these diseases and how some of the diseases still lack proper models. We urge the establishment of subsequent international consortiums to cooperatively plan and carry out future human disease modeling strategies. Detailed information on disease mechanisms brings along broader understanding of the molecular pathways they act along both parallel and transverse to the proteins affected in rare diseases, therefore also aiding understanding of common disease pathologies.
Collapse
Affiliation(s)
- Tomáš Zárybnický
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014 Helsinki, Finland;
| | - Anne Heikkinen
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland
| | - Salla M. Kangas
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
- PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland;
- Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
| | - Marika Karikoski
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (M.K.); (G.A.M.-N.)
| | - Guillermo Antonio Martínez-Nieto
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (M.K.); (G.A.M.-N.)
- Turku Center for Disease Modelling (TCDM), Institute of Biomedicine, University of Turku, 20520 Turku, Finland
| | - Miia H. Salo
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
- PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland;
- Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
| | - Johanna Uusimaa
- PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland;
- Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
- Clinic for Children and Adolescents, Division of Pediatric Neurology, Oulu University Hospital, P.O. Box 20, 90029 Oulu, Finland
| | - Reetta Vuolteenaho
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
| | - Reetta Hinttala
- Biocenter Oulu, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland; (A.H.); (S.M.K.); (M.H.S.); (R.V.)
- PEDEGO Research Unit, University of Oulu, P.O. Box 8000, 90014 Oulu, Finland;
- Medical Research Center, Oulu University Hospital, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
- Correspondence: (R.H.); (P.S.); (S.K.)
| | - Petra Sipilä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (M.K.); (G.A.M.-N.)
- Turku Center for Disease Modelling (TCDM), Institute of Biomedicine, University of Turku, 20520 Turku, Finland
- Correspondence: (R.H.); (P.S.); (S.K.)
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014 Helsinki, Finland;
- GM-Unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, 00790 Helsinki, Finland
- Correspondence: (R.H.); (P.S.); (S.K.)
| |
Collapse
|
29
|
Blood biomarkers for assessment of mitochondrial dysfunction: An expert review. Mitochondrion 2021; 62:187-204. [PMID: 34740866 DOI: 10.1016/j.mito.2021.10.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/20/2022]
Abstract
Although mitochondrial dysfunction is the known cause of primary mitochondrial disease, mitochondrial dysfunction is often difficult to measure and prove, especially when biopsies of affected tissue are not available. In order to identify blood biomarkers of mitochondrial dysfunction, we reviewed studies that measured blood biomarkers in genetically, clinically or biochemically confirmed primary mitochondrial disease patients. In this way, we were certain that there was an underlying mitochondrial dysfunction which could validate the biomarker. We found biomarkers of three classes: 1) functional markers measured in blood cells, 2) biochemical markers of serum/plasma and 3) DNA markers. While none of the reviewed single biomarkers may perfectly reveal all underlying mitochondrial dysfunction, combining biomarkers that cover different aspects of mitochondrial impairment probably is a good strategy. This biomarker panel may assist in the diagnosis of primary mitochondrial disease patients. As mitochondrial dysfunction may also play a significant role in the pathophysiology of multifactorial disorders such as Alzheimer's disease and glaucoma, the panel may serve to assess mitochondrial dysfunction in complex multifactorial diseases as well and enable selection of patients who could benefit from therapies targeting mitochondria.
Collapse
|
30
|
Odom JD, Sutton VR. Metabolomics in Clinical Practice: Improving Diagnosis and Informing Management. Clin Chem 2021; 67:1606-1617. [PMID: 34633032 DOI: 10.1093/clinchem/hvab184] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/17/2021] [Indexed: 11/14/2022]
Abstract
BACKGROUND Metabolomics is the study of small molecules to simultaneously identify multiple low molecular weight molecules in a system. Broadly speaking, metabolomics can be subdivided into targeted and untargeted types of analysis, each type having advantages and drawbacks. Targeted metabolomics can quantify analytes but only looks for known or expected analytes related to particular disease(s), whereas untargeted metabolomics is typically nonquantitative but can detect thousands of analytes from an agnostic or nonhypothesis driven perspective, allowing for novel discoveries. CONTENT One application of metabolomics is the study of inborn errors of metabolism (IEM). The biochemical hallmark of IEMs is decreased concentrations of analytes distal to the enzymatic defect and buildup of analytes proximal to the defect. Metabolomics can detect these changes with one test and is effective in screening for and diagnosis of IEMs. Metabolomics has also been used to study many nonmetabolic diseases such as autism spectrum disorder, various cancers, and multiple congenital anomalies syndromes. Metabolomics has led to the discovery of many novel biomarkers of disease. Recent publications demonstrate how metabolomics can be useful clinically in the diagnosis and management of patients, as well as for research and clinical discovery. SUMMARY Metabolomics has proved to be a useful tool clinically for screening and diagnostic purposes and from a research perspective for the detection of novel biomarkers. In the future, metabolomics will likely become a routine part of the evaluation for many diseases as either a supplementary test or it may simply replace historical analyses that require several individual tests and sample types.
Collapse
Affiliation(s)
- John D Odom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - V Reid Sutton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX.,Baylor Genetics Laboratory, Houston, TX
| |
Collapse
|
31
|
Odom J, Amin H, Gijavanekar C, Elsea SH, Kralik S, Chinen J, Lin Y, Yates AMM, Mizerik E, Potocki L, Scaglia F. A phenotypic expansion of TRNT1 associated sideroblastic anemia with immunodeficiency, fevers, and developmental delay. Am J Med Genet A 2021; 188:259-268. [PMID: 34510712 DOI: 10.1002/ajmg.a.62482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 11/06/2022]
Abstract
Sideroblastic anemia with immunodeficiency, fevers, and developmental delay (SIFD; MIM #616084) is an autosomal recessive disorder of mitochondrial and cytosolic tRNA processing caused by pathogenic, biallelic variants in TRNT1. Other features of this disorder include central nervous system, renal, cardiac, ophthalmological features, and sensorineural hearing impairment. SIFD was first described in 2013 and to date, it has been reported in 46 patients. Herein, we review the literature and describe two siblings with SIFD and note the novel phenotype of hypoglycemia in the context of growth hormone (GH) deficiency. GH deficiency without hypoglycemia has previously been reported in three patients with SIFD, but GH deficiency had not been firmly ascribed to SIFD. We propose to expand the phenotype to include GH deficiency, hypoglycemia, and previously unreported dysmorphic features. Furthermore, we highlight the intrafamilial variability of the disease by the discordance of our patients' clinical phenotypes and biochemical profiles measured by untargeted metabolomics analysis. Several metabolomic abnormalities were observed in both patients, and these may represent a potential biochemical signature for SIFD.
Collapse
Affiliation(s)
- John Odom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Hitha Amin
- Texas Children's Hospital, Houston, Texas, USA.,Cortica Care, Irvine, California, USA.,Section of Child Neurology and Neurodevelopmental Disabilities, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Charul Gijavanekar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah H Elsea
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Stephen Kralik
- Texas Children's Hospital, Houston, Texas, USA.,Department of Radiology, Baylor College of Medicine, Houston, Texas, USA
| | - Javier Chinen
- Division of Allergy and Immunology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Yuezhen Lin
- Division of Endocrinology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Amber Meshell Mayfield Yates
- Texas Children's Hospital, Houston, Texas, USA.,Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Elizabeth Mizerik
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Lorraine Potocki
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA.,Joint BCM-CUHK Center of Medical Genetics, Prince of Wales Hospital, Shatin, Hong Kong SAR
| |
Collapse
|
32
|
Sharma R, Reinstadler B, Engelstad K, Skinner OS, Stackowitz E, Haller RG, Clish CB, Pierce K, Walker MA, Fryer R, Oglesbee D, Mao X, Shungu DC, Khatri A, Hirano M, De Vivo DC, Mootha VK. Circulating markers of NADH-reductive stress correlate with mitochondrial disease severity. J Clin Invest 2021; 131:136055. [PMID: 33463549 DOI: 10.1172/jci136055] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial disorders represent a large collection of rare syndromes that are difficult to manage both because we do not fully understand biochemical pathogenesis and because we currently lack facile markers of severity. The m.3243A>G variant is the most common heteroplasmic mitochondrial DNA mutation and underlies a spectrum of diseases, notably mitochondrial encephalomyopathy lactic acidosis and stroke-like episodes (MELAS). To identify robust circulating markers of m.3243A>G disease, we first performed discovery proteomics, targeted metabolomics, and untargeted metabolomics on plasma from a deeply phenotyped cohort (102 patients, 32 controls). In a validation phase, we measured concentrations of prioritized metabolites in an independent cohort using distinct methods. We validated 20 analytes (1 protein, 19 metabolites) that distinguish patients with MELAS from controls. The collection includes classic (lactate, alanine) and more recently identified (GDF-15, α-hydroxybutyrate) mitochondrial markers. By mining untargeted mass-spectra we uncovered 3 less well-studied metabolite families: N-lactoyl-amino acids, β-hydroxy acylcarnitines, and β-hydroxy fatty acids. Many of these 20 analytes correlate strongly with established measures of severity, including Karnofsky status, and mechanistically, nearly all markers are attributable to an elevated NADH/NAD+ ratio, or NADH-reductive stress. Our work defines a panel of organelle function tests related to NADH-reductive stress that should enable classification and monitoring of mitochondrial disease.
Collapse
Affiliation(s)
- Rohit Sharma
- Howard Hughes Medical Institute, Department of Molecular Biology, and.,Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute, Cambridge, Massachusetts, USA
| | - Bryn Reinstadler
- Howard Hughes Medical Institute, Department of Molecular Biology, and.,Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute, Cambridge, Massachusetts, USA
| | - Kristin Engelstad
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Owen S Skinner
- Howard Hughes Medical Institute, Department of Molecular Biology, and.,Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute, Cambridge, Massachusetts, USA
| | - Erin Stackowitz
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Ronald G Haller
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Institute for Exercise and Environmental Medicine of Texas Health Presbyterian Hospital, Dallas, Texas, USA
| | | | | | - Melissa A Walker
- Howard Hughes Medical Institute, Department of Molecular Biology, and.,Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute, Cambridge, Massachusetts, USA.,Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert Fryer
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiangling Mao
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Ashok Khatri
- Endocrine Division and Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michio Hirano
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Darryl C De Vivo
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Vamsi K Mootha
- Howard Hughes Medical Institute, Department of Molecular Biology, and.,Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
33
|
Maresca A, Del Dotto V, Capristo M, Scimonelli E, Tagliavini F, Morandi L, Tropeano CV, Caporali L, Mohamed S, Roberti M, Scandiffio L, Zaffagnini M, Rossi J, Cappelletti M, Musiani F, Contin M, Riva R, Liguori R, Pizza F, La Morgia C, Antelmi E, Loguercio Polosa P, Mignot E, Zanna C, Plazzi G, Carelli V. DNMT1 mutations leading to neurodegeneration paradoxically reflect on mitochondrial metabolism. Hum Mol Genet 2021; 29:1864-1881. [PMID: 31984424 PMCID: PMC7372549 DOI: 10.1093/hmg/ddaa014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
ADCA-DN and HSN-IE are rare neurodegenerative syndromes caused by dominant mutations in the replication foci targeting sequence (RFTS) of the DNA methyltransferase 1 (DNMT1) gene. Both phenotypes resemble mitochondrial disorders, and mitochondrial dysfunction was first observed in ADCA-DN. To explore mitochondrial involvement, we studied the effects of DNMT1 mutations in fibroblasts from four ADCA-DN and two HSN-IE patients. We documented impaired activity of purified DNMT1 mutant proteins, which in fibroblasts results in increased DNMT1 amount. We demonstrated that DNMT1 is not localized within mitochondria, but it is associated with the mitochondrial outer membrane. Concordantly, mitochondrial DNA failed to show meaningful CpG methylation. Strikingly, we found activated mitobiogenesis and OXPHOS with significant increase of H2O2, sharply contrasting with a reduced ATP content. Metabolomics profiling of mutant cells highlighted purine, arginine/urea cycle and glutamate metabolisms as the most consistently altered pathways, similar to primary mitochondrial diseases. The most severe mutations showed activation of energy shortage AMPK-dependent sensing, leading to mTORC1 inhibition. We propose that DNMT1 RFTS mutations deregulate metabolism lowering ATP levels, as a result of increased purine catabolism and urea cycle pathways. This is associated with a paradoxical mitochondrial hyper-function and increased oxidative stress, possibly resulting in neurodegeneration in non-dividing cells.
Collapse
Affiliation(s)
- Alessandra Maresca
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Valentina Del Dotto
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Mariantonietta Capristo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Emanuela Scimonelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Francesca Tagliavini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Luca Morandi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | | | - Leonardo Caporali
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Susan Mohamed
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy
| | - Marina Roberti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari 70126, Italy
| | - Letizia Scandiffio
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari 70126, Italy
| | - Mirko Zaffagnini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Jacopo Rossi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Martina Cappelletti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Francesco Musiani
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Manuela Contin
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Roberto Riva
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Fabio Pizza
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Chiara La Morgia
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Elena Antelmi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Paola Loguercio Polosa
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari 70126, Italy
| | - Emmanuel Mignot
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94304, USA
| | - Claudia Zanna
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Giuseppe Plazzi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna 40139, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40139, Italy
| |
Collapse
|
34
|
Ng YS, Bindoff LA, Gorman GS, Klopstock T, Kornblum C, Mancuso M, McFarland R, Sue CM, Suomalainen A, Taylor RW, Thorburn DR, Turnbull DM. Mitochondrial disease in adults: recent advances and future promise. Lancet Neurol 2021; 20:573-584. [PMID: 34146515 DOI: 10.1016/s1474-4422(21)00098-3] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/17/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Mitochondrial diseases are some of the most common inherited neurometabolic disorders, and major progress has been made in our understanding, diagnosis, and treatment of these conditions in the past 5 years. Development of national mitochondrial disease cohorts and international collaborations has changed our knowledge of the spectrum of clinical phenotypes and natural history of mitochondrial diseases. Advances in high-throughput sequencing technologies have altered the diagnostic algorithm for mitochondrial diseases by increasingly using a genetics-first approach, with more than 350 disease-causing genes identified to date. While the current management strategy for mitochondrial disease focuses on surveillance for multisystem involvement and effective symptomatic treatment, new endeavours are underway to find better treatments, including repurposing current drugs, use of novel small molecules, and gene therapies. Developments made in reproductive technology offer women the opportunity to prevent transmission of DNA-related mitochondrial disease to their children.
Collapse
Affiliation(s)
- Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Directorate of Neurosciences, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Laurence A Bindoff
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Gráinne S Gorman
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Directorate of Neurosciences, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institute, LMU Hospital, Ludwig Maximilians University, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany
| | - Cornelia Kornblum
- Department of Neurology, Neuromuscular Disease Section, University Hospital Bonn, Bonn, Germany; Centre for Rare Diseases, University Hospital Bonn, Bonn, Germany
| | - Michelangelo Mancuso
- Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Italy
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Carolyn M Sue
- Department of Neurogenetics, Kolling Institute, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Neurology, Royal North Shore Hospital, Northern Sydney Local Health District, St Leonards, NSW, Australia
| | - Anu Suomalainen
- Research Program in Stem Cells and Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Neuroscience Centre, HiLife, University of Helsinki, Helsinki, Finland; Helsinki University Hospital, HUSlab, Helsinki, Finland
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - David R Thorburn
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia; Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Doug M Turnbull
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
35
|
Maresca A, Carelli V. Molecular Mechanisms behind Inherited Neurodegeneration of the Optic Nerve. Biomolecules 2021; 11:496. [PMID: 33806088 PMCID: PMC8064499 DOI: 10.3390/biom11040496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 01/01/2023] Open
Abstract
Inherited neurodegeneration of the optic nerve is a paradigm in neurology, as many forms of isolated or syndromic optic atrophy are encountered in clinical practice. The retinal ganglion cells originate the axons that form the optic nerve. They are particularly vulnerable to mitochondrial dysfunction, as they present a peculiar cellular architecture, with axons that are not myelinated for a long intra-retinal segment, thus, very energy dependent. The genetic landscape of causative mutations and genes greatly enlarged in the last decade, pointing to common pathways. These mostly imply mitochondrial dysfunction, which leads to a similar outcome in terms of neurodegeneration. We here critically review these pathways, which include (1) complex I-related oxidative phosphorylation (OXPHOS) dysfunction, (2) mitochondrial dynamics, and (3) endoplasmic reticulum-mitochondrial inter-organellar crosstalk. These major pathogenic mechanisms are in turn interconnected and represent the target for therapeutic strategies. Thus, their deep understanding is the basis to set and test new effective therapies, an urgent unmet need for these patients. New tools are now available to capture all interlinked mechanistic intricacies for the pathogenesis of optic nerve neurodegeneration, casting hope for innovative therapies to be rapidly transferred into the clinic and effectively cure inherited optic neuropathies.
Collapse
Affiliation(s)
- Alessandra Maresca
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, 40139 Bologna, Italy;
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, 40139 Bologna, Italy;
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy
| |
Collapse
|
36
|
Gusic M, Prokisch H. Genetic basis of mitochondrial diseases. FEBS Lett 2021; 595:1132-1158. [PMID: 33655490 DOI: 10.1002/1873-3468.14068] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Mitochondrial disorders are monogenic disorders characterized by a defect in oxidative phosphorylation and caused by pathogenic variants in one of over 340 different genes. The implementation of whole-exome sequencing has led to a revolution in their diagnosis, duplicated the number of associated disease genes, and significantly increased the diagnosed fraction. However, the genetic etiology of a substantial fraction of patients exhibiting mitochondrial disorders remains unknown, highlighting limitations in variant detection and interpretation, which calls for improved computational and DNA sequencing methods, as well as the addition of OMICS tools. More intriguingly, this also suggests that some pathogenic variants lie outside of the protein-coding genes and that the mechanisms beyond the Mendelian inheritance and the mtDNA are of relevance. This review covers the current status of the genetic basis of mitochondrial diseases, discusses current challenges and perspectives, and explores the contribution of factors beyond the protein-coding regions and monogenic inheritance in the expansion of the genetic spectrum of disease.
Collapse
Affiliation(s)
- Mirjana Gusic
- Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.,Institute of Human Genetics, Technical University of Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Germany
| | - Holger Prokisch
- Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.,Institute of Human Genetics, Technical University of Munich, Germany
| |
Collapse
|
37
|
Esterhuizen K, Lindeque JZ, Mason S, van der Westhuizen FH, Rodenburg RJ, de Laat P, Smeitink JAM, Janssen MCH, Louw R. One mutation, three phenotypes: novel metabolic insights on MELAS, MIDD and myopathy caused by the m.3243A > G mutation. Metabolomics 2021; 17:10. [PMID: 33438095 DOI: 10.1007/s11306-020-01769-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/31/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The m.3243A > G mitochondrial DNA mutation is one of the most common mitochondrial disease-causing mutations, with a carrier rate as high as 1:400. This point mutation affects the MT-TL1 gene, ultimately affecting the oxidative phosphorylation system and the cell's energy production. Strikingly, the m.3243A > G mutation is associated with different phenotypes, including mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), maternally inherited diabetes and deafness (MIDD) and myopathy. OBJECTIVES We investigated urine metabolomes of MELAS, MIDD and myopathy patients in order to identify affected metabolic pathways and possible treatment options. METHODS A multiplatform metabolomics approach was used to comprehensively analyze the metabolome and compare metabolic profiles of different phenotypes caused by the m.3243A > G mutation. Our analytical array consisted of NMR spectroscopy, LC-MS/MS and GC-TOF-MS. RESULTS The investigation revealed phenotypic specific metabolic perturbations, as well as metabolic similarities between the different phenotypes. We show that glucose metabolism is highly disturbed in the MIDD phenotype, but not in MELAS or myopathy, remodeled fatty acid oxidation is characteristic of the MELAS patients, while one-carbon metabolism is strongly modified in both MELAS and MIDD, but not in the myopathy group. Lastly we identified increased creatine in the urine of the myopathy patients, but not in MELAS or MIDD. CONCLUSION We conclude by giving novel insight on the phenotypes of the m.3243A > G mutation from a metabolomics point of view. Directives are also given for future investigations that could lead to better treatment options for patients suffering from this debilitating disease.
Collapse
Affiliation(s)
- Karien Esterhuizen
- Mitochondria Research Laboratory, Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - J Zander Lindeque
- Mitochondria Research Laboratory, Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Shayne Mason
- Mitochondria Research Laboratory, Human Metabolomics, North-West University, Potchefstroom, South Africa
| | | | - Richard J Rodenburg
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Paul de Laat
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Jan A M Smeitink
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Mirian C H Janssen
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud Center for Mitochondrial Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Roan Louw
- Mitochondria Research Laboratory, Human Metabolomics, North-West University, Potchefstroom, South Africa.
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom Campus, Private Bag X6001, Potchefstroom, South Africa.
| |
Collapse
|
38
|
Mocking RJT, Naviaux JC, Li K, Wang L, Monk JM, Bright AT, Figueroa CA, Schene AH, Ruhé HG, Assies J, Naviaux RK. Metabolic features of recurrent major depressive disorder in remission, and the risk of future recurrence. Transl Psychiatry 2021; 11:37. [PMID: 33431800 PMCID: PMC7801396 DOI: 10.1038/s41398-020-01182-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/08/2020] [Accepted: 12/15/2020] [Indexed: 01/29/2023] Open
Abstract
Recurrent major depressive disorder (rMDD) is a relapsing-remitting disease with high morbidity and a 5-year risk of recurrence of up to 80%. This was a prospective pilot study to examine the potential diagnostic and prognostic value of targeted plasma metabolomics in the care of patients with rMDD in remission. We used an established LC-MS/MS platform to measure 399 metabolites in 68 subjects with rMDD (n = 45 females and 23 males) in antidepressant-free remission and 59 age- and sex-matched controls (n = 40 females and 19 males). Patients were then followed prospectively for 2.5 years. Metabolomics explained up to 43% of the phenotypic variance. The strongest biomarkers were gender specific. 80% of the metabolic predictors of recurrence in both males and females belonged to 6 pathways: (1) phospholipids, (2) sphingomyelins, (3) glycosphingolipids, (4) eicosanoids, (5) microbiome, and (6) purines. These changes traced to altered mitochondrial regulation of cellular redox, signaling, energy, and lipid metabolism. Metabolomics identified a chemical endophenotype that could be used to stratify rrMDD patients at greatest risk for recurrence with an accuracy over 0.90 (95%CI = 0.69-1.0). Power calculations suggest that a validation study of at least 198 females and 198 males (99 cases and 99 controls each) will be needed to confirm these results. Although a small study, these results are the first to show the potential utility of metabolomics in assisting with the important clinical challenge of prospectively identifying the patients at greatest risk of recurrence of a depressive episode and those who are at lower risk.
Collapse
Affiliation(s)
- Roel J T Mocking
- Department of Psychiatry, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Meibergdreef 5, 1105 AZ, Amsterdam, The Netherlands.
| | - Jane C Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
- Department of Neurosciences, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
| | - Kefeng Li
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
| | - Lin Wang
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
| | - Jonathan M Monk
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
| | - A Taylor Bright
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
- Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA
- Colt Neck Labs, 838 E High St 202., Lexington, KY, 40503, USA
| | - Caroline A Figueroa
- Department of Psychiatry, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Meibergdreef 5, 1105 AZ, Amsterdam, The Netherlands
- School of Social Welfare, University of California, Berkeley, CA, 94720, USA
| | - Aart H Schene
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Henricus G Ruhé
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Johanna Assies
- Department of Psychiatry, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Meibergdreef 5, 1105 AZ, Amsterdam, The Netherlands.
| | - Robert K Naviaux
- The Mitochondrial and Metabolic Disease Center, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA.
- Department of Medicine, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA.
- Department of Pediatrics, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA.
- Department of Pathology, University of California, San Diego School of Medicine, 214 Dickinson St., Bldg CTF, Rm C107, San Diego, CA, 92103-8467, USA.
| |
Collapse
|
39
|
Maresca A, Del Dotto V, Romagnoli M, La Morgia C, Di Vito L, Capristo M, Valentino ML, Carelli V. Expanding and validating the biomarkers for mitochondrial diseases. J Mol Med (Berl) 2020; 98:1467-1478. [PMID: 32851462 PMCID: PMC7524861 DOI: 10.1007/s00109-020-01967-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/05/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
Mitochondrial diseases are highly heterogeneous metabolic disorders caused by genetic alterations in the mitochondrial DNA (mtDNA) or in the nuclear genome. In this study, we investigated a panel of blood biomarkers in a cohort of 123 mitochondrial patients, with prominent neurological and muscular manifestations. These biomarkers included creatine, fibroblast growth factor 21 (FGF21) and growth/differentiation factor 15 (GDF-15), and the novel cell free circulating-mtDNA (ccf-mtDNA). All biomarkers were significantly increased in the patient group. After stratification by the specific phenotypes, ccf-mtDNA was significantly increased in the Mitochondrial Encephalomyopathy Lactic Acidosis Stroke-like episodes syndrome (MELAS) group, and FGF21 and GDF-15 were significantly elevated in patients with MELAS and Myoclonic Epilepsy Ragged Red Fibers syndrome. On the contrary, in our cohort, creatine was not associated to a specific clinical phenotype. Longitudinal assessment in four MELAS patients showed increased levels of ccf-mtDNA in relation to acute events (stroke-like episodes/status epilepticus) or progression of neurodegeneration. Our results confirm the association of FGF21 and GDF-15 with mitochondrial translation defects due to tRNA mutations. Most notably, the novel ccf-mtDNA was strongly associated with MELAS and may be used for monitoring the disease course or to evaluate the efficacy of therapies, especially in the acute phase. KEY MESSAGES: • FGF21/GDF15 efficiently identifies mitochondrial diseases due to mutations in tRNA genes. • The novel ccf-mtDNA is associated with MELAS and increases during acute events. • Creatine only discriminates severe mitochondrial patients. • FGF21, GDF-15, and ccf-mtDNA are possibly useful for monitoring therapy efficacy.
Collapse
Affiliation(s)
- Alessandra Maresca
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Valentina Del Dotto
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Martina Romagnoli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Chiara La Morgia
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Lidia Di Vito
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Mariantonietta Capristo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Maria Lucia Valentino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
40
|
Liu D, Zuo X, Luo H, Zhu H. The altered metabolism profile in pathogenesis of idiopathic inflammatory myopathies. Semin Arthritis Rheum 2020; 50:627-635. [PMID: 32502727 DOI: 10.1016/j.semarthrit.2020.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/28/2020] [Accepted: 05/11/2020] [Indexed: 11/29/2022]
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of heterogeneous autoimmune diseases characterized by muscle weakness, muscle inflammation and extramuscular manifestations. Despite extensive efforts, the mechanisms of IIMs remain largely unknown, and treatment is still a challenge for physicians. Metabolism changes have emerged as a crucial player in autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). However, little is known about metabolism changes in IIMs. In this review, we focus on the alteration of metabolism profile in IIMs, and the relationships with clinical information. We highlight the potential roles of metabolism in the pathogenesis of IIMs and discuss future perspectives for metabolic checkpoint-based therapeutic interventions.
Collapse
Affiliation(s)
- Di Liu
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Hui Luo
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Honglin Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.
| |
Collapse
|
41
|
Timper K, Del Río-Martín A, Cremer AL, Bremser S, Alber J, Giavalisco P, Varela L, Heilinger C, Nolte H, Trifunovic A, Horvath TL, Kloppenburg P, Backes H, Brüning JC. GLP-1 Receptor Signaling in Astrocytes Regulates Fatty Acid Oxidation, Mitochondrial Integrity, and Function. Cell Metab 2020; 31:1189-1205.e13. [PMID: 32433922 PMCID: PMC7272126 DOI: 10.1016/j.cmet.2020.05.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 12/09/2019] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
Astrocytes represent central regulators of brain glucose metabolism and neuronal function. They have recently been shown to adapt their function in response to alterations in nutritional state through responding to the energy state-sensing hormones leptin and insulin. Here, we demonstrate that glucagon-like peptide (GLP)-1 inhibits glucose uptake and promotes β-oxidation in cultured astrocytes. Conversely, postnatal GLP-1 receptor (GLP-1R) deletion in glial fibrillary acidic protein (GFAP)-expressing astrocytes impairs astrocyte mitochondrial integrity and activates an integrated stress response with enhanced fibroblast growth factor (FGF)21 production and increased brain glucose uptake. Accordingly, central neutralization of FGF21 or astrocyte-specific FGF21 inactivation abrogates the improvements in glucose tolerance and learning in mice lacking GLP-1R expression in astrocytes. Collectively, these experiments reveal a role for astrocyte GLP-1R signaling in maintaining mitochondrial integrity, and lack of GLP-1R signaling mounts an adaptive stress response resulting in an improvement of systemic glucose homeostasis and memory formation.
Collapse
Affiliation(s)
- Katharina Timper
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Almudena Del Río-Martín
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Anna Lena Cremer
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany
| | - Stephan Bremser
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; Institute for Zoology, Biocenter, University of Cologne, Zuelpicher Str. 47B, 50674 Cologne, Germany
| | - Jens Alber
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Patrick Giavalisco
- Max Planck Institute for Biology of Aging, Joseph-Stelzmann-Str. 9b, 50931 Cologne, Germany
| | - Luis Varela
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Christian Heilinger
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Hendrik Nolte
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Aleksandra Trifunovic
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Tamas L Horvath
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anatomy and Histology, University of Veterinary Medicine, 1078 Budapest, Hungary
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; Institute for Zoology, Biocenter, University of Cologne, Zuelpicher Str. 47B, 50674 Cologne, Germany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
42
|
Emmerzaal TL, Preston G, Geenen B, Verweij V, Wiesmann M, Vasileiou E, Grüter F, de Groot C, Schoorl J, de Veer R, Roelofs M, Arts M, Hendriksen Y, Klimars E, Donti TR, Graham BH, Morava E, Rodenburg RJ, Kozicz T. Impaired mitochondrial complex I function as a candidate driver in the biological stress response and a concomitant stress-induced brain metabolic reprogramming in male mice. Transl Psychiatry 2020; 10:176. [PMID: 32488052 PMCID: PMC7266820 DOI: 10.1038/s41398-020-0858-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/05/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022] Open
Abstract
Mitochondria play a critical role in bioenergetics, enabling stress adaptation, and therefore, are central in biological stress responses and stress-related complex psychopathologies. To investigate the effect of mitochondrial dysfunction on the stress response and the impact on various biological domains linked to the pathobiology of depression, a novel mouse model was created. These mice harbor a gene trap in the first intron of the Ndufs4 gene (Ndufs4GT/GT mice), encoding the NDUFS4 protein, a structural component of complex I (CI), the first enzyme of the mitochondrial electron transport chain. We performed a comprehensive behavioral screening with a broad range of behavioral, physiological, and endocrine markers, high-resolution ex vivo brain imaging, brain immunohistochemistry, and multi-platform targeted mass spectrometry-based metabolomics. Ndufs4GT/GT mice presented with a 25% reduction of CI activity in the hippocampus, resulting in a relatively mild phenotype of reduced body weight, increased physical activity, decreased neurogenesis and neuroinflammation compared to WT littermates. Brain metabolite profiling revealed characteristic biosignatures discriminating Ndufs4GT/GT from WT mice. Specifically, we observed a reversed TCA cycle flux and rewiring of amino acid metabolism in the prefrontal cortex. Next, exposing mice to chronic variable stress (a model for depression-like behavior), we found that Ndufs4GT/GT mice showed altered stress response and coping strategies with a robust stress-associated reprogramming of amino acid metabolism. Our data suggest that impaired mitochondrial CI function is a candidate driver for altered stress reactivity and stress-induced brain metabolic reprogramming. These changes result in unique phenomic and metabolomic signatures distinguishing groups based on their mitochondrial genotype.
Collapse
Affiliation(s)
- Tim L Emmerzaal
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Graeme Preston
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
- Hayward Genetics Center, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Bram Geenen
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Vivienne Verweij
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Maximilian Wiesmann
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Elisavet Vasileiou
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Femke Grüter
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Corné de Groot
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Jeroen Schoorl
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Renske de Veer
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Monica Roelofs
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Martijn Arts
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Yara Hendriksen
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | - Eva Klimars
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands
| | | | - Brett H Graham
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard J Rodenburg
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tamas Kozicz
- Department of Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, The Netherlands.
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
43
|
Advances in the diagnosis of inherited neuromuscular diseases and implications for therapy development. Lancet Neurol 2020; 19:522-532. [PMID: 32470424 DOI: 10.1016/s1474-4422(20)30028-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/19/2022]
Abstract
Advances in DNA sequencing technologies have resulted in a near doubling, in under 10 years, of the number of causal genes identified for inherited neuromuscular disorders. However, around half of patients, whether children or adults, do not receive a molecular diagnosis after initial diagnostic workup. Massively parallel technologies targeting RNA, proteins, and metabolites are being increasingly used to diagnose these unsolved cases. The use of these technologies to delineate pathways, biomarkers, and therapeutic targets has led to new approaches entering the drug development pipeline. However, these technologies might give rise to misleading conclusions if used in isolation, and traditional techniques including comprehensive neurological evaluation, histopathology, and biochemistry continue to have a crucial role in diagnostics. For optimal diagnosis, prognosis, and precision medicine, no single ruling technology exists. Instead, an interdisciplinary approach combining novel and traditional neurological techniques with computer-aided analysis and international data sharing is needed to advance the diagnosis and treatment of neuromuscular disorders.
Collapse
|
44
|
Abnormalities of hydrogen sulfide and glutathione pathways in mitochondrial dysfunction. J Adv Res 2020; 27:79-84. [PMID: 33318868 PMCID: PMC7728579 DOI: 10.1016/j.jare.2020.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/17/2022] Open
Abstract
Background Mitochondrial disorders are genetic diseases for which therapy remains woefully inadequate. Therapy of these disorders is particularly challenging partially due to the heterogeneity and tissue-specificity of pathomechanisms involved in these disorders. Abnormalities in hydrogen sulfide (H2S) metabolism are emerging as novel mechanism in mitochondrial dysfunction. However, further studies are necessary to understand the effects, protective or detrimental, of these abnormalities, and their relevance, in mitochondrial diseases. Aim of Review To review the recent evidences of derangement of the metabolism of H2S, at biosynthesis or oxidation levels, in mitochondrial dysfunction, focusing specifically on the alterations of H2S oxidation caused by primary Coenzyme Q (CoQ) deficiency. Key Scientific Concepts of Review Mitochondria play a key role in the regulation of H2S and GSH metabolism pathways. However, further studies are needed to understand the consequences of abnormalities of H2S and GSH synthesis on the oxidation pathway, and vice versa; and on the levels of H2S and GSH, their tissue-specific detrimental effects, and their role the role in mitochondrial diseases. Beside the known H2S pathways, additional, tissue-specific, enzymatic systems, involved in H2S production and elimination, might exist.
Collapse
|
45
|
Steele H, Gomez‐Duran A, Pyle A, Hopton S, Newman J, Stefanetti RJ, Charman SJ, Parikh JD, He L, Viscomi C, Jakovljevic DG, Hollingsworth KG, Robinson AJ, Taylor RW, Bottolo L, Horvath R, Chinnery PF. Metabolic effects of bezafibrate in mitochondrial disease. EMBO Mol Med 2020; 12:e11589. [PMID: 32107855 PMCID: PMC7059007 DOI: 10.15252/emmm.201911589] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial disorders affect 1/5,000 and have no cure. Inducing mitochondrial biogenesis with bezafibrate improves mitochondrial function in animal models, but there are no comparable human studies. We performed an open-label observational experimental medicine study of six patients with mitochondrial myopathy caused by the m.3243A>G MTTL1 mutation. Our primary aim was to determine the effects of bezafibrate on mitochondrial metabolism, whilst providing preliminary evidence of safety and efficacy using biomarkers. The participants received 600-1,200 mg bezafibrate daily for 12 weeks. There were no clinically significant adverse events, and liver function was not affected. We detected a reduction in the number of complex IV-immunodeficient muscle fibres and improved cardiac function. However, this was accompanied by an increase in serum biomarkers of mitochondrial disease, including fibroblast growth factor 21 (FGF-21), growth and differentiation factor 15 (GDF-15), plus dysregulation of fatty acid and amino acid metabolism. Thus, although potentially beneficial in short term, inducing mitochondrial biogenesis with bezafibrate altered the metabolomic signature of mitochondrial disease, raising concerns about long-term sequelae.
Collapse
Affiliation(s)
- Hannah Steele
- Institute of Genetic MedicineNewcastle UniversityNewcastle upon TyneUK
| | - Aurora Gomez‐Duran
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUK
- MRC Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
| | - Angela Pyle
- Institute of Genetic MedicineNewcastle UniversityNewcastle upon TyneUK
- Wellcome Centre for Mitochondrial ResearchNewcastle UniversityNewcastle upon TyneUK
| | - Sila Hopton
- Wellcome Centre for Mitochondrial ResearchNewcastle UniversityNewcastle upon TyneUK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders of Adults and ChildrenNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Jane Newman
- Wellcome Centre for Mitochondrial ResearchNewcastle UniversityNewcastle upon TyneUK
- Institute of NeuroscienceNewcastle UniversityNewcastle upon TyneUK
| | | | - Sarah J Charman
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - Jehill D Parikh
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUK
| | - Langping He
- Wellcome Centre for Mitochondrial ResearchNewcastle UniversityNewcastle upon TyneUK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders of Adults and ChildrenNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Carlo Viscomi
- MRC Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
| | | | | | - Alan J Robinson
- MRC Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial ResearchNewcastle UniversityNewcastle upon TyneUK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders of Adults and ChildrenNewcastle upon Tyne Hospitals NHS Foundation TrustNewcastle upon TyneUK
- Institute of NeuroscienceNewcastle UniversityNewcastle upon TyneUK
| | - Leonardo Bottolo
- Department of Medical GeneticsUniversity of CambridgeCambridgeUK
- The Alan Turing InstituteLondonUK
- MRC Biostatistics UnitUniversity of CambridgeCambridgeUK
| | - Rita Horvath
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUK
| | - Patrick F Chinnery
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUK
- MRC Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
| |
Collapse
|
46
|
Khan S, Ince-Dunn G, Suomalainen A, Elo LL. Integrative omics approaches provide biological and clinical insights: examples from mitochondrial diseases. J Clin Invest 2020; 130:20-28. [PMID: 31895050 PMCID: PMC6934214 DOI: 10.1172/jci129202] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
High-throughput technologies for genomics, transcriptomics, proteomics, and metabolomics, and integrative analysis of these data, enable new, systems-level insights into disease pathogenesis. Mitochondrial diseases are an excellent target for hypothesis-generating omics approaches, as the disease group is mechanistically exceptionally complex. Although the genetic background in mitochondrial diseases is in either the nuclear or the mitochondrial genome, the typical downstream effect is dysfunction of the mitochondrial respiratory chain. However, the clinical manifestations show unprecedented variability, including either systemic or tissue-specific effects across multiple organ systems, with mild to severe symptoms, and occurring at any age. So far, the omics approaches have provided mechanistic understanding of tissue-specificity and potential treatment options for mitochondrial diseases, such as metabolome remodeling. However, no curative treatments exist, suggesting that novel approaches are needed. In this Review, we discuss omics approaches and discoveries with the potential to elucidate mechanisms of and therapies for mitochondrial diseases.
Collapse
Affiliation(s)
- Sofia Khan
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Gulayse Ince-Dunn
- Research Programs Unit, Stem Cells and Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu Suomalainen
- Research Programs Unit, Stem Cells and Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Neuroscience Center, HiLife, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, HUSlab, Helsinki, Finland
| | - Laura L. Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
47
|
Whitehall JC, Greaves LC. Aberrant mitochondrial function in ageing and cancer. Biogerontology 2019; 21:445-459. [PMID: 31802313 PMCID: PMC7347693 DOI: 10.1007/s10522-019-09853-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/23/2019] [Indexed: 12/12/2022]
Abstract
Alterations in mitochondrial metabolism have been described as one of the major hallmarks of both ageing cells and cancer. Age is the biggest risk factor for the development of a significant number of cancer types and this therefore raises the question of whether there is a link between age-related mitochondrial dysfunction and the advantageous changes in mitochondrial metabolism prevalent in cancer cells. A common underlying feature of both ageing and cancer cells is the presence of somatic mutations of the mitochondrial genome (mtDNA) which we postulate may drive compensatory alterations in mitochondrial metabolism that are advantageous for tumour growth. In this review, we discuss basic mitochondrial functions, mechanisms of mtDNA mutagenesis and their metabolic consequences, and review the evidence for and against a role for mtDNA mutations in cancer development.
Collapse
Affiliation(s)
- Julia C Whitehall
- The Medical School, Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Laura C Greaves
- The Medical School, Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
48
|
Chang Y, Zhang W, Chen K, Wang Z, Xia S, Li H. Metabonomics window into plateau hypoxia. J Int Med Res 2019; 47:5441-5452. [PMID: 31594434 PMCID: PMC6862876 DOI: 10.1177/0300060519879323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
Oxygen deficiency in the plateau environment weakens aerobic metabolism and reduces the energy supply, leading to high-altitude diseases including decreased circulatory function, decreased nutrient and energy supply to tissues and organs, and decreased waste discharge. The involvement of many metabolic pathways is reflected in dramatic changes in levels of endogenous small molecule metabolites. Metabolomics represents a promising technique for mechanistic studies and drug screening, and metabonomics, or quantitative metabolomics, has been increasingly applied to the study of hypoxic diseases and their pathogenesis, as well as to pharmacodynamics at high altitudes. In this article, we review the recent literature on the pathogenesis of altitude hypoxia and the clinical and preclinical metabonomics of drug interventions. Endogenous metabolites and metabolic pathways change significantly under high-altitude hypoxia. Some drug interventions have also been shown to regulate pathway metabolism, and the problems of applying metabonomics to hypoxic diseases at high altitude and the prospects for its future application are summarized.
Collapse
Affiliation(s)
- Yue Chang
- Department of Hepatopancreatobiliary and Splenic Medicine, Characteristic Medical Center of People’s Armed Police Force, Tianjin, China
- Tianjin Key Laboratory of Hepatopancreatic Fibrosis and Molecular Diagnosis and Treatment, Tianjin, China
| | - Wen Zhang
- Department of Hepatopancreatobiliary and Splenic Medicine, Characteristic Medical Center of People’s Armed Police Force, Tianjin, China
- Tianjin Key Laboratory of Hepatopancreatic Fibrosis and Molecular Diagnosis and Treatment, Tianjin, China
| | - Kai Chen
- Department of Hepatopancreatobiliary and Splenic Medicine, Characteristic Medical Center of People’s Armed Police Force, Tianjin, China
- Tianjin Key Laboratory of Hepatopancreatic Fibrosis and Molecular Diagnosis and Treatment, Tianjin, China
| | - Zhenguo Wang
- Department of Hepatopancreatobiliary and Splenic Medicine, Characteristic Medical Center of People’s Armed Police Force, Tianjin, China
| | - Shihai Xia
- Department of Hepatopancreatobiliary and Splenic Medicine, Characteristic Medical Center of People’s Armed Police Force, Tianjin, China
- Tianjin Key Laboratory of Hepatopancreatic Fibrosis and Molecular Diagnosis and Treatment, Tianjin, China
| | - Hai Li
- Tianjin Key Laboratory of Hepatopancreatic Fibrosis and Molecular Diagnosis and Treatment, Tianjin, China
- Division of Gastroenterology and Hepatology, Tianjin Xiqing Hospital, Tianjin, China
| |
Collapse
|
49
|
Anzmann AF, Pinto S, Busa V, Carlson J, McRitchie S, Sumner S, Pandey A, Vernon HJ. Multi-omics studies in cellular models of methylmalonic acidemia and propionic acidemia reveal dysregulation of serine metabolism. Biochim Biophys Acta Mol Basis Dis 2019; 1865:165538. [PMID: 31449969 DOI: 10.1016/j.bbadis.2019.165538] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/06/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Methylmalonic acidemia (MMA) and propionic acidemia (PA) are related disorders of mitochondrial propionate metabolism, caused by defects in methylmalonyl-CoA mutase (MUT) and propionyl-CoA carboxylase (PCC), respectively. These biochemical defects lead to a complex cascade of downstream metabolic abnormalities, and identification of these abnormal pathways has important implications for understanding disease pathophysiology. Using a multi-omics approach in cellular models of MMA and PA, we identified serine and thiol metabolism as important areas of metabolic dysregulation. METHODS We performed global proteomic analysis of fibroblasts and untargeted metabolomics analysis of plasma from individuals with MMA to identify novel pathways of dysfunction. We probed these novel pathways in CRISPR-edited, MUT and PCCA null HEK293 cell lines via targeted metabolomics, gene expression analysis, and flux metabolomics tracing utilization of 13C-glucose. RESULTS Proteomic analysis of fibroblasts identified upregulation of multiple proteins involved in serine synthesis and thiol metabolism including: phosphoserine amino transferase (PSAT1), cystathionine beta synthase (CBS), and mercaptopyruvate sulfurtransferase (MPST). Metabolomics analysis of plasma revealed significantly increased levels of cystathionine and glutathione, central metabolites in thiol metabolism. CRISPR-edited MUT and PCCA HEK293 cells recapitulate primary defects of MMA and PA and have upregulation of transcripts associated with serine and thiol metabolism including PSAT1. 13C-glucose flux metabolomics in MUT and PCCA null HEK293 cells identified increases in serine de novo biosynthesis, serine transport, and abnormal downstream TCA cycle utilization. CONCLUSION We identified abnormal serine metabolism as a novel area of cellular dysfunction in MMA and PA, thus introducing a potential new target for therapeutic investigation.
Collapse
Affiliation(s)
- Arianna Franca Anzmann
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Sneha Pinto
- Institute of Bioinformatics, Bengalaru, India; Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Veronica Busa
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - James Carlson
- LECO Corporation, St. Joseph, MI, United States of America; RTI International, Research Triangle Park, NC, USA
| | - Susan McRitchie
- RTI International, Research Triangle Park, NC, USA; University of North Carolina at Chapel Hill, Nutrition Research Institute, Eastern Regional Comprehensive Metabolomics Resource Core, University of North Carolina at Chapel Hill, United States of America
| | - Susan Sumner
- RTI International, Research Triangle Park, NC, USA; University of North Carolina at Chapel Hill, Nutrition Research Institute, Eastern Regional Comprehensive Metabolomics Resource Core, University of North Carolina at Chapel Hill, United States of America
| | - Akhilesh Pandey
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States of America
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, United States of America.
| |
Collapse
|
50
|
De Paepe B. Sporadic Inclusion Body Myositis: An Acquired Mitochondrial Disease with Extras. Biomolecules 2019; 9:biom9010015. [PMID: 30621041 PMCID: PMC6359202 DOI: 10.3390/biom9010015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022] Open
Abstract
The sporadic form of inclusion body myositis (IBM) is the most common late-onset myopathy. Its complex pathogenesis includes degenerative, inflammatory and mitochondrial aspects. However, which of those mechanisms are cause and which effect, as well as their interrelations, remain partly obscured to this day. In this review the nature of the mitochondrial dysregulation in IBM muscle is explored and comparison is made with other muscle disorders. Mitochondrial alterations in IBM are evidenced by histological and serum biomarkers. Muscular mitochondrial dynamics is disturbed, with deregulated organelle fusion leading to subsequent morphological alterations and muscle displays abnormal mitophagy. The tissue increases mitochondrial content in an attempt to compensate dysfunction, yet mitochondrial DNA (mtDNA) alterations and mild mtDNA depletion are also present. Oxidative phosphorylation defects have repeatedly been shown, most notably a reduction in complex IV activities and levels of mitokines and regulatory RNAs are perturbed. Based on the cumulating evidence of mitochondrial abnormality as a disease contributor, it is therefore warranted to regard IBM as a mitochondrial disease, offering a feasible therapeutic target to be developed for this yet untreatable condition.
Collapse
Affiliation(s)
- Boel De Paepe
- Neuromuscular Reference Centre, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| |
Collapse
|