1
|
Antico O, Thompson PW, Hertz NT, Muqit MMK, Parton LE. Targeting mitophagy in neurodegenerative diseases. Nat Rev Drug Discov 2025; 24:276-299. [PMID: 39809929 DOI: 10.1038/s41573-024-01105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Mitochondrial dysfunction is a hallmark of idiopathic neurodegenerative diseases, including Parkinson disease, amyotrophic lateral sclerosis, Alzheimer disease and Huntington disease. Familial forms of Parkinson disease and amyotrophic lateral sclerosis are often characterized by mutations in genes associated with mitophagy deficits. Therefore, enhancing the mitophagy pathway may represent a novel therapeutic approach to targeting an underlying pathogenic cause of neurodegenerative diseases, with the potential to deliver neuroprotection and disease modification, which is an important unmet need. Accumulating genetic, molecular and preclinical model-based evidence now supports targeting mitophagy in neurodegenerative diseases. Despite clinical development challenges, small-molecule-based approaches for selective mitophagy enhancement - namely, USP30 inhibitors and PINK1 activators - are entering phase I clinical trials for the first time.
Collapse
Affiliation(s)
- Odetta Antico
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Paul W Thompson
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK
| | | | - Miratul M K Muqit
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura E Parton
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
2
|
Chakraborty K, Burman R, Nisar S, Miller S, Loschinskey Z, Wu S, Li Y, Bag AK, Khan A, Goodenough C, Wilson N, Haris M, McCormack SE, Reddy R, Ness K, Finkel R, Bagga P. Reliability of In Vivo Creatine-Weighted Chemical Exchange Saturation Transfer (CrCEST) MRI in Calf Skeletal Muscle of Healthy Volunteers at 3 T. J Magn Reson Imaging 2025; 61:1681-1692. [PMID: 39212126 PMCID: PMC11868465 DOI: 10.1002/jmri.29566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Skeletal muscle mitochondrial oxidative phosphorylation (mtOXPHOS) is important for ATP generation and its dysfunction leads to exercise intolerance. Phosphorus magnetic resonance spectroscopy (31P-MRS) is a useful, noninvasive technique for mtOXPHOS assessment but has limitations. Creatine-weighted chemical exchange saturation transfer (CrCEST) MRI is a potential alternative to assess muscle bioenergetics. PURPOSE To evaluate the interscan repeatability, intra- and interobserver reproducibility of CrCEST during mild plantar flexion exercise. STUDY TYPE Retrospective. SUBJECTS Twenty healthy volunteers (age 37.6 ± 12.4 years, 11 females). FIELD STRENGTH/SEQUENCE 3 T/CEST imaging using gradient echo readout. ASSESSMENT τCrCEST (postexercise Cr recovery time) was assessed in two scans for each participant, following mild plantar flexion exercises targeting the medial gastrocnemius (MG), lateral gastrocnemius (LG), and soleus (Sol) muscles. Three observers measured τCrCEST for interobserver reproducibility. Three readings by one observer were used to measure intraobserver reproducibility. Two scans were used for within-participant interscan repeatability. STATISTICAL TESTS Paired t tests, intraclass correlation coefficient (ICC), and Pearson correlation were conducted. Bland-Altman plots were used to analyze the interobserver variability. A P-value of 0.05 was considered statistically significant. RESULTS There was excellent intra- (ICC ∈ 0.94 - 0.98 ) and interobserver (ICC ∈ 0.9 - 0.98 ) reproducibility, with moderate interscan repeatability for τCrCEST in LG and MG (ICC ∈ 0.54 - 0.74 ) and poor-to-moderate interscan repeatability in Sol (ICC ∈ 0.24 - 0.53 ). Excellent interobserver reproducibility was confirmed by Bland-Altman plots (fixed bias P-value ∈ 0.08 - 0.87 ). DATA CONCLUSION CrCEST MRI shows promise in assessing muscle bioenergetics by evaluating τCrCEST during mild plantar flexion exercise with reasonable reliability, particularly in LG and MG. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Kasturee Chakraborty
- Department of Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ritambhar Burman
- Department of Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sabah Nisar
- Department of Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Saorla Miller
- Department of Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Zachary Loschinskey
- Department of Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Shengjie Wu
- Department of Biostatistics, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yimei Li
- Department of Biostatistics, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Asim K Bag
- Department of Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ayaz Khan
- Department of Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Chelsea Goodenough
- Department of Epidemiology & Cancer Control, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Neil Wilson
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mohammad Haris
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shana E. McCormack
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kirsten Ness
- Department of Epidemiology & Cancer Control, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Richard Finkel
- Department of Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Puneet Bagga
- Department of Diagnostic Imaging, St Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
3
|
Lopez FV, O'Shea A, Huo Z, DeKosky ST, Trouard TP, Alexander GE, Woods AJ, Bowers D. Neurocognitive correlates of cerebral mitochondrial function and energy metabolism using phosphorus magnetic resonance spectroscopy in older adults. GeroScience 2025; 47:2223-2234. [PMID: 39477865 PMCID: PMC11978590 DOI: 10.1007/s11357-024-01403-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/15/2024] [Indexed: 04/09/2025] Open
Abstract
The goal of the current study was to learn about the role of cerebral mitochondrial function on cognition. Based on established cognitive neuroscience, clinical neuropsychology, and cognitive aging literature, we hypothesized mitochondrial function within a focal brain region would map onto cognitive behaviors linked to that brain region. To test this hypothesis, we used phosphorous (31P) magnetic resonance spectroscopy (MRS) to derive indirect markers of mitochondrial function and energy metabolism across two regions of the brain (bifrontal, left temporal). We administered cognitive tasks sensitive to frontal-executive or temporal-hippocampal systems to a sample of 70 cognitively unimpaired older adults with subjective memory complaints and a first-degree family history of Alzheimer's disease and predicted better executive function and recent memory performance would be related to greater frontal and temporal 31P MRS indirect markers, respectively. Results of separate hierarchical linear regressions indicated better recent memory scores were related to 31P MRS indirect markers of lower static energy and higher energy reserve within the left temporal voxel; these findings were associated with moderate effect sizes. Contrary to predictions, executive function performance was unrelated to 31P MRS indirect markers within the bilateral frontal voxel, which may reflect a combination of theoretical and/or methodological issues. Findings represent a snapshot of the relationship between cognition and 31P MRS indirect markers of mitochondrial function, providing potential avenues for future work investigating mitochondrial underpinnings of cognition. 31P MRS may provide a sensitive neuroimaging marker for differences in aspects of memory among persons at-risk for mild cognitive impairment or dementia.
Collapse
Affiliation(s)
- Francesca V Lopez
- Department of Clinical and Health Psychology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Andrew O'Shea
- Center for Cognitive Aging and Memory, Evelyn F. McKnight Brain Institute, University of Florida, Gainesville, Gainesville, FL, USA
| | - Zhiguang Huo
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Steven T DeKosky
- Department of Neurology and Fixel Center for Neurological Diseases, College of Medicine, University of Florida and Evelyn F. McKnight Brain Institute, Gainesville, FL, USA
| | - Theodore P Trouard
- Department of Biomedical Engineering, College of Engineering, and Evelyn F. McKnight Brain Institute, University of Arizona and Alzheimer's Disease Consortium, Tucson, AZ, USA
| | - Gene E Alexander
- Department of Psychology and Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
- Department of Psychiatry, Neuroscience and Physiological Sciences Graduate Interdisciplinary Programs, and BIO5 Institute, University of Arizona and Arizona Alzheimer's Disease Consortium, Tucson, AZ, USA
| | - Adam J Woods
- Department of Clinical and Health Psychology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Cognitive Aging and Memory, Evelyn F. McKnight Brain Institute, University of Florida, Gainesville, Gainesville, FL, USA
| | - Dawn Bowers
- Department of Clinical and Health Psychology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neurology, Fixel Center of Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
4
|
Sassani M, Ghafari T, Arachchige PRW, Idrees I, Gao Y, Waitt A, Weaver SRC, Mazaheri A, Lyons HS, Grech O, Thaller M, Witton C, Bagshaw AP, Wilson M, Park H, Brookes M, Novak J, Mollan SP, Hill LJ, Lucas SJE, Mitchell JL, Sinclair AJ, Mullinger K, Fernández-Espejo D. Current and prospective roles of magnetic resonance imaging in mild traumatic brain injury. Brain Commun 2025; 7:fcaf120. [PMID: 40241788 PMCID: PMC12001801 DOI: 10.1093/braincomms/fcaf120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 11/26/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
There is unmet clinical need for biomarkers to predict recovery or the development of long-term sequelae of mild traumatic brain injury, a highly prevalent condition causing a constellation of disabling symptoms. A substantial proportion of patients live with long-lasting sequelae affecting their quality of life and ability to work. At present, symptoms can be assessed through clinical tests; however, there are no imaging or laboratory tests fully reflective of pathophysiology routinely used by clinicians to characterize post-concussive symptoms. Magnetic resonance imaging has potential to link subtle pathophysiological alterations to clinical outcomes. Here, we review the state of the art of MRI research in adults with mild traumatic brain injury and provide recommendations to facilitate transition into clinical practice. Studies utilizing MRI can inform on pathophysiology of mild traumatic brain injury. They suggest presence of early cytotoxic and vasogenic oedema. They also show that mild traumatic brain injury results in cellular injury and microbleeds affecting the integrity of myelin and white matter tracts, all processes that appear to induce delayed vascular reactions and functional changes. Crucially, correlates between MRI parameters and post-concussive symptoms are emerging. Clinical sequences such as T1-weighted MRI, susceptibility-weighted MRI or fluid attenuation inversion recovery could be easily implementable in clinical practice, but are not sufficient, in isolation for prognostication. Diffusion sequences have shown promises and, although in need of analysis standardization, are a research priority. Lastly, arterial spin labelling is emerging as a high-utility research as it could become useful to assess delayed neurovascular response and possible long-term symptoms.
Collapse
Affiliation(s)
- Matilde Sassani
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Tara Ghafari
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Pradeepa R W Arachchige
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Iman Idrees
- College of Health and Life Sciences, Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK
| | - Yidian Gao
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Alice Waitt
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
- College of Health and Life Sciences, Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK
| | - Samuel R C Weaver
- Centre for Human Brain Health and School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ali Mazaheri
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Hannah S Lyons
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Olivia Grech
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - Mark Thaller
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Caroline Witton
- College of Health and Life Sciences, Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK
| | - Andrew P Bagshaw
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Martin Wilson
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Hyojin Park
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Matthew Brookes
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jan Novak
- College of Health and Life Sciences, Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK
| | - Susan P Mollan
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Birmingham Neuro-ophthalmology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust Birmingham, Birmingham B15 2WB, UK
| | - Lisa J Hill
- Department of Biomedical Sciences, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Samuel J E Lucas
- Centre for Human Brain Health and School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - James L Mitchell
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Alexandra J Sinclair
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Karen Mullinger
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Davinia Fernández-Espejo
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
5
|
Nibrad D, Shiwal A, Tadas M, Katariya R, Kale M, Kotagale N, Umekar M, Taksande B. Therapeutic modulation of mitochondrial dynamics by agmatine in neurodegenerative disorders. Neuroscience 2025; 569:43-57. [PMID: 39890051 DOI: 10.1016/j.neuroscience.2025.01.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Mitochondrial dysfunction is a pivotal factor in the pathogenesis of neurodegenerative disorders, driving neuronal degeneration through mechanisms involving oxidative stress, impaired energy production, and dysregulated calcium homeostasis. Agmatine, an endogenous polyamine derived from arginine, has garnered attention for its neuroprotective properties, including anti-inflammatory, anti-oxidative, and antiapoptotic effects. Recent studies have highlighted the potential of agmatine in preserving mitochondrial function and mitigating neurodegeneration, making it a promising candidate for therapeutic intervention. One of the key mechanisms by which agmatine exerts its neuroprotective effects is through the maintenance of mitochondrial homeostasis. Agmatine has been shown to modulate mitochondrial dynamics, promoting mitochondrial fusion and fission balance essential for cellular energy metabolism and signaling. Moreover, agmatine acts as a regulator of mitochondrial permeability transition pore (mPTP) opening, preventing excessive calcium influx and subsequent mitochondrial dysfunction. Despite promising findings, challenges such as optimizing agmatine's pharmacokinetics, determining optimal dosing regimens, and elucidating its precise molecular targets within mitochondria remain to be addressed. Future research directions should focus on developing targeted delivery systems for agmatine, investigating its interactions with mitochondrial proteins, and conducting well-designed clinical trials to evaluate its therapeutic efficacy and safety profile in neurodegenerative disorders. Overall, agmatine emerges as a novel therapeutic agent with the potential to modulate mitochondrial homeostasis and alleviate neurodegenerative pathology, offering new avenues for treating these debilitating conditions.
Collapse
Affiliation(s)
- Dhanshree Nibrad
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Amit Shiwal
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Manasi Tadas
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Raj Katariya
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Mayur Kale
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy, Kathora Naka, VMV Road, Amravati, (M.S.) 444604, India
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India.
| |
Collapse
|
6
|
Jellinger KA. Mild cognitive impairment in amyotrophic lateral sclerosis: current view. J Neural Transm (Vienna) 2025; 132:357-368. [PMID: 39470847 DOI: 10.1007/s00702-024-02850-7] [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: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal multi-system neurodegenerative disorder with no effective treatment or cure. Although primarily characterized by motor degeneration, cognitive dysfunction is an important non-motor symptom that has a negative impact on patient and caregiver burden. Mild cognitive deficits are present in a subgroup of non-demented patients with ALS, often preceding motor symptoms. Detailed neuropsychological assessments reveal deficits in a variety of cognitive domains, including those of verbal fluency and retrieval, language, executive function, attention and verbal memory. Mild cognitive impairment (MCI), a risk factor for developing dementia, affects between 10% and over 50% of ALS patients. Neuroimaging revealed atrophy of frontal and temporal cortices, disordered white matter Integrity, volume reduction in amygdala and thalamus, hypometabolism in the frontal and superior temporal gyrus and anterior insula. Neuronal loss in non-motor brain areas, associated with TDP-43 deposition, one of the morphological hallmarks of ALS, is linked to functional disruption of frontostriatal and frontotemporo-limbic connectivities as markers for cognitive deficits in ALS, the pathogenesis of which is still poorly understood. Early diagnosis by increased cerebrospinal fluid or serum levels of neurofilament light/heavy chain or glial fibrillary acidic protein awaits confirmation for MCI in ALS. These fluid biomarkers and early detection of brain connectivity signatures before structural changes will be helpful not only in establishing early premature diagnosis but also in clarifying the pathophysiological mechanisms of MCI in ALS, which might serve as novel targets for prohibition/delay and future adequate treatment of this debilitating disorder.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, Vienna, A-1150, Austria.
| |
Collapse
|
7
|
Jellinger KA. The spectrum of behavioral disorders in amyotrophic lateral sclerosis: current view. J Neural Transm (Vienna) 2025; 132:217-236. [PMID: 39402174 DOI: 10.1007/s00702-024-02841-8] [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: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 02/02/2025]
Abstract
Behavioral disorders, with an average prevalence of 30-60% are important non-motor symptoms in amyotrophic lateral sclerosis (ALS) that have a negative impact on prognosis, management and quality of life, yet the underlying neurobiology is poorly understood. Among people with ALS, apathy, fatigue, anxiety, irritability and other behavioral symptoms are the most prominent, although less frequent than cognitive impairment. The present review explores the current understanding of behavioral changes in ALS with particular emphasis on our current knowledge about their structural and functional brain correlates, substantiating a multisystem degeneration with particular dysfunction of frontal-subcortical circuits and dysfunction of fronto-striatal, frontotemporal and other essential brain systems. The natural history of behavioral dysfunctions in ALS and their relationship to frontotemporal lobe degeneration (FTLD) are not fully understood, although they form a clinical continuum, suggesting a differential vulnerability of non-motor brain networks, ALS being considered a brain network disorder. An assessment of risks or the early detection of brain connectivity signatures before structural changes may be helpful in investigating the pathophysiological mechanisms of behavioral impairment in ALS. Treatment of both ALS and co-morbid behavioral disorders is a multidisciplinary task, but whereas no causal or disease-modifying therapies for ALS are available, symptomatic treatment of a variety of behavioral symptoms plays a pivotal role in patient care, although the management of behavioral symptoms in clinical care still remains limited.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, Vienna, A-1150, Austria.
| |
Collapse
|
8
|
Jing Y, Haeger A, Boumezbeur F, Binkofski F, Reetz K, Romanzetti S. Neuroenergetic alterations in neurodegenerative diseases: A systematic review and meta-analysis of in vivo 31P-MRS studies. Ageing Res Rev 2024; 101:102488. [PMID: 39243891 DOI: 10.1016/j.arr.2024.102488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Phosphorus magnetic resonance spectroscopy (31P-MRS) is applied for non-invasive studies of neuroenergetic metabolism in neurodegenerative diseases. However, the findings are inconsistent and have not yet been tested in meta-analyses. To address this gap, we performed a systematic review of 29 studies and conducted meta-analyses for 9 studies on Alzheimer's disease (AD, n = 140 patients), 9 studies on Parkinson's disease (PD, n = 183 patients), 3 studies on Progressive Supranuclear Palsy (PSP, n = 42 patients), and 2 studies on Multiple System Atrophy (MSA, n = 24 patients). Compared to controls, AD patients had a higher ratio of phosphomonoesters/phosphodiesters (PME/PDE) in the frontal lobe (MD = 0.049, p = 0.0003); PD patients showed decreases in PME/PDE in the putamen (MD = -0.050, p = 0.023) and adenosine triphosphate/inorganic phosphate (ATP/Pi) in the midbrain (MD = -0.274, p = 0.002); PSP patients presented increased phosphocreatine (PCr)/Pi in the basal ganglia (MD = 0.556, p = 0.030) and adenosine diphosphate (ADP)/Pi in the occipital lobe (MD = 0.005, p = 0.009); no significant effects were observed in MSA. Here, our review underlines the importance of 31P-MRS in the characterization of distinct neuroenergetic changes and its potential to improve the diagnosis and follow-up of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yinghua Jing
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-Brain Institute Molecular Neuroscience and Neuroimaging (INM-11), Research Centre Jülich and RWTH Aachen University, Aachen, Germany
| | - Alexa Haeger
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-Brain Institute Molecular Neuroscience and Neuroimaging (INM-11), Research Centre Jülich and RWTH Aachen University, Aachen, Germany
| | - Fawzi Boumezbeur
- NeuroSpin, CEA, CNRS UMR9027, Paris-Saclay University, Gif-sur-Yvette, France
| | - Ferdinand Binkofski
- Division for Clinical Cognitive Sciences, Department of Neurology, University Hospital RWTH Aachen, Germany
| | - Kathrin Reetz
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-Brain Institute Molecular Neuroscience and Neuroimaging (INM-11), Research Centre Jülich and RWTH Aachen University, Aachen, Germany
| | - Sandro Romanzetti
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-Brain Institute Molecular Neuroscience and Neuroimaging (INM-11), Research Centre Jülich and RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
9
|
Sun H, Yang B, Li Q, Zhu X, Song E, Liu C, Song Y, Jiang G. Polystyrene nanoparticles trigger aberrant condensation of TDP-43 and amyotrophic lateral sclerosis-like symptoms. NATURE NANOTECHNOLOGY 2024; 19:1354-1365. [PMID: 38849544 DOI: 10.1038/s41565-024-01683-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 04/23/2024] [Indexed: 06/09/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the dysfunction and progressive death of cerebral and spinal motor neurons. Preliminary epidemiological research has hinted at a relationship between environmental risks and the escalation of ALS, but the underlying reasons remain mostly mysterious. Here we show that nanosize polystyrene plastics (PS) induce ALS-like symptoms and illustrate the related molecular mechanism. When exposed to PS, cells endure internal oxidative stress, which leads to the aggregation of TAR DNA-binding protein 43 kDa (TDP-43), triggering ALS-like characteristics. In addition, the oxidized heat shock protein 70 fails to escort TDP-43 back to the nucleus. The cytoplasmic accumulation of TDP-43 facilitates the formation of a complex between PS and TDP-43, enhancing the condensation and solidification of TDP-43. These findings are corroborated through in silico and in vivo assays. Altogether, our work illustrates a unique toxicological mechanism induced by nanoparticles and provides insights into the connection between environmental pollution and neurodegenerative disorders.
Collapse
Affiliation(s)
- Hang Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Bingwei Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Qiong Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Xiaokang Zhu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
10
|
Lopez FV, O'Shea A, Huo Z, DeKosky ST, Trouard TP, Alexander GE, Woods AJ, Bowers D. Frontal-temporal regional differences in brain energy metabolism and mitochondrial function using 31P MRS in older adults. GeroScience 2024; 46:3185-3195. [PMID: 38225480 PMCID: PMC11009166 DOI: 10.1007/s11357-023-01046-3] [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: 10/19/2023] [Accepted: 12/07/2023] [Indexed: 01/17/2024] Open
Abstract
Aging is a major risk for cognitive decline and transition to dementia. One well-known age-related change involves decreased brain efficiency and energy production, mediated in part by changes in mitochondrial function. Damaged or dysfunctional mitochondria have been implicated in the pathogenesis of age-related neurodegenerative conditions like Alzheimer's disease (AD). The aim of the current study was to investigate mitochondrial function over frontal and temporal regions in a sample of 70 cognitively normal older adults with subjective memory complaints and a first-degree family history of AD. We hypothesized cerebral mitochondrial function and energy metabolism would be greater in temporal as compared to frontal regions based on the high energy consumption in the temporal lobes (i.e., hippocampus). To test this hypothesis, we used phosphorous (31P) magnetic resonance spectroscopy (MRS) which is a non-invasive and powerful method for investigating in vivo mitochondrial function via high energy phosphates and phospholipid metabolism ratios. We used a single voxel method (left temporal and bilateral prefrontal) to achieve optimal sensitivity. Results of separate repeated measures analyses of variance showed 31P MRS ratios of static energy, energy reserve, energy consumption, energy demand, and phospholipid membrane metabolism were greater in the left temporal than bilateral prefrontal voxels. Our findings that all 31P MRS ratios were greater in temporal than bifrontal regions support our hypothesis. Future studies are needed to determine whether findings are related to cognition in older adults.
Collapse
Affiliation(s)
- Francesca V Lopez
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, PO Box 100165, Gainesville, FL, 32610, USA.
| | - Andrew O'Shea
- Center for Cognitive Aging and Memory, Evelyn F. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Zhiguang Huo
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Steven T DeKosky
- Department of Neurology, Fixel Center for Neurological Diseases, College of Medicine, and Evelyn F. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Theodore P Trouard
- Department of Biomedical Engineering, College of Engineering, and Evelyn F. McKnight Brain Institute, University of Arizona and Alzheimer's Disease Consortium, Tucson, AZ, USA
| | - Gene E Alexander
- Department of Psychology and Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
- Department of Psychiatry, Neuroscience and Physiological Sciences Graduate Interdisciplinary Programs, and BIO5 Institute, University of Arizona and Arizona Alzheimer's Disease Consortium, Tucson, AZ, USA
| | - Adam J Woods
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, PO Box 100165, Gainesville, FL, 32610, USA
- Center for Cognitive Aging and Memory, Evelyn F. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Dawn Bowers
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, PO Box 100165, Gainesville, FL, 32610, USA
- Department of Neurology, Fixel Center of Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
11
|
Evans LJ, O'Brien D, Shaw PJ. Current neuroprotective therapies and future prospects for motor neuron disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:327-384. [PMID: 38802178 DOI: 10.1016/bs.irn.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Four medications with neuroprotective disease-modifying effects are now in use for motor neuron disease (MND). With FDA approvals for tofersen, relyvrio and edaravone in just the past year, 2022 ended a quarter of a century when riluzole was the sole such drug to offer to patients. The acceleration of approvals may mean we are witnessing the beginning of a step-change in how MND can be treated. Improvements in understanding underlying disease biology has led to more therapies being developed to target specific and multiple disease mechanisms. Consideration for how the pipeline of new therapeutic agents coming through in clinical and preclinical development can be more effectively evaluated with biomarkers, advances in patient stratification and clinical trial design pave the way for more successful translation for this archetypal complex neurodegenerative disease. While it must be cautioned that only slowed rates of progression have so far been demonstrated, pre-empting rapid neurodegeneration by using neurofilament biomarkers to signal when to treat, as is currently being trialled with tofersen, may be more effective for patients with known genetic predisposition to MND. Early intervention with personalized medicines could mean that for some patients at least, in future we may be able to substantially treat what is considered by many to be one of the most distressing diseases in medicine.
Collapse
Affiliation(s)
- Laura J Evans
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - David O'Brien
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - Pamela J Shaw
- The Sheffield Institute for Translational Neuroscience, and the NIHR Sheffield Biomedical Research Centre, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
12
|
Sun W, Liu SH, Wei XJ, Sun H, Ma ZW, Yu XF. Potential of neuroimaging as a biomarker in amyotrophic lateral sclerosis: from structure to metabolism. J Neurol 2024; 271:2238-2257. [PMID: 38367047 DOI: 10.1007/s00415-024-12201-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 02/19/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by motor neuron degeneration. The development of ALS involves metabolite alterations leading to tissue lesions in the nervous system. Recent advances in neuroimaging have significantly improved our understanding of the underlying pathophysiology of ALS, with findings supporting the corticoefferent axonal disease progression theory. Current studies on neuroimaging in ALS have demonstrated inconsistencies, which may be due to small sample sizes, insufficient statistical power, overinterpretation of findings, and the inherent heterogeneity of ALS. Deriving meaningful conclusions solely from individual imaging metrics in ALS studies remains challenging, and integrating multimodal imaging techniques shows promise for detecting valuable ALS biomarkers. In addition to giving an overview of the principles and techniques of different neuroimaging modalities, this review describes the potential of neuroimaging biomarkers in the diagnosis and prognostication of ALS. We provide an insight into the underlying pathology, highlighting the need for standardized protocols and multicenter collaborations to advance ALS research.
Collapse
Affiliation(s)
- Wei Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Si-Han Liu
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiao-Jing Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hui Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhen-Wei Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xue-Fan Yu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
13
|
Du R, Chen P, Li M, Zhu Y, He Z, Huang X. Developing a novel immune infiltration-associated mitophagy prediction model for amyotrophic lateral sclerosis using bioinformatics strategies. Front Immunol 2024; 15:1360527. [PMID: 38601155 PMCID: PMC11005030 DOI: 10.3389/fimmu.2024.1360527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease, which leads to muscle weakness and eventual paralysis. Numerous studies have indicated that mitophagy and immune inflammation have a significant impact on the onset and advancement of ALS. Nevertheless, the possible diagnostic and prognostic significance of mitophagy-related genes associated with immune infiltration in ALS is uncertain. The purpose of this study is to create a predictive model for ALS using genes linked with mitophagy-associated immune infiltration. Methods ALS gene expression profiles were downloaded from the Gene Expression Omnibus (GEO) database. Univariate Cox analysis and machine learning methods were applied to analyze mitophagy-associated genes and develop a prognostic risk score model. Subsequently, functional and immune infiltration analyses were conducted to study the biological attributes and immune cell enrichment in individuals with ALS. Additionally, validation of identified feature genes in the prediction model was performed using ALS mouse models and ALS patients. Results In this study, a comprehensive analysis revealed the identification of 22 mitophagy-related differential expression genes and 40 prognostic genes. Additionally, an 18-gene prognostic signature was identified with machine learning, which was utilized to construct a prognostic risk score model. Functional enrichment analysis demonstrated the enrichment of various pathways, including oxidative phosphorylation, unfolded proteins, KRAS, and mTOR signaling pathways, as well as other immune-related pathways. The analysis of immune infiltration revealed notable distinctions in certain congenital immune cells and adaptive immune cells between the low-risk and high-risk groups, particularly concerning the T lymphocyte subgroup. ALS mouse models and ALS clinical samples demonstrated consistent expression levels of four mitophagy-related immune infiltration genes (BCKDHA, JTB, KYNU, and GTF2H5) with the results of bioinformatics analysis. Conclusion This study has successfully devised and verified a pioneering prognostic predictive risk score for ALS, utilizing eighteen mitophagy-related genes. Furthermore, the findings indicate that four of these genes exhibit promising roles in the context of ALS prognostic.
Collapse
Affiliation(s)
- Rongrong Du
- School of Medicine, Nankai University, Tianjin, China
- Department of Neurology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Peng Chen
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
- Department of General Surgery & Institute of General Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Mao Li
- Department of Neurology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yahui Zhu
- Department of Neurology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Zhengqing He
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xusheng Huang
- School of Medicine, Nankai University, Tianjin, China
- Department of Neurology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| |
Collapse
|
14
|
Jellinger KA. Understanding depression with amyotrophic lateral sclerosis: a short assessment of facts and perceptions. J Neural Transm (Vienna) 2024; 131:107-115. [PMID: 37922093 DOI: 10.1007/s00702-023-02714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/19/2023] [Indexed: 11/05/2023]
Abstract
Depression with an average prevalence of 25-40% is a serious condition in amyotrophic lateral sclerosis (ALS) that can impact quality of life and survival of patients and caregiver burden, yet the underlying neurobiology is poorly understood. Preexisting depression has been associated with a higher risk of developing ALS, while people with ALS have a significantly higher risk of developing depression that can cause multiple complications. Depression may be a prodromal or subclinical symptom prior to motor involvement, although its relations with disease progression and impairment of quality of life are under discussion. Unfortunately, there are no studies existing that explore the pathogenic mechanisms of depression associated with the basic neurodegenerative process, and no specific neuroimaging data or postmortem findings for the combination of ALS and depression are currently available. Experience from other neurodegenerative processes suggests that depressive symptoms in ALS may be the consequence of cortical thinning in prefrontal regions and other cortex areas, disruption of mood-related brain networks, dysfunction of neurotransmitter systems, changing cortisol levels and other, hitherto unknown mechanisms. Treatment of both ALS and depression is a multidisciplinary task, depression generally being treated with a combination of antidepressant medication, physiotherapy, psychological and other interventions, while electroconvulsive therapy and deep brain stimulation might not be indicated in the majority of patients in view of their poor prognosis. Since compared to depression in other neurodegenerative diseases, our knowledge of its molecular basis in ALS is missing, multidisciplinary clinicopathological studies to elucidate the pathomechanism of depression in motor system disorders including ALS are urgently warranted.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
15
|
Silaidos CV, Reutzel M, Wachter L, Dieter F, Ludin N, Blum WF, Wudy SA, Matura S, Pilatus U, Hattingen E, Pantel J, Eckert GP. Age-related changes in energy metabolism in peripheral mononuclear blood cells (PBMCs) and the brains of cognitively healthy seniors. GeroScience 2024; 46:981-998. [PMID: 37308768 PMCID: PMC10828287 DOI: 10.1007/s11357-023-00810-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/25/2023] [Indexed: 06/14/2023] Open
Abstract
Mitochondrial dysfunction is a hallmark of cellular senescence and many age-related neurodegenerative diseases. We therefore investigated the relationship between mitochondrial function in peripheral blood cells and cerebral energy metabolites in young and older sex-matched, physically and mentally healthy volunteers. Cross-sectional observational study involving 65 young (26.0 ± 0.49 years) and 65 older (71.7 ± 0.71 years) women and men recruited. Cognitive health was evaluated using established psychometric methods (MMSE, CERAD). Blood samples were collected and analyzed, and fresh peripheral blood mononuclear cells (PBMCs) were isolated. Mitochondrial respiratory complex activity was measured using a Clarke electrode. Adenosine triphosphate (ATP) and citrate synthase activity (CS) were determined by bioluminescence and photometrically. N-aspartyl-aspartate (tNAA), ATP, creatine (Cr), and phosphocreatine (PCr) were quantified in brains using 1H- and 31P-magnetic resonance spectroscopic imaging (MRSI). Levels of insulin-like growth factor 1 (IGF-1) were determined using a radio-immune assay (RIA). Complex IV activity (CIV) (- 15%) and ATP levels (- 11%) were reduced in PBMCs isolated from older participants. Serum levels of IGF-1 were significantly reduced (- 34%) in older participants. Genes involved in mitochondrial activity, antioxidant mechanisms, and autophagy were unaffected by age. tNAA levels were reduced (- 5%), Cr (+ 11%), and PCr (+ 14%) levels were increased, and ATP levels were unchanged in the brains of older participants. Markers of energy metabolism in blood cells did not significantly correlate with energy metabolites in the brain. Age-related bioenergetic changes were detected in peripheral blood cells and the brains of healthy older people. However, mitochondrial function in peripheral blood cells does not reflect energy related metabolites in the brain. While ATP levels in PBMCs may be be a valid marker for age-related mitochondrial dysfunction in humans, cerebral ATP remained constant.
Collapse
Affiliation(s)
- Carmina V Silaidos
- Laboratory for Nutrition in Prevention and Therapy, Biomedical Research Center Seltersberg (BFS), Institute of Nutritional Sciences, Justus-Liebig-University of Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Martina Reutzel
- Laboratory for Nutrition in Prevention and Therapy, Biomedical Research Center Seltersberg (BFS), Institute of Nutritional Sciences, Justus-Liebig-University of Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Lena Wachter
- Laboratory for Nutrition in Prevention and Therapy, Biomedical Research Center Seltersberg (BFS), Institute of Nutritional Sciences, Justus-Liebig-University of Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Fabian Dieter
- Laboratory for Nutrition in Prevention and Therapy, Biomedical Research Center Seltersberg (BFS), Institute of Nutritional Sciences, Justus-Liebig-University of Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Nasir Ludin
- Institute for Neuroradiology, University Hospital, Goethe University, Schleusenweg 2-16, Frankfurt, Germany
| | - Werner F Blum
- Laboratory for Translational Hormone Analytics in Pediatric Endocrinology, Peptide Hormone Research Unit Division of Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Stefan A Wudy
- Laboratory for Translational Hormone Analytics in Pediatric Endocrinology, Peptide Hormone Research Unit Division of Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Silke Matura
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Ulrich Pilatus
- Institute for Neuroradiology, University Hospital, Goethe University, Schleusenweg 2-16, Frankfurt, Germany
- Brain Imaging Center (BIC), University Hospital Frankfurt, Frankfurt a. M, Germany
| | - Elke Hattingen
- Institute for Neuroradiology, University Hospital, Goethe University, Schleusenweg 2-16, Frankfurt, Germany
| | - Johannes Pantel
- Geriatric Medicine, Institute of General Practice, Goethe University, Frankfurt a. M, Germany
| | - Gunter P Eckert
- Laboratory for Nutrition in Prevention and Therapy, Biomedical Research Center Seltersberg (BFS), Institute of Nutritional Sciences, Justus-Liebig-University of Giessen, Schubertstrasse 81, 35392, Giessen, Germany.
| |
Collapse
|
16
|
Payne T, Burgess T, Bradley S, Roscoe S, Sassani M, Dunning MJ, Hernandez D, Scholz S, McNeill A, Taylor R, Su L, Wilkinson I, Jenkins T, Mortiboys H, Bandmann O. Multimodal assessment of mitochondrial function in Parkinson's disease. Brain 2024; 147:267-280. [PMID: 38059801 PMCID: PMC10766247 DOI: 10.1093/brain/awad364] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/02/2023] [Accepted: 09/27/2023] [Indexed: 12/08/2023] Open
Abstract
The heterogenous aetiology of Parkinson's disease is increasingly recognized; both mitochondrial and lysosomal dysfunction have been implicated. Powerful, clinically applicable tools are required to enable mechanistic stratification for future precision medicine approaches. The aim of this study was to characterize bioenergetic dysfunction in Parkinson's disease by applying a multimodal approach, combining standardized clinical assessment with midbrain and putaminal 31-phosphorus magnetic resonance spectroscopy (31P-MRS) and deep phenotyping of mitochondrial and lysosomal function in peripheral tissue in patients with recent-onset Parkinson's disease and control subjects. Sixty participants (35 patients with Parkinson's disease and 25 healthy controls) underwent 31P-MRS for quantification of energy-rich metabolites [ATP, inorganic phosphate (Pi) and phosphocreatine] in putamen and midbrain. In parallel, skin biopsies were obtained from all research participants to establish fibroblast cell lines for subsequent quantification of total intracellular ATP and mitochondrial membrane potential (MMP) as well as mitochondrial and lysosomal morphology, using high content live cell imaging. Lower MMP correlated with higher intracellular ATP (r = -0.55, P = 0.0016), higher mitochondrial counts (r = -0.72, P < 0.0001) and higher lysosomal counts (r = -0.62, P = 0.0002) in Parkinson's disease patient-derived fibroblasts only, consistent with impaired mitophagy and mitochondrial uncoupling. 31P-MRS-derived posterior putaminal Pi/ATP ratio variance was considerably greater in Parkinson's disease than in healthy controls (F-tests, P = 0.0036). Furthermore, elevated 31P-MRS-derived putaminal, but not midbrain Pi/ATP ratios (indicative of impaired oxidative phosphorylation) correlated with both greater mitochondrial (r = 0.37, P = 0.0319) and lysosomal counts (r = 0.48, P = 0.0044) as well as lower MMP in both short (r = -0.52, P = 0.0016) and long (r = -0.47, P = 0.0052) mitochondria in Parkinson's disease. Higher 31P-MRS midbrain phosphocreatine correlated with greater risk of rapid disease progression (r = 0.47, P = 0.0384). Our data suggest that impaired oxidative phosphorylation in the striatal dopaminergic nerve terminals exceeds mitochondrial dysfunction in the midbrain of patients with early Parkinson's disease. Our data further support the hypothesis of a prominent link between impaired mitophagy and impaired striatal energy homeostasis as a key event in early Parkinson's disease.
Collapse
Affiliation(s)
- Thomas Payne
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Toby Burgess
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Stephen Bradley
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Sarah Roscoe
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Matilde Sassani
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, The University of Birmingham, Birmingham B15 2TT, UK
| | - Mark J Dunning
- The Bioinformatics Core, Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Dena Hernandez
- Molecular Genetics Section, Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD 20814, USA
| | - Sonja Scholz
- Neurodegenerative Diseases Research Unit, Laboratory of Neurogenetics, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD 21287, USA
| | - Alisdair McNeill
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Rosie Taylor
- Statistical Services Unit, The University of Sheffield, Shefield S3 7RH, UK
| | - Li Su
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Department of Psychiatry, University of Cambridge, Cambridge CB2 0SP, UK
| | - Iain Wilkinson
- Academic Unit of Radiology, University of Sheffield, Sheffield S10 2JF, UK
| | - Thomas Jenkins
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Department of Neurology, Royal Perth Hospital, Perth WA6000, Australia
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield S10 2HQ, UK
| |
Collapse
|
17
|
Moyle DB, Kudiersky MN, Totton MN, Sassani DM, Nichols DS, Jenkins DT, Redgrave DJ, Baig DS, Nair DKPS, Majid PA, Ali DAN. Remote ischaemic conditioning for fatigue after stroke (RICFAST): A pilot randomised controlled trial. J Stroke Cerebrovasc Dis 2023; 32:107420. [PMID: 37832270 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107420] [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: 03/14/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Post stroke fatigue (PSF) affects 50 % of stroke survivors, and can be disabling. Remote ischaemic conditioning (RIC), can preserve mitochondrial function, improve tissue perfusion and may mitigate PSF. This pilot randomised controlled trial evaluates the safety and feasibility of using RIC for PSF and evaluated measures of cellular bioenergetics. METHODS 24 people with debilitating PSF (7 item Fatigue Severity Score, FSS-7 > 4) were randomised (1:1) in this single-centre phase 2 study to RIC (blood pressure cuff inflation around the upper arm 200 mmHg for 5 min followed by 5 min of deflation), or sham (inflation pressure 20 mmHg), repeated 4 cycles, 3 times per week for 6 weeks. Primary outcomes were safety, acceptability, and compliance. Secondary outcomes included FSS-7, 6 min walking test (6MWT), peak oxygen consumption (V̇O2peak), ventilatory anaerobic threshold (VAT), and muscle adenosine triphosphate (ATP) content measured using 31-phosphorous magnetic resonance spectroscopy of tibialis anterior. RESULTS RIC was safe (no serious adverse events, adverse events mild) and adherence excellent (91 % sessions completed). Exploratory analysis revealed lower FSS-7 scores in the RIC group compared to sham at 6 weeks (between group difference FSS-7 -0.7, 95 %CI -2.0 to 0.6), 3 months (-1.0, 95 %CI -2.2 to 0.2) and 6 months (-0.9, 95 %CI -2.0 to 0.2). There were trends towards increased VAT, increased muscle ATP content and improved 6MWT in the RIC group. DISCUSSION RIC is safe and acceptable for people with PSF and may result in clinically meaningful improvements in fatigue and muscle bioenergetics that require further investigation in larger studies.
Collapse
Affiliation(s)
| | | | | | - Dr Matilde Sassani
- Translational Brain Science, Institute of Metabolism and Systems Research, UK
| | | | - Dr Tom Jenkins
- Sheffield Institute for Translational Neurology, UK; Royal Perth Hospital, Western Australia, UK
| | | | | | | | | | - Dr Ali N Ali
- Sheffield Teaching Hospitals NIHR Biomedical Research Centre, University of Sheffield, UK.
| |
Collapse
|
18
|
Harrington JS, Ryter SW, Plataki M, Price DR, Choi AMK. Mitochondria in health, disease, and aging. Physiol Rev 2023; 103:2349-2422. [PMID: 37021870 PMCID: PMC10393386 DOI: 10.1152/physrev.00058.2021] [Citation(s) in RCA: 231] [Impact Index Per Article: 115.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Mitochondria are well known as organelles responsible for the maintenance of cellular bioenergetics through the production of ATP. Although oxidative phosphorylation may be their most important function, mitochondria are also integral for the synthesis of metabolic precursors, calcium regulation, the production of reactive oxygen species, immune signaling, and apoptosis. Considering the breadth of their responsibilities, mitochondria are fundamental for cellular metabolism and homeostasis. Appreciating this significance, translational medicine has begun to investigate how mitochondrial dysfunction can represent a harbinger of disease. In this review, we provide a detailed overview of mitochondrial metabolism, cellular bioenergetics, mitochondrial dynamics, autophagy, mitochondrial damage-associated molecular patterns, mitochondria-mediated cell death pathways, and how mitochondrial dysfunction at any of these levels is associated with disease pathogenesis. Mitochondria-dependent pathways may thereby represent an attractive therapeutic target for ameliorating human disease.
Collapse
Affiliation(s)
- John S Harrington
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | | | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | - David R Price
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| |
Collapse
|
19
|
Jellinger KA. The Spectrum of Cognitive Dysfunction in Amyotrophic Lateral Sclerosis: An Update. Int J Mol Sci 2023; 24:14647. [PMID: 37834094 PMCID: PMC10572320 DOI: 10.3390/ijms241914647] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Cognitive dysfunction is an important non-motor symptom in amyotrophic lateral sclerosis (ALS) that has a negative impact on survival and caregiver burden. It shows a wide spectrum ranging from subjective cognitive decline to frontotemporal dementia (FTD) and covers various cognitive domains, mainly executive/attention, language and verbal memory deficits. The frequency of cognitive impairment across the different ALS phenotypes ranges from 30% to 75%, with up to 45% fulfilling the criteria of FTD. Significant genetic, clinical, and pathological heterogeneity reflects deficits in various cognitive domains. Modern neuroimaging studies revealed frontotemporal degeneration and widespread involvement of limbic and white matter systems, with hypometabolism of the relevant areas. Morphological substrates are frontotemporal and hippocampal atrophy with synaptic loss, associated with TDP-43 and other co-pathologies, including tau deposition. Widespread functional disruptions of motor and extramotor networks, as well as of frontoparietal, frontostriatal and other connectivities, are markers for cognitive deficits in ALS. Cognitive reserve may moderate the effect of brain damage but is not protective against cognitive decline. The natural history of cognitive dysfunction in ALS and its relationship to FTD are not fully understood, although there is an overlap between the ALS variants and ALS-related frontotemporal syndromes, suggesting a differential vulnerability of motor and non-motor networks. An assessment of risks or the early detection of brain connectivity signatures before structural changes may be helpful in investigating the pathophysiological mechanisms of cognitive impairment in ALS, which might even serve as novel targets for effective disease-modifying therapies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| |
Collapse
|
20
|
Bustamante-Barrientos FA, Luque-Campos N, Araya MJ, Lara-Barba E, de Solminihac J, Pradenas C, Molina L, Herrera-Luna Y, Utreras-Mendoza Y, Elizondo-Vega R, Vega-Letter AM, Luz-Crawford P. Mitochondrial dysfunction in neurodegenerative disorders: Potential therapeutic application of mitochondrial transfer to central nervous system-residing cells. J Transl Med 2023; 21:613. [PMID: 37689642 PMCID: PMC10493034 DOI: 10.1186/s12967-023-04493-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
Mitochondrial dysfunction is reiteratively involved in the pathogenesis of diverse neurodegenerative diseases. Current in vitro and in vivo approaches support that mitochondrial dysfunction is branded by several molecular and cellular defects, whose impact at different levels including the calcium and iron homeostasis, energetic balance and/or oxidative stress, makes it difficult to resolve them collectively given their multifactorial nature. Mitochondrial transfer offers an overall solution since it contains the replacement of damage mitochondria by healthy units. Therefore, this review provides an introducing view on the structure and energy-related functions of mitochondria as well as their dynamics. In turn, we summarize current knowledge on how these features are deregulated in different neurodegenerative diseases, including frontotemporal dementia, multiple sclerosis, amyotrophic lateral sclerosis, Friedreich ataxia, Alzheimer´s disease, Parkinson´s disease, and Huntington's disease. Finally, we analyzed current advances in mitochondrial transfer between diverse cell types that actively participate in neurodegenerative processes, and how they might be projected toward developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliana Lara-Barba
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Javiera de Solminihac
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Carolina Pradenas
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Luis Molina
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ana María Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaiso, Valparaiso, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Mons. Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| |
Collapse
|
21
|
Lopez FV, O’Shea A, Rosenberg JT, Leeuwenburgh C, Anton S, Bowers D, Woods AJ. Frontal adenosine triphosphate markers from 31P MRS are associated with cognitive performance in healthy older adults: preliminary findings. Front Aging Neurosci 2023; 15:1180994. [PMID: 37614473 PMCID: PMC10442546 DOI: 10.3389/fnagi.2023.1180994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/17/2023] [Indexed: 08/25/2023] Open
Abstract
Aging is associated with declines in mitochondrial efficiency and energy production which directly impacts the availability of adenosine triphosphate (ATP), which contains high energy phosphates critical for a variety of cellular functions. Previous phosphorous magnetic resonance spectroscopy (31P MRS) studies demonstrate cerebral ATP declines with age. The purpose of this study was to explore the functional relationships of frontal and posterior ATP levels with cognition in healthy aging. Here, we measured frontal and posterior ATP levels using 31P MRS at 3 Tesla (3 T) and assessed cognition using the Montreal Cognitive Assessment (MoCA) in 30 healthy older adults. We found that greater frontal, but not posterior, ATP levels were significantly associated with better MoCA performance. This relationship remained significant after controlling for age, sex, years of education, and brain atrophy. In conclusion, our findings indicate that cognition is related to ATP in the frontal cortex. These preliminary findings may have important implications in the search for non-invasive markers of in vivo mitochondrial function and the impact of ATP availability on cognition. Future studies are needed to confirm the functional significance of regional ATP and cognition across the lifespan.
Collapse
Affiliation(s)
- Francesca V. Lopez
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Andrew O’Shea
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Jens T. Rosenberg
- Advanced Magnetic Resonance Imaging and Spectroscopy Facility, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, College of Medicine, University of Florida, Gainesville, FL, United States
- College of Medicine, Institute on Aging, University of Florida, Gainesville, FL, United States
| | - Stephen Anton
- Department of Aging and Geriatric Research, College of Medicine, University of Florida, Gainesville, FL, United States
- College of Medicine, Institute on Aging, University of Florida, Gainesville, FL, United States
| | - Dawn Bowers
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Department of Neurology, College of Medicine, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Adam J. Woods
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
22
|
Xie X, Shi Y, Ma L, Yang W, Pu J, Shen Y, Liu Y, Zhang H, Lv F, Hu L. Altered neurometabolite levels in the brains of patients with depression: A systematic analysis of magnetic resonance spectroscopy studies. J Affect Disord 2023; 328:95-102. [PMID: 36521666 DOI: 10.1016/j.jad.2022.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Numerous magnetic resonance spectroscopy (MRS) studies have reported metabolic abnormalities in the brains of patients with depression, although inconsistent results have been reported. The aim of this study was to explore changes in neurometabolite levels in patients with depression across large-scale MRS studies. METHOD A total of 307 differential metabolite entries associated with depression were retrieved from 180 MRS studies retrieved from the Metabolite Network of Depression Database. The vote-counting method was used to identify consistently altered metabolites in the whole brain and specific brain regions of patients with depression. RESULTS Only few differential neurometabolites showed a stable change trend. The levels of total choline (tCho) and the tCho/N-acetyl aspartate (NAA) ratio were consistently higher in the brains of patients with depression, and that the levels of NAA, glutamate and glutamine (Glx), and gamma-aminobutyric acid (GABA) were lower. For specific brain regions, we found lower Glx levels in the prefrontal cortex and lower GABA concentrations in the occipital cortex. We also found lower concentrations of NAA in the anterior cingulate cortex and prefrontal cortex. The levels of tCho were higher in the prefrontal cortex and putamen. CONCLUSION Our results revealed that most altered neurometabolites in previous studies lack of adequate reproducibility. Through vote-counting method with large-scale studies, downregulation of glutamatergic neurometabolites, impaired neuronal integrity, and disturbed membrane metabolism were found in the pathobiology of depression, which contribute to existing knowledge of neurometabolic changes in depression. Further studies based on a larger dataset are needed to confirm our findings.
Collapse
Affiliation(s)
- Xiongfei Xie
- Department of Radiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Shi
- Department of Radiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Ma
- Department of Radiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Wenqin Yang
- Department of Radiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Juncai Pu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiqing Shen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hanping Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fajin Lv
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Liangbo Hu
- Department of Radiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
23
|
Mead RJ, Shan N, Reiser HJ, Marshall F, Shaw PJ. Amyotrophic lateral sclerosis: a neurodegenerative disorder poised for successful therapeutic translation. Nat Rev Drug Discov 2023; 22:185-212. [PMID: 36543887 PMCID: PMC9768794 DOI: 10.1038/s41573-022-00612-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2022] [Indexed: 12/24/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating disease caused by degeneration of motor neurons. As with all major neurodegenerative disorders, development of disease-modifying therapies has proven challenging for multiple reasons. Nevertheless, ALS is one of the few neurodegenerative diseases for which disease-modifying therapies are approved. Significant discoveries and advances have been made in ALS preclinical models, genetics, pathology, biomarkers, imaging and clinical readouts over the last 10-15 years. At the same time, novel therapeutic paradigms are being applied in areas of high unmet medical need, including neurodegenerative disorders. These developments have evolved our knowledge base, allowing identification of targeted candidate therapies for ALS with diverse mechanisms of action. In this Review, we discuss how this advanced knowledge, aligned with new approaches, can enable effective translation of therapeutic agents from preclinical studies through to clinical benefit for patients with ALS. We anticipate that this approach in ALS will also positively impact the field of drug discovery for neurodegenerative disorders more broadly.
Collapse
Affiliation(s)
- Richard J Mead
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Keapstone Therapeutics, The Innovation Centre, Broomhall, Sheffield, UK
| | - Ning Shan
- Aclipse Therapeutics, Radnor, PA, US
| | | | - Fiona Marshall
- MSD UK Discovery Centre, Merck, Sharp and Dohme (UK) Limited, London, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, UK.
- Keapstone Therapeutics, The Innovation Centre, Broomhall, Sheffield, UK.
| |
Collapse
|
24
|
Weil EL, Nakawah MO, Masdeu JC. Advances in the neuroimaging of motor disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:359-381. [PMID: 37562878 DOI: 10.1016/b978-0-323-98818-6.00039-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Neuroimaging is a valuable adjunct to the history and examination in the evaluation of motor system disorders. Conventional imaging with computed tomography or magnetic resonance imaging depicts important anatomic information and helps to identify imaging patterns which may support diagnosis of a specific motor disorder. Advanced imaging techniques can provide further detail regarding volume, functional, or metabolic changes occurring in nervous system pathology. This chapter is an overview of the advances in neuroimaging with particular emphasis on both standard and less well-known advanced imaging techniques and findings, such as diffusion tensor imaging or volumetric studies, and their application to specific motor disorders. In addition, it provides reference to emerging imaging biomarkers in motor system disorders such as Parkinson disease, amyotrophic lateral sclerosis, and Huntington disease, and briefly reviews the neuroimaging findings in different causes of myelopathy and peripheral nerve disorders.
Collapse
Affiliation(s)
- Erika L Weil
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States; Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, TX, United States.
| | - Mohammad Obadah Nakawah
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, TX, United States; Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| | - Joseph C Masdeu
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, TX, United States; Department of Neurology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
25
|
Alterations in metabolic flux in migraine and the translational relevance. J Headache Pain 2022; 23:127. [PMID: 36175833 PMCID: PMC9523955 DOI: 10.1186/s10194-022-01494-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/10/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Migraine is a highly prevalent disorder with significant economical and personal burden. Despite the development of effective therapeutics, the causes which precipitate migraine attacks remain elusive. Clinical studies have highlighted altered metabolic flux and mitochondrial function in patients. In vivo animal experiments can allude to the metabolic mechanisms which may underlie migraine susceptibility. Understanding the translational relevance of these studies are important to identifying triggers, biomarkers and therapeutic targets in migraine. MAIN BODY Functional imaging studies have suggested that migraineurs feature metabolic syndrome, exhibiting hallmark features including upregulated oxidative phosphorylation yet depleted available free energy. Glucose hypometabolism is also evident in migraine patients and can lead to altered neuronal hyperexcitability such as the incidence of cortical spreading depression (CSD). The association between obesity and increased risk, frequency and worse prognosis of migraine also highlights lipid dysregulation in migraine pathology. Calcitonin gene related peptide (CGRP) has demonstrated an important role in sensitisation and nociception in headache, however its role in metabolic regulation in connection with migraine has not been thoroughly explored. Whether impaired metabolic function leads to increased release of peptides such as CGRP or excessive nociception leads to altered flux is yet unknown. CONCLUSION Migraine susceptibility may be underpinned by impaired metabolism resulting in depleted energy stores and altered neuronal function. This review discusses both clinical and in vivo studies which provide evidence of altered metabolic flux which contribute toward pathophysiology. It also reviews the translational relevance of animal studies in identifying targets of biomarker or therapeutic development.
Collapse
|
26
|
Hertel N, Kuzma-Kozakiewicz M, Gromicho M, Grosskreutz J, de Carvalho M, Uysal H, Dengler R, Petri S, Körner S. Analysis of routine blood parameters in patients with amyotrophic lateral sclerosis and evaluation of a possible correlation with disease progression—a multicenter study. Front Neurol 2022; 13:940375. [PMID: 35968316 PMCID: PMC9364810 DOI: 10.3389/fneur.2022.940375] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Amyotrophic lateral sclerosis (ALS) pathogenesis is still unclear, its course is considerably variable, and prognosis is hard to determine. Despite much research, there is still a lack of easily accessible markers predicting prognosis. We investigated routine blood parameters in ALS patients regarding correlations with disease severity, progression rate, and survival. Additionally, we analyzed disease and patients' characteristics relating to baseline blood parameter levels. Methods We analyzed creatine kinase (CK), albumin (ALB), creatinine (CREA), total cholesterol (TC), high-density lipoprotein cholesterol (HDL), low-density lipoprotein cholesterol (LDL), and triglycerides (TG) levels around time of diagnosis in 1,084 ALS patients. We carried out linear regression analyses including disease and patients' characteristics with each blood parameter to detect correlations with them. Linear regression models were performed for ALSFRS-R at study entry, its retrospectively defined rate of decay and prospectively collected progression rate. Different survival analysis methods were used to examine associations between blood parameters and survival. Results We found higher CK (p-value 0.001), ALB (p-value <0.001), CREA (p-value <0.001), and HDL levels (p-value 0.044) at time of diagnosis being associated with better functional status according to ALSFRS-R scores at study entry. Additionally, higher CREA levels were associated with lower risk of death (p-value 0.003). Conclusions Our results indicate potential of CK, ALB, CREA, and HDL as disease severity or progression markers, and may also provide clues to ALS pathogenesis. However, these values are highly dependent on other variables, and further careful, longitudinal analyses will be necessary to prove the relevance of our findings.
Collapse
Affiliation(s)
- Nora Hertel
- Department of Neurology, Hannover Medical School, Hanover, Germany
| | | | - Marta Gromicho
- Institute of Physiology-Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | - Mamede de Carvalho
- Institute of Physiology-Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Hilmi Uysal
- Department of Neurology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Reinhard Dengler
- Department of Neurology, Hannover Medical School, Hanover, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hanover, Germany
- Center for Systems Neuroscience (ZSN), Hanover, Germany
| | - Sonja Körner
- Department of Neurology, Hannover Medical School, Hanover, Germany
- *Correspondence: Sonja Körner
| |
Collapse
|
27
|
El Mendili MM, Grapperon AM, Dintrich R, Stellmann JP, Ranjeva JP, Guye M, Verschueren A, Attarian S, Zaaraoui W. Alterations of Microstructure and Sodium Homeostasis in Fast Amyotrophic Lateral Sclerosis Progressors: A Brain DTI and Sodium MRI Study. AJNR Am J Neuroradiol 2022; 43:984-990. [PMID: 35772800 DOI: 10.3174/ajnr.a7559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/10/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE While conventional MR imaging has limited value in amyotrophic lateral sclerosis, nonconventional MR imaging has shown alterations of microstructure using diffusion MR imaging and recently sodium homeostasis with sodium MR imaging. We aimed to investigate the topography of brain regions showing combined microstructural and sodium homeostasis alterations in amyotrophic lateral sclerosis subgroups according to their disease-progression rates. MATERIALS AND METHODS Twenty-nine patients with amyotrophic lateral sclerosis and 24 age-matched healthy controls were recruited. Clinical assessments included disease duration and the Revised Amyotrophic Lateral Sclerosis Functional Rating Scale. Patients were clinically differentiated into fast (n = 13) and slow (n = 16) progressors according to the Revised Amyotrophic Lateral Sclerosis Functional Rating Scale progression rate. 3T MR imaging brain protocol included 1H T1-weighted and diffusion sequences and a 23Na density-adapted radial sequence. Quantitative maps of diffusion with fractional anisotropy, mean diffusivity, and total sodium concentration were measured. The topography of diffusion and sodium abnormalities was assessed by voxelwise analyses. RESULTS Patients with amyotrophic lateral sclerosis showed significantly higher sodium concentrations and lower fractional anisotropy, along with higher sodium concentrations and higher mean diffusivity compared with healthy controls, primarily within the corticospinal tracts, corona radiata, and body and genu of the corpus callosum. Fast progressors showed wider-spread abnormalities mainly in the frontal areas. In slow progressors, only fractional anisotropy measures showed abnormalities compared with healthy controls, localized in focal regions of the corticospinal tracts, the body of corpus callosum, corona radiata, and thalamic radiation. CONCLUSIONS The present study evidenced widespread combined microstructural and sodium homeostasis brain alterations in fast amyotrophic lateral sclerosis progressors.
Collapse
Affiliation(s)
- M M El Mendili
- From the Aix Marseille University (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), Centre national de la recherche scientifique, The Center for Magnetic Resonance in Biology and Medicine, Marseille, France .,APHM, Hopital de la Timone (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), CEMEREM, Marseille, France
| | - A-M Grapperon
- From the Aix Marseille University (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), Centre national de la recherche scientifique, The Center for Magnetic Resonance in Biology and Medicine, Marseille, France.,APHM, Hopital de la Timone (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), CEMEREM, Marseille, France.,APHM, Hôpital de la Timone (A.-M.G., R.D., S.A.), Referral Centre for Neuromuscular Diseases and ALS, Marseille, France
| | - R Dintrich
- From the Aix Marseille University (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), Centre national de la recherche scientifique, The Center for Magnetic Resonance in Biology and Medicine, Marseille, France.,APHM, Hopital de la Timone (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), CEMEREM, Marseille, France.,APHM, Hôpital de la Timone (A.-M.G., R.D., S.A.), Referral Centre for Neuromuscular Diseases and ALS, Marseille, France
| | - J-P Stellmann
- From the Aix Marseille University (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), Centre national de la recherche scientifique, The Center for Magnetic Resonance in Biology and Medicine, Marseille, France.,APHM, Hopital de la Timone (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), CEMEREM, Marseille, France
| | - J-P Ranjeva
- From the Aix Marseille University (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), Centre national de la recherche scientifique, The Center for Magnetic Resonance in Biology and Medicine, Marseille, France.,APHM, Hopital de la Timone (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), CEMEREM, Marseille, France
| | - M Guye
- From the Aix Marseille University (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), Centre national de la recherche scientifique, The Center for Magnetic Resonance in Biology and Medicine, Marseille, France.,APHM, Hopital de la Timone (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), CEMEREM, Marseille, France
| | - A Verschueren
- From the Aix Marseille University (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), Centre national de la recherche scientifique, The Center for Magnetic Resonance in Biology and Medicine, Marseille, France.,APHM, Hopital de la Timone (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), CEMEREM, Marseille, France
| | - S Attarian
- APHM, Hôpital de la Timone (A.-M.G., R.D., S.A.), Referral Centre for Neuromuscular Diseases and ALS, Marseille, France
| | - W Zaaraoui
- From the Aix Marseille University (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), Centre national de la recherche scientifique, The Center for Magnetic Resonance in Biology and Medicine, Marseille, France.,APHM, Hopital de la Timone (M.M.E.M., A.-M.G., R.D., J.-P.S., J.-P.R., M.G., A.V., W.Z.), CEMEREM, Marseille, France
| |
Collapse
|
28
|
Lanznaster D, Bruno C, Bourgeais J, Emond P, Zemmoura I, Lefèvre A, Reynier P, Eymieux S, Blanchard E, Vourc’h P, Andres CR, Bakkouche SE, Herault O, Favard L, Corcia P, Blasco H. Metabolic Profile and Pathological Alterations in the Muscle of Patients with Early-Stage Amyotrophic Lateral Sclerosis. Biomedicines 2022; 10:biomedicines10061307. [PMID: 35740329 PMCID: PMC9220134 DOI: 10.3390/biomedicines10061307] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 12/26/2022] Open
Abstract
Diverse biomarkers and pathological alterations have been found in muscle of patients with Amyotrophic lateral sclerosis (ALS), but the relation between such alterations and dysfunction in energetic metabolism remains to be investigated. We established the metabolome of muscle and serum of ALS patients and correlated these findings with the clinical status and pathological alterations observed in the muscle. We obtained data from 20 controls and 17 ALS patients (disease duration: 9.4 ± 6.8 months). Multivariate metabolomics analysis identified a distinct serum metabolome for ALS compared to controls (p-CV-ANOVA < 0.035) and revealed an excellent discriminant profile for muscle metabolome (p-CV-ANOVA < 0.0012). Citramalate was discriminant for both muscle and serum. High lauroylcarnitine levels in muscle were associated with low Forced Vital Capacity. Transcriptomics analysis of key antioxidant enzymes showed an upregulation of SOD3 (p = 0.0017) and GLRX2(1) (p = 0.0022) in ALS muscle. Analysis of mitochondrial enzymatic activity in muscle revealed higher complex II/CS (p = 0.04) and lower LDH (p = 0.03) activity in ALS than in controls. Our study showed, for the first time, a global dysfunction in the muscle of early-stage ALS patients. Furthermore, we identified novel metabolites to be employed as biomarkers for diagnosis and prognosis of ALS patients.
Collapse
Affiliation(s)
- Débora Lanznaster
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Correspondence:
| | - Clément Bruno
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37000 Tours, France
| | - Jérôme Bourgeais
- CNRS ERL7001, EA 7501 GICC, Université de Tours, 37000 Tours, France; (J.B.); (O.H.)
| | - Patrick Emond
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Médecine Nucléaire In Vitro, CHU de Tours, 37000 Tours, France
| | - Ilyess Zemmoura
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Neurochirurgie, CHU de Tours, 37000 Tours, France
| | - Antoine Lefèvre
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
| | - Pascal Reynier
- Service de Biochimie et Biologie Moléculaire, CHU d’Angers, 49000 Angers, France;
- Mitovasc-Mitolab, UMR CNRS6015-INSERM1083, 49000 Angers, France
| | - Sébastien Eymieux
- Plateforme IBiSA de Microscopie Electronique, Université de Tours et CHU de Tours, 37000 Tours, France; (S.E.); (E.B.)
- INSERM U1259, Université de Tours, 37000 Tours, France
| | - Emmanuelle Blanchard
- Plateforme IBiSA de Microscopie Electronique, Université de Tours et CHU de Tours, 37000 Tours, France; (S.E.); (E.B.)
- INSERM U1259, Université de Tours, 37000 Tours, France
| | - Patrick Vourc’h
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37000 Tours, France
| | - Christian R. Andres
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37000 Tours, France
| | | | - Olivier Herault
- CNRS ERL7001, EA 7501 GICC, Université de Tours, 37000 Tours, France; (J.B.); (O.H.)
| | - Luc Favard
- Service de Neurologie, CHU de Tours, 37000 Tours, France;
| | - Philippe Corcia
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Neurologie, CHU de Tours, 37000 Tours, France;
| | - Hélène Blasco
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37000 Tours, France
| |
Collapse
|
29
|
Hippocampal Mitochondrial Abnormalities Induced the Dendritic Complexity Reduction and Cognitive Decline in a Rat Model of Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9253916. [PMID: 35571236 PMCID: PMC9095360 DOI: 10.1155/2022/9253916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/19/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022]
Abstract
Spinal cord injury (SCI) is a progressive neurodegenerative disease in addition to a traumatic event. Cognitive dysfunction following SCI has been widely reported in patients and animal models. However, the neuroanatomical changes affecting cognitive function after SCI, as well as the mechanisms behind these changes, have so far remained elusive. Herein, we found that SCI accelerates oxidative stress damage of hippocampal neuronal mitochondria. Then, for the first time, we presented a three-dimensional morphological atlas of rat hippocampal neurons generated using a fluorescence Micro-Optical Sectioning Tomography system, a method that accurately identifies the spatial localization of neurons and trace neurites. We showed that the number of dendritic branches and dendritic length was decreased in late stage of SCI. Western blot and transmission electron microscopy analyses also showed a decrease in synaptic communication. In addition, a battery of behavioral tests in these animals revealed hippocampal based cognitive dysfunction, which could be attributed to changes in the dendritic complexity of hippocampal neurons. Taken together, these results suggested that mitochondrial abnormalities in hippocampal neurons induced the dendritic complexity reduction and cognitive decline following SCI. Our study highlights the neuroanatomical basis and importance of mitochondria in brain degeneration following SCI, which might contribute to propose new therapeutic strategies.
Collapse
|
30
|
Mechanistic Insights of Mitochondrial Dysfunction in Amyotrophic Lateral Sclerosis: An Update on a Lasting Relationship. Metabolites 2022; 12:metabo12030233. [PMID: 35323676 PMCID: PMC8951432 DOI: 10.3390/metabo12030233] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive loss of the upper and lower motor neurons. Despite the increasing effort in understanding the etiopathology of ALS, it still remains an obscure disease, and no therapies are currently available to halt its progression. Following the discovery of the first gene associated with familial forms of ALS, Cu–Zn superoxide dismutase, it appeared evident that mitochondria were key elements in the onset of the pathology. However, as more and more ALS-related genes were discovered, the attention shifted from mitochondria impairment to other biological functions such as protein aggregation and RNA metabolism. In recent years, mitochondria have again earned central, mechanistic roles in the pathology, due to accumulating evidence of their derangement in ALS animal models and patients, often resulting in the dysregulation of the energetic metabolism. In this review, we first provide an update of the last lustrum on the molecular mechanisms by which the most well-known ALS-related proteins affect mitochondrial functions and cellular bioenergetics. Next, we focus on evidence gathered from human specimens and advance the concept of a cellular-specific mitochondrial “metabolic threshold”, which may appear pivotal in ALS pathogenesis.
Collapse
|
31
|
Lazzarino G, Mangione R, Belli A, Di Pietro V, Nagy Z, Barnes NM, Bruce L, Ropero BM, Persson LI, Manca B, Saab MW, Amorini AM, Tavazzi B, Lazzarino G, Logan A. ILB ® Attenuates Clinical Symptoms and Serum Biomarkers of Oxidative/Nitrosative Stress and Mitochondrial Dysfunction in Patients with Amyotrophic Lateral Sclerosis. J Pers Med 2021; 11:794. [PMID: 34442438 PMCID: PMC8399678 DOI: 10.3390/jpm11080794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 01/22/2023] Open
Abstract
Oxidative/nitrosative stress and mitochondrial dysfunction is a hallmark of amyotrophic lateral sclerosis (ALS), an invariably fatal progressive neurodegenerative disease. Here, as an exploratory arm of a phase II clinical trial (EudraCT Number 2017-005065-47), we used high performance liquid chromatography(HPLC) to investigate changes in the metabolic profiles of serum from ALS patients treated weekly for 4 weeks with a repeated sub-cutaneous dose of 1 mg/kg of a proprietary low molecular weight dextran sulphate, called ILB®. A significant normalization of the serum levels of several key metabolites was observed over the treatment period, including N-acetylaspartate (NAA), oxypurines, biomarkers of oxidative/nitrosative stress and antioxidants. An improved serum metabolic profile was accompanied by significant amelioration of the patients' clinical conditions, indicating a response to ILB® treatment that appears to be mediated by improvement of tissue bioenergetics, decrease of oxidative/nitrosative stress and attenuation of (neuro)inflammatory processes.
Collapse
Affiliation(s)
- Giacomo Lazzarino
- UniCamillus, Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Renata Mangione
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Rome, 00168 Rome, Italy;
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Belli
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.B.); (V.D.P.); (Z.N.); (N.M.B.)
| | - Valentina Di Pietro
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.B.); (V.D.P.); (Z.N.); (N.M.B.)
| | - Zsuzsanna Nagy
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.B.); (V.D.P.); (Z.N.); (N.M.B.)
| | - Nicholas M. Barnes
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (A.B.); (V.D.P.); (Z.N.); (N.M.B.)
| | | | - Bernardo M. Ropero
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden; (B.M.R.); (L.I.P.)
| | - Lennart I. Persson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden; (B.M.R.); (L.I.P.)
| | - Benedetta Manca
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy;
| | - Miriam Wissam Saab
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, 95123 Catania, Italy; (M.W.S.); (A.M.A.)
| | - Angela M. Amorini
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, 95123 Catania, Italy; (M.W.S.); (A.M.A.)
| | - Barbara Tavazzi
- UniCamillus, Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Rome, 00168 Rome, Italy;
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, 95123 Catania, Italy; (M.W.S.); (A.M.A.)
| | - Ann Logan
- Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Axolotl Consulting Ltd., Droitwich WR9 0JS, UK
| |
Collapse
|
32
|
Obrador E, Salvador-Palmer R, López-Blanch R, Jihad-Jebbar A, Vallés SL, Estrela JM. The Link between Oxidative Stress, Redox Status, Bioenergetics and Mitochondria in the Pathophysiology of ALS. Int J Mol Sci 2021; 22:ijms22126352. [PMID: 34198557 PMCID: PMC8231819 DOI: 10.3390/ijms22126352] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common neurodegenerative disease of the motor system. It is characterized by the degeneration of both upper and lower motor neurons, which leads to muscle weakness and paralysis. ALS is incurable and has a bleak prognosis, with median survival of 3-5 years after the initial symptomatology. In ALS, motor neurons gradually degenerate and die. Many features of mitochondrial dysfunction are manifested in neurodegenerative diseases, including ALS. Mitochondria have shown to be an early target in ALS pathophysiology and contribute to disease progression. Disruption of their axonal transport, excessive generation of reactive oxygen species, disruption of the mitochondrial structure, dynamics, mitophagy, energy production, calcium buffering and apoptotic triggering have all been directly involved in disease pathogenesis and extensively reported in ALS patients and animal model systems. Alterations in energy production by motor neurons, which severely limit their survival capacity, are tightly linked to the redox status and mitochondria. The present review focuses on this link. Placing oxidative stress as a main pathophysiological mechanism, the molecular interactions and metabolic flows involved are analyzed. This leads to discussing potential therapeutic approaches targeting mitochondrial biology to slow disease progression.
Collapse
Affiliation(s)
- Elena Obrador
- Correspondence: (E.O.); (J.M.E.); Tel.: +34-963864646 (J.M.E.)
| | | | | | | | | | - José M. Estrela
- Correspondence: (E.O.); (J.M.E.); Tel.: +34-963864646 (J.M.E.)
| |
Collapse
|
33
|
Scaricamazza S, Salvatori I, Ferri A, Valle C. Skeletal Muscle in ALS: An Unappreciated Therapeutic Opportunity? Cells 2021; 10:525. [PMID: 33801336 PMCID: PMC8000428 DOI: 10.3390/cells10030525] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the selective degeneration of upper and lower motor neurons and by the progressive weakness and paralysis of voluntary muscles. Despite intense research efforts and numerous clinical trials, it is still an incurable disease. ALS had long been considered a pure motor neuron disease; however, recent studies have shown that motor neuron protection is not sufficient to prevent the course of the disease since the dismantlement of neuromuscular junctions occurs before motor neuron degeneration. Skeletal muscle alterations have been described in the early stages of the disease, and they seem to be mainly involved in the "dying back" phenomenon of motor neurons and metabolic dysfunctions. In recent years, skeletal muscles have been considered crucial not only for the etiology of ALS but also for its treatment. Here, we review clinical and preclinical studies that targeted skeletal muscles and discuss the different approaches, including pharmacological interventions, supplements or diets, genetic modifications, and training programs.
Collapse
Affiliation(s)
- Silvia Scaricamazza
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Illari Salvatori
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Alberto Ferri
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Cristiana Valle
- Fondazione Santa Lucia IRCCS, c/o CERC, 00143 Rome, Italy; (S.S.); (I.S.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| |
Collapse
|
34
|
Mitochondrial Dysfunction in Alzheimer's Disease: A Biomarker of the Future? Biomedicines 2021; 9:biomedicines9010063. [PMID: 33440662 PMCID: PMC7827030 DOI: 10.3390/biomedicines9010063] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide and is characterised pathologically by the accumulation of amyloid beta and tau protein aggregates. Currently, there are no approved disease modifying therapies for clearance of either of these proteins from the brain of people with AD. As well as abnormalities in protein aggregation, other pathological changes are seen in this condition. The function of mitochondria in both the nervous system and rest of the body is altered early in this disease, and both amyloid and tau have detrimental effects on mitochondrial function. In this review article, we describe how the function and structure of mitochondria change in AD. This review summarises current imaging techniques that use surrogate markers of mitochondrial function in both research and clinical practice, but also how mitochondrial functions such as ATP production, calcium homeostasis, mitophagy and reactive oxygen species production are affected in AD mitochondria. The evidence reviewed suggests that the measurement of mitochondrial function may be developed into a future biomarker for early AD. Further work with larger cohorts of patients is needed before mitochondrial functional biomarkers are ready for clinical use.
Collapse
|