1
|
Belosludtseva NV, Ilzorkina AI, Dubinin MV, Mikheeva IB, Belosludtsev KN. Comparative Study of Structural and Functional Rearrangements in Skeletal Muscle Mitochondria of SOD1-G93A Transgenic Mice at Pre-, Early-, and Late-Symptomatic Stages of ALS Progression. FRONT BIOSCI-LANDMRK 2025; 30:28260. [PMID: 40152389 DOI: 10.31083/fbl28260] [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: 11/16/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive multisystem disease characterized by limb and trunk muscle weakness that is attributed, in part, to abnormalities in mitochondrial ultrastructure and impaired mitochondrial functions. This study investigated the time course of structural and functional rearrangements in skeletal muscle mitochondria in combination with motor impairments in Tg (copper-zinc superoxide dismutase enzyme (SOD1) G93A) dl1/GurJ (referred to as SOD1-G93A/low) male mice, a familial ALS model, as compared with non-transgenic littermates. METHODS The neurological status and motor functions were assessed weekly using the paw grip endurance method and the grid suspension test with two-limb and four-limb suspension tasks. Transmission electron microscopy followed by quantitative analysis was performed to study ultrastructural alterations in the quadriceps femoris. Functional analysis of skeletal muscle mitochondria was performed using high-resolution Oxygraph-2k (O2K) respirometry and methods for assessing the calcium retention capacity index and the content of lipid peroxidation products in freshly isolated preparations. RESULTS Based on the behavioral phenotyping data, specific age groups were identified: postnatal day 56 (P56) (n = 10-11), 84 (P84) (n = 10-11), and 156 (P154) (n = 10-12), representing the pre-symptomatic, early-symptomatic and late-symptomatic stages of ALS progression in SOD1-G93A/low mice, respectively. Electron microscopy showed mosaic destructive changes in subsarcolemmal mitochondria in fibers of the quadriceps femoris from 84-day-old SOD1-G93A/low mice. Morphometric analysis revealed an elevation in the mean size of the mitochondria in SOD1-G93A mice at P84 and P154. In addition, the P154 transgenic group demonstrated a decrease in sarcomere width and the number of mitochondria per unit area. At the symptomatic stage, SOD1-G93A mice exhibited a decreased respiratory control ratio, ADP-stimulated, and uncoupled respiration rates of mitochondria isolated from the quadriceps femoris muscle, as measured by high-resolution respirometry. In parallel, the mitochondria showed lower calcium retention capacity and increased levels of lipid peroxidation products compared with the control. CONCLUSIONS Taken together, these results indicate stage-dependent changes in skeletal muscle mitochondrial ultrastructure and functions associated with defective oxidative phosphorylation, impaired calcium homeostasis, and oxidative damage in the SOD1-G93A/low mouse model, which appears to be a promising direction for the development of combination therapies for ALS.
Collapse
Affiliation(s)
- Natalia V Belosludtseva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Anna I Ilzorkina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Mikhail V Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, 424001 Yoshkar-Ola, Russia
| | - Irina B Mikheeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Konstantin N Belosludtsev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| |
Collapse
|
2
|
Rosina M, Scaricamazza S, Fenili G, Nesci V, Valle C, Ferri A, Paronetto MP. Hidden players in the metabolic vulnerabilities of amyotrophic lateral sclerosis. Trends Endocrinol Metab 2025:S1043-2760(25)00044-X. [PMID: 40090808 DOI: 10.1016/j.tem.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/18/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex and rapidly progressive motor neuron disorder with a fatal outcome. Despite the remarkable progress in understanding ALS pathophysiology, which has significantly contributed to clinical trial design, ALS remains a rapidly disabling and life-shortening condition. The non-motor neuron features of ALS, including nutritional status, energy expenditure, and metabolic imbalance, are increasingly gaining attention. Indeed, the bioenergetic failure and mitochondrial dysfunction of patients with ALS impact not only the high energy-demanding motor neurons but also organs and brain areas long considered irrelevant to the disease. As such, here we discuss how considering energy balance in ALS is reshaping research on this disease, opening the path to novel targetable opportunities for its treatment.
Collapse
Affiliation(s)
- Marco Rosina
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; Department of Neuroscience, Italian National Institute of Health (ISS), Rome, Italy
| | - Silvia Scaricamazza
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Gianmarco Fenili
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; Department of Movement, Human, and Health Sciences, University of Rome 'Foro Italico', Rome, Italy
| | - Valentina Nesci
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, University of Roma 'Tor Vergata', Rome, Italy
| | - Cristiana Valle
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Alberto Ferri
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy.
| | - Maria Paola Paronetto
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, Rome, Italy; Department of Movement, Human, and Health Sciences, University of Rome 'Foro Italico', Rome, Italy.
| |
Collapse
|
3
|
Rosina M, Scaricamazza S, Riggio F, Fenili G, Giannessi F, Matteocci A, Nesci V, Salvatori I, Angelini DF, Aquilano K, Chiurchiù V, Barbato DL, Mercuri NB, Valle C, Ferri A. Brown Adipose Tissue undergoes pathological perturbations and shapes C2C12 myoblast homeostasis in the SOD1-G93A mouse model of Amyotrophic Lateral Sclerosis. Heliyon 2025; 11:e41801. [PMID: 39916853 PMCID: PMC11800085 DOI: 10.1016/j.heliyon.2025.e41801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/04/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by the selective loss of motor neurons. The contribution of peripheral organs remains incompletely understood. We focused our attention on brown adipose tissue (BAT) and its secreted extracellular vesicles (EVs) given their role in regulating systemic energy balance. In this study, we employed a multi-omics approach, including RNA sequencing (GEO identifier GSE273052) and proteomics (ProteomeXchange identifier PXD054147), to investigate the alterations in BAT and its EVs in the SOD1-G93A mouse model of ALS. Our results revealed consistent changes in the proteomic and transcriptomic profiles of BAT from SOD1-G93A mice, highlighting alterations such as mitochondrial dysfunction and impaired differentiation capacity. Specifically, primary brown adipocytes (PBAs) from SOD1-G93A mice exhibited differentiation impairment, respiratory defects, and alterations in mitochondrial dynamics. Furthermore, the BAT-derived EVs from SOD1-G93A mice displayed distinct changes in size distribution and cargo content. In parallel, such EVs negatively impacted the differentiation and homeostasis of C2C12 murine myoblasts, as well as induced atrophy in C2C12-derived myotubes. These findings suggest that BAT undergoes pathological perturbations in ALS mouse model and could impact on skeletal muscle homeostasis through the secretion of dysfunctional EVs.
Collapse
Affiliation(s)
- Marco Rosina
- Unit of Neurology, Fondazione PTV Policlinico Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Silvia Scaricamazza
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Via del fosso del cavaliere 100, 00133, Rome, Italy
| | - Flaminia Riggio
- Department of Biology and Biotechnology “Charles Darwin”, University of Roma “La Sapienza”, 00161, Rome, Italy
| | - Gianmarco Fenili
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Department of Movement, Human and Health Sciences University of Rome "Foro Italico”, Piazza Lauro de Bosis 6, 00135, Rome, Italy
| | - Flavia Giannessi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146, Rome, Italy
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- PhD program in Immunology, Molecular Medicine and Applied biotechnologies, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Valentina Nesci
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Department of Systems Medicine, University of Roma "Tor Vergata", 00133, Rome, Italy
| | - Illari Salvatori
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Department of Experimental Medicine, University of Roma "La Sapienza", 00161, Rome, Italy
| | - Daniela F. Angelini
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome “Tor Vergata”, via della ricerca scientifica, 00133, Rome, Italy
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, Via del fosso del cavaliere 100, 00133, Rome, Italy
- Laboratory of Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Daniele Lettieri Barbato
- Department of Biology, University of Rome “Tor Vergata”, via della ricerca scientifica, 00133, Rome, Italy
| | - Nicola Biagio Mercuri
- Unit of Neurology, Fondazione PTV Policlinico Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
- Department of Systems Medicine, University of Roma "Tor Vergata", 00133, Rome, Italy
- Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
| | - Cristiana Valle
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Via del fosso del cavaliere 100, 00133, Rome, Italy
| | - Alberto Ferri
- Laboratory of Neurochemistry, IRCCS Fondazione Santa Lucia, Via del fosso di fiorano 64, 00143, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Via del fosso del cavaliere 100, 00133, Rome, Italy
| |
Collapse
|
4
|
van Eijk RPA, Steyn FJ, Janse van Mantgem MR, Schmidt A, Meyjes M, Allen S, Daygon DV, Loeffler JP, Al-Chalabi A, van den Berg LH, Henderson RD, Ngo ST. An open-label Phase 2a study to assess the safety and tolerability of trimetazidine in patients with amyotrophic lateral sclerosis. Brain Commun 2025; 7:fcaf063. [PMID: 40008327 PMCID: PMC11851067 DOI: 10.1093/braincomms/fcaf063] [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: 11/08/2024] [Revised: 01/15/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic imbalance is associated with amyotrophic lateral sclerosis progression. Impaired glucose oxidation and increased reliance on fatty acid oxidation contribute to reduced metabolic flexibility and faster disease progression in amyotrophic lateral sclerosis. We sought to evaluate the safety and tolerability, and explore the pharmacodynamic response of trimetazidine, a partial fatty acid oxidation inhibitor, on oxidative stress markers and energy expenditure in amyotrophic lateral sclerosis. The study was conducted between June 29, 2021 and May 24, 2023. People living with amyotrophic lateral sclerosis, recruited in Australia and the Netherlands, received open-label oral trimetazidine for 12 weeks after an initial 4-week lead-in period. The primary outcome measures were safety and tolerability, as well as the change from baseline in oxidative stress markers malondialdehyde (MDA) and 8-hydroxy-2'-deoxyguanosine (8-OHdG). Secondary outcome measures were change from baseline in energy expenditure, amyotrophic lateral sclerosis functional rating scale-revised, and slow vital capacity (SVC). Linear mixed effects were used to estimate the mean difference in MDA and 8-OHdG between the on- and off-treatment periods. This trial is registered under ClinicalTrial.gov National Clinical Trial (NCT) number NCT04788745 and European Union Drug Regulating Authorities Clinical Trials (EudraCT) number 2020-005018-17. Twenty-one participants received trimetazidine; 19 (90%) completed the treatment period. Trimetazidine was well tolerated; there were 57 adverse events reported, of which 7 (11%) were deemed potentially drug-related, including hot flushes (2), nausea (2), paraesthesia (2) and fatigue (1). MDA was numerically lower during treatment [-0.29 uM; 95% confidence interval (CI) -0.90 to 0.33, P = 0.36]; 8-OHdG was significantly lower during treatment (-0.12 nM; 95% CI -0.23 to -0.01, P = 0.0245). The decrease in oxidative stress markers was accompanied by a reduction in resting energy expenditure (95 kcal, 95% CI 36.8-154, P = 0.0014). The absence of a placebo group prevented the interpretation of the clinical parameters. Oral trimetazidine was safe and well tolerated among patients with amyotrophic lateral sclerosis. This, combined with the significant reduction in markers of oxidative stress and resting energy expenditure, warrants a larger double-blind placebo-controlled efficacy study.
Collapse
Affiliation(s)
- Ruben P A van Eijk
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Biostatistics and Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Frederik J Steyn
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane 4072, Australia
- Department of Neurology, Royal Brisbane and Women’s Hospital, Herston, Brisbane 4006, Australia
| | - Mark R Janse van Mantgem
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Angela Schmidt
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane 4072, Australia
| | - Myrte Meyjes
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Sally Allen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane 4072, Australia
| | - Dara V Daygon
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane 4072, Australia
- Queensland Metabolomics and Proteomics Facility, The University of Queensland, St Lucia, Brisbane 4072, Australia
| | - Jean-Philippe Loeffler
- Centre de Recherche de Biomédecine de Strasbourg (CRBS), Université de Strasbourg, 67000 Strasbourg, France
- INSERM, U1118, Central and Peripheral Mechanisms of Neurodegeneration, 67085 Strasbourg, France
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, King's College London, London SE5 9RX, UK
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Robert D Henderson
- Department of Neurology, Royal Brisbane and Women’s Hospital, Herston, Brisbane 4006, Australia
| | - Shyuan T Ngo
- Department of Neurology, Royal Brisbane and Women’s Hospital, Herston, Brisbane 4006, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane 4072, Australia
| |
Collapse
|
5
|
Singh D, Oladimeji-Salami J, Akindele AJ. New insights on pharmacological and therapeutic potentials of trimetazidine beyond anti-anginal drug: A comprehensive review. Eur J Pharmacol 2024; 985:177062. [PMID: 39427862 DOI: 10.1016/j.ejphar.2024.177062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Trimetazidine (TMZ) is a beneficial and well-tolerable anti-anginal drug which has protective action towards ischemia and reperfusion injury. TMZ performs its anti-ischemic effect by modifying cardiac metabolism without shifting the hemodynamic functions, so it represents an outstanding complementary perspective to the general angina treatment. TMZ possesses a positive impact on the inflammatory profile, and also endothelial function furthermore displays various benefits through minimising the number, as well as the intensity of angina strikes and ameliorating the clinical indication and symptoms of myocardium ischemia. It is administrated as monotherapy along with a combination of different antianginal drugs. Apart from anti-angina action, in recent years TMZ has shown various pharmacological activities such as neuroprotective, renal protective, hepato-protective, cardio-protective effects, and other beneficial pharmacological activities. We select to write the present review article to cover the different pharmacological and therapeutic potentials of TMZ.
Collapse
Affiliation(s)
- Dhirendra Singh
- Department of Pharmacology, M.M College of Pharmacy, Maharishi Markandeshwar Mullana, Ambala, Haryana, India.
| | - Joy Oladimeji-Salami
- Medical Biotechnology Department, National Biotechnology Research and Development Agency, Abuja, Nigeria
| | - Abidemi James Akindele
- Department of Pharmacology, Therapeutics & Toxicology, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Idi-Araba, P.M.B. 12003, Lagos, Nigeria.
| |
Collapse
|
6
|
Baduini IR, Castro Vildosola JE, Kavehmoghaddam S, Kiliç F, Nadeem SA, Nizama JJ, Rowand MA, Annapureddy D, Bryan CA, Do LH, Hsiao S, Jonnalagadda SA, Kasturi A, Mandava N, Muppavaram S, Ramirez B, Siner A, Suoto CN, Tamajal N, Scoma ER, Da Costa RT, Solesio ME. Type 2 diabetes mellitus and neurodegenerative disorders: The mitochondrial connection. Pharmacol Res 2024; 209:107439. [PMID: 39357690 DOI: 10.1016/j.phrs.2024.107439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
The incidence of type 2 diabetes mellitus (T2DM) has increased in our society in recent decades as the population ages, and this trend is not expected to revert. This is the same for the incidence of the main neurodegenerative disorders, including the two most common ones, which are, Alzheimer's and Parkinson's disease. Currently, no pharmacological therapies have been developed to revert or cure any of these pathologies. Interestingly, in recent years, an increased number of studies have shown a high co-morbidity between T2DM and neurodegeneration, as well as some common molecular pathways that are affected in both types of diseases. For example, while the etiopathology of T2DM and neurodegenerative disorders is highly complex, mitochondrial dysfunction has been broadly described in the early steps of both diseases; accordingly, this dysfunction has emerged as a plausible molecular link between them. In fact, the prominent role played by mitochondria in the mammalian metabolism of glucose places the physiology of the organelle in a central position to regulate many cellular processes that are affected in both T2DM and neurodegenerative disorders. In this collaborative review, we critically describe the relationship between T2DM and neurodegeneration; making a special emphasis on the mitochondrial mechanisms that could link these diseases. A better understanding of the role of mitochondria on the etiopathology of T2DM and neurodegeneration could pave the way for the development of new pharmacological therapies focused on the regulation of the physiology of the organelle. These therapies could, ultimately, contribute to increase healthspan.
Collapse
Affiliation(s)
- Isabella R Baduini
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Jose E Castro Vildosola
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Sheida Kavehmoghaddam
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Fatmanur Kiliç
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - S Aiman Nadeem
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Juan J Nizama
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Marietta A Rowand
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Dileep Annapureddy
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Chris-Ann Bryan
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Lisa H Do
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Samuel Hsiao
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Sai A Jonnalagadda
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Akhila Kasturi
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Nikhila Mandava
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Sachin Muppavaram
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Bryan Ramirez
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Aleece Siner
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Christina N Suoto
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Nasira Tamajal
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Ernest R Scoma
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Renata T Da Costa
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Maria E Solesio
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA.
| |
Collapse
|
7
|
Mao M, Zeng W, Zheng Y, Fan W, Yao Y. Fasudil attenuates syncytin-1-mediated activation of microglia and impairments of motor neurons and motor function in mice. Drug Dev Res 2024; 85:e22254. [PMID: 39234934 DOI: 10.1002/ddr.22254] [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: 04/15/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease. Syncytin-1 (Syn), an envelope glycoprotein encoded by the env gene of the human endogenous retrovirus-W family, has been resorted to be highly expressed in biopsies from the muscles from ALS patients; however, the specific regulatory role of Syn during ALS progression remains uncovered. In this study, C57BL/6 mice were injected with adeno-associated virus-overexpressing Syn, with or without Fasudil administration. The Syn expression was assessed by quantitative real-time polymerase chain reaction and immunohistochemistry analysis. The histological change of anterior tibial muscles was determined by hematoxylin-eosin staining. Qualitative ultrastructural analysis of electron micrographs obtained from lumbar spinal cords was carried out. Serum inflammatory cytokines were assessed by enzyme linked immunosorbent assay (ELISA) assay and motor function was recorded using Basso, Beattie, and Bresnahan (BBB) scoring, climbing test and treadmill running test. Immunofluorescence and western blot assays were conducted to examine microglial- and motor neurons-related proteins. Syn overexpression significantly caused systemic inflammatory response, muscle tissue lesions, and motor dysfunction in mice. Meanwhile, Syn overexpression promoted the impairment of motor neuron, evidenced by the damaged structure of the neurons and reduced expression of microtubule-associated protein 2, HB9, neuronal nuclei and neuron-specific enolase in Syn-induced mice. In addition, Syn overexpression greatly promoted the expression of CD16/CD32 and inducible nitric oxide synthase (M1 phenotype markers), and reduced the expression of CD206 and arginase 1 (M2 phenotype markers). Importantly, the above changes caused by Syn overexpression were partly abolished by Fasudil administration. This study provides evidence that Syn-activated microglia plays a pivotal role during the progression of ALS.
Collapse
Affiliation(s)
- Mei Mao
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Wen Zeng
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yan Zheng
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Wen Fan
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yuanrong Yao
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
8
|
Gu D, Xia Y, Ding Z, Qian J, Gu X, Bai H, Jiang M, Yao D. Inflammation in the Peripheral Nervous System after Injury. Biomedicines 2024; 12:1256. [PMID: 38927464 PMCID: PMC11201765 DOI: 10.3390/biomedicines12061256] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Nerve injury is a common condition that occurs as a result of trauma, iatrogenic injury, or long-lasting stimulation. Unlike the central nervous system (CNS), the peripheral nervous system (PNS) has a strong capacity for self-repair and regeneration. Peripheral nerve injury results in the degeneration of distal axons and myelin sheaths. Macrophages and Schwann cells (SCs) can phagocytose damaged cells. Wallerian degeneration (WD) makes the whole axon structure degenerate, creating a favorable regenerative environment for new axons. After nerve injury, macrophages, neutrophils and other cells are mobilized and recruited to the injury site to phagocytose necrotic cells and myelin debris. Pro-inflammatory and anti-inflammatory factors involved in the inflammatory response provide a favorable microenvironment for peripheral nerve regeneration and regulate the effects of inflammation on the body through relevant signaling pathways. Previously, inflammation was thought to be detrimental to the body, but further research has shown that appropriate inflammation promotes nerve regeneration, axon regeneration, and myelin formation. On the contrary, excessive inflammation can cause nerve tissue damage and pathological changes, and even lead to neurological diseases. Therefore, after nerve injury, various cells in the body interact with cytokines and chemokines to promote peripheral nerve repair and regeneration by inhibiting the negative effects of inflammation and harnessing the positive effects of inflammation in specific ways and at specific times. Understanding the interaction between neuroinflammation and nerve regeneration provides several therapeutic ideas to improve the inflammatory microenvironment and promote nerve regeneration.
Collapse
Affiliation(s)
- Dandan Gu
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Yiming Xia
- Medical School, Nantong University, Nantong 226001, China
| | - Zihan Ding
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Jiaxi Qian
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Xi Gu
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Huiyuan Bai
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Maorong Jiang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Dengbing Yao
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| |
Collapse
|
9
|
Janse van Mantgem MR, Soors D'Ancona ML, Meyjes M, Van Den Berg LH, Steenhagen E, Kok A, Van Eijk RPA. A comparison between bioelectrical impedance analysis and air-displacement plethysmography in assessing fat-free mass in patients with motor neurone diseases: a cross-sectional study. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:326-335. [PMID: 38265049 DOI: 10.1080/21678421.2023.2300963] [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: 09/07/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024]
Abstract
AIM To determine the validity of bioelectrical impedance analysis (BIA) in quantifying fat-free mass (FFM) compared to air-displacement plethysmography (ADP) in patients with a motor neurone disease (MND). METHODS FFM of 140 patients diagnosed with MND was determined by ADP using the BodPod (i.e. the gold standard), and by BIA using the whole-body Bodystat. FFM values were translated to predicted resting energy expenditure (REE); the actual REE was measured using indirect calorimetry, resulting in a metabolic index. Validity of the BIA compared to the ADP was assessed using Bland-Altman analysis and Pearson's r. To assess the clinical relevance of differences, we evaluated changes in metabolic index and in individualized protein demand. RESULTS Despite the high correlation between ADP and BIA (r = 0.93), averaged across patients, the assessed mean fat-free mass was 51.7 kg (± 0.9) using ADP and 54.2 kg (± 1.0) using BIA. Hence, BIA overestimated fat-free mass by 2.5 kg (95% CI 1.8-3.2, p < 0.001). Clinically, an increased metabolic index would be more often underdiagnosed in patients with MND using BIA (31.4% according to BIA versus 44.2% according to ADP, p = 0.048). A clinically relevant overestimation of ≥ 15 g in protein demand was observed for 4 (2.9%) patients using BIA. CONCLUSIONS BIA systematically overestimates FFM in patients with MND. Although the differences are limited with ADP, underscoring the utility of BIA for research, overestimation of fat-free mass may have consequences for clinical decision-making, especially when interest lies in determining the metabolic index.
Collapse
Affiliation(s)
- Mark R Janse van Mantgem
- Department of Neurology, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Maaike L Soors D'Ancona
- Department of Neurology, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Dietetics, University Medical Centre Utrecht, Utrecht, The Netherlands, and
| | - Myrte Meyjes
- Department of Neurology, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Leonard H Van Den Berg
- Department of Dietetics, University Medical Centre Utrecht, Utrecht, The Netherlands, and
| | - Elles Steenhagen
- Department of Dietetics, University Medical Centre Utrecht, Utrecht, The Netherlands, and
| | - Annemieke Kok
- Department of Dietetics, University Medical Centre Utrecht, Utrecht, The Netherlands, and
| | - Ruben P A Van Eijk
- Department of Dietetics, University Medical Centre Utrecht, Utrecht, The Netherlands, and
- Biostatistics & Research Support, Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
10
|
Scaricamazza S, Nesci V, Salvatori I, Fenili G, Rosina M, Gloriani M, Paronetto MP, Madaro L, Ferri A, Valle C. Endurance exercise has a negative impact on the onset of SOD1-G93A ALS in female mice and affects the entire skeletal muscle-motor neuron axis. Front Pharmacol 2024; 15:1360099. [PMID: 38590640 PMCID: PMC10999529 DOI: 10.3389/fphar.2024.1360099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/04/2024] [Indexed: 04/10/2024] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease characterized by the degeneration of motor neurons that leads to muscle wasting and atrophy. Epidemiological and experimental evidence suggests a causal relationship between ALS and physical activity (PA). However, the impact of PA on motor neuron loss and sarcopenia is still debated, probably because of the heterogeneity and intensities of the proposed exercises. With this study, we aimed to clarify the effect of intense endurance exercise on the onset and progression of ALS in the SOD1-G93A mouse model. Methods We randomly selected four groups of twelve 35-day-old female mice. SOD1-G93A and WT mice underwent intense endurance training on a motorized treadmill for 8 weeks, 5 days a week. During the training, we measured muscle strength, weight, and motor skills and compared them with the corresponding sedentary groups to define the disease onset. At the end of the eighth week, we analyzed the skeletal muscle-motor neuron axis by histological and molecular techniques. Results Intense endurance exercise anticipates the onset of the disease by 1 week (age of the onset: trained SOD1-G93A = 63.17 ± 2.25 days old; sedentary SOD1-G93A = 70.75 ± 2.45 days old). In SOD1-G93A mice, intense endurance exercise hastens the muscular switch to a more oxidative phenotype and worsens the denervation process by dismantling neuromuscular junctions in the tibialis anterior, enhancing the Wallerian degeneration in the sciatic nerve, and promoting motor neuron loss in the spinal cord. The training exacerbates neuroinflammation, causing immune cell infiltration in the sciatic nerve and a faster activation of astrocytes and microglia in the spinal cord. Conclusion Intense endurance exercise, acting on skeletal muscles, worsens the pathological hallmarks of ALS, such as denervation and neuroinflammation, brings the onset forward, and accelerates the progression of the disease. Our findings show the potentiality of skeletal muscle as a target for both prognostic and therapeutic strategies; the preservation of skeletal muscle health by specific intervention could counteract the dying-back process and protect motor neurons from death. The physiological characteristics and accessibility of skeletal muscle further enhance its appeal as a therapeutic target.
Collapse
Affiliation(s)
| | - Valentina Nesci
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Systems Medicine, University of Roma “Tor Vergata”, Rome, Italy
| | - Illari Salvatori
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Experimental Medicine, University of Roma “La Sapienza”, Rome, Italy
| | - Gianmarco Fenili
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Marco Rosina
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Neurology Unit, PTV Foundation Tor Vergata University Hospital, Rome, Italy
| | - Michela Gloriani
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Roma “La Sapienza”, Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Maria Paola Paronetto
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Luca Madaro
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Roma “La Sapienza”, Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alberto Ferri
- IRCCS Fondazione Santa Lucia, Rome, Italy
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Cristiana Valle
- IRCCS Fondazione Santa Lucia, Rome, Italy
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| |
Collapse
|
11
|
Hassan FE, Aboulhoda BE, Ali IH, Elwi HM, Matter LM, Abdallah HA, Khalifa MM, Selmy A, Alghamdi MA, Morsy SA, Al Dreny BA. Evaluating the protective role of trimetazidine versus nano-trimetazidine in amelioration of bilateral renal ischemia/reperfusion induced neuro-degeneration: Implications of ERK1/2, JNK and Galectin-3 /NF-κB/TNF-α/HMGB-1 signaling. Tissue Cell 2023; 85:102241. [PMID: 37865040 DOI: 10.1016/j.tice.2023.102241] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/24/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Renal ischemia/reperfusion (I/R) is a primary culprit of acute kidney injury. Neurodegeneration can result from I/R, but the mechanisms are still challenging. We studied the implications of bilateral renal I/R on brain and potential involvement of the oxidative stress (OS) driven extracellular signal-regulated kinase1/2, c-Jun N-terminal kinase (ERK1/2, JNK) and Galectin-3 (Gal-3)/nuclear factor Kappa B (NF-қB)/tumor necrosis factor-alpha (TNF-α), high mobility group box-1 (HMGB-1), and caspase-3 paths upregulation. We tested the impact of Nano-trimetazidine (Nano-TMZ) on these pathways being a target of its neuroprotective effects. METHODS Study groups; Sham, I/R, TMZ+I/R, and Nano-TMZ+I/R. Kidney functions, cognition, hippocampal OS markers, Gal-3, NF-қB, p65 and HMGB-1 gene expression, TNF-α level, t-JNK/p-JNK and t-ERK/p-ERK proteins, caspase-3, glial fibrillary acidic protein (GFAP) and ionized calcium binding protein-1 (Iba-1) were assessed. RESULTS Nano-TMZ averted renal I/R-induced hippocampal impairment by virtue of its anti: oxidative, inflammatory, and apoptotic properties. CONCLUSION Nano-TMZ is more than anti-ischemic.
Collapse
Affiliation(s)
- Fatma E Hassan
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt; General Medicine Practice Program, Department of Physiology, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Basma Emad Aboulhoda
- Anatomy and Embryology Department, Faculty of Medicine, Cairo University, Egypt.
| | - Isra H Ali
- Department of Pharmaceutics, Faculty of Pharmacy, University of Sadat City, P.O. Box 32897, Sadat City, Egypt; Nanomedicine Laboratory, Faculty of Pharmacy, University of Sadat City, P.O. Box 32897, Sadat City, Egypt
| | - Heba M Elwi
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Lamiaa M Matter
- Medical pharmacology, Kasr Alainy, Faculty of Medicine, Cairo University, Egypt
| | - Hend Ahmed Abdallah
- Anatomy and Embryology Department, Faculty of Medicine, Cairo University, Egypt
| | - Mohamed Mansour Khalifa
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt; Department of Human Physiology, College of Medicine, King Saud University, Saudi Arabia
| | - Asmaa Selmy
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt
| | - Mansour A Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia; Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | - Suzan Awad Morsy
- Fakeeh College For Medical Sciences, Jeddah, Saudi Arabia; Faculty of Medicine, Alexandria University, Egypt
| | - Basant A Al Dreny
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt
| |
Collapse
|
12
|
Piano I, Votta A, Colucci P, Corsi F, Vitolo S, Cerri C, Puppi D, Lai M, Maya-Vetencourt JF, Leigheb M, Gabellini C, Ferraro E. Anti-inflammatory reprogramming of microglia cells by metabolic modulators to counteract neurodegeneration; a new role for Ranolazine. Sci Rep 2023; 13:20138. [PMID: 37978212 PMCID: PMC10656419 DOI: 10.1038/s41598-023-47540-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
Microglia chronic activation is a hallmark of several neurodegenerative diseases, including the retinal ones, possibly contributing to their etiopathogenesis. However, some microglia sub-populations have anti-inflammatory and neuroprotective functions, thus making arduous deciphering the role of these cells in neurodegeneration. Since it has been proposed that functionally different microglia subsets also rely on different metabolic routes, we hypothesized that modulating microglia metabolism might be a tool to enhance their anti-inflammatory features. This would have a preventive and therapeutic potential in counteracting neurodegenerative diseases. For this purpose, we tested various molecules known to act on cell metabolism, and we revealed the anti-inflammatory effect of the FDA-approved piperazine derivative Ranolazine on microglia cells, while confirming the one of the flavonoids Quercetin and Naringenin, both in vitro and in vivo. We also demonstrated the synergistic anti-inflammatory effect of Quercetin and Idebenone, and the ability of Ranolazine, Quercetin and Naringenin to counteract the neurotoxic effect of LPS-activated microglia on 661W neuronal cells. Overall, these data suggest that using the selected molecules -also in combination therapies- might represent a valuable approach to reduce inflammation and neurodegeneration while avoiding long term side effects of corticosteroids.
Collapse
Affiliation(s)
- Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Arianna Votta
- Department of Biology, University of Pisa, Pisa, Italy
| | | | | | - Sara Vitolo
- Department of Biology, University of Pisa, Pisa, Italy
| | - Chiara Cerri
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Dario Puppi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Michele Lai
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - José Fernando Maya-Vetencourt
- Department of Biology, University of Pisa, Pisa, Italy
- Centre for Synaptic Neuroscience, Italian Institute of Technology (IIT), Genova, Italy
| | - Massimiliano Leigheb
- Orthopaedics and Traumatology Unit, "Maggiore della Carità" Hospital, Department of Health Sciences, University of Piemonte Orientale (UPO), Novara, Italy
| | | | | |
Collapse
|
13
|
Shefner JM, Musaro A, Ngo ST, Lunetta C, Steyn FJ, Robitaille R, De Carvalho M, Rutkove S, Ludolph AC, Dupuis L. Skeletal muscle in amyotrophic lateral sclerosis. Brain 2023; 146:4425-4436. [PMID: 37327376 PMCID: PMC10629757 DOI: 10.1093/brain/awad202] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS), the major adult-onset motor neuron disease, has been viewed almost exclusively as a disease of upper and lower motor neurons, with muscle changes interpreted as a consequence of the progressive loss of motor neurons and neuromuscular junctions. This has led to the prevailing view that the involvement of muscle in ALS is only secondary to motor neuron loss. Skeletal muscle and motor neurons reciprocally influence their respective development and constitute a single functional unit. In ALS, multiple studies indicate that skeletal muscle dysfunction might contribute to progressive muscle weakness, as well as to the final demise of neuromuscular junctions and motor neurons. Furthermore, skeletal muscle has been shown to participate in disease pathogenesis of several monogenic diseases closely related to ALS. Here, we move the narrative towards a better appreciation of muscle as a contributor of disease in ALS. We review the various potential roles of skeletal muscle cells in ALS, from passive bystanders to active players in ALS pathophysiology. We also compare ALS to other motor neuron diseases and draw perspectives for future research and treatment.
Collapse
Affiliation(s)
- Jeremy M Shefner
- Barrow Neurological Institute, Phoenix, AZ, USA
- College of Medicine, University of Arizona, Phoenix, AZ, USA
- College of Medicine, Creighton University, Phoenix, AZ, USA
| | - Antonio Musaro
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Scuola Superiore di Studi Avanzati Sapienza (SSAS), Rome, Italy
| | - Shyuan T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Christian Lunetta
- Neurorehabilitation Department, Istituti Clinici Scientifici Maugeri IRCCS, Milan, Italy
| | - Frederik J Steyn
- Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Richard Robitaille
- Département de neurosciences, CIRCA, Université de Montréal, Montréal H7G 1T7, Canada
| | - Mamede De Carvalho
- Instituto de Fisiologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Seward Rutkove
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Albert C Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany
- Deutsches Zentrum für neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Luc Dupuis
- Université de Strasbourg, Inserm, UMR-S1118, Mécanismes centraux et périphériques de la neurodégénérescence, CRBS, Strasbourg, France
| |
Collapse
|
14
|
Ludolph A, Dupuis L, Kasarskis E, Steyn F, Ngo S, McDermott C. Nutritional and metabolic factors in amyotrophic lateral sclerosis. Nat Rev Neurol 2023; 19:511-524. [PMID: 37500993 DOI: 10.1038/s41582-023-00845-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/29/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disease that is classically thought to impact the motor system. Over the past 20 years, research has started to consider the contribution of non-motor symptoms and features of the disease, and how they might affect ALS prognosis. Of the non-motor features of the disease, nutritional status (for example, malnutrition) and metabolic balance (for example, weight loss and hypermetabolism) have been consistently shown to contribute to more rapid disease progression and/or earlier death. Several complex cellular changes observed in ALS, including mitochondrial dysfunction, are also starting to be shown to contribute to bioenergetic failure. The resulting energy depletion in high energy demanding neurons makes them sensitive to apoptosis. Given that nutritional and metabolic stressors at the whole-body and cellular level can impact the capacity to maintain optimal function, these factors present avenues through which we can identify novel targets for treatment in ALS. Several clinical trials are now underway evaluating the effectiveness of modifying energy balance in ALS, making this article timely in reviewing the evidence base for metabolic and nutritional interventions.
Collapse
Affiliation(s)
- Albert Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Site Ulm, Ulm, Germany
| | - Luc Dupuis
- Université de Strasbourg, Inserm, Mécanismes Centraux et Périphériques de la Neurodégénérescence, UMR-S1118, Centre de Recherches en Biomédecine, Strasbourg, France
| | - Edward Kasarskis
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Frederik Steyn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Shyuan Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | | |
Collapse
|
15
|
Kamranian H, Asoudeh H, Sharif RK, Taheri F, Hayes AW, Gholami M, Alavi A, Motaghinejad M. Neuroprotective potential of trimetazidine against tramadol-induced neurotoxicity: role of PI3K/Akt/mTOR signaling pathways. Toxicol Mech Methods 2023; 33:607-623. [PMID: 37051630 DOI: 10.1080/15376516.2023.2202785] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 04/14/2023]
Abstract
Tramadol (TRA) causes neurotoxicity whereas trimetazidine (TMZ) is neuroprotective. The potential involvement of the PI3K/Akt/mTOR signaling pathway in the neuroprotection of TMZ against TRA-induced neurotoxicity was evaluated. Seventy male Wistar rats were divided into groups. Groups 1 and 2 received saline or TRA (50 mg/kg). Groups 3, 4, and 5 received TRA (50 mg/kg) and TMZ (40, 80, or 160 mg/kg) for 14 days. Group 6 received TMZ (160 mg/kg). Hippocampal neurodegenerative, mitochondrial quadruple complex enzymes, phosphatidylinositol-3-kinases (PI3Ks)/protein kinase B levels, oxidative stress, inflammatory, apoptosis, autophagy, and histopathology were evaluated. TMZ decreased anxiety and depressive-like behavior induced by TRA. TMZ in tramadol-treated animals inhibited lipid peroxidation, GSSG, TNF-α, and IL-1β while increasing GSH, SOD, GPx, GR, and mitochondrial quadruple complex enzymes in the hippocampus. TRA inhibited Glial fibrillary acidic protein expression and increased pyruvate dehydrogenase levels. TMZ reduced these changes. TRA decreased the level of JNK and increased Beclin-1 and Bax. TMZ decreased phosphorylated Bcl-2 while increasing the unphosphorylated form in tramadol-treated rats. TMZ activated phosphorylated PI3Ks, Akt, and mTOR proteins. TMZ inhibited tramadol-induced neurotoxicity by modulating the PI3K/Akt/mTOR signaling pathways and its downstream inflammatory, apoptosis, and autophagy-related cascades.
Collapse
Affiliation(s)
- Houman Kamranian
- Department of Psychiatry, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Hadi Asoudeh
- Faculty of Pharmacy, Central Branch of Islamic Azad University, Tehran, Iran
| | | | - Fereshteh Taheri
- Department of Medicine, Islamic Azad University, Qom Branch, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA and Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Mina Gholami
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Alavi
- Department of Medicine, Islamic Azad University, Qom Branch, Iran
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Guareschi S, Ravasi M, Baldessari D, Pozzi S, Zaffino T, Melazzini M, Ambrosini A. The positive impact on translational research of Fondazione italiana di ricerca per la Sclerosi Laterale Amiotrofica (AriSLA), a non-profit foundation focused on amyotrophic lateral sclerosis. Convergence of ex-ante evaluation and ex-post outcomes when goals are set upfront. Front Res Metr Anal 2023; 8:1067981. [PMID: 37601533 PMCID: PMC10436489 DOI: 10.3389/frma.2023.1067981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
Charities investing on rare disease research greatly contribute to generate ground-breaking knowledge with the clear goal of finding a cure for their condition of interest. Although the amount of their investments may be relatively small compared to major funders, the advocacy groups' clear mission promotes innovative research and aggregates highly motivated and mission-oriented scientists. Here, we illustrate the case of Fondazione italiana di ricerca per la Sclerosi Laterale Amiotrofica (AriSLA), the main Italian funding agency entirely dedicated to amyotrophic lateral sclerosis research. An international benchmark analysis of publications derived from AriSLA-funded projects indicated that their mean relative citation ratio values (iCite dashboard, National Institutes of Health, U.S.) were very high, suggesting a strong influence on the referring international scientific community. An interesting trend of research toward translation based on the "triangle of biomedicine" and paper citations (iCite) was also observed. Qualitative analysis on researchers' accomplishments was convergent with the bibliometric data, indicating a high level of performance of several working groups, lines of research that speak of progression toward clinical translation, and one study that has progressed from the investigation of cellular mechanisms to a Phase 2 international clinical trial. The key elements of the success of the AriSLA investment lie in: (i) the clear definition of the objectives (research with potential impact on patients, no matter how far), (ii) a rigorous peer-review process entrusted to an international panel of experts, (iii) diversification of the portfolio with ad hoc selection criteria, which also contributed to bringing new experts and younger scientists to the field, and (iv) a close interaction of AriSLA stakeholders with scientists, who developed a strong sense of belonging. Periodic review of the portfolio of investments is a vital practice for funding agencies. Sharing information between funding agencies about their own policies and research assessment methods and outcomes help guide the international debate on funding strategies and research directions to be undertaken, particularly in the field of rare diseases, where synergy is a relevant enabling factor.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anna Ambrosini
- Fondazione AriSLA ETS, Milan, Italy
- Fondazione Telethon ETS, Milan, Italy
| |
Collapse
|
17
|
Barone C, Qi X. Altered Metabolism in Motor Neuron Diseases: Mechanism and Potential Therapeutic Target. Cells 2023; 12:1536. [PMID: 37296656 PMCID: PMC10252517 DOI: 10.3390/cells12111536] [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: 04/08/2023] [Revised: 05/21/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Motor Neuron Diseases (MND) are neurological disorders characterized by a loss of varying motor neurons resulting in decreased physical capabilities. Current research is focused on hindering disease progression by determining causes of motor neuron death. Metabolic malfunction has been proposed as a promising topic when targeting motor neuron loss. Alterations in metabolism have also been noted at the neuromuscular junction (NMJ) and skeletal muscle tissue, emphasizing the importance of a cohesive system. Finding metabolism changes consistent throughout both neurons and skeletal muscle tissue could pose as a target for therapeutic intervention. This review will focus on metabolic deficits reported in MNDs and propose potential therapeutic targets for future intervention.
Collapse
Affiliation(s)
| | - Xin Qi
- Department of Physiology and Biophysics, School of Medicine Case Western Reserve University, Cleveland, OH 44106-4970, USA;
| |
Collapse
|
18
|
Bolborea M, Vercruysse P, Daria T, Reiners JC, Alami NO, Guillot SJ, Dieterlé S, Sinniger J, Scekic-Zahirovic J, Londo A, Arcay H, Goy MA, de Tapia CN, Thal DR, Shibuya K, Otani R, Arai K, Kuwabara S, Ludolph AC, Roselli F, Yilmazer-Hanke D, Dupuis L. Loss of hypothalamic MCH decreases food intake in amyotrophic lateral sclerosis. Acta Neuropathol 2023; 145:773-791. [PMID: 37058170 PMCID: PMC10175407 DOI: 10.1007/s00401-023-02569-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is associated with impaired energy metabolism, including weight loss and decreased appetite which are negatively correlated with survival. Neural mechanisms underlying metabolic impairment in ALS remain unknown. ALS patients and presymptomatic gene carriers have early hypothalamic atrophy. The lateral hypothalamic area (LHA) controls metabolic homeostasis through the secretion of neuropeptides such as orexin/hypocretin and melanin-concentrating hormone (MCH). Here, we show loss of MCH-positive neurons in three mouse models of ALS based on SOD1 or FUS mutations. Supplementation with MCH (1.2 µg/d) through continuous intracerebroventricular delivery led to weight gain in male mutant Sod1G86R mice. MCH supplementation increased food intake, rescued expression of the key appetite-related neuropeptide AgRP (agouti-related protein) and modified respiratory exchange ratio, suggesting increased carbohydrate usage during the inactive phase. Importantly, we document pTDP-43 pathology and neurodegeneration in the LHA of sporadic ALS patients. Neuronal cell loss was associated with pTDP-43-positive inclusions and signs of neurodegeneration in MCH-positive neurons. These results suggest that hypothalamic MCH is lost in ALS and contributes to the metabolic changes, including weight loss and decreased appetite.
Collapse
Affiliation(s)
- Matei Bolborea
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France.
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK.
| | - Pauline Vercruysse
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France
| | - Tselmen Daria
- Clinical Neuroanatomy Section, Department of Neurology, Ulm University, Ulm, Germany
| | - Johanna C Reiners
- Clinical Neuroanatomy Section, Department of Neurology, Ulm University, Ulm, Germany
- Institute for Neurobiochemistry, Ulm University, Ulm, Germany
| | - Najwa Ouali Alami
- Clinical Neuroanatomy Section, Department of Neurology, Ulm University, Ulm, Germany
| | - Simon J Guillot
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France
| | - Stéphane Dieterlé
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France
| | - Jérôme Sinniger
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France
| | - Jelena Scekic-Zahirovic
- Department of Neurology, Neurology Clinic, Ulm University, Ulm, Germany
- Laboratory for Neuropathology, Institute for Pathology, Ulm University, Ulm, Germany
| | - Amela Londo
- Department of Neurology, Neurology Clinic, Ulm University, Ulm, Germany
- Laboratory for Neuropathology, Institute for Pathology, Ulm University, Ulm, Germany
| | - Hippolyte Arcay
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France
| | - Marc-Antoine Goy
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France
| | - Claudia Nelson de Tapia
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France
| | - Dietmar R Thal
- Laboratory for Neuropathology, Institute for Pathology, Ulm University, Ulm, Germany
- Laboratory for Neuropathology, Department of Imaging and Pathology, and Leuven Brain Institute, KU louvain, Belgium
- Department of Pathology, UZ Leuven, Japan
| | - Kazumoto Shibuya
- Department of Neurology, Chiba University School of Medicine, Chiba, Japan
| | - Ryo Otani
- Department of Neurology, Chiba University School of Medicine, Chiba, Japan
| | - Kimihito Arai
- Department of Neurology, Chiba University School of Medicine, Chiba, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Chiba University School of Medicine, Chiba, Japan
| | - Albert C Ludolph
- Department of Neurology, Neurology Clinic, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Neurology Clinic, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Deniz Yilmazer-Hanke
- Clinical Neuroanatomy Section, Department of Neurology, Ulm University, Ulm, Germany.
| | - Luc Dupuis
- Université de Strasbourg, INSERM, Mécanismes centraux et périphériques de la neurodégénérescence, UMR-S1118, Strasbourg, France.
| |
Collapse
|
19
|
Martinez-Gonzalez L, Martinez A. Emerging clinical investigational drugs for the treatment of amyotrophic lateral sclerosis. Expert Opin Investig Drugs 2023; 32:141-160. [PMID: 36762798 DOI: 10.1080/13543784.2023.2178416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder caused by motoneuron death with a median survival time of 3-5 years since disease onset. There are no effective treatments to date. However, a variety of innovative investigational drugs and biological-based therapies are under clinical development. AREAS COVERED This review provides an overview of the clinical investigational small molecules as well as a brief summary of the biological-based therapies that are currently undergoing clinical trials for the treatment of ALS. All the data were obtained from ClinicalTrials.gov (registered through November 1). EXPERT OPINION Drug discovery for ALS is an active and evolving field, where many investigational clinical drugs are in different trials. There are several mechanisms of action supporting all these new therapies, although proteostasis is gaining stage. Probably, small orally bioavailable molecules able to recover functional TDP-43 homeostasis may have solid chances to modify ALS progression.
Collapse
Affiliation(s)
- Loreto Martinez-Gonzalez
- Centro de Investigaciones Biológicas "Margarita Salas"-CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red en enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas "Margarita Salas"-CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red en enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
20
|
Aghajani A, Khakpourian Z, Bakhthiarzadeh S, Adibipour F, Sadr M, Coleman-Fuller N, Jamaati H, Motaghinejad M. Trimetazidine May Potentially Confer Neuroprotective Effects against COVID-19-Induced Neurological Sequelae via Inhibition of Death-Associated Protein Kinase 1 (DAPK1) Signaling Pathways: An Evidenced-Based Hypothesis. TANAFFOS 2023; 22:182-186. [PMID: 38628884 PMCID: PMC11016919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Affiliation(s)
- Ali Aghajani
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Khakpourian
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soodeh Bakhthiarzadeh
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Adibipour
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Makan Sadr
- Virology Research Center, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Natalie Coleman-Fuller
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN 55108.
| | - Hamidreza Jamaati
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Zhou Y, Tang J, Lan J, Zhang Y, Wang H, Chen Q, Kang Y, Sun Y, Feng X, Wu L, Jin H, Chen S, Peng Y. Honokiol alleviated neurodegeneration by reducing oxidative stress and improving mitochondrial function in mutant SOD1 cellular and mouse models of amyotrophic lateral sclerosis. Acta Pharm Sin B 2023; 13:577-597. [PMID: 36873166 PMCID: PMC9979194 DOI: 10.1016/j.apsb.2022.07.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/12/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease affecting both upper and lower motor neurons (MNs) with large unmet medical needs. Multiple pathological mechanisms are considered to contribute to the progression of ALS, including neuronal oxidative stress and mitochondrial dysfunction. Honokiol (HNK) has been reported to exert therapeutic effects in several neurologic disease models including ischemia stroke, Alzheimer's disease and Parkinson's disease. Here we found that honokiol also exhibited protective effects in ALS disease models both in vitro and in vivo. Honokiol improved the viability of NSC-34 motor neuron-like cells that expressed the mutant G93A SOD1 proteins (SOD1-G93A cells for short). Mechanistical studies revealed that honokiol alleviated cellular oxidative stress by enhancing glutathione (GSH) synthesis and activating the nuclear factor erythroid 2-related factor 2 (NRF2)-antioxidant response element (ARE) pathway. Also, honokiol improved both mitochondrial function and morphology via fine-tuning mitochondrial dynamics in SOD1-G93A cells. Importantly, honokiol extended the lifespan of the SOD1-G93A transgenic mice and improved the motor function. The improvement of antioxidant capacity and mitochondrial function was further confirmed in the spinal cord and gastrocnemius muscle in mice. Overall, honokiol showed promising preclinical potential as a multiple target drug for ALS treatment.
Collapse
Affiliation(s)
- Yujun Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jingshu Tang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jiaqi Lan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yong Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hongyue Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qiuyu Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuying Kang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yang Sun
- Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinhong Feng
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hongtao Jin
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.,NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drug, Beijing 100050, China
| | - Shizhong Chen
- Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ying Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
22
|
Pattern of Mitochondrial Respiration in Peripheral Blood Cells of Patients with Parkinson's Disease. Int J Mol Sci 2022; 23:ijms231810863. [PMID: 36142777 PMCID: PMC9506016 DOI: 10.3390/ijms231810863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Mitochondria are central in the pathogenesis of Parkinson’s disease (PD), as they are involved in oxidative stress, synaptopathy, and other immunometabolic pathways. Accordingly, they are emerging as a potential neuroprotection target, although further human-based evidence is needed for therapeutic advancements. This study aims to shape the pattern of mitochondrial respiration in the blood leukocytes of PD patients in relation to both clinical features and the profile of cerebrospinal fluid (CSF) biomarkers of neurodegeneration. Mitochondrial respirometry on the peripheral blood mononucleate cells (PBMCs) of 16 PD patients and 14 controls was conducted using Seahorse Bioscience technology. Bioenergetic parameters were correlated either with standard clinical scores for motor and non-motor disturbances or with CSF levels of α-synuclein, amyloid-β peptides, and tau proteins. In PD, PBMC mitochondrial basal respiration was normal; maximal and spare respiratory capacities were both increased; and ATP production was higher, although not significantly. Maximal and spare respiratory capacity was directly correlated with disease duration, MDS-UPDRS part III and Hoehn and Yahr motor scores; spare respiratory capacity was correlated with the CSF amyloid-β-42 to amyloid-β-42/40 ratio. We provided preliminary evidence showing that mitochondrial respiratory activity increases in the PBMCs of PD patients, probably following the compensatory adaptations to disease progression, in contrast to the bases of the neuropathological substrate.
Collapse
|
23
|
Altered TDP-43 Structure and Function: Key Insights into Aberrant RNA, Mitochondrial, and Cellular and Systemic Metabolism in Amyotrophic Lateral Sclerosis. Metabolites 2022; 12:metabo12080709. [PMID: 36005581 PMCID: PMC9415507 DOI: 10.3390/metabo12080709] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal neuromuscular disorder with no cure available and limited treatment options. ALS is a highly heterogeneous disease, whereby patients present with vastly different phenotypes. Despite this heterogeneity, over 97% of patients will exhibit pathological TAR-DNA binding protein-43 (TDP-43) cytoplasmic inclusions. TDP-43 is a ubiquitously expressed RNA binding protein with the capacity to bind over 6000 RNA and DNA targets—particularly those involved in RNA, mitochondrial, and lipid metabolism. Here, we review the unique structure and function of TDP-43 and its role in affecting the aforementioned metabolic processes in ALS. Considering evidence published specifically in TDP-43-relevant in vitro, in vivo, and ex vivo models we posit that TDP-43 acts in a positive feedback loop with mRNA transcription/translation, stress granules, cytoplasmic aggregates, and mitochondrial proteins causing a relentless cycle of disease-like pathology eventuating in neuronal toxicity. Given its undeniable presence in ALS pathology, TDP-43 presents as a promising target for mechanistic disease modelling and future therapeutic investigations.
Collapse
|
24
|
Torcinaro A, Cappetta D, De Santa F, Telesca M, Leigheb M, Berrino L, Urbanek K, De Angelis A, Ferraro E. Ranolazine Counteracts Strength Impairment and Oxidative Stress in Aged Sarcopenic Mice. Metabolites 2022; 12:663. [PMID: 35888787 PMCID: PMC9316887 DOI: 10.3390/metabo12070663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 12/02/2022] Open
Abstract
Sarcopenia is defined as the loss of muscle mass associated with reduced strength leading to poor quality of life in elderly people. The decline of skeletal muscle performance is characterized by bioenergetic impairment and severe oxidative stress, and does not always strictly correlate with muscle mass loss. We chose to investigate the ability of the metabolic modulator Ranolazine to counteract skeletal muscle dysfunctions that occur with aging. For this purpose, we treated aged C57BL/6 mice with Ranolazine/vehicle for 14 days and collected the tibialis anterior and gastrocnemius muscles for histological and gene expression analyses, respectively. We found that Ranolazine treatment significantly increased the muscle strength of aged mice. At the histological level, we found an increase in centrally nucleated fibers associated with an up-regulation of genes encoding MyoD, Periostin and Osteopontin, thus suggesting a remodeling of the muscle even in the absence of physical exercise. Notably, these beneficial effects of Ranolazine were also accompanied by an up-regulation of antioxidant and mitochondrial genes as well as of NADH-dehydrogenase activity, together with a more efficient protection from oxidative damage in the skeletal muscle. These data indicate that the protection of muscle from oxidative stress by Ranolazine might represent a valuable approach to increase skeletal muscle strength in elderly populations.
Collapse
Affiliation(s)
- Alessio Torcinaro
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Via Ercole Ramarini, 32, Monterotondo, 00015 Rome, Italy; (A.T.); (F.D.S.)
- Istituto Dermopatico dell’Immacolata (IDI), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Immunology Laboratory, Via Monti di Creta, 104, 00167 Rome, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (D.C.); (M.T.); (L.B.); (A.D.A.)
| | - Francesca De Santa
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Via Ercole Ramarini, 32, Monterotondo, 00015 Rome, Italy; (A.T.); (F.D.S.)
| | - Marialucia Telesca
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (D.C.); (M.T.); (L.B.); (A.D.A.)
| | - Massimiliano Leigheb
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Department of Health Sciences, University of Piemonte Orientale (UPO), 28100 Novara, Italy;
| | - Liberato Berrino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (D.C.); (M.T.); (L.B.); (A.D.A.)
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80138 Naples, Italy;
- CEINGE-Advanced Biotechnologies, 80138 Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (D.C.); (M.T.); (L.B.); (A.D.A.)
| | | |
Collapse
|
25
|
de Sire A, Ferraro E, Leigheb M. Advance in the Diagnostics and Management of Musculoskeletal Diseases. Diagnostics (Basel) 2022; 12:1588. [PMID: 35885494 PMCID: PMC9325206 DOI: 10.3390/diagnostics12071588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/02/2022] Open
Abstract
Musculoskeletal disorders are a broad spectrum of diseases, affecting muscles, bones, ligaments, and tendons worldwide [...].
Collapse
Affiliation(s)
- Alessandro de Sire
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
| | - Elisabetta Ferraro
- Cell and Developmental Biology Unit, Department of Biology, University of Pisa, 56127 Pisa, Italy;
| | - Massimiliano Leigheb
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy;
| |
Collapse
|
26
|
Alteration of the Neuromuscular Junction and Modifications of Muscle Metabolism in Response to Neuron-Restricted Expression of the CHMP2Bintron5 Mutant in a Mouse Model of ALS-FTD Syndrome. Biomolecules 2022; 12:biom12040497. [PMID: 35454086 PMCID: PMC9025139 DOI: 10.3390/biom12040497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/12/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
CHMP2B is a protein that coordinates membrane scission events as a core component of the ESCRT machinery. Mutations in CHMP2B are an uncommon cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two neurodegenerative diseases with clinical, genetic, and pathological overlap. Different mutations have now been identified across the ALS-FTD spectrum. Disruption of the neuromuscular junction is an early pathogenic event in ALS. Currently, the links between neuromuscular junction functionality and ALS-associated genes, such as CHMP2B, remain poorly understood. We have previously shown that CHMP2B transgenic mice expressing the CHMP2Bintron5 mutant specifically in neurons develop a progressive motor phenotype reminiscent of ALS. In this study, we used complementary approaches (behavior, histology, electroneuromyography, and biochemistry) to determine the extent to which neuron-specific expression of CHMP2Bintron5 could impact the skeletal muscle characteristics. We show that neuronal expression of the CHMP2Bintron5 mutant is sufficient to trigger progressive gait impairment associated with structural and functional changes in the neuromuscular junction. Indeed, CHMP2Bintron5 alters the pre-synaptic terminal organization and the synaptic transmission that ultimately lead to a switch of fast-twitch glycolytic muscle fibers to more oxidative slow-twitch muscle fibers. Taken together these data indicate that neuronal expression of CHMP2Bintron5 is sufficient to induce a synaptopathy with molecular and functional changes in the motor unit reminiscent of those found in ALS patients.
Collapse
|
27
|
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
|
28
|
Tarantino N, Canfora I, Camerino GM, Pierno S. Therapeutic Targets in Amyotrophic Lateral Sclerosis: Focus on Ion Channels and Skeletal Muscle. Cells 2022; 11:cells11030415. [PMID: 35159225 PMCID: PMC8834084 DOI: 10.3390/cells11030415] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Amyotrophic Lateral Sclerosis is a neurodegenerative disease caused by progressive loss of motor neurons, which severely compromises skeletal muscle function. Evidence shows that muscle may act as a molecular powerhouse, whose final signals generate in patients a progressive loss of voluntary muscle function and weakness leading to paralysis. This pathology is the result of a complex cascade of events that involves a crosstalk among motor neurons, glia, and muscles, and evolves through the action of converging toxic mechanisms. In fact, mitochondrial dysfunction, which leads to oxidative stress, is one of the mechanisms causing cell death. It is a common denominator for the two existing forms of the disease: sporadic and familial. Other factors include excitotoxicity, inflammation, and protein aggregation. Currently, there are limited cures. The only approved drug for therapy is riluzole, that modestly prolongs survival, with edaravone now waiting for new clinical trial aimed to clarify its efficacy. Thus, there is a need of effective treatments to reverse the damage in this devastating pathology. Many drugs have been already tested in clinical trials and are currently under investigation. This review summarizes the already tested drugs aimed at restoring muscle-nerve cross-talk and on new treatment options targeting this tissue.
Collapse
|