1
|
Zhong R, Rua MT, Wei-LaPierre L. Targeting mitochondrial Ca 2+ uptake for the treatment of amyotrophic lateral sclerosis. J Physiol 2024; 602:1519-1549. [PMID: 38010626 PMCID: PMC11032238 DOI: 10.1113/jp284143] [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: 09/06/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare adult-onset neurodegenerative disease characterized by progressive motor neuron (MN) loss, muscle denervation and paralysis. Over the past several decades, researchers have made tremendous efforts to understand the pathogenic mechanisms underpinning ALS, with much yet to be resolved. ALS is described as a non-cell autonomous condition with pathology detected in both MNs and non-neuronal cells, such as glial cells and skeletal muscle. Studies in ALS patient and animal models reveal ubiquitous abnormalities in mitochondrial structure and function, and disturbance of intracellular calcium homeostasis in various tissue types, suggesting a pivotal role of aberrant mitochondrial calcium uptake and dysfunctional calcium signalling cascades in ALS pathogenesis. Calcium signalling and mitochondrial dysfunction are intricately related to the manifestation of cell death contributing to MN loss and skeletal muscle dysfunction. In this review, we discuss the potential contribution of intracellular calcium signalling, particularly mitochondrial calcium uptake, in ALS pathogenesis. Functional consequences of excessive mitochondrial calcium uptake and possible therapeutic strategies targeting mitochondrial calcium uptake or the mitochondrial calcium uniporter, the main channel mediating mitochondrial calcium influx, are also discussed.
Collapse
Affiliation(s)
- Renjia Zhong
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611
- Department of Emergency Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China, 110001
| | - Michael T. Rua
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611
| | - Lan Wei-LaPierre
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611
- Myology Institute, University of Florida, Gainesville, FL 32611
| |
Collapse
|
2
|
Cunha-Oliveira T, Montezinho L, Simões RF, Carvalho M, Ferreiro E, Silva FSG. Mitochondria: A Promising Convergent Target for the Treatment of Amyotrophic Lateral Sclerosis. Cells 2024; 13:248. [PMID: 38334639 PMCID: PMC10854804 DOI: 10.3390/cells13030248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the progressive loss of motor neurons, for which current treatment options are limited. Recent studies have shed light on the role of mitochondria in ALS pathogenesis, making them an attractive therapeutic intervention target. This review contains a very comprehensive critical description of the involvement of mitochondria and mitochondria-mediated mechanisms in ALS. The review covers several key areas related to mitochondria in ALS, including impaired mitochondrial function, mitochondrial bioenergetics, reactive oxygen species, metabolic processes and energy metabolism, mitochondrial dynamics, turnover, autophagy and mitophagy, impaired mitochondrial transport, and apoptosis. This review also highlights preclinical and clinical studies that have investigated various mitochondria-targeted therapies for ALS treatment. These include strategies to improve mitochondrial function, such as the use of dichloroacetate, ketogenic and high-fat diets, acetyl-carnitine, and mitochondria-targeted antioxidants. Additionally, antiapoptotic agents, like the mPTP-targeting agents minocycline and rasagiline, are discussed. The paper aims to contribute to the identification of effective mitochondria-targeted therapies for ALS treatment by synthesizing the current understanding of the role of mitochondria in ALS pathogenesis and reviewing potential convergent therapeutic interventions. The complex interplay between mitochondria and the pathogenic mechanisms of ALS holds promise for the development of novel treatment strategies to combat this devastating disease.
Collapse
Affiliation(s)
- Teresa Cunha-Oliveira
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Liliana Montezinho
- Center for Investigation Vasco da Gama (CIVG), Escola Universitária Vasco da Gama, 3020-210 Coimbra, Portugal;
| | - Rui F. Simões
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Marcelo Carvalho
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Elisabete Ferreiro
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Filomena S. G. Silva
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Mitotag Lda, Biocant Park, 3060-197 Cantanhede, Portugal
| |
Collapse
|
3
|
Mikhail AI, Ng SY, Mattina SR, Ljubicic V. AMPK is mitochondrial medicine for neuromuscular disorders. Trends Mol Med 2023:S1471-4914(23)00070-9. [PMID: 37080889 DOI: 10.1016/j.molmed.2023.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/22/2023]
Abstract
Duchenne muscular dystrophy (DMD), myotonic dystrophy type 1 (DM1), and spinal muscular atrophy (SMA) are the most prevalent neuromuscular disorders (NMDs) in children and adults. Central to a healthy neuromuscular system are the processes that govern mitochondrial turnover and dynamics, which are regulated by AMP-activated protein kinase (AMPK). Here, we survey mitochondrial stresses that are common between, as well as unique to, DMD, DM1, and SMA, and which may serve as potential therapeutic targets to mitigate neuromuscular disease. We also highlight recent advances that leverage a mutation-agnostic strategy featuring physiological or pharmacological AMPK activation to enhance mitochondrial health in these conditions, as well as identify outstanding questions and opportunities for future pursuit.
Collapse
Affiliation(s)
- Andrew I Mikhail
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, Ontario, Canada.
| | - Sean Y Ng
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, Ontario, Canada.
| | - Stephanie R Mattina
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, Ontario, Canada.
| | - Vladimir Ljubicic
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
4
|
Lépine S, Castellanos-Montiel MJ, Durcan TM. TDP-43 dysregulation and neuromuscular junction disruption in amyotrophic lateral sclerosis. Transl Neurodegener 2022; 11:56. [PMID: 36575535 PMCID: PMC9793560 DOI: 10.1186/s40035-022-00331-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/29/2022] [Indexed: 12/28/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a disease characterized by upper and lower motor neuron (MN) loss with a signature feature of cytoplasmic aggregates containing TDP-43, which are detected in nearly all patients. Mutations in the gene that encodes TDP-43 (TARBDP) are known to result in both familial and sporadic ALS. In ALS, disruption of neuromuscular junctions (NMJs) constitutes a critical event in disease pathogenesis, leading to denervation atrophy, motor impairments and disability. Morphological defects and impaired synaptic transmission at NMJs have been reported in several TDP-43 animal models and in vitro, linking TDP-43 dysregulation to the loss of NMJ integrity in ALS. Through the lens of the dying-back and dying-forward hypotheses of ALS, this review discusses the roles of TDP-43 related to synaptic function, with a focus on the potential molecular mechanisms occurring within MNs, skeletal muscles and glial cells that may contribute to NMJ disruption in ALS.
Collapse
Affiliation(s)
- Sarah Lépine
- grid.14709.3b0000 0004 1936 8649The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, QC H3A 2B4 Canada ,grid.14709.3b0000 0004 1936 8649Faculty of Medicine and Health Sciences, McGill University, 3605 De La Montagne, Montreal, QC H3G 2M1 Canada
| | - Maria José Castellanos-Montiel
- grid.14709.3b0000 0004 1936 8649The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, QC H3A 2B4 Canada
| | - Thomas Martin Durcan
- grid.14709.3b0000 0004 1936 8649The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, QC H3A 2B4 Canada
| |
Collapse
|
5
|
Anakor E, Duddy WJ, Duguez S. The Cellular and Molecular Signature of ALS in Muscle. J Pers Med 2022; 12:1868. [PMID: 36579600 PMCID: PMC9692882 DOI: 10.3390/jpm12111868] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022] Open
Abstract
Amyotrophic lateral sclerosis is a disease affecting upper and lower motor neurons. Although motor neuron death is the core event of ALS pathology, it is increasingly recognized that other tissues and cell types are affected in the disease, making potentially major contributions to the occurrence and progression of pathology. We review here the known cellular and molecular characteristics of muscle tissue affected by ALS. Evidence of toxicity in skeletal muscle tissue is considered, including metabolic dysfunctions, impaired proteostasis, and deficits in muscle regeneration and RNA metabolism. The role of muscle as a secretory organ, and effects on the skeletal muscle secretome are also covered, including the increase in secretion of toxic factors or decrease in essential factors that have consequences for neuronal function and survival.
Collapse
Affiliation(s)
| | | | - Stephanie Duguez
- Northern Ireland Center for Personalised Medicine, School of Medicine, Ulster University, Derry-Londonderry BT47 6SB, UK
| |
Collapse
|
6
|
Zilio E, Piano V, Wirth B. Mitochondrial Dysfunction in Spinal Muscular Atrophy. Int J Mol Sci 2022; 23:10878. [PMID: 36142791 PMCID: PMC9503857 DOI: 10.3390/ijms231810878] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating neuromuscular disorder caused by recessive mutations in the SMN1 gene, globally affecting ~8-14 newborns per 100,000. The severity of the disease depends on the residual levels of functional survival of motor neuron protein, SMN. SMN is a ubiquitously expressed RNA binding protein involved in a plethora of cellular processes. In this review, we discuss the effects of SMN loss on mitochondrial functions in the neuronal and muscular systems that are the most affected in patients with spinal muscular atrophy. Our aim is to highlight how mitochondrial defects may contribute to disease progression and how restoring mitochondrial functionality may be a promising approach to develop new therapies. We also collected from previous studies a list of transcripts encoding mitochondrial proteins affected in various SMA models. Moreover, we speculate that in adulthood, when motor neurons require only very low SMN levels, the natural deterioration of mitochondria associated with aging may be a crucial triggering factor for adult spinal muscular atrophy, and this requires particular attention for therapeutic strategies.
Collapse
Affiliation(s)
- Eleonora Zilio
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Valentina Piano
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
7
|
Strope TA, Birky CJ, Wilkins HM. The Role of Bioenergetics in Neurodegeneration. Int J Mol Sci 2022; 23:9212. [PMID: 36012480 PMCID: PMC9409169 DOI: 10.3390/ijms23169212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/10/2022] [Accepted: 08/13/2022] [Indexed: 11/18/2022] Open
Abstract
Bioenergetic and mitochondrial dysfunction are common hallmarks of neurodegenerative diseases. Decades of research describe how genetic and environmental factors initiate changes in mitochondria and bioenergetics across Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Mitochondria control many cellular processes, including proteostasis, inflammation, and cell survival/death. These cellular processes and pathologies are common across neurodegenerative diseases. Evidence suggests that mitochondria and bioenergetic disruption may drive pathological changes, placing mitochondria as an upstream causative factor in neurodegenerative disease onset and progression. Here, we discuss evidence of mitochondrial and bioenergetic dysfunction in neurodegenerative diseases and address how mitochondria can drive common pathological features of these diseases.
Collapse
Affiliation(s)
- Taylor A. Strope
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS 66205, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Cole J. Birky
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS 66205, USA
| | - Heather M. Wilkins
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS 66205, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
The Cell Autonomous and Non-Cell Autonomous Aspects of Neuronal Vulnerability and Resilience in Amyotrophic Lateral Sclerosis. BIOLOGY 2022; 11:biology11081191. [PMID: 36009818 PMCID: PMC9405388 DOI: 10.3390/biology11081191] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/14/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Amyotrophic lateral sclerosis (ALS) is a fatal disease characterized by a progressive paralysis due to the loss of particular neurons in our nervous system called motor neurons, that exert voluntary control of all our skeletal muscles. It is not entirely understood why motor neurons are particularly vulnerable in ALS, neither is it completely clear why certain groups of motor neurons, including those that regulate eye movement, are rather resilient to this disease. However, both vulnerability and resilience to ALS likely reflect cell intrinsic properties of different motor neuron subpopulations as well as non-cell autonomous events regulated by surrounding cell types. In this review we dissect the particular properties of different motor neuron types and their responses to disease that may underlie their respective vulnerabilities and resilience. Disease progression in ALS involves multiple cell types that are closely connected to motor neurons and we here also discuss their contributions to the differential vulnerability of motor neurons. Abstract Amyotrophic lateral sclerosis (ALS) is defined by the loss of upper motor neurons (MNs) that project from the cerebral cortex to the brain stem and spinal cord and of lower MNs in the brain stem and spinal cord which innervate skeletal muscles, leading to spasticity, muscle atrophy, and paralysis. ALS involves several disease stages, and multiple cell types show dysfunction and play important roles during distinct phases of disease initiation and progression, subsequently leading to selective MN loss. Why MNs are particularly vulnerable in this lethal disease is still not entirely clear. Neither is it fully understood why certain MNs are more resilient to degeneration in ALS than others. Brain stem MNs of cranial nerves III, IV, and VI, which innervate our eye muscles, are highly resistant and persist until the end-stage of the disease, enabling paralyzed patients to communicate through ocular tracking devices. MNs of the Onuf’s nucleus in the sacral spinal cord, that innervate sphincter muscles and control urogenital functions, are also spared throughout the disease. There is also a differential vulnerability among MNs that are intermingled throughout the spinal cord, that directly relate to their physiological properties. Here, fast-twitch fatigable (FF) MNs, which innervate type IIb muscle fibers, are affected early, before onset of clinical symptoms, while slow-twitch (S) MNs, that innervate type I muscle fibers, remain longer throughout the disease progression. The resilience of particular MN subpopulations has been attributed to intrinsic determinants and multiple studies have demonstrated their unique gene regulation and protein content in health and in response to disease. Identified factors within resilient MNs have been utilized to protect more vulnerable cells. Selective vulnerability may also, in part, be driven by non-cell autonomous processes and the unique surroundings and constantly changing environment close to particular MN groups. In this article, we review in detail the cell intrinsic properties of resilient and vulnerable MN groups, as well as multiple additional cell types involved in disease initiation and progression and explain how these may contribute to the selective MN resilience and vulnerability in ALS.
Collapse
|
9
|
Cunha-Oliveira T, Carvalho M, Sardão V, Ferreiro E, Mena D, Pereira FB, Borges F, Oliveira PJ, Silva FSG. Integrative Profiling of Amyotrophic Lateral Sclerosis Lymphoblasts Identifies Unique Metabolic and Mitochondrial Disease Fingerprints. Mol Neurobiol 2022; 59:6373-6396. [PMID: 35933467 DOI: 10.1007/s12035-022-02980-7] [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: 12/23/2021] [Accepted: 07/26/2022] [Indexed: 11/26/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease with a rapid progression and no effective treatment. Metabolic and mitochondrial alterations in peripheral tissues of ALS patients may present diagnostic and therapeutic interest. We aimed to identify mitochondrial fingerprints in lymphoblast from ALS patients harboring SOD1 mutations (mutSOD1) or with unidentified mutations (undSOD1), compared with age-/sex-matched controls. Three groups of lymphoblasts, from mutSOD1 or undSOD1 ALS patients and age-/sex-matched controls, were obtained from Coriell Biobank and divided into 3 age-/sex-matched cohorts. Mitochondria-associated metabolic pathways were analyzed using Seahorse MitoStress and ATP Rate assays, complemented with metabolic phenotype microarrays, metabolite levels, gene expression, and protein expression and activity. Pooled (all cohorts) and paired (intra-cohort) analyses were performed by using bioinformatic tools, and the features with higher information gain values were selected and used for principal component analysis and Naïve Bayes classification. Considering the group as a target, the features that contributed to better segregation of control, undSOD1, and mutSOD1 were found to be the protein levels of Tfam and glycolytic ATP production rate. Metabolic phenotypic profiles in lymphoblasts from ALS patients with mutSOD1 and undSOD1 revealed unique age-dependent different substrate oxidation profiles. For most parameters, different patterns of variation in experimental endpoints in lymphoblasts were found between cohorts, which may be due to the age or sex of the donor. In the present work, we investigated several metabolic and mitochondrial hallmarks in lymphoblasts from each donor, and although a high heterogeneity of results was found, we identified specific metabolic and mitochondrial fingerprints, especially protein levels of Tfam and glycolytic ATP production rate, that may have a diagnostic and therapeutic interest.
Collapse
Affiliation(s)
- Teresa Cunha-Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| | - Marcelo Carvalho
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Vilma Sardão
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Elisabete Ferreiro
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Débora Mena
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Francisco B Pereira
- CISUC-Center for Informatics & Systems, University of Coimbra, Coimbra, Portugal
- Polytechnic Institute of Coimbra, Coimbra Institute of Engineering, Coimbra, Portugal
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Filomena S G Silva
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- Mitotag Lda, Biocant Park, Cantanhede, Portugal.
| |
Collapse
|
10
|
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: 3.0] [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
|
11
|
Yu M, Zhao X, Wu W, Wang Q, Liu J, Zhang W, Yuan Y, Hong D, Wang Z, Deng J. Widespread Mislocalization of FUS Is Associated With Mitochondrial Abnormalities in Skeletal Muscle in Amyotrophic Lateral Sclerosis With FUS Mutations. J Neuropathol Exp Neurol 2022; 81:172-181. [PMID: 35139534 DOI: 10.1093/jnen/nlac004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mutations in the fused in sarcoma (FUS) gene have been reported to be the most common genetic cause of early-onset amyotrophic lateral sclerosis (ALS); cytoplasmic inclusions containing FUS protein are the predominant pathological feature. Recent studies indicated that mutant FUS impaired neuromuscular junctions and induced muscle intrinsic toxicity in cell and animal models. However, the role of FUS in muscle degeneration remains unclear. In this study, we investigated FUS protein distribution in skeletal muscle fibers in ALS-FUS. Our data show that cytoplasmic mislocalized FUS in the unaggregated form represented a remarkable pathological feature in affected muscle fibers in ALS-FUS. Additional studies found that cytoplasmic FUS colocalized with some mitochondria and was associated with mitochondrial swelling and disorganized cristae. RNA sequencing and quantitative real-time polymerase chain reaction analyses indicated downregulation of the key subunits of mitochondrial oxidative phosphorylation complexes in the affected skeletal muscle in ALS-FUS patients. Further immunoblot analysis showed increased levels of FUS, but decreased levels of Cox I (subunit of complex IV) in ALS-FUS patients compared with age-matched controls. This is the first demonstration of the close association of cytoplasmic mislocalized FUS with mitochondrial dysfunction in skeletal muscle, implicating the presence of a cell-autonomous mechanism in muscle degeneration in ALS.
Collapse
Affiliation(s)
- Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Xutong Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wei Wu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Qingqing Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jing Liu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Daojun Hong
- Department of Medical Genetics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhaoxia Wang
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| | - Jianwen Deng
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| |
Collapse
|
12
|
Verma S, Khurana S, Vats A, Sahu B, Ganguly NK, Chakraborti P, Gourie-Devi M, Taneja V. Neuromuscular Junction Dysfunction in Amyotrophic Lateral Sclerosis. Mol Neurobiol 2022; 59:1502-1527. [PMID: 34997540 DOI: 10.1007/s12035-021-02658-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by progressive degeneration of motor neurons leading to skeletal muscle denervation. Earlier studies have shown that motor neuron degeneration begins in motor cortex and descends to the neuromuscular junction (NMJ) in a dying forward fashion. However, accumulating evidences support that ALS is a distal axonopathy where early pathological changes occur at the NMJ, prior to onset of clinical symptoms and propagates towards the motor neuron cell body supporting "dying back" hypothesis. Despite several evidences, series of events triggering NMJ disassembly in ALS are still obscure. Neuromuscular junction is a specialized tripartite chemical synapse which involves a well-coordinated communication among the presynaptic motor neuron, postsynaptic skeletal muscle, and terminal Schwann cells. This review provides comprehensive insight into the role of NMJ in ALS pathogenesis. We have emphasized the molecular alterations in cellular components of NMJ leading to loss of effective neuromuscular transmission in ALS. Further, we provide a preview into research involved in exploring NMJ as potential target for designing effective therapies for ALS.
Collapse
Affiliation(s)
- Sagar Verma
- Department of Research, Sir Ganga Ram Hospital, Delhi, India.,Department of Biotechnology, Jamia Hamdard, Delhi, India
| | - Shiffali Khurana
- Department of Research, Sir Ganga Ram Hospital, Delhi, India.,Department of Biomedical Science, Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi, India
| | - Abhishek Vats
- Department of Ophthalmology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bandana Sahu
- Department of Research, Sir Ganga Ram Hospital, Delhi, India
| | | | | | | | - Vibha Taneja
- Department of Research, Sir Ganga Ram Hospital, Delhi, India.
| |
Collapse
|
13
|
Massopust R, Juros D, Shapiro D, Lopes M, Haldar SM, Taetzsch T, Valdez G. KLF15 overexpression in myocytes fails to ameliorate ALS-related pathology or extend the lifespan of SOD1G93A mice. Neurobiol Dis 2022; 162:105583. [PMID: 34902552 PMCID: PMC8750438 DOI: 10.1016/j.nbd.2021.105583] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/19/2021] [Accepted: 12/10/2021] [Indexed: 01/22/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a currently incurable disease that causes progressive motor neuron loss, paralysis and death. Skeletal muscle pathology occurs early during the course of ALS. It is characterized by impaired mitochondrial biogenesis, metabolic dysfunction and deterioration of the neuromuscular junction (NMJ), the synapse through which motor neurons communicate with muscles. Therefore, a better understanding of the molecules that underlie this pathology may lead to therapies that slow motor neuron loss and delay ALS progression. Kruppel Like Factor 15 (KLF15) has been identified as a transcription factor that activates alternative metabolic pathways and NMJ maintenance factors, including Fibroblast Growth Factor Binding Protein 1 (FGFBP1), in skeletal myocytes. In this capacity, KLF15 has been shown to play a protective role in Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA), however its role in ALS has not been evaluated. Here, we examined whether muscle-specific KLF15 overexpression promotes the health of skeletal muscles and NMJs in the SOD1G93A ALS mouse model. We show that muscle-specific KLF15 overexpression did not elicit a significant beneficial effect on skeletal muscle atrophy, NMJ health, motor function, or survival in SOD1G93A ALS mice. Our findings suggest that, unlike in mouse models of DMD and SMA, KLF15 overexpression has a minimal impact on ALS disease progression in SOD1G93A mice.
Collapse
Affiliation(s)
- Ryan Massopust
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Devin Juros
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Dillon Shapiro
- Molecular Biology, Cell Biology, & Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Mikayla Lopes
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Saptarsi M. Haldar
- Gladstone Institutes, San Francisco, California, USA,Department of Medicine, Cardiology Division, UCSF School of Medicine, San Francisco, California, USA,Current address: Amgen Research, South San Francisco, California, USA
| | - Thomas Taetzsch
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA,Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, USA,Department of Neurology, Warren Alpert Medical School of Brown University, Providence, United States
| |
Collapse
|
14
|
Abstract
The design of the energy metabolism system in striated muscle remains a major area of investigation. Here, we review our current understanding and emerging hypotheses regarding the metabolic support of muscle contraction. Maintenance of ATP free energy, so called energy homeostasis, via mitochondrial oxidative phosphorylation is critical to sustained contractile activity, and this major design criterion is the focus of this review. Cell volume invested in mitochondria reduces the space available for generating contractile force, and this spatial balance between mitochondria acontractile elements to meet the varying sustained power demands across muscle types is another important design criterion. This is accomplished with remarkably similar mass-specific mitochondrial protein composition across muscle types, implying that it is the organization of mitochondria within the muscle cell that is critical to supporting sustained muscle function. Beyond the production of ATP, ubiquitous distribution of ATPases throughout the muscle requires rapid distribution of potential energy across these large cells. Distribution of potential energy has long been thought to occur primarily through facilitated metabolite diffusion, but recent analysis has questioned the importance of this process under normal physiological conditions. Recent structural and functional studies have supported the hypothesis that the mitochondrial reticulum provides a rapid energy distribution system via the conduction of the mitochondrial membrane potential to maintain metabolic homeostasis during contractile activity. We extensively review this aspect of the energy metabolism design contrasting it with metabolite diffusion models and how mitochondrial structure can play a role in the delivery of energy in the striated muscle.
Collapse
Affiliation(s)
- Brian Glancy
- Muscle Energetics Laboratory, National Heart, Lung, and Blood Insititute and National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, Maryland
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Insititute, Bethesda, Maryland
| | - Robert S Balaban
- Muscle Energetics Laboratory, National Heart, Lung, and Blood Insititute and National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, Maryland
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Insititute, Bethesda, Maryland
| |
Collapse
|
15
|
Moya GE, Rivera PD, Dittenhafer-Reed KE. Evidence for the Role of Mitochondrial DNA Release in the Inflammatory Response in Neurological Disorders. Int J Mol Sci 2021; 22:7030. [PMID: 34209978 PMCID: PMC8268735 DOI: 10.3390/ijms22137030] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are regarded as the metabolic centers of cells and are integral in many other cell processes, including the immune response. Each mitochondrion contains numerous copies of mitochondrial DNA (mtDNA), a small, circular, and bacterial-like DNA. In response to cellular damage or stress, mtDNA can be released from the mitochondrion and trigger immune and inflammatory responses. mtDNA release into the cytosol or bloodstream can occur as a response to hypoxia, sepsis, traumatic injury, excitatory cytotoxicity, or drastic mitochondrial membrane potential changes, some of which are hallmarks of neurodegenerative and mood disorders. Released mtDNA can mediate inflammatory responses observed in many neurological and mood disorders by driving the expression of inflammatory cytokines and the interferon response system. The current understanding of the role of mtDNA release in affective mood disorders and neurodegenerative diseases will be discussed.
Collapse
Affiliation(s)
| | - Phillip D. Rivera
- Department of Chemistry and Biology, Hope College, Holland, MI 49423, USA;
| | | |
Collapse
|
16
|
Skeletal Muscle Metabolism: Origin or Prognostic Factor for Amyotrophic Lateral Sclerosis (ALS) Development? Cells 2021; 10:cells10061449. [PMID: 34207859 PMCID: PMC8226541 DOI: 10.3390/cells10061449] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/26/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive and selective loss of motor neurons, amyotrophy and skeletal muscle paralysis usually leading to death due to respiratory failure. While generally considered an intrinsic motor neuron disease, data obtained in recent years, including our own, suggest that motor neuron protection is not sufficient to counter the disease. The dismantling of the neuromuscular junction is closely linked to chronic energy deficit found throughout the body. Metabolic (hypermetabolism and dyslipidemia) and mitochondrial alterations described in patients and murine models of ALS are associated with the development and progression of disease pathology and they appear long before motor neurons die. It is clear that these metabolic changes participate in the pathology of the disease. In this review, we summarize these changes seen throughout the course of the disease, and the subsequent impact of glucose–fatty acid oxidation imbalance on disease progression. We also highlight studies that show that correcting this loss of metabolic flexibility should now be considered a major goal for the treatment of ALS.
Collapse
|
17
|
Sassani M, Alix JJ, McDermott CJ, Baster K, Hoggard N, Wild JM, Mortiboys HJ, Shaw PJ, Wilkinson ID, Jenkins TM. Magnetic resonance spectroscopy reveals mitochondrial dysfunction in amyotrophic lateral sclerosis. Brain 2021; 143:3603-3618. [PMID: 33439988 DOI: 10.1093/brain/awaa340] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/16/2020] [Accepted: 08/08/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial dysfunction is postulated to be central to amyotrophic lateral sclerosis (ALS) pathophysiology. Evidence comes primarily from disease models and conclusive data to support bioenergetic dysfunction in vivo in patients is currently lacking. This study is the first to assess mitochondrial dysfunction in brain and muscle in individuals living with ALS using 31P-magnetic resonance spectroscopy (MRS), the modality of choice to assess energy metabolism in vivo. We recruited 20 patients and 10 healthy age and gender-matched control subjects in this cross-sectional clinico-radiological study. 31P-MRS was acquired from cerebral motor regions and from tibialis anterior during rest and exercise. Bioenergetic parameter estimates were derived including: ATP, phosphocreatine, inorganic phosphate, adenosine diphosphate, Gibbs free energy of ATP hydrolysis (ΔGATP), phosphomonoesters, phosphodiesters, pH, free magnesium concentration, and muscle dynamic recovery constants. Linear regression was used to test for associations between brain data and clinical parameters (revised amyotrophic functional rating scale, slow vital capacity, and upper motor neuron score) and between muscle data and clinico-neurophysiological measures (motor unit number and size indices, force of contraction, and speed of walking). Evidence for primary dysfunction of mitochondrial oxidative phosphorylation was detected in the brainstem where ΔGATP and phosphocreatine were reduced. Alterations were also detected in skeletal muscle in patients where resting inorganic phosphate, pH, and phosphomonoesters were increased, whereas resting ΔGATP, magnesium, and dynamic phosphocreatine to inorganic phosphate recovery were decreased. Phosphocreatine in brainstem correlated with respiratory dysfunction and disability; in muscle, energy metabolites correlated with motor unit number index, muscle power, and speed of walking. This study provides in vivo evidence for bioenergetic dysfunction in ALS in brain and skeletal muscle, which appears clinically and electrophysiologically relevant. 31P-MRS represents a promising technique to assess the pathophysiology of mitochondrial function in vivo in ALS and a potential tool for future clinical trials targeting bioenergetic dysfunction.
Collapse
Affiliation(s)
- Matilde Sassani
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - James J Alix
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Christopher J McDermott
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Kathleen Baster
- Statistical Service Unit, University of Sheffield, Sheffield, UK
| | - Nigel Hoggard
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Jim M Wild
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Heather J Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Iain D Wilkinson
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Thomas M Jenkins
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| |
Collapse
|
18
|
Martin LJ, Wong M. Skeletal Muscle-Restricted Expression of Human SOD1 in Transgenic Mice Causes a Fatal ALS-Like Syndrome. Front Neurol 2020; 11:592851. [PMID: 33381076 PMCID: PMC7767933 DOI: 10.3389/fneur.2020.592851] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal heterogeneous neurodegenerative disease that causes motor neuron (MN) loss and skeletal muscle paralysis. It is uncertain whether this degeneration of MNs is triggered intrinsically and is autonomous, or if the disease initiating mechanisms are extrinsic to MNs. We hypothesized that skeletal muscle is a primary site of pathogenesis in ALS that triggers MN degeneration. Some inherited forms of ALS are caused by mutations in the superoxide dismutase-1 (SOD1) gene, that encodes an antioxidant protein, so we created transgenic (tg) mice expressing wild-type-, G37R-, and G93A-human SOD1 gene variants only in skeletal muscle. Presence of human SOD1 (hSOD1) protein in skeletal muscle was verified by western blotting, enzyme activity gels, and immunofluorescence in myofibers and satellite cells. These tg mice developed limb weakness and paresis with motor deficits, limb and chest muscle wasting, diaphragm atrophy, and age-related fatal disease with a lifespan shortening of 10–16%. Brown and white adipose tissue also became wasted. Myofibers of tg mice developed crystalline-like inclusions, individualized sarcomere destruction, mitochondriopathy with vesiculation, DNA damage, and activated p53. Satellite cells became apoptotic. The diaphragm developed severe loss of neuromuscular junction presynaptic and postsynaptic integrity, including decreased innervation, loss of synaptophysin, nitration of synaptophysin, and loss of nicotinic acetylcholine receptor and scaffold protein rapsyn. Co-immunoprecipitation identified hSOD1 interaction with rapsyn. Spinal cords of tg mice developed gross atrophy. Spinal MNs formed cytoplasmic and nuclear inclusions, axonopathy, mitochondriopathy, accumulated DNA damage, activated p53 and cleaved caspase-3, and died. Tg mice had a 40–50% loss of MNs. This work shows that hSOD1 in skeletal muscle is a driver of pathogenesis in ALS, that involves myofiber and satellite cell toxicity, and apparent muscle-adipose tissue disease relationships. It also identifies a non-autonomous mechanism for MN degeneration explaining their selective vulnerability as likely a form of target-deprivation retrograde neurodegeneration.
Collapse
Affiliation(s)
- Lee J Martin
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Margaret Wong
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
Marini C, Cossu V, Bonifacino T, Bauckneht M, Torazza C, Bruno S, Castellani P, Ravera S, Milanese M, Venturi C, Carlone S, Piccioli P, Emionite L, Morbelli S, Orengo AM, Donegani MI, Miceli A, Raffa S, Marra S, Signori A, Cortese K, Grillo F, Fiocca R, Bonanno G, Sambuceti G. Mechanisms underlying the predictive power of high skeletal muscle uptake of FDG in amyotrophic lateral sclerosis. EJNMMI Res 2020; 10:76. [PMID: 32638178 PMCID: PMC7340686 DOI: 10.1186/s13550-020-00666-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
Background We recently reported that enhanced [18F]-fluorodeoxyglucose (FDG) uptake in skeletal muscles predicts disease aggressiveness in patients with amyotrophic lateral sclerosis (ALS). The present experimental study aimed to assess whether this predictive potential reflects the link between FDG uptake and redox stress that has been previously reported in different tissues and disease models. Methods The study included 15 SOD1G93A mice (as experimental ALS model) and 15 wildtype mice (around 120 days old). Mice were submitted to micro-PET imaging. Enzymatic pathways and response to oxidative stress were evaluated in harvested quadriceps and hearts by biochemical, immunohistochemical, and immunofluorescence analysis. Colocalization between the endoplasmic reticulum (ER) and the fluorescent FDG analog 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxyglucose (2-NBDG) was performed in fresh skeletal muscle sections. Finally, mitochondrial ultrastructure and bioenergetics were evaluated in harvested quadriceps and hearts. Results FDG retention was significantly higher in hindlimb skeletal muscles of symptomatic SOD1G93A mice with respect to control ones. This difference was not explained by any acceleration in glucose degradation through glycolysis or cytosolic pentose phosphate pathway (PPP). Similarly, it was independent of inflammatory infiltration. Rather, the high FDG retention in SOD1G93A skeletal muscle was associated with an accelerated generation of reactive oxygen species. This redox stress selectively involved the ER and the local PPP triggered by hexose-6P-dehydrogenase. ER involvement was confirmed by the colocalization of the 2-NBDG with a vital ER tracker. The oxidative damage in transgenic skeletal muscle was associated with a severe impairment in the crosstalk between ER and mitochondria combined with alterations in mitochondrial ultrastructure and fusion/fission balance. The expected respiratory damage was confirmed by a deceleration in ATP synthesis and oxygen consumption rate. These same abnormalities were represented to a markedly lower degree in the myocardium, as a sample of non-voluntary striated muscle. Conclusion Skeletal muscle of SOD1G93A mice reproduces the increased FDG uptake observed in ALS patients. This finding reflects the selective activation of the ER-PPP in response to significant redox stress associated with alterations of mitochondrial ultrastructure, networking, and connection with the ER itself. This scenario is less severe in cardiomyocytes suggesting a relevant role for either communication with synaptic plaque or contraction dynamics.
Collapse
Affiliation(s)
- Cecilia Marini
- CNR Institute of Molecular Bioimaging and Physiology (IBFM), Milano, Italy. .,Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.
| | - Vanessa Cossu
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.,Department of Health Sciences, University of Genoa, Genova, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Section of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.,Department of Health Sciences, University of Genoa, Genova, Italy
| | - Carola Torazza
- Department of Pharmacy, Section of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Silvia Bruno
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova, Italy
| | | | - Silvia Ravera
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova, Italy
| | - Marco Milanese
- Department of Pharmacy, Section of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Consuelo Venturi
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova, Italy
| | | | | | - Laura Emionite
- Animal Facility, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvia Morbelli
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.,Department of Health Sciences, University of Genoa, Genova, Italy
| | - Anna Maria Orengo
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy
| | | | - Alberto Miceli
- Department of Health Sciences, University of Genoa, Genova, Italy
| | - Stefano Raffa
- Department of Health Sciences, University of Genoa, Genova, Italy
| | - Stefano Marra
- Department of Health Sciences, University of Genoa, Genova, Italy
| | - Alessio Signori
- Department of Health Sciences, University of Genoa, Genova, Italy
| | - Katia Cortese
- Department of Experimental Medicine, Human Anatomy, University of Genoa, Genova, Italy
| | - Federica Grillo
- Department of Surgical Sciences and Integrated Diagnostics, Pathology Unit, University of Genoa, Genova, Italy
| | - Roberto Fiocca
- Department of Surgical Sciences and Integrated Diagnostics, Pathology Unit, University of Genoa, Genova, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Section of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy.,Pharmacology and Toxicology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genova, Italy.,Department of Health Sciences, University of Genoa, Genova, Italy
| |
Collapse
|
20
|
Vianello C, Cocetta V, Caicci F, Boldrin F, Montopoli M, Martinuzzi A, Carelli V, Giacomello M. Interaction Between Mitochondrial DNA Variants and Mitochondria/Endoplasmic Reticulum Contact Sites: A Perspective Review. DNA Cell Biol 2020; 39:1431-1443. [PMID: 32598172 DOI: 10.1089/dna.2020.5614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mitochondria contain their own genome, mitochondrial DNA (mtDNA), essential to support their fundamental intracellular role in ATP production and other key metabolic and homeostatic pathways. Mitochondria are highly dynamic organelles that communicate with all the other cellular compartments, through sites of high physical proximity. Among all, their crosstalk with the endoplasmic reticulum (ER) appears particularly important as its derangement is tightly implicated with several human disorders. Population-specific mtDNA variants clustered in defining the haplogroups have been shown to exacerbate or mitigate these pathological conditions. The exact mechanisms of the mtDNA background-modifying effect are not completely clear and a possible explanation is the outcome of mitochondrial efficiency on retrograde signaling to the nucleus. However, the possibility that different haplogroups shape the proximity and crosstalk between mitochondria and the ER has never been proposed neither investigated. In this study, we pose and discuss this question and provide preliminary data to answer it. Besides, we also address the possibility that single, disease-causing mtDNA point mutations may act also by reshaping organelle communication. Overall, this perspective review provides a theoretical platform for future studies on the interaction between mtDNA variants and organelle contact sites.
Collapse
Affiliation(s)
| | - Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | | | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.,VIMM-Veneto Institute of Molecular Medicine, Padova, Italy
| | - Andrea Martinuzzi
- Department of Neurorehabilitation, IRCCS "E. Medea" Scientific Institute, Conegliano Research Center, Treviso, Italy
| | - Valerio Carelli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Marta Giacomello
- Department of Biology, University of Padova, Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
21
|
Shteinfer-Kuzmine A, Argueti S, Gupta R, Shvil N, Abu-Hamad S, Gropper Y, Hoeber J, Magrì A, Messina A, Kozlova EN, Shoshan-Barmatz V, Israelson A. A VDAC1-Derived N-Terminal Peptide Inhibits Mutant SOD1-VDAC1 Interactions and Toxicity in the SOD1 Model of ALS. Front Cell Neurosci 2019; 13:346. [PMID: 31474832 PMCID: PMC6702328 DOI: 10.3389/fncel.2019.00346] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Mutations in superoxide dismutase (SOD1) are the second most common cause of familial amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease caused by the death of motor neurons in the brain and spinal cord. SOD1 neurotoxicity has been attributed to aberrant accumulation of misfolded SOD1, which in its soluble form binds to intracellular organelles, such as mitochondria and ER, disrupting their functions. Here, we demonstrate that mutant SOD1 binds specifically to the N-terminal domain of the voltage-dependent anion channel (VDAC1), an outer mitochondrial membrane protein controlling cell energy, metabolic and survival pathways. Mutant SOD1G93A and SOD1G85R, but not wild type SOD1, directly interact with VDAC1 and reduce its channel conductance. No such interaction with N-terminal-truncated VDAC1 occurs. Moreover, a VDAC1-derived N-terminal peptide inhibited mutant SOD1-induced toxicity. Incubation of motor neuron-like NSC-34 cells expressing mutant SOD1 or mouse embryonic stem cell-derived motor neurons with different VDAC1 N-terminal peptides resulted in enhanced cell survival. Taken together, our results establish a direct link between mutant SOD1 toxicity and the VDAC1 N-terminal domain and suggest that VDAC1 N-terminal peptides targeting mutant SOD1 provide potential new therapeutic strategies for ALS.
Collapse
Affiliation(s)
- Anna Shteinfer-Kuzmine
- Department of Life Sciences, The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Shirel Argueti
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Rajeev Gupta
- Department of Life Sciences, The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Neta Shvil
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Salah Abu-Hamad
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Yael Gropper
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Jan Hoeber
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Andrea Magrì
- Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Angela Messina
- Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Elena N Kozlova
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
22
|
Zhou J, Li A, Li X, Yi J. Dysregulated mitochondrial Ca 2+ and ROS signaling in skeletal muscle of ALS mouse model. Arch Biochem Biophys 2019; 663:249-258. [PMID: 30682329 PMCID: PMC6506190 DOI: 10.1016/j.abb.2019.01.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/24/2018] [Accepted: 01/18/2019] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neuromuscular disease characterized by motor neuron loss and prominent skeletal muscle wasting. Despite more than one hundred years of research efforts, the pathogenic mechanisms underlying neuromuscular degeneration in ALS remain elusive. While the death of motor neuron is a defining hallmark of ALS, accumulated evidences suggested that in addition to being a victim of motor neuron axonal withdrawal, the intrinsic skeletal muscle degeneration may also actively contribute to ALS disease pathogenesis and progression. Examination of spinal cord and muscle autopsy/biopsy samples of ALS patients revealed similar mitochondrial abnormalities in morphology, quantity and disposition, which are accompanied by defective mitochondrial respiratory chain complex and elevated oxidative stress. Detailing the molecular/cellular mechanisms and the role of mitochondrial dysfunction in ALS relies on ALS animal model studies. This review article discusses the dysregulated mitochondrial Ca2+ and reactive oxygen species (ROS) signaling revealed in live skeletal muscle derived from ALS mouse models, and a potential role of the vicious cycle formed between the dysregulated mitochondrial Ca2+ signaling and excessive ROS production in promoting muscle wasting during ALS progression.
Collapse
Affiliation(s)
- Jingsong Zhou
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA.
| | - Ang Li
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Xuejun Li
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Jianxun Yi
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
23
|
Abstract
SIGNIFICANCE NAD+ and NADP+ are important cosubstrates in redox reactions and participate in regulatory networks operating in adjustment of metabolic pathways. Moreover, NAD+ is a cosubstrate in post-translational modification of proteins and is involved in DNA repair. NADPH is indispensable for reductive syntheses and the redox chemistry involved in attaining and maintaining correct protein conformation. Recent Advances: Within a couple of decades, a wealth of information has been gathered on NAD(H)+/NADP(H) redox imaging, regulatory role of redox potential in assembly of spatial protein structures, and the role of ADP-ribosylation of regulatory proteins affecting both gene expression and metabolism. All these have a bearing also on disease, healthy aging, and longevity. CRITICAL ISSUES Knowledge of the signal propagation pathways of NAD+-dependent post-translational modifications is still fragmentary for explaining the mechanism of cellular stress effects and nutritional state on these actions. Evaluation of the cosubstrate and regulator roles of NAD(H) and NADP(H) still suffers from some controversies in experimental data. FUTURE DIRECTIONS Activating or inhibiting interventions in NAD+-dependent protein modifications for medical purposes has shown promise, but restraining tumor growth by inhibiting DNA repair in tumors by means of interference in sirtuins is still in the early stage. The same is true for the use of this technology in improving health and healthy aging. New genetically encoded specific NAD and NADP probes are expected to modernize the research on redox biology.
Collapse
Affiliation(s)
- Ilmo E Hassinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
24
|
Amyotrophic Lateral Sclerosis Associated with Statin Use: A Disproportionality Analysis of the FDA's Adverse Event Reporting System. Drug Saf 2018; 41:403-413. [PMID: 29427042 DOI: 10.1007/s40264-017-0620-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Apparent elevations in reporting of amyotrophic lateral sclerosis (ALS)-like conditions associated with statin use have been previously described from data obtained via US and European databases. OBJECTIVE The aim of this study was to examine US FDA Adverse Event Reporting System (FAERS) data to compare reporting odds ratios (RORs) of ALS and ALS-like conditions between statins and other drugs, for each statin agent. METHODS We assessed for disproportional rates of reported ALS and ALS-related conditions for each statin agent separately by using the ROR formula. FAERS data were analyzed through September 2015. RESULTS RORs for ALS were elevated for all statins, with elevations possibly stronger for lipophilic statins. RORs ranged from 9.09 (6.57-12.6) and 16.2 (9.56-27.5) for rosuvastatin and pravastatin (hydrophilic) to 17.0 (14.1-20.4), 23.0 (18.3-29.1), and 107 (68.5-167) for atorvastatin, simvastatin, and lovastatin (lipophilic), respectively. For simvastatin, an ROR of 57.1 (39.5-82.7) was separately present for motor neuron disease. CONCLUSION These findings extend previous evidence showing that significantly elevated ALS reporting extends to individual statin agents, and add to concerns about potential elevated occurrence of ALS-like conditions in association with statin usage.
Collapse
|
25
|
Bojungikgi-tang Improves Muscle and Spinal Cord Function in an Amyotrophic Lateral Sclerosis Model. Mol Neurobiol 2018; 56:2394-2407. [PMID: 30030751 DOI: 10.1007/s12035-018-1236-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a motor neuron disease characterized by progressive motor function impairment, dysphagia, and respiratory failure. Owing to the complexity of its pathogenic mechanisms, an effective therapy for ALS is lacking. Herbal medicines with multiple targets have good efficacy and low adverse reactions for the treatment of neurodegenerative diseases. In this study, the effects of Bojungikgi-tang (BJIGT), an herbal medicine with eight component herbs, on muscle and spinal cord function were evaluated in an ALS animal model. Animals were randomly divided into three groups: a non-transgenic group (nTg, n = 24), a hSOD1G93A transgenic group (Tg, n = 24), and a hSOD1G93A transgenic group in which 8-week-old mice were orally administered BJIGT (1 mg/g) once daily for 6 weeks (Tg+BJIGT, n = 24). The effects of BJIGT were evaluated using a rotarod test, foot-printing, and survival analyses based on Kaplan-Meier survival curves. To determine the biological mechanism underlying the effects of BJIGT in hSOD1G93A mice, western blotting, transmission electron microscopy, and Bungarotoxin staining were used. BJIGT improved motor function and extended the survival duration of hSOD1G93A mice. In addition, BJIGT had protective effects, including anti-oxidative and anti-inflammatory effects, in both the spinal cord and muscle of hSOD1G93A mice. Our results demonstrated that BJIGT causes muscle atrophy and the denervation of neuromuscular junctions in the gastrocnemius of hSOD1G93A mice. The components of BJIGT may alleviate the symptoms of ALS via different mechanisms, and accordingly, BJIGT treatment may be an effective therapeutic approach.
Collapse
|
26
|
Ramalho TC, de Castro AA, Tavares TS, Silva MC, Silva DR, Cesar PH, Santos LA, da Cunha EFF, Nepovimova E, Kuca K. Insights into the pharmaceuticals and mechanisms of neurological orphan diseases: Current Status and future expectations. Prog Neurobiol 2018; 169:135-157. [PMID: 29981392 DOI: 10.1016/j.pneurobio.2018.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 06/30/2018] [Indexed: 12/20/2022]
Abstract
Several rare or orphan diseases have been characterized that singly affect low numbers of people, but cumulatively reach ∼6%-10% of the population in Europe and in the United States. Human genetics has shown to be broadly effective when evaluating subjacent genetic defects such as orphan genetic diseases, but on the other hand, a modest progress has been achieved toward comprehending the molecular pathologies and designing new therapies. Chemical genetics, placed at the interface of chemistry and genetics, could be employed to understand the molecular mechanisms of subjacent illnesses and for the discovery of new remediation processes. This review debates current progress in chemical genetics, and how a variety of compounds and reaction mechanisms can be used to study and ultimately treat rare genetic diseases. We focus here on a study involving Amyotrophic lateral sclerosis (ALS), Duchenne Muscular Dystrophy (DMD), Spinal muscular atrophy (SMA) and Familial Amyloid Polyneuropathy (FAP), approaching different treatment methods and the reaction mechanisms of several compounds, trying to elucidate new routes capable of assisting in the treatment profile.
Collapse
Affiliation(s)
- Teodorico C Ramalho
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil; Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| | | | - Tássia S Tavares
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Maria C Silva
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Daniela R Silva
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Pedro H Cesar
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Lucas A Santos
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Elaine F F da Cunha
- Department of Chemistry, Federal University of Lavras, 37200-000, Lavras, Brazil
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| |
Collapse
|
27
|
Wilkins HM, Morris JK. New Therapeutics to Modulate Mitochondrial Function in Neurodegenerative Disorders. Curr Pharm Des 2018; 23:731-752. [PMID: 28034353 DOI: 10.2174/1381612822666161230144517] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mitochondrial function and energy metabolism are impaired in neurodegenerative diseases. There is evidence for these functional declines both within the brain and systemically in Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis. Due to these observations, therapeutics targeted to alter mitochondrial function and energy pathways are increasingly studied in pre-clinical and clinical settings. METHODS The goal of this article was to review therapies with specific implications on mitochondrial energy metabolism published through May 2016 that have been tested for treatment of neurodegenerative diseases. RESULTS We discuss implications for mitochondrial dysfunction in neurodegenerative diseases and how this drives new therapeutic initiatives. CONCLUSION Thus far, treatments have achieved varying degrees of success. Further investigation into the mechanisms driving mitochondrial dysfunction and bioenergetic failure in neurodegenerative diseases is warranted.
Collapse
Affiliation(s)
- Heather M Wilkins
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jill K Morris
- University of Kansas School of Medicine, University of Kansas Alzheimer's Disease Center MS 6002, 3901 Rainbow Blvd, Kansas City, KS 66160. United States
| |
Collapse
|
28
|
Cykowski MD, Powell SZ, Appel JW, Arumanayagam AS, Rivera AL, Appel SH. Phosphorylated TDP-43 (pTDP-43) aggregates in the axial skeletal muscle of patients with sporadic and familial amyotrophic lateral sclerosis. Acta Neuropathol Commun 2018; 6:28. [PMID: 29653597 PMCID: PMC5899326 DOI: 10.1186/s40478-018-0528-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 03/19/2018] [Indexed: 01/08/2023] Open
Abstract
Muscle atrophy with weakness is a core feature of amyotrophic lateral sclerosis (ALS) that has long been attributed to motor neuron loss alone. However, several studies in ALS patients, and more so in animal models, have challenged this assumption with the latter providing direct evidence that muscle can play an active role in the disease. Here, we examined the possible role of cell autonomous pathology in 148 skeletal muscle samples from 57 ALS patients, identifying phosphorylated TAR DNA-binding protein (pTDP-43) inclusions in the muscle fibers of 19 patients (33.3%) and 24 tissue samples (16.2% of specimens). A muscle group-specific difference was identified with pTDP-43 pathology being significantly more common in axial (paraspinous, diaphragm) than appendicular muscles (P = 0.0087). This pathology was not significantly associated with pertinent clinical, genetic (c9ALS) or nervous system pathologic data, suggesting it is not limited to any particular subgroup of ALS patients. Among 25 non-ALS muscle samples, pTDP-43 inclusions were seen only in the autophagy-related disorder inclusion body myositis (IBM) (n = 4), where they were more diffuse than in positive ALS samples (P = 0.007). As in IBM samples, pTDP-43 aggregates in ALS were p62/ sequestosome-1-positive, potentially indicating induction of autophagy. Phospho-TDP-43-positive ALS and IBM samples also showed significant up-regulation of TARDBP and SQSTM1 expression. These findings implicate axial skeletal muscle as an additional site of pTDP-43 pathology in some ALS patients, including sporadic and familial cases, which is deserving of further investigation.
Collapse
Affiliation(s)
- Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA.
- Institute of Academic Medicine (IAM) in the Houston Methodist Research Institute (HMRI), Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA.
| | - Suzanne Z Powell
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Institute of Academic Medicine (IAM) in the Houston Methodist Research Institute (HMRI), Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Houston Methodist Neurological Institute, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Joan W Appel
- Houston Methodist Neurological Institute, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Department of Neurology, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Anithachristy S Arumanayagam
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Andreana L Rivera
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Institute of Academic Medicine (IAM) in the Houston Methodist Research Institute (HMRI), Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Houston Methodist Neurological Institute, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Stanley H Appel
- Institute of Academic Medicine (IAM) in the Houston Methodist Research Institute (HMRI), Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Houston Methodist Neurological Institute, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Department of Neurology, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| |
Collapse
|
29
|
Vandoorne T, De Bock K, Van Den Bosch L. Energy metabolism in ALS: an underappreciated opportunity? Acta Neuropathol 2018; 135:489-509. [PMID: 29549424 PMCID: PMC5978930 DOI: 10.1007/s00401-018-1835-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a relentlessly progressive and fatal neurodegenerative disorder that primarily affects motor neurons. Despite our increased understanding of the genetic factors contributing to ALS, no effective treatment is available. A growing body of evidence shows disturbances in energy metabolism in ALS. Moreover, the remarkable vulnerability of motor neurons to ATP depletion has become increasingly clear. Here, we review metabolic alterations present in ALS patients and models, discuss the selective vulnerability of motor neurons to energetic stress, and provide an overview of tested and emerging metabolic approaches to treat ALS. We believe that a further understanding of the metabolic biology of ALS can lead to the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Tijs Vandoorne
- Department of Neurosciences, Experimental Neurology, KU Leuven-University of Leuven, Campus Gasthuisberg O&N 4, Herestraat 49, PB 602, 3000, Leuven, Belgium
- Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000, Leuven, Belgium
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, KU Leuven-University of Leuven, Campus Gasthuisberg O&N 4, Herestraat 49, PB 602, 3000, Leuven, Belgium.
- Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000, Leuven, Belgium.
| |
Collapse
|
30
|
Fibroblast bioenergetics to classify amyotrophic lateral sclerosis patients. Mol Neurodegener 2017; 12:76. [PMID: 29065921 PMCID: PMC5655870 DOI: 10.1186/s13024-017-0217-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/17/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The objective of this study was to investigate cellular bioenergetics in primary skin fibroblasts derived from patients with amyotrophic lateral sclerosis (ALS) and to determine if they can be used as classifiers for patient stratification. METHODS We assembled a collection of unprecedented size of fibroblasts from patients with sporadic ALS (sALS, n = 171), primary lateral sclerosis (PLS, n = 34), ALS/PLS with C9orf72 mutations (n = 13), and healthy controls (n = 91). In search for novel ALS classifiers, we performed extensive studies of fibroblast bioenergetics, including mitochondrial membrane potential, respiration, glycolysis, and ATP content. Next, we developed a machine learning approach to determine whether fibroblast bioenergetic features could be used to stratify patients. RESULTS Compared to controls, sALS and PLS fibroblasts had higher average mitochondrial membrane potential, respiration, and glycolysis, suggesting that they were in a hypermetabolic state. Only membrane potential was elevated in C9Orf72 lines. ATP steady state levels did not correlate with respiration and glycolysis in sALS and PLS lines. Based on bioenergetic profiles, a support vector machine (SVM) was trained to classify sALS and PLS with 99% specificity and 70% sensitivity. CONCLUSIONS sALS, PLS, and C9Orf72 fibroblasts share hypermetabolic features, while presenting differences of bioenergetics. The absence of correlation between energy metabolism activation and ATP levels in sALS and PLS fibroblasts suggests that in these cells hypermetabolism is a mechanism to adapt to energy dissipation. Results from SVM support the use of metabolic characteristics of ALS fibroblasts and multivariate analysis to develop classifiers for patient stratification.
Collapse
|
31
|
Ngo ST, Mi JD, Henderson RD, McCombe PA, Steyn FJ. Exploring targets and therapies for amyotrophic lateral sclerosis: current insights into dietary interventions. Degener Neurol Neuromuscul Dis 2017; 7:95-108. [PMID: 30050381 PMCID: PMC6053104 DOI: 10.2147/dnnd.s120607] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A growing number of preclinical and human studies demonstrate a disease-modifying effect of nutritional state in amyotrophic lateral sclerosis (ALS). The management of optimal nutrition in ALS is complicated, as physiological, physical, and psychological effects of the disease need to be considered and addressed accordingly. In this regard, multidisciplinary care teams play an integral role in providing dietary guidance to ALS patients and their carers. However, with an increasing research focus on the use of dietary intervention strategies to manage disease symptoms and improve prognosis in ALS, many ALS patients are now seeking or are actively engaged in using complementary and alternative therapies that are dietary in nature. In this article, we review the aspects of appetite control, energy balance, and the physiological effects of ALS relative to their impact on overall nutrition. We then provide current insights into dietary interventions for ALS, considering the mechanisms of action of some of the common dietary interventions used in ALS, discussing their validity in the context of clinical trials.
Collapse
Affiliation(s)
- Shyuan T Ngo
- School of Biomedical Sciences.,Queensland Brain Institute.,Centre for Clinical Research, The University of Queensland, .,Department of Neurology, Royal Brisbane & Women's Hospital, .,Wesley Medical Research, The Wesley Hospital, Auchenflower, Brisbane, QLD, Australia,
| | | | - Robert D Henderson
- Centre for Clinical Research, The University of Queensland, .,Department of Neurology, Royal Brisbane & Women's Hospital,
| | - Pamela A McCombe
- Centre for Clinical Research, The University of Queensland, .,Department of Neurology, Royal Brisbane & Women's Hospital,
| | - Frederik J Steyn
- Centre for Clinical Research, The University of Queensland, .,Department of Neurology, Royal Brisbane & Women's Hospital, .,Wesley Medical Research, The Wesley Hospital, Auchenflower, Brisbane, QLD, Australia,
| |
Collapse
|
32
|
Triheptanoin Protects Motor Neurons and Delays the Onset of Motor Symptoms in a Mouse Model of Amyotrophic Lateral Sclerosis. PLoS One 2016; 11:e0161816. [PMID: 27564703 PMCID: PMC5001695 DOI: 10.1371/journal.pone.0161816] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 08/14/2016] [Indexed: 12/12/2022] Open
Abstract
There is increasing evidence that energy metabolism is disturbed in Amyotrophic Lateral Sclerosis (ALS) patients and animal models. Treatment with triheptanoin, the triglyceride of heptanoate, is a promising approach to provide alternative fuel to improve oxidative phosphorylation and aid ATP generation. Heptanoate can be metabolized to propionyl-CoA, which after carboxylation can produce succinyl-CoA and thereby re-fill the tricarboxylic acid (TCA) cycle (anaplerosis). Here we tested the hypothesis that treatment with triheptanoin prevents motor neuron loss and delays the onset of disease symptoms in female mice overexpressing the mutant human SOD1G93A (hSOD1G93A) gene. When oral triheptanoin (35% of caloric content) was initiated at P35, motor neuron loss at 70 days of age was attenuated by 33%. In untreated hSOD1G93A mice, the loss of hind limb grip strength began at 16.7 weeks. Triheptanoin maintained hind limb grip strength for 2.8 weeks longer (p<0.01). Loss of balance on the rotarod and reduction of body weight were delayed by 13 and 11 days respectively (both p<0.01). Improved motor function occurred in parallel with alterations in the expression of genes associated with muscle metabolism. In gastrocnemius muscles, the mRNA levels of pyruvate, 2-oxoglutarate and succinate dehydrogenases and methyl-malonyl mutase were reduced by 24–33% in 10 week old hSOD1G93A mice when compared to wild-type mice, suggesting that TCA cycling in skeletal muscle may be slowed in this ALS mouse model at a stage when muscle strength is still normal. At 25 weeks of age, mRNA levels of succinate dehydrogenases, glutamic pyruvic transaminase 2 and the propionyl carboxylase β subunit were reduced by 69–84% in control, but not in triheptanoin treated hSOD1G93A animals. Taken together, our results suggest that triheptanoin slows motor neuron loss and the onset of motor symptoms in ALS mice by improving TCA cycling.
Collapse
|
33
|
Casas C, Manzano R, Vaz R, Osta R, Brites D. Synaptic Failure: Focus in an Integrative View of ALS. Brain Plast 2016; 1:159-175. [PMID: 29765840 PMCID: PMC5928542 DOI: 10.3233/bpl-140001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
From early description by Charcot, the classification of the Amyotrophic Lateral Sclerosis (ALS) is evolving from a subtype of Motor Neuron (MN) Disease to be considered rather a multi-systemic, non-cell autonomous and complex neurodegenerative disease. In the last decade, the huge amount of knowledge acquired has shed new insights on the pathological mechanisms underlying ALS from different perspectives. However, a whole vision on the multiple dysfunctional pathways is needed with the inclusion of information often excluded in other published revisions. We propose an integrative view of ALS pathology, although centered on the synaptic failure as a converging and crucial player to the etiology of the disease. Homeostasis of input and output synaptic activity of MNs has been proved to be severely and early disrupted and to definitively contribute to microcircuitry alterations at the spinal cord. Several cells play roles in synaptic communication across the MNs network system such as interneurons, astrocytes, microglia, Schwann and skeletal muscle cells. Microglia are described as highly dynamic surveying cells of the nervous system but also as determinant contributors to the synaptic plasticity linked to neuronal activity. Several signaling axis such as TNFα/TNFR1 and CX3CR1/CX3CL1 that characterize MN-microglia cross talk contribute to synaptic scaling and maintenance, have been found altered in ALS. The presence of dystrophic and atypical microglia in late stages of ALS, with a decline in their dynamic motility and phagocytic ability, together with less synaptic and neuronal contacts disrupts the MN-microglia dialogue, decreases homeostatic regulation of neuronal activity, perturbs “on/off” signals and accelerates disease progression associated to impaired synaptic function and regeneration. Other hotspot in the ALS affected network system is the unstable neuromuscular junction (NMJ) leading to distal axonal degeneration. Reduced neuromuscular spontaneous synaptic activity in ALS mice models was also suggested to account for the selective vulnerability of MNs and decreased regenerative capability. Synaptic destabilization may as well derive from increased release of molecules by muscle cells (e.g. NogoA) and by terminal Schwann cells (e.g. semaphorin 3A) conceivably causing nerve terminal retraction and denervation, as well as inhibition of re-connection to muscle fibers. Indeed, we have overviewed the alterations on the metabolic pathways and self-regenerative capacity presented in skeletal muscle cells that contribute to muscle wasting in ALS. Finally, a detailed footpath of pathologic changes on MNs and associated dysfunctional and synaptic alterations is provided. The oriented motivation in future ALS studies as outlined in the present article will help in fruitful novel achievements on the mechanisms involved and in developing more target-driven therapies that will bring new hope in halting or delaying disease progression in ALS patients.
Collapse
Affiliation(s)
- Caty Casas
- Group of Neuroplasticity and Regeneration, Institut de Neurociències and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Barcelona, Spain
| | - Raquel Manzano
- Laboratory of Genetic Biochemistry (LAGENBIO-I3A), Aragón Institute of Health Sciences, Universidad de Zaragoza, Zaragoza, Spain
| | - Rita Vaz
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| | - Rosario Osta
- Laboratory of Genetic Biochemistry (LAGENBIO-I3A), Aragón Institute of Health Sciences, Universidad de Zaragoza, Zaragoza, Spain
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| |
Collapse
|
34
|
Loeffler J, Picchiarelli G, Dupuis L, Gonzalez De Aguilar J. The Role of Skeletal Muscle in Amyotrophic Lateral Sclerosis. Brain Pathol 2016; 26:227-36. [PMID: 26780251 PMCID: PMC8029271 DOI: 10.1111/bpa.12350] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/14/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset disease primarily characterized by upper and lower motor neuron degeneration, muscle wasting and paralysis. It is increasingly accepted that the pathological process leading to ALS is the result of multiple disease mechanisms that operate within motor neurons and other cell types both inside and outside the central nervous system. The implication of skeletal muscle has been the subject of a number of studies conducted on patients and related animal models. In this review, we describe the features of ALS muscle pathology and discuss on the contribution of muscle to the pathological process. We also give an overview of the therapeutic strategies proposed to alleviate muscle pathology or to deliver curative agents to motor neurons. ALS muscle mainly suffers from oxidative stress, mitochondrial dysfunction and bioenergetic disturbances. However, the way by which the disease affects different types of myofibers depends on their contractile and metabolic features. Although the implication of muscle in nourishing the degenerative process is still debated, there is compelling evidence suggesting that it may play a critical role. Detailed understanding of the muscle pathology in ALS could, therefore, lead to the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Jean‐Philippe Loeffler
- Université de Strasbourg, UMR_S 1118StrasbourgFrance
- INSERM, U1118, Mécanismes Centraux et Péripheriques de la NeurodégénérescenceStrasbourgFrance
| | - Gina Picchiarelli
- Université de Strasbourg, UMR_S 1118StrasbourgFrance
- INSERM, U1118, Mécanismes Centraux et Péripheriques de la NeurodégénérescenceStrasbourgFrance
| | - Luc Dupuis
- Université de Strasbourg, UMR_S 1118StrasbourgFrance
- INSERM, U1118, Mécanismes Centraux et Péripheriques de la NeurodégénérescenceStrasbourgFrance
| | - Jose‐Luis Gonzalez De Aguilar
- Université de Strasbourg, UMR_S 1118StrasbourgFrance
- INSERM, U1118, Mécanismes Centraux et Péripheriques de la NeurodégénérescenceStrasbourgFrance
| |
Collapse
|
35
|
Sharma A, Varghese AM, Vijaylakshmi K, Sumitha R, Prasanna VK, Shruthi S, Chandrasekhar Sagar BK, Datta KK, Gowda H, Nalini A, Alladi PA, Christopher R, Sathyaprabha TN, Raju TR, Srinivas Bharath MM. Cerebrospinal Fluid from Sporadic Amyotrophic Lateral Sclerosis Patients Induces Mitochondrial and Lysosomal Dysfunction. Neurochem Res 2015; 41:965-84. [DOI: 10.1007/s11064-015-1779-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 10/15/2015] [Accepted: 11/17/2015] [Indexed: 12/14/2022]
|
36
|
Zarei S, Carr K, Reiley L, Diaz K, Guerra O, Altamirano PF, Pagani W, Lodin D, Orozco G, Chinea A. A comprehensive review of amyotrophic lateral sclerosis. Surg Neurol Int 2015; 6:171. [PMID: 26629397 PMCID: PMC4653353 DOI: 10.4103/2152-7806.169561] [Citation(s) in RCA: 395] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/12/2015] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset fatal neurodegenerative disease affecting motor neurons with an incidence of about 1/100,000. Most ALS cases are sporadic, but 5–10% of the cases are familial ALS. Both sporadic and familial ALS (FALS) are associated with degeneration of cortical and spinal motor neurons. The etiology of ALS remains unknown. However, mutations of superoxide dismutase 1 have been known as the most common cause of FALS. In this study, we provide a comprehensive review of ALS. We cover all aspects of the disease including epidemiology, comorbidities, environmental risk factor, molecular mechanism, genetic factors, symptoms, diagnostic, treatment, and even the available supplement and management of ALS. This will provide the reader with an advantage of receiving a broad range of information about the disease.
Collapse
Affiliation(s)
- Sara Zarei
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Karen Carr
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Luz Reiley
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Kelvin Diaz
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Orleiquis Guerra
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | | | - Wilfredo Pagani
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Daud Lodin
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Gloria Orozco
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Angel Chinea
- Neurologist, Caribbean Neurological Center, Caguas, USA
| |
Collapse
|
37
|
The interplay between metabolic homeostasis and neurodegeneration: insights into the neurometabolic nature of amyotrophic lateral sclerosis. CELL REGENERATION 2015; 4:5. [PMID: 26322226 PMCID: PMC4551561 DOI: 10.1186/s13619-015-0019-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 07/23/2015] [Indexed: 02/06/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, neurodegenerative disease that is characterized by the selective degeneration of upper motor neurons and lower spinal motor neurons, resulting in the progressive paralysis of all voluntary muscles. Approximately 10 % of ALS cases are linked to known genetic mutations, with the remaining 90 % of cases being sporadic. While the primary pathology in ALS is the selective death of upper and lower motor neurons, numerous studies indicate that an imbalance in whole body and/or cellular metabolism influences the rate of progression of disease. This review summarizes current research surrounding the impact of impaired metabolic physiology in ALS. We extend ideas to consider prospects that lie ahead in terms of how metabolic alterations may impact the selective degeneration of neurons in ALS and how targeting of adenosine triphosphate-sensitive potassium (KATP) channels may represent a promising approach for obtaining neuroprotection in ALS.
Collapse
|
38
|
Mancuso R, Navarro X. Amyotrophic lateral sclerosis: Current perspectives from basic research to the clinic. Prog Neurobiol 2015; 133:1-26. [PMID: 26253783 DOI: 10.1016/j.pneurobio.2015.07.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive degeneration of upper and lower motoneurons, leading to muscle weakness and paralysis, and finally death. Considerable recent advances have been made in basic research and preclinical therapeutic attempts using experimental models, leading to increasing clinical and translational research in the context of this disease. In this review we aim to summarize the most relevant findings from a variety of aspects about ALS, including evaluation methods, animal models, pathophysiology, and clinical findings, with particular emphasis in understanding the role of every contributing mechanism to the disease for elucidating the causes underlying degeneration of motoneurons and the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Renzo Mancuso
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Xavier Navarro
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain.
| |
Collapse
|
39
|
Netzahualcoyotzi C, Tapia R. Degeneration of spinal motor neurons by chronic AMPA-induced excitotoxicity in vivo and protection by energy substrates. Acta Neuropathol Commun 2015; 3:27. [PMID: 25968178 PMCID: PMC4429664 DOI: 10.1186/s40478-015-0205-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Several data suggest that excitotoxicity due to excessive glutamatergic neurotransmission may be an important factor in the mechanisms of motor neuron (MN) death occurring in amyotrophic lateral sclerosis (ALS). We have previously shown that the overactivation of the Ca(2+)-permeable α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) glutamate receptor type, through the continuous infusion of AMPA in the lumbar spinal cord of adult rats during several days, results in progressive rear limb paralysis and bilateral MN degeneration. Because it has been shown that energy failure and oxidative stress are involved in MN degeneration, in both ALS and experimental models of spinal MN degeneration, including excitotoxicity, in this work we tested the protective effect of the energy substrates pyruvate and β-hydroxybutyrate (βHB) and the antioxidants glutathione ethyl ester (GEE) and ascorbate in this chronic AMPA-induced neurodegeneration. RESULTS AMPA infusion induced remarkable progressive motor deficits, assessed by two motor tasks, that by day seven reach bilateral rear limb paralysis. These effects correlate with the death of >80% of lumbar spinal MNs in the infused and the neighbor spinal cord segments, as well as with notable astrogliosis in the ventral horns, detected by glial fibrillary acidic protein immunohistochemistry. Co-infusion with pyruvate or βHB notably prevented the motor deficits and paralysis, decreased MN loss to <25% and completely prevented the induction of astrogliosis. In contrast, the antioxidants tested were ineffective regarding all parameters analyzed. CONCLUSIONS Chronic progressive excitotoxicity due to AMPA receptors overactivation results in MN death and astrogliosis, with consequent motor deficits and paralysis. Because of the notable protection against these effects exerted by pyruvate and βHB, which are well established mitochondrial energy substrates, we conclude that deficits in mitochondrial energy metabolism are an important factor in the mechanisms of this slowly developed excitotoxic MN death, while the lack of protective effect of the antioxidants indicates that oxidative stress seems to be less significant factor. Because excitotoxicity may be involved in MN degeneration in ALS, these findings suggest possible preventive or therapeutic strategies for the disease.
Collapse
|
40
|
Boyer JG, Ferrier A, Kothary R. More than a bystander: the contributions of intrinsic skeletal muscle defects in motor neuron diseases. Front Physiol 2013; 4:356. [PMID: 24391590 PMCID: PMC3866803 DOI: 10.3389/fphys.2013.00356] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 11/20/2013] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy (SMA), amyotrophic lateral sclerosis (ALS), and spinal-bulbar muscular atrophy (SBMA) are devastating diseases characterized by the degeneration of motor neurons. Although the molecular causes underlying these diseases differ, recent findings have highlighted the contribution of intrinsic skeletal muscle defects in motor neuron diseases. The use of cell culture and animal models has led to the important finding that muscle defects occur prior to and independently of motor neuron degeneration in motor neuron diseases. In SMA for instance, the muscle specific requirements of the SMA disease-causing gene have been demonstrated by a series of genetic rescue experiments in SMA models. Conditional ALS mouse models expressing a muscle specific mutant SOD1 gene develop atrophy and muscle degeneration in the absence of motor neuron pathology. Treating SBMA mice by over-expressing IGF-1 in a skeletal muscle-specific manner attenuates disease severity and improves motor neuron pathology. In the present review, we provide an in depth description of muscle intrinsic defects, and discuss how they impact muscle function in these diseases. Furthermore, we discuss muscle-specific therapeutic strategies used to treat animal models of SMA, ALS, and SBMA. The study of intrinsic skeletal muscle defects is crucial for the understanding of the pathophysiology of these diseases and will open new therapeutic options for the treatment of motor neuron diseases.
Collapse
Affiliation(s)
- Justin G Boyer
- Ottawa Hospital Research Institute, Regenerative Medicine Program Ottawa ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa, ON, Canada
| | - Andrew Ferrier
- Ottawa Hospital Research Institute, Regenerative Medicine Program Ottawa ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa, ON, Canada
| | - Rashmi Kothary
- Ottawa Hospital Research Institute, Regenerative Medicine Program Ottawa ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa, ON, Canada ; Department of Medicine, University of Ottawa Ottawa, ON, Canada
| |
Collapse
|
41
|
Chaturvedi RK, Flint Beal M. Mitochondrial diseases of the brain. Free Radic Biol Med 2013; 63:1-29. [PMID: 23567191 DOI: 10.1016/j.freeradbiomed.2013.03.018] [Citation(s) in RCA: 325] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 03/21/2013] [Accepted: 03/22/2013] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders are debilitating diseases of the brain, characterized by behavioral, motor and cognitive impairments. Ample evidence underpins mitochondrial dysfunction as a central causal factor in the pathogenesis of neurodegenerative disorders including Parkinson's disease, Huntington's disease, Alzheimer's disease, Amyotrophic lateral sclerosis, Friedreich's ataxia and Charcot-Marie-Tooth disease. In this review, we discuss the role of mitochondrial dysfunction such as bioenergetics defects, mitochondrial DNA mutations, gene mutations, altered mitochondrial dynamics (mitochondrial fusion/fission, morphology, size, transport/trafficking, and movement), impaired transcription and the association of mutated proteins with mitochondria in these diseases. We highlight the therapeutic role of mitochondrial bioenergetic agents in toxin and in cellular and genetic animal models of neurodegenerative disorders. We also discuss clinical trials of bioenergetics agents in neurodegenerative disorders. Lastly, we shed light on PGC-1α, TORC-1, AMP kinase, Nrf2-ARE, and Sirtuins as novel therapeutic targets for neurodegenerative disorders.
Collapse
Affiliation(s)
- Rajnish K Chaturvedi
- CSIR-Indian Institute of Toxicology Research, 80 MG Marg, Lucknow 226001, India.
| | | |
Collapse
|
42
|
Martin LJ, Wong M. Aberrant regulation of DNA methylation in amyotrophic lateral sclerosis: a new target of disease mechanisms. Neurotherapeutics 2013; 10:722-33. [PMID: 23900692 PMCID: PMC3805862 DOI: 10.1007/s13311-013-0205-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the third most common adult-onset neurodegenerative disease. A diagnosis is fatal owing to degeneration of motor neurons in brain and spinal cord that control swallowing, breathing, and movement. ALS can be inherited, but most cases are not associated with a family history of the disease. The mechanisms causing motor neuron death in ALS are still unknown. Given the suspected complex interplay between multiple genes, the environment, metabolism, and lifestyle in the pathogenesis of ALS, we have hypothesized that the mechanisms of disease in ALS involve epigenetic contributions that can drive motor neuron degeneration. DNA methylation is an epigenetic mechanism for gene regulation engaged by DNA methyltransferase (Dnmt)-catalyzed methyl group transfer to carbon-5 in cytosine residues in gene regulatory promoter and nonpromoter regions. Recent genome-wide analyses have found differential gene methylation in human ALS. Neuropathologic assessments have revealed that motor neurons in human ALS show significant abnormalities in Dnmt1, Dnmt3a, and 5-methylcytosine. Similar changes are seen in mice with motor neuron degeneration, and Dnmt3a was found abundantly at synapses and in mitochondria. During apoptosis of cultured motor neuron-like cells, Dnmt1 and Dnmt3a protein levels increase, and 5-methylcytosine accumulates. Enforced expression of Dnmt3a, but not Dnmt1, induces degeneration of cultured neurons. Truncation mutation of the Dnmt3a catalytic domain and Dnmt3a RNAi blocks apoptosis of cultured neurons. Inhibition of Dnmt catalytic activity with small molecules RG108 and procainamide protects motor neurons from excessive DNA methylation and apoptosis in cell culture and in a mouse model of ALS. Thus, motor neurons can engage epigenetic mechanisms to cause their degeneration, involving Dnmts and increased DNA methylation. Aberrant DNA methylation in vulnerable cells is a new direction for discovering mechanisms of ALS pathogenesis that could be relevant to new disease target identification and therapies for ALS.
Collapse
Affiliation(s)
- Lee J Martin
- Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, 558 Ross Building, 720 Rutland Avenue, Baltimore, MD, 21205-2196, USA,
| | | |
Collapse
|
43
|
Katsetos CD, Koutzaki S, Melvin JJ. Mitochondrial dysfunction in neuromuscular disorders. Semin Pediatr Neurol 2013; 20:202-15. [PMID: 24331362 DOI: 10.1016/j.spen.2013.10.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This review deciphers aspects of mitochondrial (mt) dysfunction among nosologically, pathologically, and genetically diverse diseases of the skeletal muscle, lower motor neuron, and peripheral nerve, which fall outside the traditional realm of mt cytopathies. Special emphasis is given to well-characterized mt abnormalities in collagen VI myopathies (Ullrich congenital muscular dystrophy and Bethlem myopathy), megaconial congenital muscular dystrophy, limb-girdle muscular dystrophy type 2 (calpainopathy), centronuclear myopathies, core myopathies, inflammatory myopathies, spinal muscular atrophy, Charcot-Marie-Tooth neuropathy type 2, and drug-induced peripheral neuropathies. Among inflammatory myopathies, mt abnormalities are more prominent in inclusion body myositis and a subset of polymyositis with mt pathology, both of which are refractory to corticosteroid treatment. Awareness is raised about instances of phenotypic mimicry between cases harboring primary mtDNA depletion, in the context of mtDNA depletion syndrome, and established neuromuscular disorders such as spinal muscular atrophy. A substantial body of experimental work, derived from animal models, attests to a major role of mitochondria (mt) in the early process of muscle degeneration. Common mechanisms of mt-related cell injury include dysregulation of the mt permeability transition pore opening and defective autophagy. The therapeutic use of mt permeability transition pore modifiers holds promise in various neuromuscular disorders, including muscular dystrophies.
Collapse
Affiliation(s)
- Christos D Katsetos
- Department of Pediatrics, Drexel University College of Medicine, St. Christopher's Hospital for Children, Philadelphia, PA; Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA; Department of Neurology, Drexel University College of Medicine, Philadelphia, PA.
| | - Sirma Koutzaki
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA
| | - Joseph J Melvin
- Department of Pediatrics, Drexel University College of Medicine, St. Christopher's Hospital for Children, Philadelphia, PA; Department of Neurology, Drexel University College of Medicine, Philadelphia, PA
| |
Collapse
|
44
|
Su YC, Qi X. Impairment of mitochondrial dynamics: a target for the treatment of neurological disorders? FUTURE NEUROLOGY 2013. [DOI: 10.2217/fnl.13.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondrial dysfunction has long been appreciated in the pathogenesis of various neurological disorders. However, the molecular basis underlying the decline in mitochondrial function is not fully understood. Mitochondria are highly dynamic organelles that frequently undergo fusion and fission. In healthy cells, the delicate balance between fusion and fission is required for maintaining normal mitochondrial and cellular function. However, under pathological conditions, the balance is disrupted, resulting in excessive mitochondrial fragmentation and mitochondrial dysfunction. The impaired fusion and fission processes can lead to apoptosis, necrosis and autophagic cell death and seem to play causal roles in the progression of acute and chronic neuronal injuries. In this article, important aspects of what is currently known about the molecular machinery regulating mitochondrial fission and fusion in mammalian cells is summarized. Special emphasis will be given to the consequences of disregulated mitochondrial morphology in the pathogenesis of neurological diseases.
Collapse
Affiliation(s)
- Yu-Chin Su
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xin Qi
- Center for Mitochondrial Diseases, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, E516, Cleveland, OH, 44106-44970, USA
| |
Collapse
|
45
|
Kumar A, Ghosh D, Singh RL. Amyotrophic Lateral Sclerosis and Metabolomics: Clinical Implication and Therapeutic Approach. J Biomark 2013; 2013:538765. [PMID: 26317018 PMCID: PMC4437352 DOI: 10.1155/2013/538765] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 02/02/2013] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is one of the most common motor neurodegenerative disorders, primarily affecting upper and lower motor neurons in the brain, brainstem, and spinal cord, resulting in paralysis due to muscle weakness and atrophy. The majority of patients die within 3-5 years of symptom onset as a consequence of respiratory failure. Due to relatively fast progression of the disease, early diagnosis is essential. Metabolomics offer a unique opportunity to understand the spatiotemporal metabolic crosstalks through the assessment of body fluids and tissue. So far, one of the most challenging issues related to ALS is to understand the variation of metabolites in body fluids and CNS with the progression of disease. In this paper we will review the changes in metabolic profile in response to disease progression condition and also see the therapeutic implication of various drugs in ALS patients.
Collapse
Affiliation(s)
- Alok Kumar
- Center for Shock, Trauma and Anesthesiology Research (STAR) and the Department of Anesthesiology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Devlina Ghosh
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - R. L. Singh
- Department of Biochemistry, Dr. Ram Manohar Lohia Avadh University, Faizabad 224001, India
| |
Collapse
|
46
|
Bernardini C, Censi F, Lattanzi W, Barba M, Calcagnini G, Giuliani A, Tasca G, Sabatelli M, Ricci E, Michetti F. Mitochondrial network genes in the skeletal muscle of amyotrophic lateral sclerosis patients. PLoS One 2013; 8:e57739. [PMID: 23469062 PMCID: PMC3585165 DOI: 10.1371/journal.pone.0057739] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 01/24/2013] [Indexed: 12/14/2022] Open
Abstract
Recent evidence suggested that muscle degeneration might lead and/or contribute to neurodegeneration, thus it possibly play a key role in the etiopathogenesis and progression of amyotrophic lateral sclerosis (ALS). To test this hypothesis, this study attempted to categorize functionally relevant genes within the genome-wide expression profile of human ALS skeletal muscle, using microarray technology and gene regulatory network analysis. The correlation network structures significantly change between patients and controls, indicating an increased inter-gene connection in patients compared to controls. The gene network observed in the ALS group seems to reflect the perturbation of muscle homeostasis and metabolic balance occurring in affected individuals. In particular, the network observed in the ALS muscles includes genes (PRKR1A, FOXO1, TRIM32, ACTN3, among others), whose functions connect the sarcomere integrity to mitochondrial oxidative metabolism. Overall, the analytical approach used in this study offer the possibility to observe higher levels of correlation (i.e. common expression trends) among genes, whose function seems to be aberrantly activated during the progression of muscle atrophy.
Collapse
Affiliation(s)
- Camilla Bernardini
- Institute of Anatomy and Cell Biology, School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
- * E-mail:
| | - Federica Censi
- Department of Technologies and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Wanda Lattanzi
- Institute of Anatomy and Cell Biology, School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marta Barba
- Institute of Anatomy and Cell Biology, School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Calcagnini
- Department of Technologies and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandro Giuliani
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Mario Sabatelli
- Institute of Neurology, Università Cattolica del Sacro Cuore, School of Medicine, Rome, Italy
| | - Enzo Ricci
- Institute of Neurology, Università Cattolica del Sacro Cuore, School of Medicine, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
47
|
Artuso L, Zoccolella S, Favia P, Amati A, Capozzo R, Logroscino G, Serlenga L, Simone I, Gasparre G, Petruzzella V. Mitochondrial genome aberrations in skeletal muscle of patients with motor neuron disease. Amyotroph Lateral Scler Frontotemporal Degener 2012; 14:261-6. [DOI: 10.3109/21678421.2012.735239] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Manzano R, Toivonen JM, Calvo AC, Oliván S, Zaragoza P, Muñoz MJ, Montarras D, Osta R. Quantity and activation of myofiber-associated satellite cells in a mouse model of amyotrophic lateral sclerosis. Stem Cell Rev Rep 2012; 8:279-87. [PMID: 21537993 DOI: 10.1007/s12015-011-9268-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Raquel Manzano
- LAGENBIO-I3A, Instituto Aragonés de Ciencias de la Salud (IACS), Universidad de Zaragoza, Miguel Servet 177, 50013, Zaragoza, Spain
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Ghiasi P, Hosseinkhani S, Noori A, Nafissi S, Khajeh K. Mitochondrial complex I deficiency and ATP/ADP ratio in lymphocytes of amyotrophic lateral sclerosis patients. Neurol Res 2012; 34:297-303. [PMID: 22450425 DOI: 10.1179/1743132812y.0000000012] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Several lines of evidence suggest that mitochondrial dysfunction is involved in amyotrophic lateral sclerosis (ALS), but despite the fact that mitochondria play a central role in excitotoxicity, oxidative stress, and apoptosis, the intimate underlying mechanism linking mitochondrial defects to motor neuron degeneration in ALS still remains elusive. This study was performed to assess the mitochondrial respiratory chain dysfunction and cellular energy index (ATP/ADP ratio) in lymphocytes of ALS patients. METHODS In this study, activity of mitochondrial respiratory chain complex I (measured as NADH-ferricyanide reductase) and both intracellular ATP and ADP measurements were performed on lymphocytes of ALS patients (n = 14) and control subjects (n = 26). Then, ATP/ADP ratio was calculated. RESULTS Our finding showed that in patients compared with controls, complex I activity and intracellular ATP were significantly reduced (P = 0·001) and intracellular ADP content was increased (P<0·005) and ATP/ADP ratio subsequently was decreased and also we found strong correlation between complex I activity and intracellular ATP content and strong reverse correlation between complex I activity and intracellular ADP content in the patients with ALS (r(2) = 0·90). DISCUSSION This study suggests that complex I deficiency and both reduction in intracellular ATP and increase in intracellular ADP content may be involved in the progression and pathogenesis of ALS.
Collapse
Affiliation(s)
- Parisa Ghiasi
- Department of Biochemistry, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | |
Collapse
|
50
|
Mezzani A, Pisano F, Cavalli A, Tommasi MA, Corrà U, Colombo S, Grassi B, Marzorati M, Porcelli S, Morandi L, Giannuzzi P. Reduced exercise capacity in early-stage amyotrophic lateral sclerosis: Role of skeletal muscle. ACTA ACUST UNITED AC 2011; 13:87-94. [DOI: 10.3109/17482968.2011.601463] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|