1
|
Ratano P, Cocozza G, Pinchera C, Busdraghi LM, Cantando I, Martinello K, Scioli M, Rosito M, Bezzi P, Fucile S, Wulff H, Limatola C, D’Alessandro G. Reduction of inflammation and mitochondrial degeneration in mutant SOD1 mice through inhibition of voltage-gated potassium channel Kv1.3. Front Mol Neurosci 2024; 16:1333745. [PMID: 38292023 PMCID: PMC10824952 DOI: 10.3389/fnmol.2023.1333745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/31/2023] [Indexed: 02/01/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with no effective therapy, causing progressive loss of motor neurons in the spinal cord, brainstem, and motor cortex. Regardless of its genetic or sporadic origin, there is currently no cure for ALS or therapy that can reverse or control its progression. In the present study, taking advantage of a human superoxide dismutase-1 mutant (hSOD1-G93A) mouse that recapitulates key pathological features of human ALS, we investigated the possible role of voltage-gated potassium channel Kv1.3 in disease progression. We found that chronic administration of the brain-penetrant Kv1.3 inhibitor, PAP-1 (40 mg/Kg), in early symptomatic mice (i) improves motor deficits and prolongs survival of diseased mice (ii) reduces astrocyte reactivity, microglial Kv1.3 expression, and serum pro-inflammatory soluble factors (iii) improves structural mitochondrial deficits in motor neuron mitochondria (iv) restores mitochondrial respiratory dysfunction. Taken together, these findings underscore the potential significance of Kv1.3 activity as a contributing factor to the metabolic disturbances observed in ALS. Consequently, targeting Kv1.3 presents a promising avenue for modulating disease progression, shedding new light on potential therapeutic strategies for ALS.
Collapse
Affiliation(s)
| | - Germana Cocozza
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | | | | | - Iva Cantando
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | | | | | - Maria Rosito
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - Paola Bezzi
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Sergio Fucile
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| | - Heike Wulff
- Department of Pharmacology, University of California Davis, Health Sciences Drive, Davis, CA, United States
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur, Sapienza University, Rome, Italy
| | - Giuseppina D’Alessandro
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy
| |
Collapse
|
2
|
Hnath B, Chen J, Reynolds J, Choi E, Wang J, Zhang D, Sha CM, Dokholyan NV. Big versus small: The impact of aggregate size in disease. Protein Sci 2023; 32:e4686. [PMID: 37243896 PMCID: PMC10273386 DOI: 10.1002/pro.4686] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Protein aggregation results in an array of different size soluble oligomers and larger insoluble fibrils. Insoluble fibrils were originally thought to cause neuronal cell deaths in neurodegenerative diseases due to their prevalence in tissue samples and disease models. Despite recent studies demonstrating the toxicity associated with soluble oligomers, many therapeutic strategies still focus on fibrils or consider all types of aggregates as one group. Oligomers and fibrils require different modeling and therapeutic strategies, targeting the toxic species is crucial for successful study and therapeutic development. Here, we review the role of different-size aggregates in disease, and how factors contributing to aggregation (mutations, metals, post-translational modifications, and lipid interactions) may promote oligomers opposed to fibrils. We review two different computational modeling strategies (molecular dynamics and kinetic modeling) and how they are used to model both oligomers and fibrils. Finally, we outline the current therapeutic strategies targeting aggregating proteins and their strengths and weaknesses for targeting oligomers versus fibrils. Altogether, we aim to highlight the importance of distinguishing the difference between oligomers and fibrils and determining which species is toxic when modeling and creating therapeutics for protein aggregation in disease.
Collapse
Affiliation(s)
- Brianna Hnath
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jiaxing Chen
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Joshua Reynolds
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Esther Choi
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jian Wang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Dongyan Zhang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Congzhou M. Sha
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
| | - Nikolay V. Dokholyan
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of Biochemistry & Molecular BiologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of ChemistryPenn State UniversityUniversity ParkPennsylvaniaUSA
| |
Collapse
|
3
|
Damaged DNA Is an Early Event of Neurodegeneration in Induced Pluripotent Stem Cell-Derived Motoneurons with UBQLN2P497H Mutation. Int J Mol Sci 2022; 23:ijms231911333. [PMID: 36232630 PMCID: PMC9570184 DOI: 10.3390/ijms231911333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 12/03/2022] Open
Abstract
Ubiquilin-2 (UBQLN2) mutations lead to familial amyotrophic lateral sclerosis (FALS)/and frontotemporal dementia (FTLD) through unknown mechanisms. The combination of iPSC technology and CRISPR-mediated genome editing technology can generate an iPSC-derived motor neuron (iPSC-MN) model with disease-relevant mutations, which results in increased opportunities for disease mechanism research and drug screening. In this study, we introduced a UBQLN2-P497H mutation into a healthy control iPSC line using CRISPR/Cas9, and differentiated into MNs to study the pathology of UBQLN2-related ALS. Our in vitro MN model faithfully recapitulated specific aspects of the disease, including MN apoptosis. Under sodium arsenite (SA) treatment, we found differences in the number and the size of UBQLN2+ inclusions in UBQLN2P497H MNs and wild-type (WT) MNs. We also observed cytoplasmic TAR DNA-binding protein (TARDBP, also known as TDP-43) aggregates in UBQLN2P497H MNs, but not in WT MNs, as well as the recruitment of TDP-43 into stress granules (SGs) upon SA treatment. We noted that UBQLN2-P497H mutation induced MNs DNA damage, which is an early event in UBQLN2-ALS. Additionally, DNA damage led to an increase in compensation for FUS, whereas UBQLN2-P497H mutation impaired this function. Therefore, FUS may be involved in DNA damage repair signaling.
Collapse
|
4
|
Kaur J, Gulati M, Kapoor B, Jha NK, Gupta PK, Gupta G, Chellappan DK, Devkota HP, Prasher P, Ansari MS, Aba Alkhayl FF, Arshad MF, Morris A, Choonara YE, Adams J, Dua K, Singh SK. Advances in designing of polymeric micelles for biomedical application in brain related diseases. Chem Biol Interact 2022; 361:109960. [PMID: 35533733 DOI: 10.1016/j.cbi.2022.109960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/11/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022]
|
5
|
Dhasmana S, Dhasmana A, Narula AS, Jaggi M, Yallapu MM, Chauhan SC. The panoramic view of amyotrophic lateral sclerosis: A fatal intricate neurological disorder. Life Sci 2022; 288:120156. [PMID: 34801512 DOI: 10.1016/j.lfs.2021.120156] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal neurological disease affecting both upper and lower motor neurons. In the United States alone, there are 16,000-20,000 established cases of ALS. The early disease diagnosis is challenging due to many overlapping pathophysiologies with other neurological diseases. The etiology of ALS is unknown; however, it is divided into two categories: familial ALS (fALS) which occurs due to gene mutations & contributes to 5-10% of ALS, and sporadic ALS (sALS) which is due to environmental factors & contributes to 90-95% of ALS. There is still no curative treatment for ALS: palliative care and symptomatic treatment are therefore essential components in the management of these patients. In this review, we provide a panoramic view of ALS, which includes epidemiology, risk factors, pathophysiologies, biomarkers, diagnosis, therapeutics (natural, synthetic, gene-based, pharmacological, stem cell, extracellular vesicles, and physical therapy), controversies (in the clinical trials of ALS), the scope of nanomedicine in ALS, and future perspectives.
Collapse
Affiliation(s)
- Swati Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Acharan S Narula
- Narula Research LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA.
| |
Collapse
|
6
|
Scaricamazza S, Salvatori I, Amadio S, Nesci V, Torcinaro A, Giacovazzo G, Primiano A, Gloriani M, Candelise N, Pieroni L, Loeffler JP, Renè F, Quessada C, Tefera TW, Wang H, Steyn FJ, Ngo ST, Dobrowolny G, Lepore E, Urbani A, Musarò A, Volonté C, Ferraro E, Coccurello R, Valle C, Ferri A. Repurposing of Trimetazidine for Amyotrophic Lateral Sclerosis: a study in SOD1 G93A mice. Br J Pharmacol 2021; 179:1732-1752. [PMID: 34783031 PMCID: PMC9305494 DOI: 10.1111/bph.15738] [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: 07/16/2021] [Revised: 09/09/2021] [Accepted: 10/23/2021] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose Amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by the degeneration of upper and lower motor neurons, progressive wasting and paralysis of voluntary muscles and is currently incurable. Although considered to be a pure motor neuron disease, increasing evidence indicates that the sole protection of motor neurons by a single targeted drug is not sufficient to improve the pathological phenotype. We therefore evaluated the therapeutic potential of the multi‐target drug used to treatment of coronary artery disease, trimetazidine, in SOD1G93A mice. Experimental Approach As a metabolic modulator, trimetazidine improves glucose metabolism. Furthermore, trimetazidine enhances mitochondrial metabolism and promotes nerve regeneration, exerting an anti‐inflammatory and antioxidant effect. We orally treated SOD1G93A mice with trimetazidine, solubilized in drinking water at a dose of 20 mg kg−1, from disease onset. We assessed the impact of trimetazidine on disease progression by studying metabolic parameters, grip strength and histological alterations in skeletal muscle, peripheral nerves and the spinal cord. Key Results Trimetazidine administration delays motor function decline, improves muscle performance and metabolism, and significantly extends overall survival of SOD1G93A mice (increased median survival of 16 days and 12.5 days for male and female respectively). Moreover, trimetazidine prevents the degeneration of neuromuscular junctions, attenuates motor neuron loss and reduces neuroinflammation in the spinal cord and in peripheral nerves. Conclusion and Implications In SOD1G93A mice, therapeutic effect of trimetazidine is underpinned by its action on mitochondrial function in skeletal muscle and spinal cord.
Collapse
Affiliation(s)
- Silvia Scaricamazza
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Illari Salvatori
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Experimental Medicine, University of Roma "La Sapienza", Rome, Italy
| | | | | | - Alessio Torcinaro
- National Council of Research (CNR), Institute of Cell Biology and Neurology (IBCN), Rome, Italy
| | - Giacomo Giacovazzo
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Aniello Primiano
- Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | | | - Niccolò Candelise
- IRCCS Fondazione Santa Lucia, Rome, Italy.,National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | | | - Jean-Philippe Loeffler
- Université de Strasbourg, UMR_S 1118, Strasbourg, France.,INSERM, U1118, Central and Peripheral Mechanisms of Neurodegeneration, Strasbourg, France
| | - Frederique Renè
- Université de Strasbourg, UMR_S 1118, Strasbourg, France.,INSERM, U1118, Central and Peripheral Mechanisms of Neurodegeneration, Strasbourg, France
| | - Cyril Quessada
- Université de Strasbourg, UMR_S 1118, Strasbourg, France.,INSERM, U1118, Central and Peripheral Mechanisms of Neurodegeneration, Strasbourg, France
| | - Tesfaye W Tefera
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Hao Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Frederik J Steyn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.,Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Shyuan T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Gabriella Dobrowolny
- University of Roma "La Sapienza", DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Elisa Lepore
- University of Roma "La Sapienza", DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Andrea Urbani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy.,National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Antonio Musarò
- University of Roma "La Sapienza", DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Cinzia Volonté
- IRCCS Fondazione Santa Lucia, Rome, Italy.,National Research Council (CNR), Institute for Systems Analysis and Computer Science (IASI), Rome, Italy
| | | | - Roberto Coccurello
- IRCCS Fondazione Santa Lucia, Rome, Italy.,National Research Council (CNR), Institute for Complex System (ISC), Rome, Italy
| | - Cristiana Valle
- IRCCS Fondazione Santa Lucia, Rome, Italy.,National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Alberto Ferri
- IRCCS Fondazione Santa Lucia, Rome, Italy.,National Research Council (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
| |
Collapse
|
7
|
Zhang M, Gelfman S, McCarthy J, Harms MB, Moreno CAM, Goldstein DB, Allen AS. Incorporating external information to improve sparse signal detection in rare-variant gene-set-based analyses. Genet Epidemiol 2020; 44:330-338. [PMID: 32043633 DOI: 10.1002/gepi.22283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/17/2019] [Accepted: 01/27/2020] [Indexed: 01/30/2023]
Abstract
Gene-set analyses are used to assess whether there is any evidence of association with disease among a set of biologically related genes. Such an analysis typically treats all genes within the sets similarly, even though there is substantial, external, information concerning the likely importance of each gene within each set. For example, for traits that are under purifying selection, we would expect genes showing extensive genic constraint to be more likely to be trait associated than unconstrained genes. Here we improve gene-set analyses by incorporating such external information into a higher-criticism-based signal detection analysis. We show that when this external information is predictive of whether a gene is associated with disease, our approach can lead to a significant increase in power. Further, our approach is particularly powerful when the signal is sparse, that is when only a small number of genes within the set are associated with the trait. We illustrate our approach with a gene-set analysis of amyotrophic lateral sclerosis (ALS) and implicate a number of gene-sets containing SOD1 and NEK1 as well as showing enrichment of small p values for gene-sets containing known ALS genes. We implement our approach in the R package wHC.
Collapse
Affiliation(s)
- Mengqi Zhang
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina.,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina.,Center for Statistical Genetics and Genomics, Duke University, Durham, North Carolina
| | - Sahar Gelfman
- Institute of Genomic Medicine, Columbia University, New York City, New York
| | - Janice McCarthy
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Matthew B Harms
- Institute of Genomic Medicine, Columbia University, New York City, New York.,Department of Neurology, Columbia University, New York City, New York.,Center for Motor Neuron Biology and Disease, Columbia University, New York City, New York
| | - Cristiane A M Moreno
- Institute of Genomic Medicine, Columbia University, New York City, New York.,Center for Motor Neuron Biology and Disease, Columbia University, New York City, New York
| | - David B Goldstein
- Institute of Genomic Medicine, Columbia University, New York City, New York
| | - Andrew S Allen
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina.,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina.,Center for Statistical Genetics and Genomics, Duke University, Durham, North Carolina
| |
Collapse
|
8
|
Apolloni S, Amadio S, Fabbrizio P, Morello G, Spampinato AG, Latagliata EC, Salvatori I, Proietti D, Ferri A, Madaro L, Puglisi-Allegra S, Cavallaro S, Volonté C. Histaminergic transmission slows progression of amyotrophic lateral sclerosis. J Cachexia Sarcopenia Muscle 2019; 10:872-893. [PMID: 31020811 PMCID: PMC6711424 DOI: 10.1002/jcsm.12422] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Histamine is an immune modulator, neuroprotective, and remyelinating agent, beneficially acting on skeletal muscles and promoting anti-inflammatory features in amyotrophic lateral sclerosis (ALS) microglia. Drugs potentiating the endogenous release of histamine are in trial for neurological diseases, with a role not systematically investigated in ALS. Here, we examine histamine pathway associations in ALS patients and the efficacy of a histamine-mediated therapeutic strategy in ALS mice. METHODS We adopted an integrative multi-omics approach combining gene expression profiles, copy number variants, and single nucleotide polymorphisms of ALS patients. We treated superoxide dismutase 1 (SOD1)-G93A mice that recapitulate key ALS features, with the brain-permeable histamine precursor histidine in the symptomatic phase of the disease and analysed the rescue from disease pathological signs. We examined the action of histamine in cultured SOD1-G93A motor neuron-like cells. RESULTS We identified 13 histamine-related genes deregulated in the spinal cord of two ALS patient subgroups, among which genes involved in histamine metabolism, receptors, transport, and secretion. Some histamine-related genes overlapped with genomic regions disrupted by DNA copy number and with ALS-linked pathogenic variants. Histidine treatment in SOD1-G93A mice proved broad efficacy in ameliorating ALS features, among which most importantly lifespan, motor performance, microgliosis, muscle atrophy, and motor neurons survival in vivo and in vitro. CONCLUSIONS Our gene set/pathway enrichment analyses and preclinical studies started at the onset of symptoms establish that histamine-related genes are modifiers in ALS, supporting their role as candidate biomarkers and therapeutic targets. We disclose a novel important role for histamine in the characterization of the multi-gene network responsible for ALS and, furthermore, in the drug development process.
Collapse
Affiliation(s)
| | | | - Paola Fabbrizio
- IRCCS Fondazione Santa Lucia, Rome, Italy.,National Research Council, Institute of Cell Biology and Neurobiology, Rome, Italy
| | - Giovanna Morello
- National Research Council, Institute of Neurological Sciences, Catania, Italy
| | | | | | | | | | - Alberto Ferri
- IRCCS Fondazione Santa Lucia, Rome, Italy.,National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | | | | | | | - Cinzia Volonté
- IRCCS Fondazione Santa Lucia, Rome, Italy.,National Research Council, Institute of Cell Biology and Neurobiology, Rome, Italy
| |
Collapse
|
9
|
Zhang F, Liu M, Li Q, Song FX. Exploration of attractor modules for sporadic amyotrophic lateral sclerosis via systemic module inference and attract method. Exp Ther Med 2019; 17:2575-2580. [PMID: 30906448 PMCID: PMC6425136 DOI: 10.3892/etm.2019.7264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 02/01/2019] [Indexed: 12/01/2022] Open
Abstract
Sporadic amyotrophic lateral sclerosis (SALS) is a devastating neurodegenerative disorder. However, the understanding of SALS is still poor. This research aimed to excavate attractor modules for SALS by integrating the systemic module inference and attract method. To achieve this, gene expression data and protein-protein data were recruited and preprocessed. Then, based on the Spearman's correlation coefficient (SCC) of the interactions under these two conditions, two PPI networks separately with 870 nodes (979 interactions) in normal control group and 601 nodes (777 interactions) in SALS group were built. Systemic module inference method was performed to identify the modules, and attract method was used to identify attractor modules. Finally, pathway enrichment analysis was performed to disclose the functional enrichment of these attractor modules. In total 44 and 118 modules were identified for normal control and SALS groups, respectively. Among them, 6 modules were with similar gene composition between the two groups, and all 6 modules were considered as the attractor module via attract method. These attractor modules might be potential biomarkers for early diagnosis and therapy of SALS, which could provide insight into the disease biology and suggest possible directions for drug screening programs.
Collapse
Affiliation(s)
- Fang Zhang
- Department of Rehabilitation, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Mei Liu
- Department of Rehabilitation, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Qun Li
- Department of Rehabilitation, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Fei-Xue Song
- Department of Oncology, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
10
|
Raloxifene, a promising estrogen replacement, limits TDP-25 cell death by enhancing autophagy and suppressing apoptosis. Brain Res Bull 2018; 140:281-290. [DOI: 10.1016/j.brainresbull.2018.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/29/2018] [Accepted: 05/21/2018] [Indexed: 12/11/2022]
|
11
|
Handley EE, Pitman KA, Dawkins E, Young KM, Clark RM, Jiang TC, Turner BJ, Dickson TC, Blizzard CA. Synapse Dysfunction of Layer V Pyramidal Neurons Precedes Neurodegeneration in a Mouse Model of TDP-43 Proteinopathies. Cereb Cortex 2018; 27:3630-3647. [PMID: 27496536 DOI: 10.1093/cercor/bhw185] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
TDP-43 is a major protein component of pathological neuronal inclusions that are present in frontotemporal dementia and amyotrophic lateral sclerosis. We report that TDP-43 plays an important role in dendritic spine formation in the cortex. The density of spines on YFP+ pyramidal neurons in both the motor and somatosensory cortex of Thy1-YFP mice, increased significantly from postnatal day 30 (P30), to peak at P60, before being pruned by P90. By comparison, dendritic spine density was significantly reduced in the motor cortex of Thy1-YFP::TDP-43A315T transgenic mice prior to symptom onset (P60), and in the motor and somatosensory cortex at symptom onset (P90). Morphological spine-type analysis revealed that there was a significant impairment in the development of basal mushroom spines in the motor cortex of Thy1-YFP::TDP-43A315T mice compared to Thy1-YFP control. Furthermore, reductions in spine density corresponded to mislocalisation of TDP-43 immunoreactivity and lowered efficacy of synaptic transmission as determined by electrophysiology at P60. We conclude that mutated TDP-43 has a significant pathological effect at the dendritic spine that is associated with attenuated neural transmission.
Collapse
Affiliation(s)
- Emily E Handley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Edgar Dawkins
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Rosemary M Clark
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Tongcui C Jiang
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Bradley J Turner
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Tracey C Dickson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Catherine A Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
12
|
Oberstadt M, Stieler J, Simpong DL, Römuß U, Urban N, Schaefer M, Arendt T, Holzer M. TDP-43 self-interaction is modulated by redox-active compounds Auranofin, Chelerythrine and Riluzole. Sci Rep 2018; 8:2248. [PMID: 29396541 PMCID: PMC5797228 DOI: 10.1038/s41598-018-20565-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 01/19/2018] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) represents a fatal neurodegenerative disease, which is characterized by a rapid loss of lower and upper motor neurons. As a major neuropathological hallmark, protein aggregates containing the Transactivating Response Region (TAR) DNA Binding Protein (TDP-43) are detectable in about 95% of sporadic ALS patients. TDP-43 interacts with itself physiologically to form liquid droplets, which may progress to pathological aggregates. In this study, we established the NanoBit luciferase complementation assay to measure TDP-43 self-interaction and found the fusion of the split luciferase subunits to the N-terminus of the protein as the strongest interacting partners. A screen of pharmacologically active compounds from the LOPAC®1280 library identified auranofin, chelerythrine and riluzole as dose-dependent inhibitors of TDP-43 self-interaction. Further analysis of drug action of the gold-containing thioredoxin reductase inhibitor auranofin revealed a redistribution from insoluble TDP-43 protein pool to PBS-soluble protein pool in N2a cells. In addition, auranofin treatment diminished reduced glutathione as a sign for oxidative modulation.
Collapse
Affiliation(s)
- Moritz Oberstadt
- Department of Neurology, University of Leipzig, Liebigstraße 20, 04103, Leipzig, Germany.
| | - Jens Stieler
- Department for Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Liebigstraße 19, 04103, Leipzig, Germany
| | - David Larbi Simpong
- Department for Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Liebigstraße 19, 04103, Leipzig, Germany
| | - Ute Römuß
- Department of Neurology, University of Leipzig, Liebigstraße 20, 04103, Leipzig, Germany
| | - Nicole Urban
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstraße 16-18, 04107, Leipzig, Germany
| | - Michael Schaefer
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, Härtelstraße 16-18, 04107, Leipzig, Germany
| | - Thomas Arendt
- Department for Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Liebigstraße 19, 04103, Leipzig, Germany
| | - Max Holzer
- Department for Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Liebigstraße 19, 04103, Leipzig, Germany
| |
Collapse
|
13
|
Ciervo Y, Ning K, Jun X, Shaw PJ, Mead RJ. Advances, challenges and future directions for stem cell therapy in amyotrophic lateral sclerosis. Mol Neurodegener 2017; 12:85. [PMID: 29132389 PMCID: PMC5683324 DOI: 10.1186/s13024-017-0227-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/02/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative condition where loss of motor neurons within the brain and spinal cord leads to muscle atrophy, weakness, paralysis and ultimately death within 3–5 years from onset of symptoms. The specific molecular mechanisms underlying the disease pathology are not fully understood and neuroprotective treatment options are minimally effective. In recent years, stem cell transplantation as a new therapy for ALS patients has been extensively investigated, becoming an intense and debated field of study. In several preclinical studies using the SOD1G93A mouse model of ALS, stem cells were demonstrated to be neuroprotective, effectively delayed disease onset and extended survival. Despite substantial improvements in stem cell technology and promising results in preclinical studies, several questions still remain unanswered, such as the identification of the most suitable and beneficial cell source, cell dose, route of delivery and therapeutic mechanisms. This review will cover publications in this field and comprehensively discuss advances, challenges and future direction regarding the therapeutic potential of stem cells in ALS, with a focus on mesenchymal stem cells. In summary, given their high proliferation activity, immunomodulation, multi-differentiation potential, and the capacity to secrete neuroprotective factors, adult mesenchymal stem cells represent a promising candidate for clinical translation. However, technical hurdles such as optimal dose, differentiation state, route of administration, and the underlying potential therapeutic mechanisms still need to be assessed.
Collapse
Affiliation(s)
- Yuri Ciervo
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, 385a Glossop Rd S10 2HQ, Sheffield, UK.,Tongji University School of Medicine, 1239 Siping Rd, Yangpu Qu, Shanghai, China
| | - Ke Ning
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, 385a Glossop Rd S10 2HQ, Sheffield, UK.,Tongji University School of Medicine, 1239 Siping Rd, Yangpu Qu, Shanghai, China
| | - Xu Jun
- Tongji University School of Medicine, 1239 Siping Rd, Yangpu Qu, Shanghai, China
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, 385a Glossop Rd S10 2HQ, Sheffield, UK
| | - Richard J Mead
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, 385a Glossop Rd S10 2HQ, Sheffield, UK.
| |
Collapse
|
14
|
HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients. Nat Commun 2017; 8:861. [PMID: 29021520 PMCID: PMC5636840 DOI: 10.1038/s41467-017-00911-y] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 08/04/2017] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder due to selective loss of motor neurons (MNs). Mutations in the fused in sarcoma (FUS) gene can cause both juvenile and late onset ALS. We generated and characterized induced pluripotent stem cells (iPSCs) from ALS patients with different FUS mutations, as well as from healthy controls. Patient-derived MNs show typical cytoplasmic FUS pathology, hypoexcitability, as well as progressive axonal transport defects. Axonal transport defects are rescued by CRISPR/Cas9-mediated genetic correction of the FUS mutation in patient-derived iPSCs. Moreover, these defects are reproduced by expressing mutant FUS in human embryonic stem cells (hESCs), whereas knockdown of endogenous FUS has no effect, confirming that these pathological changes are mutant FUS dependent. Pharmacological inhibition as well as genetic silencing of histone deacetylase 6 (HDAC6) increase α-tubulin acetylation, endoplasmic reticulum (ER)–mitochondrial overlay, and restore the axonal transport defects in patient-derived MNs. Amyotrophic lateral sclerosis (ALS) leads to selective loss of motor neurons. Using motor neurons derived from induced pluripotent stem cells from patients with ALS and FUS mutations, the authors demonstrate that axonal transport deficits that are observed in these cells can be rescued by HDAC6 inhibition.
Collapse
|
15
|
Oxidative stress induced by cumene hydroperoxide produces synaptic depression and transient hyperexcitability in rat primary motor cortex neurons. Mol Cell Neurosci 2017. [DOI: 10.1016/j.mcn.2017.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
16
|
Paganoni S, Macklin EA, Karam C, Yu H, Gonterman F, Fetterman KA, Cudkowicz M, Berry J, Wills A. Vitamin D levels are associated with gross motor function in amyotrophic lateral sclerosis. Muscle Nerve 2017; 56:726-731. [DOI: 10.1002/mus.25555] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/27/2016] [Accepted: 12/30/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Sabrina Paganoni
- Neurological Clinical Research Institute, Harvard Medical School, Massachusetts General Hospital165 Cambridge Street, Suite 600, Boston Massachusetts02114 USA
- Department of NeurologyNeurological Clinical Research Institute, Massachusetts General HospitalBoston Massachusetts USA
- Department of Physical Medicine and RehabilitationSpaulding Rehabilitation HospitalCambridge Massachusetts USA
| | - Eric A. Macklin
- Neurological Clinical Research Institute, Harvard Medical School, Massachusetts General Hospital165 Cambridge Street, Suite 600, Boston Massachusetts02114 USA
- Biostatistics Center, Massachusetts General HospitalBoston Massachusetts USA
| | - Chafic Karam
- Department of NeurologyOregon Health and Science UniversityPortland Oregon USA
| | - Hong Yu
- Department of NeurologyNeurological Clinical Research Institute, Massachusetts General HospitalBoston Massachusetts USA
| | - Fernando Gonterman
- Department of NeurologyNeurological Clinical Research Institute, Massachusetts General HospitalBoston Massachusetts USA
| | - K. Ashley Fetterman
- Department of NeurologyNeurological Clinical Research Institute, Massachusetts General HospitalBoston Massachusetts USA
| | - Merit Cudkowicz
- Neurological Clinical Research Institute, Harvard Medical School, Massachusetts General Hospital165 Cambridge Street, Suite 600, Boston Massachusetts02114 USA
- Department of NeurologyNeurological Clinical Research Institute, Massachusetts General HospitalBoston Massachusetts USA
| | - James Berry
- Neurological Clinical Research Institute, Harvard Medical School, Massachusetts General Hospital165 Cambridge Street, Suite 600, Boston Massachusetts02114 USA
- Department of NeurologyNeurological Clinical Research Institute, Massachusetts General HospitalBoston Massachusetts USA
| | - Anne‐Marie Wills
- Neurological Clinical Research Institute, Harvard Medical School, Massachusetts General Hospital165 Cambridge Street, Suite 600, Boston Massachusetts02114 USA
- Department of NeurologyNeurological Clinical Research Institute, Massachusetts General HospitalBoston Massachusetts USA
| |
Collapse
|
17
|
Gonzalez-Freire M, Semba RD, Ubaida-Mohien C, Fabbri E, Scalzo P, Højlund K, Dufresne C, Lyashkov A, Ferrucci L. The Human Skeletal Muscle Proteome Project: a reappraisal of the current literature. J Cachexia Sarcopenia Muscle 2017; 8:5-18. [PMID: 27897395 PMCID: PMC5326819 DOI: 10.1002/jcsm.12121] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/11/2016] [Accepted: 04/05/2016] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle is a large organ that accounts for up to half the total mass of the human body. A progressive decline in muscle mass and strength occurs with ageing and in some individuals configures the syndrome of 'sarcopenia', a condition that impairs mobility, challenges autonomy, and is a risk factor for mortality. The mechanisms leading to sarcopenia as well as myopathies are still little understood. The Human Skeletal Muscle Proteome Project was initiated with the aim to characterize muscle proteins and how they change with ageing and disease. We conducted an extensive review of the literature and analysed publically available protein databases. A systematic search of peer-reviewed studies was performed using PubMed. Search terms included 'human', 'skeletal muscle', 'proteome', 'proteomic(s)', and 'mass spectrometry', 'liquid chromatography-mass spectrometry (LC-MS/MS)'. A catalogue of 5431 non-redundant muscle proteins identified by mass spectrometry-based proteomics from 38 peer-reviewed scientific publications from 2002 to November 2015 was created. We also developed a nosology system for the classification of muscle proteins based on localization and function. Such inventory of proteins should serve as a useful background reference for future research on changes in muscle proteome assessed by quantitative mass spectrometry-based proteomic approaches that occur with ageing and diseases. This classification and compilation of the human skeletal muscle proteome can be used for the identification and quantification of proteins in skeletal muscle to discover new mechanisms for sarcopenia and specific muscle diseases that can be targeted for the prevention and treatment.
Collapse
Affiliation(s)
| | - Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Elisa Fabbri
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Paul Scalzo
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Kurt Højlund
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research and Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Alexey Lyashkov
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
18
|
Li XY, Liang ZH, Han C, Wei WJ, Song CL, Zhou LN, Liu Y, Li Y, Ji XF, Liu J. Transplantation of autologous peripheral blood mononuclear cells in the subarachnoid space for amyotrophic lateral sclerosis: a safety analysis of 14 patients. Neural Regen Res 2017; 12:493-498. [PMID: 28469667 PMCID: PMC5399730 DOI: 10.4103/1673-5374.202918] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
There is a small amount of clinical data regarding the safety and feasibility of autologous peripheral blood mononuclear cell transplantation into the subarachnoid space for the treatment of amyotrophic lateral sclerosis. The objectives of this retrospective study were to assess the safety and efficacy of peripheral blood mononuclear cell transplantation in 14 amyotrophic lateral sclerosis patients to provide more objective data for future clinical trials. After stem cell mobilization and collection, autologous peripheral blood mononuclear cells (1 × 109) were isolated and directly transplanted into the subarachnoid space of amyotrophic lateral sclerosis patients. The primary outcome measure was incidence of adverse events. Secondary outcome measures were electromyography 1 week before operation and 4 weeks after operation, Functional Independence Measurement, Berg Balance Scale, and Dysarthria Assessment Scale 1 week preoperatively and 1, 2, 4 and 12 weeks postoperatively. There was no immediate or delayed transplant-related cytotoxicity. The number of leukocytes, serum alanine aminotransferase and creatinine levels, and body temperature were within the normal ranges. Radiographic evaluation showed no serious transplant-related adverse events. Muscle strength grade, results of Functional Independence Measurement, Berg Balance Scale, and Dysarthria Assessment Scale were not significantly different before and after treatment. These findings suggest that peripheral blood mononuclear cell transplantation into the subarachnoid space for the treatment of amyotrophic lateral sclerosis is safe, but its therapeutic effect is not remarkable. Thus, a large-sample investigation is needed to assess its efficacy further.
Collapse
Affiliation(s)
- Xiao-Yan Li
- Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Zhan-Hua Liang
- Neurological Department, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Chao Han
- Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Wen-Juan Wei
- Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Chun-Li Song
- Electromyography Department, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Li-Na Zhou
- Electromyography Department, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yang Liu
- Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Ying Li
- Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Xiao-Fei Ji
- Neurological Department, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Jing Liu
- Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
19
|
Wang W, Duan W, Wang Y, Wen D, Liu Y, Li Z, Hu H, Cui H, Cui C, Lin H, Li C. Intrathecal Delivery of ssAAV9-DAO Extends Survival in SOD1 G93A ALS Mice. Neurochem Res 2016; 42:986-996. [PMID: 28025800 DOI: 10.1007/s11064-016-2131-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/24/2016] [Accepted: 12/02/2016] [Indexed: 01/18/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset, irreversible neurodegenerative disease that leads to progressive paralysis and inevitable death 3-5 years after diagnosis. The mechanisms underlying this process remain unknown, but new evidence indicates that accumulating levels of D-serine result from the downregulation of D-amino acid oxidase (DAO) and that this is a novel mechanism that leads to motoneuronal death in ALS via N-methyl-D-aspartate receptor-mediated cell toxicity. Here, we explored a new therapeutic approach to ALS by overexpressing DAO in the lumbar region of the mouse spinal cord using a single stranded adeno-associated virus serotype 9 (ssAAV9) vector. A single intrathecal injection of ssAAV9-DAO was made in SOD1G93A mice, a well-established mouse model of ALS. Treatment resulted in moderate expression of exogenous DAO in motorneurons in the lumbar spinal cord, reduced immunoreactivity of D-serine, alleviated motoneuronal loss and glial activation, and extended survival. The potential mechanisms underlying these effects were associated with the down-regulation of NF-κB and the restoration of the phosphorylation of Akt. In conclusion, administering ssAAV9-DAO may be an effective complementary approach to gene therapy to extend lifespans in symptomatic ALS.
Collapse
Affiliation(s)
- Wan Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Weisong Duan
- Neurological Laboratory of Hebei Province, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Ying Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Di Wen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Yakun Liu
- Neurological Laboratory of Hebei Province, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Zhongyao Li
- Neurological Laboratory of Hebei Province, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Haojie Hu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Hongying Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Can Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Huiqian Lin
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Chunyan Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China.
- Institute of Cardiocerebrovascular Disease, West Heping Road 215, Shijiazhuang, 050000, Hebei, People's Republic of China.
- Neurological Laboratory of Hebei Province, Shijiazhuang, 050000, Hebei, People's Republic of China.
| |
Collapse
|
20
|
Heitzer M, Kaiser S, Kanagaratnam M, Zendedel A, Hartmann P, Beyer C, Johann S. Administration of 17β-Estradiol Improves Motoneuron Survival and Down-regulates Inflammasome Activation in Male SOD1(G93A) ALS Mice. Mol Neurobiol 2016; 54:8429-8443. [PMID: 27957680 DOI: 10.1007/s12035-016-0322-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease manifested by the progressive loss of upper and lower motoneurons. The pathomechanism of ALS is complex and not yet fully understood. Neuroinflammation is believed to significantly contribute to disease progression. Inflammasome activation was recently shown in the spinal cord of human sporadic ALS patients and in the SOD1(G93A) mouse model for ALS. In the present study, we investigated the neuroprotective and anti-inflammatory effects of 17β-estradiol (E2) treatment in pre-symptomatic and symptomatic male SOD1(G93A) mice. Symptomatic mice with E2 substitution exhibited improved motor performance correlating with an increased survival of motoneurons in the lumbar spinal cord. Expression of NLRP3 inflammasome proteins and levels of activated caspase 1 and mature interleukin 1 beta were significantly reduced in SOD1(G93A) mice supplemented with E2.
Collapse
Affiliation(s)
- Marius Heitzer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Sarah Kaiser
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Mithila Kanagaratnam
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,Department of Anatomical Sciences, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Philipp Hartmann
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,JARA-BRAIN, 52074, Aachen, Germany
| | - Sonja Johann
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
21
|
Corsini S, Tortora M, Nistri A. Nicotinic receptor activation contrasts pathophysiological bursting and neurodegeneration evoked by glutamate uptake block on rat hypoglossal motoneurons. J Physiol 2016; 594:6777-6798. [PMID: 27374167 PMCID: PMC5108918 DOI: 10.1113/jp272591] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/21/2016] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Impaired uptake of glutamate builds up the extracellular level of this excitatory transmitter to trigger rhythmic neuronal bursting and delayed cell death in the brainstem motor nucleus hypoglossus. This process is the expression of the excitotoxicity that underlies motoneuron degeneration in diseases such as amyotrophic lateral sclerosis affecting bulbar motoneurons. In a model of motoneuron excitotoxicity produced by pharmacological block of glutamate uptake in vitro, rhythmic bursting is suppressed by activation of neuronal nicotinic receptors with their conventional agonist nicotine. Emergence of bursting is facilitated by nicotinic receptor antagonists. Following excitotoxicity, nicotinic receptor activity decreases mitochondrial energy dysfunction, endoplasmic reticulum stress and production of toxic radicals. Globally, these phenomena synergize to provide motoneuron protection. Nicotinic receptors may represent a novel target to contrast pathological overactivity of brainstem motoneurons and therefore to prevent their metabolic distress and death. ABSTRACT Excitotoxicity is thought to be one of the early processes in the onset of amyotrophic lateral sclerosis (ALS) because high levels of glutamate have been detected in the cerebrospinal fluid of such patients due to dysfunctional uptake of this transmitter that gradually damages brainstem and spinal motoneurons. To explore potential mechanisms to arrest ALS onset, we used an established in vitro model of rat brainstem slice preparation in which excitotoxicity is induced by the glutamate uptake blocker dl-threo-β-benzyloxyaspartate (TBOA). Because certain brain neurons may be neuroprotected via activation of nicotinic acetylcholine receptors (nAChRs) by nicotine, we investigated if nicotine could arrest excitotoxic damage to highly ALS-vulnerable hypoglossal motoneurons (HMs). On 50% of patch-clamped HMs, TBOA induced intense network bursts that were inhibited by 1-10 μm nicotine, whereas nAChR antagonists facilitated burst emergence in non-burster cells. Furthermore, nicotine inhibited excitatory transmission and enhanced synaptic inhibition. Strong neuroprotection by nicotine prevented the HM loss observed after 4 h of TBOA exposure. This neuroprotective action was due to suppression of downstream effectors of neurotoxicity such as increased intracellular levels of reactive oxygen species, impaired energy metabolism and upregulated genes involved in endoplasmic reticulum (ER) stress. In addition, HMs surviving TBOA toxicity often expressed UDP-glucose glycoprotein glucosyltransferase, a key element in repair of misfolded proteins: this phenomenon was absent after nicotine application, indicative of ER stress prevention. Our results suggest nAChRs to be potential targets for inhibiting excitotoxic damage of motoneurons at an early stage of the neurodegenerative process.
Collapse
Affiliation(s)
- Silvia Corsini
- Department of NeuroscienceInternational School for Advanced Studies (SISSA)TriesteItaly
| | - Maria Tortora
- Department of NeuroscienceInternational School for Advanced Studies (SISSA)TriesteItaly
| | - Andrea Nistri
- Department of NeuroscienceInternational School for Advanced Studies (SISSA)TriesteItaly
| |
Collapse
|
22
|
Fujimaki N, Nishiya K, Miura T, Nakabayashi T. Acquisition of pro-oxidant activity of fALS-linked SOD1 mutants as revealed using circular dichroism and UV-resonance Raman spectroscopy. Chem Phys 2016. [DOI: 10.1016/j.chemphys.2016.08.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
Deane CAS, Brown IR. Induction of heat shock proteins in differentiated human neuronal cells following co-application of celastrol and arimoclomol. Cell Stress Chaperones 2016; 21:837-48. [PMID: 27273088 PMCID: PMC5003800 DOI: 10.1007/s12192-016-0708-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 01/19/2023] Open
Abstract
Few effective therapies exist for the treatment of neurodegenerative diseases that have been characterized as protein misfolding disorders. Upregulation of heat shock proteins (Hsps) mitigates against the accumulation of misfolded, aggregation-prone proteins and synaptic dysfunction, which is recognized as an early event in neurodegenerative diseases. Enhanced induction of a set of Hsps in differentiated human SH-SY5Y neuronal cells was observed following co-application of celastrol and arimoclomol, compared to their individual application. The dosages employed did not affect cell viability or neuronal process morphology. The induced Hsps included the little studied HSPA6 (Hsp70B'), a potentially neuroprotective protein that is present in the human genome but not in rat and mouse and hence is missing in current animal models of neurodegenerative disease. Enhanced induction of HSPA1A (Hsp70-1), DNAJB1 (Hsp40), HO-1 (Hsp32), and HSPB1 (Hsp27) was also observed. Celastrol activates heat shock transcription factor 1 (HSF1), the master regulator of Hsp gene transcription, and also exhibits potent anti-inflammatory and anti-oxidant activities. Arimoclomol is a co-activator that prolongs the binding of activated HSF1 to heat shock elements (HSEs) in the promoter regions of inducible Hsp genes. Elevated Hsp levels peaked at 10 to 12 h for HSPA6, HSPA1A, DNAJB1, and HO-1 and at 24 h for HSPB1. Co-application of celastrol and arimoclomol induced higher Hsp levels compared to heat shock paired with arimoclomol. The co-application strategy of celastrol and arimoclomol targets multiple neurodegenerative disease-associated pathologies including protein misfolding and protein aggregation, inflammatory and oxidative stress, and synaptic dysfunction.
Collapse
Affiliation(s)
- Catherine A S Deane
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, M1C 1A4, Canada
| | - Ian R Brown
- Centre for the Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, M1C 1A4, Canada.
| |
Collapse
|
24
|
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) or motor neuron disease is a rapidly progressive neurodegenerative disorder. The primary involvement is of motor neurons in the brain, spinal cord and peripherally. There is secondary weakness of muscles and primary involvement of other brain regions, especially involving cognition. SOURCES OF DATA Peer-reviewed journal articles and reviews. PubMed.gov AREAS OF AGREEMENT The pathogenesis of ALS remains largely unknown. There are a wide range of potential mechanisms related to neurodegeneration. An increasing number of genetic factors are recognized. AREAS OF CONTROVERSY There remains controversy, or lack of knowledge, in explaining how cellular events manifest as the complex human disease. There is controversy as to how well cellular and animal models of disease relate to the human disease. GROWING POINTS Large-scale international collaborative genetic epidemiological studies are replacing local studies. Therapies related to pathogenesis remain elusive, with the greatest advances to date relating to provision of care (including multidisciplinary management) and supportive care (nutrition and respiratory support). AREAS TIMELY FOR DEVELOPING RESEARCH The identification of C9orf72 hexanucleotide repeats as the most frequent genetic background to ALS, and the association with frontotemporal dementia, gives the potential of a genetic background against which to study other risk factors, triggers and pathogenic mechanisms, and to develop potential therapies.
Collapse
Affiliation(s)
- Sarah Morgan
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Richard W Orrell
- Department of Clinical Neuroscience, UCL Institute of Neurology, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
25
|
Qosa H, Mohamed LA, Alqahtani S, Abuasal BS, Hill RA, Kaddoumi A. Transporters as Drug Targets in Neurological Diseases. Clin Pharmacol Ther 2016; 100:441-453. [PMID: 27447939 DOI: 10.1002/cpt.435] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/13/2016] [Accepted: 07/15/2016] [Indexed: 12/13/2022]
Abstract
Membrane transport proteins have central physiological function in maintaining cerebral homeostasis. These transporters are expressed in almost all cerebral cells in which they regulate the movement of a wide range of solutes, including endogenous substrates, xenobiotic, and therapeutic drugs. Altered activity/expression of central nervous system (CNS) transporters has been implicated in the onset and progression of multiple neurological diseases. Neurological diseases are heterogeneous diseases that involve complex pathological alterations with only a few treatment options; therefore, there is a great need for the development of novel therapeutic treatments. To that end, transporters have emerged recently to be promising therapeutic targets to halt or slow the course of neurological diseases. The objective of this review is to discuss implications of transporters in neurological diseases and summarize available evidence for targeting transporters as decent therapeutic approach in the treatment of neurological diseases.
Collapse
Affiliation(s)
- H Qosa
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - L A Mohamed
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - S Alqahtani
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - B S Abuasal
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - R A Hill
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - A Kaddoumi
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA.
| |
Collapse
|
26
|
Staats KA, Humblet-Baron S, Bento-Abreu A, Scheveneels W, Nikolaou A, Deckers K, Lemmens R, Goris A, Van Ginderachter JA, Van Damme P, Hisatsune C, Mikoshiba K, Liston A, Robberecht W, Van Den Bosch L. Genetic ablation of IP3 receptor 2 increases cytokines and decreases survival of SOD1G93A mice. Hum Mol Genet 2016; 25:3491-3499. [PMID: 27378687 PMCID: PMC5179944 DOI: 10.1093/hmg/ddw190] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating progressive neurodegenerative disease characterized by the selective death of motor neurons. Disease pathophysiology is complex and not yet fully understood. Higher gene expression of the inositol 1,4,5-trisphosphate receptor 2 gene (ITPR2), encoding the IP3 receptor 2 (IP3R2), was detected in sporadic ALS patients. Here, we demonstrate that IP3R2 gene expression was also increased in spinal cords of ALS mice. Moreover, an increase of IP3R2 expression was observed in other models of chronic and acute neurodegeneration. Upregulation of IP3R2 gene expression could be induced by lipopolysaccharide (LPS) in murine astrocytes, murine macrophages and human fibroblasts indicating that it may be a compensatory response to inflammation. Preventing this response by genetic deletion of ITPR2 from SOD1G93A mice had a dose-dependent effect on disease duration, resulting in a significantly shorter lifespan of these mice. In addition, the absence of IP3R2 led to increased innate immunity, which may contribute to the decreased survival of the SOD1G93A mice. Besides systemic inflammation, IP3R2 knockout mice also had increased IFNγ, IL-6 and IL1α expression. Altogether, our data indicate that IP3R2 protects against the negative effects of inflammation, suggesting that the increase in IP3R2 expression in ALS patients is a protective response.
Collapse
Affiliation(s)
- Kim A Staats
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND).,VIB, Vesalius Research Center, Laboratory of Neurobiology
| | | | - Andre Bento-Abreu
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND).,VIB, Vesalius Research Center, Laboratory of Neurobiology
| | - Wendy Scheveneels
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND).,VIB, Vesalius Research Center, Laboratory of Neurobiology
| | - Alexandros Nikolaou
- Molecular and Biochemical Pharmacology Laboratory, Vrije Universiteit Brussel.,Myeloid Cell Immunology Laboratory, VIB, Inflammation Research Center.,Cellular and Molecular Immunology Unit, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kato Deckers
- Center for Molecular and Vascular Biology, University of Leuven
| | - Robin Lemmens
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND).,VIB, Vesalius Research Center, Laboratory of Neurobiology.,University Hospitals Leuven, Department of Neurology
| | - An Goris
- KU Leuven - University of Leuven, Department of Neurosciences, Laboratory for Neuroimmunology, Leuven, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory, VIB, Inflammation Research Center.,Cellular and Molecular Immunology Unit, Vrije Universiteit Brussel, Brussels, Belgium
| | - Philip Van Damme
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND).,VIB, Vesalius Research Center, Laboratory of Neurobiology.,University Hospitals Leuven, Department of Neurology
| | - Chihiro Hisatsune
- Laboratory for Developmental Neurobiology, Brain Science Institute, RIKEN, Wako-shi, Saitama, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, Brain Science Institute, RIKEN, Wako-shi, Saitama, Japan
| | - Adrian Liston
- VIB and Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Wim Robberecht
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND).,VIB, Vesalius Research Center, Laboratory of Neurobiology.,University Hospitals Leuven, Department of Neurology
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND) .,VIB, Vesalius Research Center, Laboratory of Neurobiology
| |
Collapse
|
27
|
Qiu H, Li JH, Yin SB, Ke JQ, Qiu CL, Zheng GQ. Dihuang Yinzi, a Classical Chinese Herbal Prescription, for Amyotrophic Lateral Sclerosis: A 12-Year Follow-up Case Report. Medicine (Baltimore) 2016; 95:e3324. [PMID: 27057909 PMCID: PMC4998825 DOI: 10.1097/md.0000000000003324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating progressive neurodegenerative disease with no effective treatment and death within 2 to 5 years after symptom onset. Here, we reported a case of ALS patient using modified Dihuang Yinzi (DHYZ), a classical traditional Chinese medicine (TCM) prescription, who has survived 12 years with significant improvement in bulbar paralysis.A 41-year-old Chinese Han nationality woman was admitted to the hospital with complaints of weakened bilateral grip, slurred speech, stumbling, and muscle twitching for 3 years. The electromyography showed neurogenic injury in bilateral upper limbs and tongue. She was diagnosed with ALS according to the revised El escorial criteria. The patient was orally administrated with Riluzole 100 mg daily for 10 months and then stopped. Subsequently, she resorted to TCM. Based on the TCM theory, the patient was diagnosed with Yinfei syndrome because of kidney deficiency. DHYZ was chosen because it has the function of replenishing kidney essence to treat Yinfei syndrome. Up to now, she has been using modified DHYZ continuously for 12 years. The patient survived with ALS and did not require permanent continuous ventilator. In addition, the symptoms of choking on liquids are improved, and the utility of 30 mL water swallow test was improved with grade 2. The symptoms of muscle fibrillations of limbs are also reduced. However, muscle strength worsened slowly. The repeated electromyography showed motor conduction amplitude reducing gradually and velocity not changing more when compared with the initial electromyography.Our findings suggested that DHYZ can be potentially used in ALS patients because of its multi-targeted neuroprotection and general safety, although ALS does not have a cure. In addition, we identified the area that is worthy of further study and DHYZ as a promising candidate for further clinical application and ALS trials. Rigorous randomized controlled trials are needed in the future.
Collapse
Affiliation(s)
- Hui Qiu
- From the Department of Internal Medicine of TCM, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou (HQ); Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou (J-hL, S-bY, J-qK, G-qZ); and Department of Neurology, Zhejiang Provincial Hospital of TCM, the First Affiliated Hospital of Zhejiang Chinese Medical University (C-lQ), Hangzhou, China
| | | | | | | | | | | |
Collapse
|
28
|
Browne EC, Abbott BM. Recent progress towards an effective treatment of amyotrophic lateral sclerosis using the SOD1 mouse model in a preclinical setting. Eur J Med Chem 2016; 121:918-925. [PMID: 27012524 DOI: 10.1016/j.ejmech.2016.02.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 02/18/2016] [Accepted: 02/18/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, fatal and incurable neurodegenerative disorder. Motor neurone degeneration can be caused by genetic mutation but the exact etiology of the disease, particularly for sporadic illness, still remains unclear. Therapeutics which target known pathogenic mechanisms involved in ALS, such as protein aggregation, oxidative stress, apoptosis, inflammation, endoplasmic reticulum stress and mitochondria dysfunction, are currently being pursued in order to provide neuroprotection which may be able to slow down, or perhaps even halt, disease progression. This present review focuses on the compounds which have been recently evaluated using the SOD1 mouse model, the most widely used preclinical model for ALS research.
Collapse
Affiliation(s)
- Elisse C Browne
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Belinda M Abbott
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
29
|
Herrando-Grabulosa M, Mulet R, Pujol A, Mas JM, Navarro X, Aloy P, Coma M, Casas C. Novel Neuroprotective Multicomponent Therapy for Amyotrophic Lateral Sclerosis Designed by Networked Systems. PLoS One 2016; 11:e0147626. [PMID: 26807587 PMCID: PMC4726541 DOI: 10.1371/journal.pone.0147626] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 01/05/2016] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic Lateral Sclerosis is a fatal, progressive neurodegenerative disease characterized by loss of motor neuron function for which there is no effective treatment. One of the main difficulties in developing new therapies lies on the multiple events that contribute to motor neuron death in amyotrophic lateral sclerosis. Several pathological mechanisms have been identified as underlying events of the disease process, including excitotoxicity, mitochondrial dysfunction, oxidative stress, altered axonal transport, proteasome dysfunction, synaptic deficits, glial cell contribution, and disrupted clearance of misfolded proteins. Our approach in this study was based on a holistic vision of these mechanisms and the use of computational tools to identify polypharmacology for targeting multiple etiopathogenic pathways. By using a repositioning analysis based on systems biology approach (TPMS technology), we identified and validated the neuroprotective potential of two new drug combinations: Aliretinoin and Pranlukast, and Aliretinoin and Mefloquine. In addition, we estimated their molecular mechanisms of action in silico and validated some of these results in a well-established in vitro model of amyotrophic lateral sclerosis based on cultured spinal cord slices. The results verified that Aliretinoin and Pranlukast, and Aliretinoin and Mefloquine promote neuroprotection of motor neurons and reduce microgliosis.
Collapse
Affiliation(s)
- Mireia Herrando-Grabulosa
- 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
| | - Roger Mulet
- Anaxomics Biotech SL, Barcelona, Catalonia, Spain
| | - Albert Pujol
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
| | | | - Xavier Navarro
- 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
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Mireia Coma
- Anaxomics Biotech SL, Barcelona, Catalonia, Spain
- * E-mail: (CC); (MC)
| | - 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
- * E-mail: (CC); (MC)
| |
Collapse
|
30
|
MicroRNA-125b regulates microglia activation and motor neuron death in ALS. Cell Death Differ 2016; 23:531-41. [PMID: 26794445 PMCID: PMC5072447 DOI: 10.1038/cdd.2015.153] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/24/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022] Open
Abstract
Understanding the means by which microglia self-regulate the neuroinflammatory response helps modulating their reaction during neurodegeneration. In amyotrophic lateral sclerosis (ALS), classical NF-κB pathway is related to persistent microglia activation and motor neuron injury; however, mechanisms of negative control of NF-κB activity remain unexplored. One of the major players in the termination of classical NF-κB pathway is the ubiquitin-editing enzyme A20, which has recognized anti-inflammatory functions. Lately, microRNAs are emerging as potent fine-tuners of neuroinflammation and reported to be regulated in ALS, for instance, by purinergic P2X7 receptor activation. In this work, we uncover an interplay between miR-125b and A20 protein in the modulation of classical NF-κB signaling in microglia. In particular, we establish the existence of a pathological circuit in which termination of A20 function by miR-125b strengthens and prolongs the noxious P2X7 receptor-dependent activation of NF-κB in microglia, with deleterious consequences on motor neurons. We prove that, by restoring A20 levels, miR-125b inhibition then sustains motor neuron survival. These results introduce miR-125b as a key mediator of microglia dynamics in ALS.
Collapse
|
31
|
The ASK1-specific inhibitors K811 and K812 prolong survival in a mouse model of amyotrophic lateral sclerosis. Hum Mol Genet 2015; 25:245-53. [DOI: 10.1093/hmg/ddv467] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023] Open
|
32
|
|
33
|
Wang X, Ma M, Teng J, Che X, Zhang W, Feng S, Zhou S, Zhang Y, Wu E, Ding X. Valproate Attenuates 25-kDa C-Terminal Fragment of TDP-43-Induced Neuronal Toxicity via Suppressing Endoplasmic Reticulum Stress and Activating Autophagy. Int J Biol Sci 2015; 11:752-761. [PMID: 26078717 PMCID: PMC4466456 DOI: 10.7150/ijbs.11880] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/17/2015] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disease. To date, there is no any effective pharmacological treatment for improving patients' symptoms and quality of life. Rapidly emerging evidence suggests that C-terminal fragments (CTFs) of TAR DNA-binding protein of 43 kDa (TDP-43), including TDP-35 and TDP-25, may play an important role in ALS pathogenesis. Valproate (VPA), a widely used antiepileptic drug, has neuroprotective effects on neurodegenerative disorders. As for ALS, preclinical studies also provide encouraging evidence for multiple beneficial effects in ALS mouse models. However, the potential molecular mechanisms have not been explored. Here, we show protective effects of VPA against TDP-43 CTFs-mediated neuronal toxicity and its underlying mechanisms in vitro. Remarkably, TDP-43 CTFs induced neuronal damage via endoplastic reticulum (ER) stress-mediated apoptosis. Furthermore, autophagic self-defense system was activated to reduce TDP-43 CTFs-induced neuronal death. Finally, VPA attenuated TDP-25-induced neuronal toxicity via suppressing ER stress-mediated apoptosis and enhancing autophagy. Taken together, these results demonstrate that VPA exerts neuroprotective effects against TDP-43 CTFs-induced neuronal damage. Thus, we provide new molecular evidence for VPA treatment in patients with ALS and other TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Xuejing Wang
- 1. Department of neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mingming Ma
- 2. Department of neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Junfang Teng
- 1. Department of neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiangqian Che
- 4. Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai 200025, China
| | - Wenwen Zhang
- 1. Department of neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shuman Feng
- 2. Department of neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Shuang Zhou
- 3. Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, 58105, USA
| | - Ying Zhang
- 3. Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, 58105, USA
| | - Erxi Wu
- 3. Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, 58105, USA
| | - Xuebing Ding
- 1. Department of neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
34
|
Tokuda E, Watanabe S, Okawa E, Ono SI. Regulation of Intracellular Copper by Induction of Endogenous Metallothioneins Improves the Disease Course in a Mouse Model of Amyotrophic Lateral Sclerosis. Neurotherapeutics 2015; 12:461-76. [PMID: 25761970 PMCID: PMC4404437 DOI: 10.1007/s13311-015-0346-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS), an incurable motor neuron disease. The pathogenesis of the disease is poorly understood, but intracellular copper dyshomeostasis has been implicated as a key process in the disease. We recently observed that metallothioneins (MTs) are an excellent target for the modification of copper dyshomeostasis in a mouse model of ALS (SOD1(G93A)). Here, we offer a therapeutic strategy designed to increase the level of endogenous MTs. The upregulation of endogenous MTs by dexamethasone, a synthetic glucocorticoid, significantly improved the disease course and rescued motor neurons in SOD1(G93A) mice, even if the induction was initiated when peak body weight had decreased by 10%. Neuroprotection was associated with the normalization of copper dyshomeostasis, as well as with decreased levels of SOD1(G93A) aggregates. Importantly, these benefits were clearly mediated in a MT-dependent manner, as dexamethasone did not provide any protection when endogenous MTs were abolished from SOD1(G93A) mice. In conclusion, the upregulation of endogenous MTs represents a promising strategy for the treatment of ALS linked to mutant SOD1.
Collapse
Affiliation(s)
- Eiichi Tokuda
- />Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555 Japan
- />Department of Medical Biosciences, Clinical Chemistry, Umeå University, Building 6M, 2nd Floor, Umeå, 901-85 Sweden
| | - Shunsuke Watanabe
- />Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555 Japan
| | - Eriko Okawa
- />Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555 Japan
| | - Shin-ichi Ono
- />Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555 Japan
- />Division of Neurology, Akiru Municipal Medical Center, 78-1 Hikida, Akiru, Tokyo, 197-0834 Japan
| |
Collapse
|
35
|
Sanli A, Sengun IS, Tertemiz KC, Alpaydin AO, Karacam V, Sanli BA, Oz D, Ozalevli S, Ozdemir N. Importance of diaphragm thickness in amyotrophic lateral sclerosis patients with diaphragm pacing system implantation. Surg Endosc 2015; 30:154-8. [PMID: 25805242 DOI: 10.1007/s00464-015-4175-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/14/2015] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Severe respiratory failure develops as a result of the involvement of the respiratory muscles in patients with amyotrophic lateral sclerosis (ALS). Implantation of diaphragm pacing system (DPS) has been carried out on ALS patients since 2005 to avoid these situations, but the importance of diaphragm thickness has not yet been established clearly. MATERIAL AND METHOD We retrospectively evaluated 34 ALS patients who had previously implanted DPS to detect the importance of diaphragm thickness. We investigated the effect of diaphragm thickness, which was measured by preoperative thorax computerized tomography on preoperative respiratory function tests (RFT), arterial blood gas (ABG) analysis, postoperative 3- and 6-month oxygen saturations and mortality. RESULTS The right diaphragm thickness was calculated as 4.60 (2.95-6.00) mm, while the left diaphragm thickness was 4.10 (2.77-6.00) mm. Six patients died during the follow-up period. We did not detect a significant relationship between ABG parameters, RFT and diaphragm thickness. However, according to our observations, the diaphragm thickness was significantly related to mortality. The right diaphragm was significantly thinner in cases that required preoperative respiratory support and had percutaneous endoscopic gastrostomy. When the cut-off values for the diaphragm thickness were accepted as 3.50 mm, significantly higher mortality among patients below this was observed. CONCLUSION Diaphragm thickness is an important criterion in cases for which DPS implantation is planned. We consider that avoidance of DPS implantation is more suitable for cases with a diaphragm thickness below 3.50 mm because of mortality.
Collapse
Affiliation(s)
- Aydın Sanli
- Thoracic Surgery Department, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Ihsan Sukru Sengun
- Neurology Department, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Kemal Can Tertemiz
- Chest Diseases Department, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey.
| | - Aylin Ozgen Alpaydin
- Chest Diseases Department, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Volkan Karacam
- Thoracic Surgery Department, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Bahar Agaoglu Sanli
- Thoracic Surgery Department, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Didem Oz
- Neurology Department, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Sevgi Ozalevli
- Physiotherapy and Rehabilitation Department, Dokuz Eylul University, Izmir, Turkey
| | - Nezih Ozdemir
- Thoracic Surgery Department, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
36
|
Cirulli ET, Lasseigne BN, Petrovski S, Sapp PC, Dion PA, Leblond CS, Couthouis J, Lu YF, Wang Q, Krueger BJ, Ren Z, Keebler J, Han Y, Levy SE, Boone BE, Wimbish JR, Waite LL, Jones AL, Carulli JP, Day-Williams AG, Staropoli JF, Xin WW, Chesi A, Raphael AR, McKenna-Yasek D, Cady J, Vianney de Jong JMB, Kenna KP, Smith BN, Topp S, Miller J, Gkazi A, Al-Chalabi A, van den Berg LH, Veldink J, Silani V, Ticozzi N, Shaw CE, Baloh RH, Appel S, Simpson E, Lagier-Tourenne C, Pulst SM, Gibson S, Trojanowski JQ, Elman L, McCluskey L, Grossman M, Shneider NA, Chung WK, Ravits JM, Glass JD, Sims KB, Van Deerlin VM, Maniatis T, Hayes SD, Ordureau A, Swarup S, Landers J, Baas F, Allen AS, Bedlack RS, Harper JW, Gitler AD, Rouleau GA, Brown R, Harms MB, Cooper GM, Harris T, Myers RM, Goldstein DB. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science 2015; 347:1436-41. [PMID: 25700176 DOI: 10.1126/science.aaa3650] [Citation(s) in RCA: 752] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurological disease with no effective treatment. We report the results of a moderate-scale sequencing study aimed at increasing the number of genes known to contribute to predisposition for ALS. We performed whole-exome sequencing of 2869 ALS patients and 6405 controls. Several known ALS genes were found to be associated, and TBK1 (the gene encoding TANK-binding kinase 1) was identified as an ALS gene. TBK1 is known to bind to and phosphorylate a number of proteins involved in innate immunity and autophagy, including optineurin (OPTN) and p62 (SQSTM1/sequestosome), both of which have also been implicated in ALS. These observations reveal a key role of the autophagic pathway in ALS and suggest specific targets for therapeutic intervention.
Collapse
Affiliation(s)
- Elizabeth T Cirulli
- Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC 27708, USA
| | | | - Slavé Petrovski
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | - Peter C Sapp
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Patrick A Dion
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Claire S Leblond
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yi-Fan Lu
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | - Quanli Wang
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | - Brian J Krueger
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | - Zhong Ren
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | | | - Yujun Han
- Duke University School of Medicine, Durham, NC 27708, USA
| | - Shawn E Levy
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Braden E Boone
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Jack R Wimbish
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Lindsay L Waite
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Angela L Jones
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | | | | | - Winnie W Xin
- Neurogenetics DNA Diagnostic Laboratory, Center for Human Genetics Research, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alessandra Chesi
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alya R Raphael
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Diane McKenna-Yasek
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Janet Cady
- Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - J M B Vianney de Jong
- Department of Genome Analysis, Academic Medical Center, Meibergdreef 9, 1105AZ Amsterdam, Netherlands
| | - Kevin P Kenna
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Republic of Ireland
| | - Bradley N Smith
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London SE5 8AF, UK
| | - Simon Topp
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London SE5 8AF, UK
| | - Jack Miller
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London SE5 8AF, UK
| | - Athina Gkazi
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London SE5 8AF, UK
| | | | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London SE5 8AF, UK
| | - Leonard H van den Berg
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Centre Utrecht, 3508 GA Utrecht, Netherlands
| | - Jan Veldink
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Centre Utrecht, 3508 GA Utrecht, Netherlands
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan 20149, Italy, and Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan 20122, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan 20149, Italy, and Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan 20122, Italy
| | - Christopher E Shaw
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London SE5 8AF, UK
| | | | - Stanley Appel
- Houston Methodist Hospital, Houston, TX 77030, USA, and Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Ericka Simpson
- Houston Methodist Hospital, Houston, TX 77030, USA, and Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Clotilde Lagier-Tourenne
- Ludwig Institute for Cancer Research and Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Summer Gibson
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren Elman
- Department of Neurology, Penn ALS Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leo McCluskey
- Department of Neurology, Penn ALS Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Murray Grossman
- Department of Neurology, Penn Frontotemporal Degeneration Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Neil A Shneider
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
| | - Wendy K Chung
- Department of Pediatrics and Medicine, Columbia University, New York, NY 10032, USA
| | - John M Ravits
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jonathan D Glass
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
| | - Katherine B Sims
- Neurogenetics DNA Diagnostic Laboratory, Center for Human Genetics Research, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tom Maniatis
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, NY 10027, USA
| | - Sebastian D Hayes
- Biogen Idec, Cambridge, MA 02142, USA. Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Ordureau
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sharan Swarup
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - John Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Frank Baas
- Department of Genome Analysis, Academic Medical Center, Meibergdreef 9, 1105AZ Amsterdam, Netherlands
| | - Andrew S Allen
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27708, USA
| | | | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Guy A Rouleau
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Robert Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Matthew B Harms
- Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gregory M Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| |
Collapse
|
37
|
Cozzolino M, Rossi S, Mirra A, Carrì MT. Mitochondrial dynamism and the pathogenesis of Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2015; 9:31. [PMID: 25713513 PMCID: PMC4322717 DOI: 10.3389/fncel.2015.00031] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/19/2015] [Indexed: 12/11/2022] Open
Abstract
Research on mitochondria in the last years has been characterized by the fundamental finding that the morphology of mitochondria is deeply connected to the regulation of a vast number of different processes, including oxidative phosphorylation and ATP production, calcium buffering, and apoptosis. This has immediately focused the attention of the neuroscience community to the possible involvement of mitochondrial dynamism, the process underlying morphological features of mitochondria, in neurodegeneration, where mitochondrial dysfunction is believed to represent an important contributing event, or even a primary causative factor. Amyotrophic Lateral Sclerosis (ALS), a disease of motor neurons and their neighboring cells, has long been considered as a neurodegenerative disease with an important mitochondrial issue. Yet, whether mitochondria have a causative, primary role in the pathogenic process has always been debated, and the specific defects which account for this role are elusive. Here we discuss recent genetic advances suggesting that defective mitochondrial dynamism is primarily involved in the pathogenic mechanisms of ALS, and that foster the longstanding concept that disruption of mitochondrial function is a vulnerable factor for motor neurons.
Collapse
Affiliation(s)
| | - Simona Rossi
- Institute of Translational Pharmacology, CNR Rome, Italy ; Department of Biology, Università di Roma Tor Vergata Rome, Italy
| | - Alessia Mirra
- Department of Biology, Università di Roma Tor Vergata Rome, Italy ; Fondazione Santa Lucia IRCCS Rome, Italy
| | - Maria Teresa Carrì
- Department of Biology, Università di Roma Tor Vergata Rome, Italy ; Fondazione Santa Lucia IRCCS Rome, Italy
| |
Collapse
|
38
|
Gershoni-Emek N, Chein M, Gluska S, Perlson E. Amyotrophic lateral sclerosis as a spatiotemporal mislocalization disease: location, location, location. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 315:23-71. [PMID: 25708461 DOI: 10.1016/bs.ircmb.2014.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Spatiotemporal localization of signals is a fundamental feature impacting cell survival and proper function. The cell needs to respond in an accurate manner in both space and time to both intra- and intercellular environment cues. The regulation of this comprehensive process involves the cytoskeleton and the trafficking machinery, as well as local protein synthesis and ligand-receptor mechanisms. Alterations in such mechanisms can lead to cell dysfunction and disease. Motor neurons that can extend over tens of centimeters are a classic example for the importance of such events. Changes in spatiotemporal localization mechanisms are thought to play a role in motor neuron degeneration that occurs in amyotrophic lateral sclerosis (ALS). In this review we will discuss these mechanisms and argue that possible misregulated factors can lead to motor neuron degeneration in ALS.
Collapse
Affiliation(s)
- Noga Gershoni-Emek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Michael Chein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shani Gluska
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
39
|
Dhama K, Kesavan M, Karthik K, . A, Tiwari R, Sunkara LT, Singh R. Neuroimmunomodulation Countering Various Diseases, Disorders, Infections, Stress and Aging. INT J PHARMACOL 2015. [DOI: 10.3923/ijp.2015.76.94] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
40
|
Apolloni S, Fabbrizio P, Parisi C, Amadio S, Volonté C. Clemastine Confers Neuroprotection and Induces an Anti-Inflammatory Phenotype in SOD1(G93A) Mouse Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2014; 53:518-531. [PMID: 25482048 DOI: 10.1007/s12035-014-9019-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/18/2014] [Indexed: 12/13/2022]
Abstract
Mutations in the Cu(2+)/Zn(2+) superoxide dismutase 1 (SOD1) gene underlie 14-23 % of familial and 1-7 % of sporadic cases of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disease characterized by a specific loss of motor neurons in the brain and spinal cord. Neuroinflammation and oxidative stress are emerging as key players in the pathogenesis of ALS, thus justifying the interest in glial cells and particularly microglia, in addition to motor neurons, as novel therapeutic approaches against ALS. Recently, histamine was proven to participate in the pathogenesis of neuroinflammatory and neurodegenerative diseases, and particularly, microglia was shown to be sensitive to the histamine challenge mainly through histamine H1 receptors. Clemastine is a first-generation and CNS-penetrant H1 receptor antagonist considered as a safe antihistamine compound that was shown to possess immune suppressive properties. In order to investigate if clemastine might find promising application in the treatment of ALS, in this work, we tested its action in the SOD1(G93A) mouse model which is extensively used in ALS preclinical studies. We demonstrated that chronic clemastine administration in SOD1(G93A) mice reduces microgliosis, modulates microglia-related inflammatory genes, and enhances motor neuron survival. Moreover, in vitro, clemastine is able to modify several activation parameters of SOD1(G93A) microglia, and particularly CD68 and arginase-1 expression, as well as phospho-ERK1/2 and NADPH oxidase 2 levels. Being clemastine a drug already employed in clinical practice, our results strongly encourage its further exploitation as a candidate for preclinical trials and a new modulator of neuroinflammation in ALS.
Collapse
Affiliation(s)
- Savina Apolloni
- Cellular Biology and Neurobiology Institute, CNR, Via del Fosso di Fiorano, 65, 00143, Rome, Italy.,Division Experimental Neuroscience, Santa Lucia Foundation, IRCCS,, Via del Fosso di Fiorano, 65, Rome, 00143, Italy
| | - Paola Fabbrizio
- Division Experimental Neuroscience, Santa Lucia Foundation, IRCCS,, Via del Fosso di Fiorano, 65, Rome, 00143, Italy
| | - Chiara Parisi
- Cellular Biology and Neurobiology Institute, CNR, Via del Fosso di Fiorano, 65, 00143, Rome, Italy
| | - Susanna Amadio
- Division Experimental Neuroscience, Santa Lucia Foundation, IRCCS,, Via del Fosso di Fiorano, 65, Rome, 00143, Italy
| | - Cinzia Volonté
- Cellular Biology and Neurobiology Institute, CNR, Via del Fosso di Fiorano, 65, 00143, Rome, Italy. .,Division Experimental Neuroscience, Santa Lucia Foundation, IRCCS,, Via del Fosso di Fiorano, 65, Rome, 00143, Italy.
| |
Collapse
|
41
|
Faravelli I, Frattini E, Ramirez A, Stuppia G, Nizzardo M, Corti S. iPSC-Based Models to Unravel Key Pathogenetic Processes Underlying Motor Neuron Disease Development. J Clin Med 2014; 3:1124-45. [PMID: 26237595 PMCID: PMC4470174 DOI: 10.3390/jcm3041124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 09/19/2014] [Accepted: 09/22/2014] [Indexed: 12/12/2022] Open
Abstract
Motor neuron diseases (MNDs) are neuromuscular disorders affecting rather exclusively upper motor neurons (UMNs) and/or lower motor neurons (LMNs). The clinical phenotype is characterized by muscular weakness and atrophy leading to paralysis and almost invariably death due to respiratory failure. Adult MNDs include sporadic and familial amyotrophic lateral sclerosis (sALS-fALS), while the most common infantile MND is represented by spinal muscular atrophy (SMA). No effective treatment is ccurrently available for MNDs, as for the vast majority of neurodegenerative disorders, and cures are limited to supportive care and symptom relief. The lack of a deep understanding of MND pathogenesis accounts for the difficulties in finding a cure, together with the scarcity of reliable in vitro models. Recent progresses in stem cell field, in particular in the generation of induced Pluripotent Stem Cells (iPSCs) has made possible for the first time obtaining substantial amounts of human cells to recapitulate in vitro some of the key pathogenetic processes underlying MNDs. In the present review, recently published studies involving the use of iPSCs to unravel aspects of ALS and SMA pathogenesis are discussed with an overview of their implications in the process of finding a cure for these still orphan disorders.
Collapse
Affiliation(s)
- Irene Faravelli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Emanuele Frattini
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Agnese Ramirez
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Giulia Stuppia
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Monica Nizzardo
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
42
|
Affiliation(s)
- Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Brain Science Institute, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Don W Cleveland
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|