101
|
Wang J, Rousseau J, Kim E, Ehresmann S, Cheng YT, Duraine L, Zuo Z, Park YJ, Li-Kroeger D, Bi W, Wong LJ, Rosenfeld J, Gleeson J, Faqeih E, Alkuraya FS, Wierenga KJ, Chen J, Afenjar A, Nava C, Doummar D, Keren B, Juusola J, Grompe M, Bellen HJ, Campeau PM. Loss of Oxidation Resistance 1, OXR1, Is Associated with an Autosomal-Recessive Neurological Disease with Cerebellar Atrophy and Lysosomal Dysfunction. Am J Hum Genet 2019; 105:1237-1253. [PMID: 31785787 PMCID: PMC6904826 DOI: 10.1016/j.ajhg.2019.11.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 11/01/2019] [Indexed: 12/28/2022] Open
Abstract
We report an early-onset autosomal-recessive neurological disease with cerebellar atrophy and lysosomal dysfunction. We identified bi-allelic loss-of-function (LoF) variants in Oxidative Resistance 1 (OXR1) in five individuals from three families; these individuals presented with a history of severe global developmental delay, current intellectual disability, language delay, cerebellar atrophy, and seizures. While OXR1 is known to play a role in oxidative stress resistance, its molecular functions are not well established. OXR1 contains three conserved domains: LysM, GRAM, and TLDc. The gene encodes at least six transcripts, including some that only consist of the C-terminal TLDc domain. We utilized Drosophila to assess the phenotypes associated with loss of mustard (mtd), the fly homolog of OXR1. Strong LoF mutants exhibit late pupal lethality or pupal eclosion defects. Interestingly, although mtd encodes 26 transcripts, severe LoF and null mutations can be rescued by a single short human OXR1 cDNA that only contains the TLDc domain. Similar rescue is observed with the TLDc domain of NCOA7, another human homolog of mtd. Loss of mtd in neurons leads to massive cell loss, early death, and an accumulation of aberrant lysosomal structures, similar to what we observe in fibroblasts of affected individuals. Our data indicate that mtd and OXR1 are required for proper lysosomal function; this is consistent with observations that NCOA7 is required for lysosomal acidification.
Collapse
Affiliation(s)
- Julia Wang
- Program in Developmental Biology, Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Justine Rousseau
- Centre Hospitalier Universitaire Saint-Justine Research Center, CHU Sainte-Justine, Montreal, QC H3T 1J4, Canada
| | - Emily Kim
- Biochemistry and Cell Biology, Rice University, Houston, TX 77005, USA
| | - Sophie Ehresmann
- Centre Hospitalier Universitaire Saint-Justine Research Center, CHU Sainte-Justine, Montreal, QC H3T 1J4, Canada
| | - Yi-Ting Cheng
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lita Duraine
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ye-Jin Park
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - David Li-Kroeger
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weimin Bi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lee-Jun Wong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jill Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joseph Gleeson
- Rady Institute of Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Eissa Faqeih
- Section of Medical Genetics, Children's Hospital, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, 11525, Saudi Arabia
| | - Klaas J Wierenga
- Department of Pediatrics, Oklahoma University Health Sciences Center (OUHSC), Oklahoma City, OK 26901, USA; Department of Clinical Genomics, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Jiani Chen
- Department of Pediatrics, Oklahoma University Health Sciences Center (OUHSC), Oklahoma City, OK 26901, USA; Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandra Afenjar
- Assistance Publique des Hôpitaux de Paris, Unité de Génétique Clinique, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire Paris, 75012, France; Département de Génétique et Embryologie Médicale, CRMR des Malformations et Maladies Congénitales du Cervelet, GRC ConCer-LD, Sorbonne Universités, Hôpital Trousseau, Paris, 75012 France
| | - Caroline Nava
- Assistance Publique des Hôpitaux de Paris, Unité de Génétique Clinique, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire Paris, 75012, France
| | - Diane Doummar
- Assistance Publique des Hôpitaux de Paris, Service de Neuropédiatrie, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire Paris, 75012 France
| | - Boris Keren
- Assistance Publique des Hôpitaux de Paris, Unité de Génétique Clinique, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire Paris, 75012, France
| | | | - Markus Grompe
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon 97201, USA; Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97201, USA
| | - Hugo J Bellen
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute and Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Philippe M Campeau
- Centre Hospitalier Universitaire Saint-Justine Research Center, CHU Sainte-Justine, Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
102
|
Tam OH, Rozhkov NV, Shaw R, Kim D, Hubbard I, Fennessey S, Propp N, Fagegaltier D, Harris BT, Ostrow LW, Phatnani H, Ravits J, Dubnau J, Gale Hammell M. Postmortem Cortex Samples Identify Distinct Molecular Subtypes of ALS: Retrotransposon Activation, Oxidative Stress, and Activated Glia. Cell Rep 2019; 29:1164-1177.e5. [PMID: 31665631 PMCID: PMC6866666 DOI: 10.1016/j.celrep.2019.09.066] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/03/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. While several pathogenic mutations have been identified, the vast majority of ALS cases have no family history of disease. Thus, for most ALS cases, the disease may be a product of multiple pathways contributing to varying degrees in each patient. Using machine learning algorithms, we stratify the transcriptomes of 148 ALS postmortem cortex samples into three distinct molecular subtypes. The largest cluster, identified in 61% of patient samples, displays hallmarks of oxidative and proteotoxic stress. Another 19% of the samples shows predominant signatures of glial activation. Finally, a third group (20%) exhibits high levels of retrotransposon expression and signatures of TARDBP/TDP-43 dysfunction. We further demonstrate that TDP-43 (1) directly binds a subset of retrotransposon transcripts and contributes to their silencing in vitro, and (2) pathological TDP-43 aggregation correlates with retrotransposon de-silencing in vivo.
Collapse
Affiliation(s)
- Oliver H Tam
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Regina Shaw
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Duyang Kim
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA
| | - Isabel Hubbard
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA
| | - Samantha Fennessey
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA
| | - Nadia Propp
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA
| | - Delphine Fagegaltier
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA
| | - Brent T Harris
- The NYGC ALS Consortium; Department of Neurology, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Lyle W Ostrow
- The NYGC ALS Consortium; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hemali Phatnani
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA; The NYGC ALS Consortium
| | - John Ravits
- The NYGC ALS Consortium; Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Josh Dubnau
- The NYGC ALS Consortium; Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Molly Gale Hammell
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; The NYGC ALS Consortium.
| |
Collapse
|
103
|
Franich NR, Basso M, André EA, Ochaba J, Kumar A, Thein S, Fote G, Kachemov M, Lau AL, Yeung SY, Osmand A, Zeitlin SO, Ratan RR, Thompson LM, Steffan JS. Striatal Mutant Huntingtin Protein Levels Decline with Age in Homozygous Huntington's Disease Knock-In Mouse Models. J Huntingtons Dis 2019; 7:137-150. [PMID: 29843246 PMCID: PMC6002862 DOI: 10.3233/jhd-170274] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background: Huntington’s disease (HD) is a progressive neurodegenerative disorder associated with aging, caused by an expanded polyglutamine (polyQ) repeat within the Huntingtin (HTT) protein. In HD, degeneration of the striatum and atrophy of the cortex are observed while cerebellum is less affected. Objective: To test the hypothesis that HTT protein levels decline with age, which together with HTT mutation could influence disease progression. Methods: Using whole brain cell lysates, a unique method of SDS-PAGE and western analysis was used to quantitate HTT protein, which resolves as a monomer and as a high molecular weight species that is modulated by the presence of transglutaminase 2. HTT levels were measured in striatum, cortex and cerebellum in congenic homozygous Q140 and HdhQ150 knock-in mice and WT littermate controls. Results: Mutant HTT in both homozygous knock-in HD mouse models and WT HTT in control striatal and cortical tissues significantly declined in a progressive manner over time. Levels of mutant HTT in HD cerebellum remained high during aging. Conclusions: A general decline in mutant HTT levels in striatum and cortex is observed that may contribute to disease progression in homozygous knock-in HD mouse models through reduction of HTT function. In cerebellum, sustained levels of mutant HTT with aging may be protective to this tissue which is less overtly affected in HD.
Collapse
Affiliation(s)
- Nicholas R Franich
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Manuela Basso
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Emily A André
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Joseph Ochaba
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Amit Kumar
- Burke Medical Research Institute, White Plains, NY, USA.,Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY, USA.,Department of Neurology, Weill Medical College of Cornell University, New York, NY, USA
| | - Soe Thein
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Gianna Fote
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Marketta Kachemov
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Alice L Lau
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Sylvia Y Yeung
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Alexander Osmand
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA
| | - Scott O Zeitlin
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Rajiv R Ratan
- Burke Medical Research Institute, White Plains, NY, USA.,Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY, USA.,Department of Neurology, Weill Medical College of Cornell University, New York, NY, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.,Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Joan S Steffan
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
104
|
Abstract
Autophagy is the major cellular pathway to degrade dysfunctional organelles and protein aggregates. Autophagy is particularly important in neurons, which are terminally differentiated cells that must last the lifetime of the organism. There are both constitutive and stress-induced pathways for autophagy in neurons, which catalyze the turnover of aged or damaged mitochondria, endoplasmic reticulum, other cellular organelles, and aggregated proteins. These pathways are required in neurodevelopment as well as in the maintenance of neuronal homeostasis. Here we review the core components of the pathway for autophagosome biogenesis, as well as the cell biology of bulk and selective autophagy in neurons. Finally, we discuss the role of autophagy in neuronal development, homeostasis, and aging and the links between deficits in autophagy and neurodegeneration.
Collapse
Affiliation(s)
- Andrea K H Stavoe
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA;
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
105
|
Halpern M, Brennand KJ, Gregory J. Examining the relationship between astrocyte dysfunction and neurodegeneration in ALS using hiPSCs. Neurobiol Dis 2019; 132:104562. [PMID: 31381978 DOI: 10.1016/j.nbd.2019.104562] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/28/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex and fatal neurodegenerative disease for which the causes of disease onset and progression remain unclear. Recent advances in human induced pluripotent stem cell (hiPSC)-based models permit the study of the genetic factors associated with ALS in patient-derived neural cell types, including motor neurons and glia. While astrocyte dysfunction has traditionally been thought to exacerbate disease progression, astrocytic dysfunction may play a more direct role in disease initiation and progression. Such non-cell autonomous mechanisms expand the potential targets of therapeutic intervention, but only a handful of ALS risk-associated genes have been examined for their impact on astrocyte dysfunction and neurodegeneration. This review summarizes what is currently known about astrocyte function in ALS and suggests ways in which hiPSC-based models can be used to more effectively study the role of astrocytes in neurodegenerative disease.
Collapse
Affiliation(s)
- Madeline Halpern
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Kristen J Brennand
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| | - James Gregory
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, United States of America.
| |
Collapse
|
106
|
Carosi JM, Sargeant TJ. Rapamycin and Alzheimer disease: a double-edged sword? Autophagy 2019; 15:1460-1462. [PMID: 31066320 PMCID: PMC6613906 DOI: 10.1080/15548627.2019.1615823] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/23/2019] [Accepted: 05/02/2019] [Indexed: 01/04/2023] Open
Abstract
Numerous studies have reported that inhibition of MTOR (mechanistic target of rapamycin kinase) clearly reduces Alzheimer disease neuropathological hallmarks in mouse models. This has resulted in calls for the use of the MTOR inhibitor rapamycin for the treatment of dementia in humans. Unfortunately, intervention with rapamycin in these mouse studies commenced before or early in the appearance of these pathological hallmarks. Later in Alzheimer disease, when dementia actually manifests, the brain's lysosomal system is severely damaged and treatment with rapamycin is likely to exacerbate this damage. We reassess literature described by a recent perspective article calling for the use of MTOR inhibition in dementia and conclude that rapamycin could be useful, but only in people who are in the earliest stages of Alzheimer disease. We contend that our interpretation of preclinical data concerning use of rapamycin in Alzheimer disease models is necessary if we are to avoid another failed Alzheimer disease drug trial. Abbreviations: AD: Alzheimer disease; APP: amyloid beta precursor protein; MAPT: microtubule associated protein tau; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1.
Collapse
Affiliation(s)
- Julian M Carosi
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Timothy J Sargeant
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
107
|
The activation of Mucolipin TRP channel 1 (TRPML1) protects motor neurons from L-BMAA neurotoxicity by promoting autophagic clearance. Sci Rep 2019; 9:10743. [PMID: 31341250 PMCID: PMC6656764 DOI: 10.1038/s41598-019-46708-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 07/04/2019] [Indexed: 12/14/2022] Open
Abstract
Cellular clearance mechanisms including the autophagy-lysosome pathway are impaired in amyotrophic lateral sclerosis (ALS). One of the most important proteins involved in the regulation of autophagy is the lysosomal Ca2+ channel Mucolipin TRP channel 1 (TRPML1). Therefore, we investigated the role of TRPML1 in a neuronal model of ALS/Parkinson-dementia complex reproduced by the exposure of motor neurons to the cyanobacterial neurotoxin beta-methylamino-L-alanine (L-BMAA). Under these conditions, L-BMAA induces a dysfunction of the endoplasmic reticulum (ER) leading to ER stress and cell death. Therefore we hypothesized a dysfunctional coupling between lysosomes and ER in L-BMAA-treated motor neurons. Here, we showed that in motor neuronal cells TRPML1 as well as the lysosomal protein LAMP1 co-localized with ER. In addition, TRPML1 co-immunoprecipitated with the ER Ca2+ sensor STIM1. Functionally, the TRPML1 agonist ML-SA1 induced lysosomal Ca2+ release in a dose-dependent way in motor neuronal cells. The SERCA inhibitor thapsigargin increased the fluorescent signal associated with lysosomal Ca2+ efflux in the cells transfected with the genetically encoded Ca2+ indicator GCaMP3-ML1, thus suggesting an interplay between the two organelles. Moreover, chronic exposure to L-BMAA reduced TRPML1 protein expression and produced an impairment of both lysosomal and ER Ca2+ homeostasis in primary motor neurons. Interestingly, the preincubation of ML-SA1, by an early activation of AMPK and beclin 1, rescued motor neurons from L-BMAA-induced cell death and reduced the expression of the ER stress marker GRP78. Finally, ML-SA1 reduced the accumulation of the autophagy-related proteins p62/SQSTM1 and LC3-II in L-BMAA-treated motor neurons. Collectively, we propose that the pharmacological stimulation of TRPML1 can rescue motor neurons from L-BMAA-induced toxicity by boosting autophagy and reducing ER stress.
Collapse
|
108
|
Marrone L, Drexler HCA, Wang J, Tripathi P, Distler T, Heisterkamp P, Anderson EN, Kour S, Moraiti A, Maharana S, Bhatnagar R, Belgard TG, Tripathy V, Kalmbach N, Hosseinzadeh Z, Crippa V, Abo-Rady M, Wegner F, Poletti A, Troost D, Aronica E, Busskamp V, Weis J, Pandey UB, Hyman AA, Alberti S, Goswami A, Sterneckert J. FUS pathology in ALS is linked to alterations in multiple ALS-associated proteins and rescued by drugs stimulating autophagy. Acta Neuropathol 2019; 138:67-84. [PMID: 30937520 PMCID: PMC6570784 DOI: 10.1007/s00401-019-01998-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a lethal disease characterized by motor neuron degeneration and associated with aggregation of nuclear RNA-binding proteins (RBPs), including FUS. How FUS aggregation and neurodegeneration are prevented in healthy motor neurons remain critically unanswered questions. Here, we use a combination of ALS patient autopsy tissue and induced pluripotent stem cell-derived neurons to study the effects of FUS mutations on RBP homeostasis. We show that FUS’ tendency to aggregate is normally buffered by interacting RBPs, but this buffering is lost when FUS mislocalizes to the cytoplasm due to ALS mutations. The presence of aggregation-prone FUS in the cytoplasm causes imbalances in RBP homeostasis that exacerbate neurodegeneration. However, enhancing autophagy using small molecules reduces cytoplasmic FUS, restores RBP homeostasis and rescues motor function in vivo. We conclude that disruption of RBP homeostasis plays a critical role in FUS-ALS and can be treated by stimulating autophagy.
Collapse
|
109
|
Maniatis S, Äijö T, Vickovic S, Braine C, Kang K, Mollbrink A, Fagegaltier D, Andrusivová Ž, Saarenpää S, Saiz-Castro G, Cuevas M, Watters A, Lundeberg J, Bonneau R, Phatnani H. Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis. Science 2019; 364:89-93. [PMID: 30948552 DOI: 10.1126/science.aav9776] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/01/2019] [Indexed: 12/12/2022]
Abstract
Paralysis occurring in amyotrophic lateral sclerosis (ALS) results from denervation of skeletal muscle as a consequence of motor neuron degeneration. Interactions between motor neurons and glia contribute to motor neuron loss, but the spatiotemporal ordering of molecular events that drive these processes in intact spinal tissue remains poorly understood. Here, we use spatial transcriptomics to obtain gene expression measurements of mouse spinal cords over the course of disease, as well as of postmortem tissue from ALS patients, to characterize the underlying molecular mechanisms in ALS. We identify pathway dynamics, distinguish regional differences between microglia and astrocyte populations at early time points, and discern perturbations in several transcriptional pathways shared between murine models of ALS and human postmortem spinal cords.
Collapse
Affiliation(s)
- Silas Maniatis
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Tarmo Äijö
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Sanja Vickovic
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Catherine Braine
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Kristy Kang
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Annelie Mollbrink
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Delphine Fagegaltier
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Žaneta Andrusivová
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Sami Saarenpää
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Gonzalo Saiz-Castro
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Miguel Cuevas
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Aaron Watters
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden. .,Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, New York, NY, USA. .,Center for Data Science, New York University, New York, NY, USA
| | - Hemali Phatnani
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA. .,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| |
Collapse
|
110
|
Ochaba J, Fote G, Kachemov M, Thein S, Yeung SY, Lau AL, Hernandez S, Lim RG, Casale M, Neel MJ, Monuki ES, Reidling J, Housman DE, Thompson LM, Steffan JS. IKKβ slows Huntington's disease progression in R6/1 mice. Proc Natl Acad Sci U S A 2019; 116:10952-10961. [PMID: 31088970 PMCID: PMC6561205 DOI: 10.1073/pnas.1814246116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neuroinflammation is an important contributor to neuronal pathology and death in neurodegenerative diseases and neuronal injury. Therapeutic interventions blocking the activity of the inflammatory kinase IKKβ, a key regulator of neuroinflammatory pathways, is protective in several animal models of neurodegenerative disease and neuronal injury. In Huntington's disease (HD), however, significant questions exist as to the impact of blocking or diminishing the activity of IKKβ on HD pathology given its potential role in Huntingtin (HTT) degradation. In cell culture, IKKβ phosphorylates HTT serine (S) 13 and activates HTT degradation, a process that becomes impaired with polyQ expansion. To investigate the in vivo relationship of IKKβ to HTT S13 phosphorylation and HD progression, we crossed conditional tamoxifen-inducible IKKβ knockout mice with R6/1 HD mice. Behavioral assays in these mice showed a significant worsening of HD pathological phenotypes. The increased behavioral pathology correlated with reduced levels of endogenous mouse full-length phospho-S13 HTT, supporting the importance of IKKβ in the phosphorylation of HTT S13 in vivo. Notably, many striatal autophagy genes were up-regulated in HD vs. control mice; however, IKKβ knockout partially reduced this up-regulation in HD, increased striatal neurodegeneration, and enhanced an activated microglial response. We propose that IKKβ is protective in striatal neurons early in HD progression via phosphorylation of HTT S13. As IKKβ is also required for up-regulation of some autophagy genes and HTT is a scaffold for selective autophagy, IKKβ may influence autophagy through multiple mechanisms to maintain healthy striatal function, thereby reducing neuronal degeneration to slow HD onset.
Collapse
Affiliation(s)
- Joseph Ochaba
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| | - Gianna Fote
- Department of Biological Chemistry, University of California, Irvine, CA 92697
| | - Marketta Kachemov
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| | - Soe Thein
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697
| | - Sylvia Y Yeung
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697
| | - Alice L Lau
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697
| | - Sarah Hernandez
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697
| | - Ryan G Lim
- Department of Biological Chemistry, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697
| | - Malcolm Casale
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| | - Michael J Neel
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA 92697
| | - Edwin S Monuki
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA 92697
| | - Jack Reidling
- Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - David E Housman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139;
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Department of Biological Chemistry, University of California, Irvine, CA 92697
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697
- Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Joan S Steffan
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697;
- Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| |
Collapse
|
111
|
Xu B, Zheng C, Chen X, Zhang Z, Liu J, Spencer P, Yang X. Dysregulation of Myosin Complex and Striated Muscle Contraction Pathway in the Brains of ALS-SOD1 Model Mice. ACS Chem Neurosci 2019; 10:2408-2417. [PMID: 30889949 DOI: 10.1021/acschemneuro.8b00704] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal disease characterized by cortical and spinal motor neuron degeneration, some inherited cases of which are caused by mutations in the gene coding for copper-zinc superoxide dismutase-1 (SOD1). The SOD1G93A mutant model mouse, which expresses large amounts of mutant SOD1, develops adult-onset neurodegeneration of spinal motor neurons and progressive motor deficits leading to paralysis. We used the Tandem Mass Tag technique to investigate the proteome profile of hippocampus, cerebral cortex, and medulla oblongata of the SOD1G93A mutant model mice as compared with that of wild-type (WT) mice. Fifteen proteins were significantly increased or decreased (i.e., changed) in all three tissues. Gene ontology analysis revealed that the changed proteins were mainly enriched in negative regulation of reactive oxygen species, myosin complex and copper ion binding. In the Striated Muscle Contraction Pathway, most of the identified proteins were decreased in the SOD1G93A mice compared with the WT mice. Myosin-1 (MYH1), fructose-2,6-bisphosphatase TIGAR (TIGAR), and sarcoplasmic/endoplasmic reticulum calcium ATPase 1 (ATP2A1) were significantly reduced in mutant vs WT mice, as confirmed by Western blot analysis. Since myosins and tropomyosins are specific for synapse function and drive actin dynamics in the maturation of dendritic spines, changes in these proteins may contribute to perturbations of brain neuronal circuitry in addition to spinal motor neuron disease.
Collapse
Affiliation(s)
- Benhong Xu
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Chengyou Zheng
- School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen 518000, China
| | - Xiao Chen
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou 510632, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Peter Spencer
- Department of Neurology, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon 97201, United States
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| |
Collapse
|
112
|
Mitochondrial Transport and Turnover in the Pathogenesis of Amyotrophic Lateral Sclerosis. BIOLOGY 2019; 8:biology8020036. [PMID: 31083575 PMCID: PMC6627920 DOI: 10.3390/biology8020036] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/20/2019] [Accepted: 02/03/2019] [Indexed: 02/06/2023]
Abstract
Neurons are high-energy consuming cells, heavily dependent on mitochondria for ATP generation and calcium buffering. These mitochondrial functions are particularly critical at specific cellular sites, where ionic currents impose a large energetic burden, such as at synapses. The highly polarized nature of neurons, with extremely large axoplasm relative to the cell body, requires mitochondria to be efficiently transported along microtubules to reach distant sites. Furthermore, neurons are post-mitotic cells that need to maintain pools of healthy mitochondria throughout their lifespan. Hence, mitochondrial transport and turnover are essential processes for neuronal survival and function. In neurodegenerative diseases, the maintenance of a healthy mitochondrial network is often compromised. Numerous lines of evidence indicate that mitochondrial impairment contributes to neuronal demise in a variety of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), where degeneration of motor neurons causes a fatal muscle paralysis. Dysfunctional mitochondria accumulate in motor neurons affected by genetic or sporadic forms of ALS, strongly suggesting that the inability to maintain a healthy pool of mitochondria plays a pathophysiological role in the disease. This article critically reviews current hypotheses on mitochondrial involvement in the pathogenesis of ALS, focusing on the alterations of mitochondrial axonal transport and turnover in motor neurons.
Collapse
|
113
|
Meng T, Lin S, Zhuang H, Huang H, He Z, Hu Y, Gong Q, Feng D. Recent progress in the role of autophagy in neurological diseases. Cell Stress 2019; 3:141-161. [PMID: 31225510 PMCID: PMC6551859 DOI: 10.15698/cst2019.05.186] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Autophagy (here refers to macroautophagy) is a catabolic pathway by which large protein aggregates and damaged organelles are first sequestered into a double-membraned structure called autophago-some and then delivered to lysosome for destruction. Recently, tremen-dous progress has been made to elucidate the molecular mechanism and functions of this essential cellular metabolic process. In addition to being either a rubbish clearing system or a cellular surviving program in response to different stresses, autophagy plays important roles in a large number of pathophysiological conditions, such as cancer, diabetes, and especially neurodegenerative disorders. Here we review recent progress in the role of autophagy in neurological diseases and discuss how dysregulation of autophagy initiation, autophagosome formation, maturation, and/or au-tophagosome-lysosomal fusion step contributes to the pathogenesis of these disorders in the nervous system.
Collapse
Affiliation(s)
- Tian Meng
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University; Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Shiyin Lin
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University; Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Haixia Zhuang
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University; Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Haofeng Huang
- Institute of Neurology, Guangdong Key Laboratory of Age-Related Cardiac-Cerebral Vascular Disease, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Zhengjie He
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University; Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Yongquan Hu
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University; Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 511436, China
| | - Qing Gong
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 511436, People's Republic of China
| | - Du Feng
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University; Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
114
|
Stavoe AKH, Holzbaur ELF. Axonal autophagy: Mini-review for autophagy in the CNS. Neurosci Lett 2019; 697:17-23. [PMID: 29548988 PMCID: PMC6136980 DOI: 10.1016/j.neulet.2018.03.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/12/2018] [Indexed: 01/14/2023]
Abstract
Neurons are long-lived and highly polarized cells that depend on autophagy to maintain cellular homeostasis. The robust, constitutive biogenesis of autophagosomes in the distal axon occurs via a conserved pathway that is required to maintain functional synapses and prevent axon degeneration. Autophagosomes are formed de novo at the axon terminal in a stepwise assembly process, engulfing mitochondrial fragments, aggregated proteins, and bulk cytosol in what appears to be a nonselective uptake mechanism. Following formation, autophagosomes fuse with late endosomes/lysosomes and then are rapidly and efficiently transported along the axon toward the soma, driven by the microtubule motor cytoplasmic dynein. Motile autophagosomes mature to autolysosomes in transit by fusing with additional late endosomes/lysosomes, arriving at the soma as fully competent degradative organelles. Misregulation of neuronal autophagy leads to axonal degeneration and synaptic destabilization, and has been implicated in neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and ALS.
Collapse
Affiliation(s)
- Andrea K H Stavoe
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
115
|
Nguyen DKH, Thombre R, Wang J. Autophagy as a common pathway in amyotrophic lateral sclerosis. Neurosci Lett 2019; 697:34-48. [PMID: 29626651 PMCID: PMC6170747 DOI: 10.1016/j.neulet.2018.04.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/26/2018] [Accepted: 04/02/2018] [Indexed: 12/11/2022]
Abstract
Age-dependent neurodegenerative diseases are associated with a decline in protein quality control systems including autophagy. Amyotrophic lateral sclerosis (ALS) is a motor neuron degenerative disease of complex etiology with increasing connections to other neurodegenerative conditions such as frontotemporal dementia. Among the diverse genetic causes for ALS, a striking feature is the common connection to autophagy and its associated pathways. There is a recurring theme of protein misfolding as in other neurodegenerative diseases, but importantly there is a distinct common thread among ALS genes that connects them to the cascade of autophagy. However, the roles of autophagy in ALS remain enigmatic and it is still unclear whether activation or inhibition of autophagy would be a reliable avenue to ameliorate the disease. The main evidence that links autophagy to different genetic forms of ALS is discussed.
Collapse
Affiliation(s)
- Dao K H Nguyen
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA; Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Ravi Thombre
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA; Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Jiou Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA; Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
116
|
Beltran S, Nassif M, Vicencio E, Arcos J, Labrador L, Cortes BI, Cortez C, Bergmann CA, Espinoza S, Hernandez MF, Matamala JM, Bargsted L, Matus S, Rojas-Rivera D, Bertrand MJM, Medinas DB, Hetz C, Manque PA, Woehlbier U. Network approach identifies Pacer as an autophagy protein involved in ALS pathogenesis. Mol Neurodegener 2019; 14:14. [PMID: 30917850 PMCID: PMC6437924 DOI: 10.1186/s13024-019-0313-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a multifactorial fatal motoneuron disease without a cure. Ten percent of ALS cases can be pointed to a clear genetic cause, while the remaining 90% is classified as sporadic. Our study was aimed to uncover new connections within the ALS network through a bioinformatic approach, by which we identified C13orf18, recently named Pacer, as a new component of the autophagic machinery and potentially involved in ALS pathogenesis. METHODS Initially, we identified Pacer using a network-based bioinformatic analysis. Expression of Pacer was then investigated in vivo using spinal cord tissue from two ALS mouse models (SOD1G93A and TDP43A315T) and sporadic ALS patients. Mechanistic studies were performed in cell culture using the mouse motoneuron cell line NSC34. Loss of function of Pacer was achieved by knockdown using short-hairpin constructs. The effect of Pacer repression was investigated in the context of autophagy, SOD1 aggregation, and neuronal death. RESULTS Using an unbiased network-based approach, we integrated all available ALS data to identify new functional interactions involved in ALS pathogenesis. We found that Pacer associates to an ALS-specific subnetwork composed of components of the autophagy pathway, one of the main cellular processes affected in the disease. Interestingly, we found that Pacer levels are significantly reduced in spinal cord tissue from sporadic ALS patients and in tissues from two ALS mouse models. In vitro, Pacer deficiency lead to impaired autophagy and accumulation of ALS-associated protein aggregates, which correlated with the induction of cell death. CONCLUSIONS This study, therefore, identifies Pacer as a new regulator of proteostasis associated with ALS pathology.
Collapse
Affiliation(s)
- S Beltran
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - M Nassif
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - E Vicencio
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - J Arcos
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - L Labrador
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - B I Cortes
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - C Cortez
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - C A Bergmann
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - S Espinoza
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile
| | - M F Hernandez
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile
| | - J M Matamala
- Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia, 1027, Santiago, Chile
| | - L Bargsted
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia, 1027, Santiago, Chile
| | - S Matus
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia, 1027, Santiago, Chile.,Fundación Ciencia & Vida, Zañartu 1482, 7780272, Santiago, Chile.,Neurounion Biomedical Foundation, 7780272, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
| | - D Rojas-Rivera
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile.,VIB Center for Inflammation Research, Technologiepark 927, Zwijnaarde, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Zwijnaarde, 9052, Ghent, Belgium
| | - M J M Bertrand
- VIB Center for Inflammation Research, Technologiepark 927, Zwijnaarde, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Zwijnaarde, 9052, Ghent, Belgium
| | - D B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia, 1027, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Independencia, 1027, Santiago, Chile
| | - C Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia, 1027, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, 94945, USA.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Independencia, 1027, Santiago, Chile.,Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - P A Manque
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile. .,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile. .,Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| | - U Woehlbier
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Piramide 5750, P.O.BOX 70086, Santiago, Chile. .,Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Piramide, 5750, Santiago, Chile.
| |
Collapse
|
117
|
Řehořová M, Vargová I, Forostyak S, Vacková I, Turnovcová K, Kupcová Skalníková H, Vodička P, Kubinová Š, Syková E, Jendelová P. A Combination of Intrathecal and Intramuscular Application of Human Mesenchymal Stem Cells Partly Reduces the Activation of Necroptosis in the Spinal Cord of SOD1 G93A Rats. Stem Cells Transl Med 2019; 8:535-547. [PMID: 30802001 PMCID: PMC6525562 DOI: 10.1002/sctm.18-0223] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/03/2019] [Indexed: 12/13/2022] Open
Abstract
An increasing number of studies have demonstrated the beneficial effects of human mesenchymal stem cells (hMSC) in the treatment of amyotrophic lateral sclerosis (ALS). We compared the effect of repeated intrathecal applications of hMSC or their conditioned medium (CondM) using lumbar puncture or injection into the muscle (quadriceps femoris), or a combination of both applications in symptomatic SOD1G93A rats. We further assessed the effect of the treatment on three major cell death pathways (necroptosis, apoptosis, and autophagy) in the spinal cord tissue. All the animals were behaviorally tested (grip strength test, Basso Beattie Bresnahan (BBB) test, and rotarod), and the tissue was analyzed immunohistochemically, by qPCR and Western blot. All symptomatic SOD1 rats treated with hMSC had a significantly increased lifespan, improved motor activity and reduced number of Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) positive cells. Moreover, a combined hMSC delivery increased motor neuron survival, maintained neuromuscular junctions in quadriceps femoris and substantially reduced the levels of proteins involved in necroptosis (Rip1, mixed lineage kinase‐like protein, cl‐casp8), apoptosis (cl‐casp 9) and autophagy (beclin 1). Furthermore, astrogliosis and elevated levels of Connexin 43 were decreased after combined hMSC treatment. The repeated application of CondM, or intramuscular injections alone, improved motor activity; however, this improvement was not supported by changes at the molecular level. Our results provide new evidence that a combination of repeated intrathecal and intramuscular hMSC applications protects motor neurons and neuromuscular junctions, not only through a reduction of apoptosis and autophagy but also through the necroptosis pathway, which is significantly involved in cell death in rodent SOD1G93A model of ALS. stem cells translational medicine2019;8:535–547
Collapse
Affiliation(s)
- Monika Řehořová
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ingrid Vargová
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Serhiy Forostyak
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic.,Prime Cell Advanced Therapy A.S., Brno, Czech Republic
| | - Irena Vacková
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic
| | - Karolína Turnovcová
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic
| | | | - Petr Vodička
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Liběchov, Czech Republic
| | - Šárka Kubinová
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eva Syková
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic.,Institute of Neuroimmunology, Slovak Academy of Science, Bratislava, Slovakia
| | - Pavla Jendelová
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
118
|
Brenner D, Sieverding K, Bruno C, Lüningschrör P, Buck E, Mungwa S, Fischer L, Brockmann SJ, Ulmer J, Bliederhäuser C, Philibert CE, Satoh T, Akira S, Boillée S, Mayer B, Sendtner M, Ludolph AC, Danzer KM, Lobsiger CS, Freischmidt A, Weishaupt JH. Heterozygous Tbk1 loss has opposing effects in early and late stages of ALS in mice. J Exp Med 2019; 216:267-278. [PMID: 30635357 PMCID: PMC6363427 DOI: 10.1084/jem.20180729] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 10/24/2018] [Accepted: 11/28/2018] [Indexed: 12/11/2022] Open
Abstract
Heterozygous loss-of-function mutations of TANK-binding kinase 1 (TBK1 ) cause familial ALS, yet downstream mechanisms of TBK1 mutations remained elusive. TBK1 is a pleiotropic kinase involved in the regulation of selective autophagy and inflammation. We show that heterozygous Tbk1 deletion alone does not lead to signs of motoneuron degeneration or disturbed autophagy in mice during a 200-d observation period. Surprisingly, however, hemizygous deletion of Tbk1 inversely modulates early and late disease phases in mice additionally overexpressing ALS-linked SOD1G93A , which represents a "second hit" that induces both neuroinflammation and proteostatic dysregulation. At the early stage, heterozygous Tbk1 deletion impairs autophagy in motoneurons and prepones both the clinical onset and muscular denervation in SOD1G93A/Tbk1+/- mice. At the late disease stage, however, it significantly alleviates microglial neuroinflammation, decelerates disease progression, and extends survival. Our results indicate a profound effect of TBK1 on brain inflammatory cells under pro-inflammatory conditions and point to a complex, two-edged role of TBK1 in SOD1-linked ALS.
Collapse
Affiliation(s)
- David Brenner
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | - Clara Bruno
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Patrick Lüningschrör
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Eva Buck
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Simon Mungwa
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Lena Fischer
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | - Johannes Ulmer
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | - Clémentine E Philibert
- Institut du Cerveau et de la Moelle Épinière, Institut National de la Santé et de la Recherche Médicale Unité 1127, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7225, Sorbonne Université, Paris, France
| | - Takashi Satoh
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Séverine Boillée
- Institut du Cerveau et de la Moelle Épinière, Institut National de la Santé et de la Recherche Médicale Unité 1127, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7225, Sorbonne Université, Paris, France
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | | | - Karin M Danzer
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Christian S Lobsiger
- Institut du Cerveau et de la Moelle Épinière, Institut National de la Santé et de la Recherche Médicale Unité 1127, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7225, Sorbonne Université, Paris, France
| | | | | |
Collapse
|
119
|
Joshi AU, Saw NL, Vogel H, Cunnigham AD, Shamloo M, Mochly-Rosen D. Inhibition of Drp1/Fis1 interaction slows progression of amyotrophic lateral sclerosis. EMBO Mol Med 2019; 10:emmm.201708166. [PMID: 29335339 PMCID: PMC5840540 DOI: 10.15252/emmm.201708166] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bioenergetic failure and oxidative stress are common pathological hallmarks of amyotrophic lateral sclerosis (ALS), but whether these could be targeted effectively for novel therapeutic intervention needs to be determined. One of the reported contributors to ALS pathology is mitochondrial dysfunction associated with excessive mitochondrial fission and fragmentation, which is predominantly mediated by Drp1 hyperactivation. Here, we determined whether inhibition of excessive fission by inhibiting Drp1/Fis1 interaction affects disease progression. We observed mitochondrial excessive fragmentation and dysfunction in several familial forms of ALS patient‐derived fibroblasts as well as in cultured motor neurons expressing SOD1 mutant. In both cell models, inhibition of Drp1/Fis1 interaction by a selective peptide inhibitor, P110, led to a significant reduction in reactive oxygen species levels, and to improvement in mitochondrial structure and functions. Sustained treatment of mice expressing G93A SOD1 mutation with P110, beginning at the onset of disease symptoms at day 90, produced an improvement in motor performance and survival, suggesting that Drp1 hyperactivation may be an attractive target in the treatment of ALS patients.
Collapse
Affiliation(s)
- Amit U Joshi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nay L Saw
- Behavioral and Functional Neuroscience Laboratory, Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anna D Cunnigham
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mehrdad Shamloo
- Behavioral and Functional Neuroscience Laboratory, Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
120
|
Wu C, Watts ME, Rubin LL. MAP4K4 Activation Mediates Motor Neuron Degeneration in Amyotrophic Lateral Sclerosis. Cell Rep 2019; 26:1143-1156.e5. [PMID: 30699345 DOI: 10.1016/j.celrep.2019.01.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/03/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting both upper and lower motor neurons (MNs). To date, its underlying mechanisms have yet to be clarified completely, and there are no truly effective treatments. Here, we show that MAP4K4, a MAP kinase family member, regulates MN death, with its suppression not only promoting survival but preventing neurite degeneration and decreasing mutant SOD1 levels through autophagy activation. Moreover, we report that MAP4K4 signaling specifically modulates MN viability via phosphorylated JNK3 and activation of the canonical c-Jun apoptotic pathway. Finally, we show the feasibility of MAP4K4 as a drug target by using an available MAP4K4-specific inhibitor, which improves survival of ESC and/or iPSC-derived MNs and MNs cultured from mouse spinal cords. In summary, our studies highlight a MAP4K4-initiated signaling cascade that induces MN degeneration, shedding light on the mechanism underlying MN degeneration and providing a druggable target for ALS therapeutics.
Collapse
Affiliation(s)
- Chen Wu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Michelle E Watts
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
121
|
Gonzalez Porras MA, Sieck GC, Mantilla CB. Impaired Autophagy in Motor Neurons: A Final Common Mechanism of Injury and Death. Physiology (Bethesda) 2019; 33:211-224. [PMID: 29638184 DOI: 10.1152/physiol.00008.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Autophagy is a cellular digestion process that contributes to cellular homeostasis and adaptation by the elimination of proteins and damaged organelles. Evidence suggests that dysregulation of autophagy plays a role in neurodegenerative diseases, including motor neuron disorders. Herein, we review emerging evidence indicating the roles of autophagy in physiological motor neuron processes and its function in specific compartments. Moreover, we discuss the involvement of autophagy in the pathogenesis of motor neuron diseases, including spinal cord injury and aging, and recent developments that offer promising therapeutic approaches to mitigate effects of dysregulated autophagy in health and disease.
Collapse
Affiliation(s)
| | - Gary C Sieck
- Department of Physiology & Biomedical Engineering, Mayo Clinic , Rochester, Minnesota.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota
| | - Carlos B Mantilla
- Department of Physiology & Biomedical Engineering, Mayo Clinic , Rochester, Minnesota.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
122
|
Liang Y. Emerging Concepts and Functions of Autophagy as a Regulator of Synaptic Components and Plasticity. Cells 2019; 8:cells8010034. [PMID: 30634508 PMCID: PMC6357011 DOI: 10.3390/cells8010034] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/23/2018] [Accepted: 01/03/2019] [Indexed: 12/15/2022] Open
Abstract
Protein homeostasis (proteostasis) is crucial to the maintenance of neuronal integrity and function. As the contact sites between neurons, synapses rely heavily on precisely regulated protein-protein interactions to support synaptic transmission and plasticity processes. Autophagy is an effective degradative pathway that can digest cellular components and maintain cellular proteostasis. Perturbations of autophagy have been implicated in aging and neurodegeneration due to a failure to remove damaged proteins and defective organelles. Recent evidence has demonstrated that autophagosome formation is prominent at synaptic terminals and neuronal autophagy is regulated in a compartment-specific fashion. Moreover, synaptic components including synaptic proteins and vesicles, postsynaptic receptors and synaptic mitochondria are known to be degraded by autophagy, thereby contributing to the remodeling of synapses. Indeed, emerging studies indicate that modulation of autophagy may be required for different forms of synaptic plasticity and memory formation. In this review, I will discuss our current understanding of the important role of neuronal/synaptic autophagy in maintaining neuronal function by degrading synaptic components and try to propose a conceptual framework of how the degradation of synaptic components via autophagy might impact synaptic function and contribute to synaptic plasticity.
Collapse
Affiliation(s)
- YongTian Liang
- Neurogenetik, Institut für Biologie, Freie Universität Berlin, 14195 Berlin, Germany.
- NeuroCure, Cluster of Excellence, Charité Universitätsmedizin, 10117 Berlin, Germany.
| |
Collapse
|
123
|
Walker CL, Meadows RM, Merfeld-Clauss S, Du Y, March KL, Jones KJ. Adipose-derived stem cell conditioned medium impacts asymptomatic peripheral neuromuscular denervation in the mutant superoxide dismutase (G93A) transgenic mouse model of amyotrophic lateral sclerosis. Restor Neurol Neurosci 2018; 36:621-627. [PMID: 30010155 DOI: 10.3233/rnn-180820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is devastating, leading to paralysis and death. Disease onset begins pre-symptomatically through spinal motor neuron (MN) axon die-back from musculature at ∼47 days of age in the mutant superoxide dismutase 1 (mSOD1G93A) transgenic ALS mouse model. This period may be optimal to assess potential therapies. We previously demonstrated that post-symptomatic adipose-derived stem cell conditioned medium (ASC-CM) treatment is neuroprotective in mSOD1G93A mice. We hypothesized that early disease onset treatment could ameliorate neuromuscular junction (NMJ) disruption. OBJECTIVE To determine whether pre-symptom administration of ASC-CM prevents early NMJ disconnection. METHODS We confirmed the NMJ denervation time course in mSOD1G93A mice using co-labeling of neurofilament and post-synaptic acetylcholine receptors (AchR) by α-bungarotoxin. We determined whether ASC-CM ameliorates early NMJ loss in mSOD1G93A mice by systemically administering 200μl ASC-CM or vehicle medium daily from post-natal days 35 to 47 and quantifying intact NMJs through co-labeling of neurofilament and synaptophysin with α-bungarotoxin in gastrocnemius muscle. RESULTS Intact NMJs were significantly decreased in 47 day old mSOD1G93A mice (p < 0.05), and daily systemic ASC-CM prevented disease-induced NMJ denervation compared to vehicle treated mice (p < 0.05). CONCLUSIONS Our results lay the foundation for testing the long-term neurological benefits of systemic ASC-CM therapy in the mSOD1G93A mouse model of ALS.
Collapse
Affiliation(s)
- Chandler L Walker
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, USA.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Rena M Meadows
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Stephanie Merfeld-Clauss
- Roudebush VA Medical Center, Indianapolis, IN, USA.,Division of Cardiovascular Medicine, Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith L March
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA.,Division of Cardiovascular Medicine, Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Kathryn J Jones
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
124
|
Sánchez-Martín P, Komatsu M. p62/SQSTM1 - steering the cell through health and disease. J Cell Sci 2018; 131:131/21/jcs222836. [PMID: 30397181 DOI: 10.1242/jcs.222836] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
SQSTM1 (also known as p62) is a multifunctional stress-inducible scaffold protein involved in diverse cellular processes. Its functions are tightly regulated through an extensive pattern of post-translational modifications, and include the isolation of cargos degraded by autophagy, induction of the antioxidant response by the Keap1-Nrf2 system, as well as the regulation of endosomal trafficking, apoptosis and inflammation. Accordingly, malfunction of SQSTM1 is associated with a wide range of diseases, including bone and muscle disorders, neurodegenerative and metabolic diseases, and multiple forms of cancer. In this Review, we summarize current knowledge regarding regulation, post-translational modifications and functions of SQSTM1, as well as how they are dysregulated in various pathogenic contexts.
Collapse
Affiliation(s)
- Pablo Sánchez-Martín
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan
| | - Masaaki Komatsu
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan .,Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
125
|
Massenzio F, Peña-Altamira E, Petralla S, Virgili M, Zuccheri G, Miti A, Polazzi E, Mengoni I, Piffaretti D, Monti B. Microglial overexpression of fALS-linked mutant SOD1 induces SOD1 processing impairment, activation and neurotoxicity and is counteracted by the autophagy inducer trehalose. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3771-3785. [PMID: 30315929 DOI: 10.1016/j.bbadis.2018.10.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease. Mutations in the gene encoding copper/zinc superoxide dismutase-1 (SOD1) are responsible for most familiar cases, but the role of mutant SOD1 protein dysfunction in non-cell autonomous neurodegeneration, especially in relation to microglial activation, is still unclear. Here, we focused our study on microglial cells, which release SOD1 also through exosomes. We observed that in rat primary microglia the overexpression of the most-common SOD1 mutations linked to fALS (G93A and A4V) leads to SOD1 intracellular accumulation, which correlates to autophagy dysfunction and microglial activation. In primary contact co-cultures, fALS mutant SOD1 overexpression by microglial cells appears to be neurotoxic by itself. Treatment with the autophagy-inducer trehalose reduced mutant SOD1 accumulation in microglial cells, decreased microglial activation and abrogated neurotoxicity in the co-culture model. These data suggest that i) the alteration of the autophagic pathway due to mutant SOD1 overexpression is involved in microglial activation and neurotoxicity; ii) the induction of autophagy with trehalose reduces microglial SOD1 accumulation through proteasome degradation and activation, leading to neuroprotection. Our results provide a novel contribution towards better understanding key cellular mechanisms in non-cell autonomous ALS neurodegeneration.
Collapse
Affiliation(s)
- Francesca Massenzio
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marco Virgili
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giampaolo Zuccheri
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy; Interdepartmental Center for Industrial Research on Life and Health Sciences at the University of Bologna, Italy; S3 Center of the Institute of Nanoscience of the National Research Council (C.N.R.), Italy
| | - Andrea Miti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Elisabetta Polazzi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Ilaria Mengoni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Deborah Piffaretti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| |
Collapse
|
126
|
Paul S, Dansithong W, Figueroa KP, Scoles DR, Pulst SM. Staufen1 links RNA stress granules and autophagy in a model of neurodegeneration. Nat Commun 2018; 9:3648. [PMID: 30194296 PMCID: PMC6128856 DOI: 10.1038/s41467-018-06041-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 07/17/2018] [Indexed: 11/26/2022] Open
Abstract
Spinocerebellar ataxia type 2 (SCA2) is a neurodegenerative disease caused by expansion of polyglutamine tract in the ATXN2 protein. We identified Staufen1 (STAU1) as an interactor of ATXN2, and showed elevation in cells from SCA2 patients, amyotrophic lateral sclerosis (ALS) patients, and in SCA2 mouse models. We demonstrated recruitment of STAU1 to mutant ATXN2 aggregates in brain tissue from patients with SCA2 human brain and in an SCA2 mouse model, and association of STAU1 elevation with dysregulation of SCA2-related transcript abundances. Targeting STAU1 in vitro by RNAi restored PCP2 transcript levels and lowering mutant ATXN2 also normalized STAU1 levels. Reduction of Stau1 in vivo improved motor behavior in an SCA2 mouse model, normalized the levels of several SCA2-related proteins, and reduced aggregation of polyglutamine-expanded ATXN2. These findings suggest a function for STAU1 in aberrant RNA metabolism associated with ATXN2 mutation, suggesting STAU1 is a possible novel therapeutic target for SCA2.
Collapse
Affiliation(s)
- Sharan Paul
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA
| | - Warunee Dansithong
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA
| | - Karla P Figueroa
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA
| | - Daniel R Scoles
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA.
| |
Collapse
|
127
|
Schmeisser K, Parker JA. Nicotinamide-N-methyltransferase controls behavior, neurodegeneration and lifespan by regulating neuronal autophagy. PLoS Genet 2018; 14:e1007561. [PMID: 30192747 PMCID: PMC6191153 DOI: 10.1371/journal.pgen.1007561] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 10/16/2018] [Accepted: 07/09/2018] [Indexed: 12/14/2022] Open
Abstract
Nicotinamide N-methyl-transferase (NNMT) is an essential contributor to various metabolic and epigenetic processes, including the regulating of aging, cellular stress response, and body weight gain. Epidemiological studies show that NNMT is a risk factor for psychiatric diseases like schizophrenia and neurodegeneration, especially Parkinson's disease (PD), but its neuronal mechanisms of action remain obscure. Here, we describe the role of neuronal NNMT using C. elegans. We discovered that ANMT-1, the nematode NNMT ortholog, competes with the methyltransferase LCMT-1 for methyl groups from S-adenosyl methionine. Thereby, it regulates the catalytic capacities of LCMT-1, targeting NPRL-2, a regulator of autophagy. Autophagy is a core cellular, catabolic process for degrading cytoplasmic material, but very little is known about the regulation of autophagy during aging. We report an important role for NNMT in regulation of autophagy during aging, where high neuronal ANMT-1 activity induces autophagy via NPRL-2, which maintains neuronal function in old wild type animals and various disease models, also affecting longevity. In younger animals, however, ANMT-1 activity disturbs neuronal homeostasis and dopamine signaling, causing abnormal behavior. In summary, we provide fundamental insights into neuronal NNMT/ANMT-1 as pivotal regulator of behavior, neurodegeneration, and lifespan by controlling neuronal autophagy, potentially influencing PD and schizophrenia risk in humans.
Collapse
Affiliation(s)
- Kathrin Schmeisser
- Research Center of the Centre Hospitalier de l‘Université de Montréal (CRCHUM), Department of Neuroscience, Université de Montréal, Quebec, Canada
| | - J. Alex Parker
- Research Center of the Centre Hospitalier de l‘Université de Montréal (CRCHUM), Department of Neuroscience, Université de Montréal, Quebec, Canada
| |
Collapse
|
128
|
Protective effects of Withania somnifera extract in SOD1 G93A mouse model of amyotrophic lateral sclerosis. Exp Neurol 2018; 309:193-204. [PMID: 30134145 DOI: 10.1016/j.expneurol.2018.08.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/21/2018] [Accepted: 08/15/2018] [Indexed: 12/11/2022]
Abstract
Withania somnifera (WS; commonly known as Ashwagandha or Indian ginseng) is a medicinal plant whose extracts have been in use for centuries in various regions of the world as a rejuvenator. There is now a growing body of evidence documenting neuroprotective functions of the plant extracts or its purified compounds in several models of neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). Based on the extract's beneficial effect in a mouse model of ALS with TDP-43 proteinopathy, the current study was designed to test its efficacy in another model of familial ALS. Our results show that administration of WS extracts by gavage to mice expressing G93A mutant form of superoxide dismutase (SOD1) resulted in increased longevity, improved motor performance and increased number of motor neurons in lumbar spinal cord. The WS treatment caused substantial reduction in levels of misfolded SOD1whereas it enhanced expression of cellular chaperons in spinal cord of SOD1G93A mice. WS markedly reduced glial activation and prevented phosphorylation of nuclear factor kappaB (NF-κB). The overall immunomodulatory effect of WS was further evidenced by changes in expression of multiple cytokines/chemokines. WS also served as an autophagy inducer which may be beneficial at early stages of the disease. These results suggest that WS extracts might constitute promising therapeutics for treatment of ALS with involvement of misfolded SOD1.
Collapse
|
129
|
Bojungikgi-tang Improves Muscle and Spinal Cord Function in an Amyotrophic Lateral Sclerosis Model. Mol Neurobiol 2018; 56:2394-2407. [PMID: 30030751 DOI: 10.1007/s12035-018-1236-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a motor neuron disease characterized by progressive motor function impairment, dysphagia, and respiratory failure. Owing to the complexity of its pathogenic mechanisms, an effective therapy for ALS is lacking. Herbal medicines with multiple targets have good efficacy and low adverse reactions for the treatment of neurodegenerative diseases. In this study, the effects of Bojungikgi-tang (BJIGT), an herbal medicine with eight component herbs, on muscle and spinal cord function were evaluated in an ALS animal model. Animals were randomly divided into three groups: a non-transgenic group (nTg, n = 24), a hSOD1G93A transgenic group (Tg, n = 24), and a hSOD1G93A transgenic group in which 8-week-old mice were orally administered BJIGT (1 mg/g) once daily for 6 weeks (Tg+BJIGT, n = 24). The effects of BJIGT were evaluated using a rotarod test, foot-printing, and survival analyses based on Kaplan-Meier survival curves. To determine the biological mechanism underlying the effects of BJIGT in hSOD1G93A mice, western blotting, transmission electron microscopy, and Bungarotoxin staining were used. BJIGT improved motor function and extended the survival duration of hSOD1G93A mice. In addition, BJIGT had protective effects, including anti-oxidative and anti-inflammatory effects, in both the spinal cord and muscle of hSOD1G93A mice. Our results demonstrated that BJIGT causes muscle atrophy and the denervation of neuromuscular junctions in the gastrocnemius of hSOD1G93A mice. The components of BJIGT may alleviate the symptoms of ALS via different mechanisms, and accordingly, BJIGT treatment may be an effective therapeutic approach.
Collapse
|
130
|
Evans CS, Holzbaur ELF. Autophagy and mitophagy in ALS. Neurobiol Dis 2018; 122:35-40. [PMID: 29981842 DOI: 10.1016/j.nbd.2018.07.005] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/19/2018] [Accepted: 07/04/2018] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating and incurable disease involving the loss of motor neurons and subsequent muscle atrophy. Genetic studies have implicated deficits in autophagy and/or mitophagy in the onset of the disease. Here we review recent progress in our understanding of the pathways for autophagy and mitophagy in neurons, and how these pathways may be affected by mutations in genes including DCTN1, OPTN, TBK1, VCP, and C9ORF72. We also discuss the implications of modulating autophagy in ALS, highlighting both the potential of the approach and the concerns raised by targeting this pathway as a therapeutic strategy in neurodegenerative disease.
Collapse
Affiliation(s)
- Chantell S Evans
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, United States
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, United States.
| |
Collapse
|
131
|
Pozzi S, Thammisetty SS, Julien JP. Chronic Administration of Pimozide Fails to Attenuate Motor and Pathological Deficits in Two Mouse Models of Amyotrophic Lateral Sclerosis. Neurotherapeutics 2018; 15:715-727. [PMID: 29790082 PMCID: PMC6095790 DOI: 10.1007/s13311-018-0634-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease which presently does not have any efficient therapeutic approach. Pimozide, a Food and Drug Administration (FDA)-approved neuroepileptic drug, has been recently proposed as a promising treatment for ALS patients based on apparent stabilization of right hand muscles after a short-time administration. A new clinical trial started at the end of 2017 to recruit patients with a prolonged drug delivery schedule. Here, our aim was to investigate the effects of chronic administration of pimozide on disease progression and pathological events in two mouse models of ALS. Pimozide was administered every 2 days to transgenic mice bearing the ALS-linked A315T mutation on the human TAR DNA-binding protein 43 (TDP-43) gene and to mice carrying the human superoxide dismutase 1 (SOD1) gene with the ALS-linked G93A mutation. Chronic administration of pimozide exacerbated motor performances in both animal models and reduced survival in SOD1G93A mice. In TDP-43A315T, it decreased the percentage of innervated neuromuscular junctions (NMJs) and increased the accumulation of insoluble TDP-43. In SOD1G93A mice, pimozide had no effects on NMJ innervation or motoneuron loss, but it increased the levels of misfolded SOD1. We conclude that a chronic administration of pimozide did not confer beneficial effects on disease progression in two mouse models of ALS. In light of a new clinical trial on ALS patients with a chronic regime of pimozide, these results with mouse models suggest prudence and careful monitoring of ALS patients subjected to pimozide treatment.
Collapse
Affiliation(s)
- Silvia Pozzi
- CERVO Brain Research Center, 2601 Chemin de la Canardière, Québec, Québec, G1J 2G3, Canada
| | | | - Jean-Pierre Julien
- CERVO Brain Research Center, 2601 Chemin de la Canardière, Québec, Québec, G1J 2G3, Canada.
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, G1V 0A6, Canada.
| |
Collapse
|
132
|
Mitsui S, Otomo A, Nozaki M, Ono S, Sato K, Shirakawa R, Adachi H, Aoki M, Sobue G, Shang HF, Hadano S. Systemic overexpression of SQSTM1/p62 accelerates disease onset in a SOD1 H46R-expressing ALS mouse model. Mol Brain 2018; 11:30. [PMID: 29843805 PMCID: PMC5975400 DOI: 10.1186/s13041-018-0373-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/20/2018] [Indexed: 02/05/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by a selective loss of upper and lower motor neurons. Recent studies have shown that mutations in SQSTM1 are linked to ALS. SQSTM1 encodes SQSTM1/p62 that regulates not only autophagy via the association with MAP1LC3/LC3 and ubiquitinated proteins but also the KEAP1-NFE2L2/Nrf2 anti-oxidative stress pathway by interacting with KEAP1. Previously, we have demonstrated that loss of SQSTM1 exacerbates disease phenotypes in a SOD1H46R-expressing ALS mouse model. To clarify the effects of SQSTM1 overexpression in this model, we generated SQSTM1 and SOD1 H46R double-transgenic (SQSTM1;SOD1 H46R ) mice. SQSTM1;SOD1 H46R mice exhibited earlier disease onset and shorter lifespan than did SOD1 H46R mice. Conversely, disease progression after the onset rather slightly but significantly slowed in SQSTM1;SOD1 H46R mice. However, there were observable differences neither in the number of Nissl positive neurons nor in the distribution of ubiquitin-positive and/or SQSTM1-positive aggregates between SOD1 H46R and SQSTM1;SOD1 H46R mice. It was noted that these protein aggregates were mainly observed in neuropil, and partly localized to astrocytes and/or microglia, but not to MAP2-positive neuronal cell bodies and dendrites at the end-stage of disease. Nonetheless, the biochemically-detectable insoluble SQSTM1 and poly-ubiquitinated proteins were significantly and progressively increased in the spinal cord of SQSTM1;SOD1 H46R mice compared to SOD1 H46R mice. These results suggest that overexpression of SQSTM1 in SOD1 H46R mice accelerates disease onset by compromising the protein degradation pathways.
Collapse
Affiliation(s)
- Shun Mitsui
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Asako Otomo
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan.,The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, 259-1193, Japan.,Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Masahisa Nozaki
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan.,Department of Anesthesiology, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Suzuka Ono
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Kai Sato
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Ryohei Shirakawa
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Fukuoka, 807-0804, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shinji Hadano
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan. .,The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, 259-1193, Japan. .,Research Center for Brain and Nervous Diseases, Tokai University Graduate School of Medicine, Kanagawa, Isehara, 259-1193, Japan.
| |
Collapse
|
133
|
Bingol B. Autophagy and lysosomal pathways in nervous system disorders. Mol Cell Neurosci 2018; 91:167-208. [PMID: 29729319 DOI: 10.1016/j.mcn.2018.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an evolutionarily conserved pathway for delivering cytoplasmic cargo to lysosomes for degradation. In its classically studied form, autophagy is a stress response induced by starvation to recycle building blocks for essential cellular processes. In addition, autophagy maintains basal cellular homeostasis by degrading endogenous substrates such as cytoplasmic proteins, protein aggregates, damaged organelles, as well as exogenous substrates such as bacteria and viruses. Given their important role in homeostasis, autophagy and lysosomal machinery are genetically linked to multiple human disorders such as chronic inflammatory diseases, cardiomyopathies, cancer, and neurodegenerative diseases. Multiple targets within the autophagy and lysosomal pathways offer therapeutic opportunities to benefit patients with these disorders. Here, I will summarize the mechanisms of autophagy pathways, the evidence supporting a pathogenic role for disturbed autophagy and lysosomal degradation in nervous system disorders, and the therapeutic potential of autophagy modulators in the clinic.
Collapse
Affiliation(s)
- Baris Bingol
- Genentech, Inc., Department of Neuroscience, 1 DNA Way, South San Francisco 94080, United States.
| |
Collapse
|
134
|
Cykowski MD, Powell SZ, Appel JW, Arumanayagam AS, Rivera AL, Appel SH. Phosphorylated TDP-43 (pTDP-43) aggregates in the axial skeletal muscle of patients with sporadic and familial amyotrophic lateral sclerosis. Acta Neuropathol Commun 2018; 6:28. [PMID: 29653597 PMCID: PMC5899326 DOI: 10.1186/s40478-018-0528-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 03/19/2018] [Indexed: 01/08/2023] Open
Abstract
Muscle atrophy with weakness is a core feature of amyotrophic lateral sclerosis (ALS) that has long been attributed to motor neuron loss alone. However, several studies in ALS patients, and more so in animal models, have challenged this assumption with the latter providing direct evidence that muscle can play an active role in the disease. Here, we examined the possible role of cell autonomous pathology in 148 skeletal muscle samples from 57 ALS patients, identifying phosphorylated TAR DNA-binding protein (pTDP-43) inclusions in the muscle fibers of 19 patients (33.3%) and 24 tissue samples (16.2% of specimens). A muscle group-specific difference was identified with pTDP-43 pathology being significantly more common in axial (paraspinous, diaphragm) than appendicular muscles (P = 0.0087). This pathology was not significantly associated with pertinent clinical, genetic (c9ALS) or nervous system pathologic data, suggesting it is not limited to any particular subgroup of ALS patients. Among 25 non-ALS muscle samples, pTDP-43 inclusions were seen only in the autophagy-related disorder inclusion body myositis (IBM) (n = 4), where they were more diffuse than in positive ALS samples (P = 0.007). As in IBM samples, pTDP-43 aggregates in ALS were p62/ sequestosome-1-positive, potentially indicating induction of autophagy. Phospho-TDP-43-positive ALS and IBM samples also showed significant up-regulation of TARDBP and SQSTM1 expression. These findings implicate axial skeletal muscle as an additional site of pTDP-43 pathology in some ALS patients, including sporadic and familial cases, which is deserving of further investigation.
Collapse
Affiliation(s)
- Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA.
- Institute of Academic Medicine (IAM) in the Houston Methodist Research Institute (HMRI), Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA.
| | - Suzanne Z Powell
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Institute of Academic Medicine (IAM) in the Houston Methodist Research Institute (HMRI), Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Houston Methodist Neurological Institute, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Joan W Appel
- Houston Methodist Neurological Institute, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Department of Neurology, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Anithachristy S Arumanayagam
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Andreana L Rivera
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Institute of Academic Medicine (IAM) in the Houston Methodist Research Institute (HMRI), Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Houston Methodist Neurological Institute, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| | - Stanley H Appel
- Institute of Academic Medicine (IAM) in the Houston Methodist Research Institute (HMRI), Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Houston Methodist Neurological Institute, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
- Department of Neurology, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, 77030, USA
| |
Collapse
|
135
|
Martínez-Silva MDL, Imhoff-Manuel RD, Sharma A, Heckman CJ, Shneider NA, Roselli F, Zytnicki D, Manuel M. Hypoexcitability precedes denervation in the large fast-contracting motor units in two unrelated mouse models of ALS. eLife 2018; 7:30955. [PMID: 29580378 PMCID: PMC5922970 DOI: 10.7554/elife.30955] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/14/2018] [Indexed: 12/17/2022] Open
Abstract
Hyperexcitability has been suggested to contribute to motoneuron degeneration in amyotrophic lateral sclerosis (ALS). If this is so, and given that the physiological type of a motor unit determines the relative susceptibility of its motoneuron in ALS, then one would expect the most vulnerable motoneurons to display the strongest hyperexcitability prior to their degeneration, whereas the less vulnerable should display a moderate hyperexcitability, if any. We tested this hypothesis in vivo in two unrelated ALS mouse models by correlating the electrical properties of motoneurons with their physiological types, identified based on their motor unit contractile properties. We found that, far from being hyperexcitable, the most vulnerable motoneurons become unable to fire repetitively despite the fact that their neuromuscular junctions were still functional. Disease markers confirm that this loss of function is an early sign of degeneration. Our results indicate that intrinsic hyperexcitability is unlikely to be the cause of motoneuron degeneration. Amyotrophic lateral sclerosis (ALS), also known as Lou Gehrig's disease, is a fatal disorder of the nervous system. Early symptoms include muscle weakness, unsteadiness and slurred speech. These symptoms arise because the neurons that control muscles – the motoneurons – lose their ability to make the muscles contract. Eventually, the muscles become paralyzed, with more and more muscles affected over time. Most patients die within a few years of diagnosis when the disease destroys the muscles that control breathing. Muscles are made up of muscle fibers. Each motoneuron controls a bundle of muscle fibers, and the motoneuron and its muscle fibers together make up a motor unit. A single muscle contains hundreds of motor units. These consist of several different types, which differ in how many muscle fibers they contain, how fast those muscle fibers can contract, and how fatigable the muscle fibers are. In ALS, motoneurons become detached from their muscle fibers, causing motor units to break down. But what triggers this process? One long-standing idea is that motoneurons begin to respond excessively to commands from the brain and spinal cord. In other words, they become hyperexcitable, which ultimately leads to their death. But some more recent studies of ALS suggest the opposite, namely that motoneurons become less active, or hypoexcitable. To distinguish between these possibilities, Martinez-Silva et al. took advantage of the fact that different types of motor unit break down at different rates in ALS. Large motor units containing fast-contracting muscle fibers break down before smaller motor units. By measuring the activity of motor units in two mouse models of ALS, Martinez-Silva et al. showed that large motoneurons are hypoexcitable. In other words, the motoneurons that are most vulnerable to ALS respond too little to commands from the nervous system, rather than too much. Studies of specific proteins inside the cells confirmed that hypoexcitable motoneurons are further along in the disease process than other motoneurons. Hypoexcitability is thus a key player in the ALS disease process. Developing drugs to target this hypoexcitability may be a promising strategy for the future of this condition.
Collapse
Affiliation(s)
| | - Rebecca D Imhoff-Manuel
- Centre de Neurophysique, Physiologie et Pathologie, CNRS, Université Paris Descartes, Paris, France
| | - Aarti Sharma
- Center for Motor Neuron Biology and Disease, Department of Neurology, Columbia University, New York, United States
| | - C J Heckman
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, United States.,Department of Physical Medicine and Rehabilitation, Northwestern University, Feinberg School of Medicine, Chicago, United States.,Department of Physical Therapy and Human Movement Science, Northwestern University, Feinberg School of Medicine, Chicago, United States
| | - Neil A Shneider
- Center for Motor Neuron Biology and Disease, Department of Neurology, Columbia University, New York, United States
| | | | - Daniel Zytnicki
- Centre de Neurophysique, Physiologie et Pathologie, CNRS, Université Paris Descartes, Paris, France
| | - Marin Manuel
- Centre de Neurophysique, Physiologie et Pathologie, CNRS, Université Paris Descartes, Paris, France.,Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, United States
| |
Collapse
|
136
|
Abstract
In recent years, the role of autophagy in the pathogenesis of most neurodegenerative diseases has transitioned into a limbo of protective or detrimental effects. Genetic evidence indicates that mutations in autophagy-regulatory genes can result in the occurrence of amyotrophic lateral sclerosis (ALS), suggesting a physiological role of the pathway to motoneuron function. However, experimental manipulation of autophagy in ALS models led to conflicting results depending on the intervention strategy and the disease model used. A recent work by the Maniatis group systematically explored the role of cell-specific autophagy in motoneurons at different disease stages, revealing surprising and unexpected findings. Autophagy activity at early stages may contribute to maintaining the structure and function of neuromuscular junctions, whereas at later steps of the disease it has a pathogenic activity possibly involving cell-nonautonomous mechanisms related to glial activation. This new study adds a new layer of complexity in the field, suggesting an intricate interplay between proteostasis alterations, the time-differential function of autophagy in neurons, and muscle innervation in ALS.
Collapse
Affiliation(s)
- Vicente Valenzuela
- a Biomedical Neuroscience Institute (BNI), Faculty of Medicine , University of Chile , Santiago , Chile.,b Center for Geroscience , Brain Health and Metabolism (GERO) , Santiago , Chile.,c Program of Cellular and Molecular Biology , Institute of Biomedical Sciences, University of Chile , Santiago , Chile
| | - Melissa Nassif
- d Center for Integrative Biology (CIB), Faculty of Sciences , Universidad Mayor , Santiago , Chile
| | - Claudio Hetz
- a Biomedical Neuroscience Institute (BNI), Faculty of Medicine , University of Chile , Santiago , Chile.,b Center for Geroscience , Brain Health and Metabolism (GERO) , Santiago , Chile.,c Program of Cellular and Molecular Biology , Institute of Biomedical Sciences, University of Chile , Santiago , Chile.,e Buck Institute for Research on Aging , Novato , CA , USA.,f Department of Immunology and Infectious Diseases , Harvard School of Public Health , Boston MA , USA
| |
Collapse
|
137
|
Abstract
Neurons are particularly dependent on robust quality control pathways to maintain cellular homeostasis and functionality throughout their extended lifetime. Failure to regulate protein and organelle integrity is linked to devastating neurodegenerative diseases. Autophagy is a lysosomal degradation pathway that maintains homeostasis by recycling damaged or aged cellular components. Autophagy has important functions in development of the nervous system, as well as in neuronal function and survival. In fact, defects in autophagy underlie neurodegeneration in mice and humans. Here, we review the compartment-specific dynamics and functions for autophagy in neurons. Emerging evidence suggests novel pathways for the intercellular coordination of quality control pathways between neurons and glia to maintain homeostasis in the brain.
Collapse
Affiliation(s)
- Aditi Kulkarni
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jessica Chen
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra Maday
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
138
|
Crosstalk of Autophagy and the Secretory Pathway and Its Role in Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 337:153-184. [DOI: 10.1016/bs.ircmb.2017.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
139
|
Deng Z, Sheehan P, Chen S, Yue Z. Is amyotrophic lateral sclerosis/frontotemporal dementia an autophagy disease? Mol Neurodegener 2017; 12:90. [PMID: 29282133 PMCID: PMC5746010 DOI: 10.1186/s13024-017-0232-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative disorders that share genetic risk factors and pathological hallmarks. Intriguingly, these shared factors result in a high rate of comorbidity of these diseases in patients. Intracellular protein aggregates are a common pathological hallmark of both diseases. Emerging evidence suggests that impaired RNA processing and disrupted protein homeostasis are two major pathogenic pathways for these diseases. Indeed, recent evidence from genetic and cellular studies of the etiology and pathogenesis of ALS-FTD has suggested that defects in autophagy may underlie various aspects of these diseases. In this review, we discuss the link between genetic mutations, autophagy dysfunction, and the pathogenesis of ALS-FTD. Although dysfunction in a variety of cellular pathways can lead to these diseases, we provide evidence that ALS-FTD is, in many cases, an autophagy disease.
Collapse
Affiliation(s)
- Zhiqiang Deng
- Brain center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, China.,Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China.,Department of Neurology, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Patricia Sheehan
- Department of Neurology, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Shi Chen
- Brain center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, China. .,Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| | - Zhenyu Yue
- Department of Neurology, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, USA.
| |
Collapse
|
140
|
Kulkarni VV, Maday S. Compartment-specific dynamics and functions of autophagy in neurons. Dev Neurobiol 2017; 78:298-310. [PMID: 29197160 DOI: 10.1002/dneu.22562] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/13/2017] [Accepted: 11/16/2017] [Indexed: 02/06/2023]
Abstract
Autophagy is a lysosomal degradation pathway that is critical to maintaining neuronal homeostasis and viability. Autophagy sequesters damaged and aged cellular components from the intracellular environment, and shuttles these diverse macromolecules to lysosomes for destruction. This active surveillance of the quality of the cytoplasm and organelles is essential in neurons to sustain their long-term functionality and viability. Indeed, defective autophagy is linked to neurodevelopmental abnormalities and neurodegeneration in mammals. Here, we review the mechanisms of autophagy in neurons and functional roles for autophagy in neuronal homeostasis. We focus on the compartment-specific dynamics of autophagy in neurons, and how autophagy might perform non-canonical functions critical for neurons. We suggest the existence of multiple populations of autophagosomes with compartment-specific functions important for neural activity and function. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 298-310, 2018.
Collapse
Affiliation(s)
- Vineet Vinay Kulkarni
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Sandra Maday
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| |
Collapse
|
141
|
To be or not to be cell autonomous? Autophagy says both. Essays Biochem 2017; 61:649-661. [PMID: 29233875 DOI: 10.1042/ebc20170025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/17/2022]
Abstract
Although cells are a part of the whole organism, classical dogma emphasizes that individual cells function autonomously. Many physiological and pathological conditions, including cancer, and metabolic and neurodegenerative diseases, have been considered mechanistically as cell-autonomous pathologies, meaning those that damage or defect within a selective population of affected cells suffice to produce disease. It is becoming clear, however, that cells and cellular processes cannot be considered in isolation. Best known for shuttling cytoplasmic content to the lysosome for degradation and repurposing of recycled building blocks such as amino acids, nucleotides, and fatty acids, autophagy serves a housekeeping function in every cell and plays key roles in cell development, immunity, tissue remodeling, and homeostasis with the surrounding environment and the distant organs. In this review, we underscore the importance of taking interactions with the microenvironment into consideration while addressing the cell autonomous and non-autonomous functions of autophagy between cells of the same and different types and in physiological and pathophysiological situations.
Collapse
|