1801
|
Liscic RM, Breljak D. Molecular basis of amyotrophic lateral sclerosis. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:370-2. [PMID: 20655970 DOI: 10.1016/j.pnpbp.2010.07.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/10/2010] [Accepted: 07/15/2010] [Indexed: 10/19/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disorder of motor neuron degeneration with unclear etiology and no effective treatment to date. ALS is, however, increasingly recognized as a multisystem disorder associated with impaired cognition. The overlap between ALS and dementia at clinical, genetic and neuropathologic levels indicates a spectrum of clinical phenotypes that may include features of frontotemporal lobar degeneration (FTLD). Most cases of ALS are sporadic (SALS), but approximately 10% of all ALS cases are familial ALS (FALS). Mutations in the Cu/Zn superoxide dismutase-1 gene (SOD-1) occur in about 20% of FALS cases. Mutations in the TAR DNA-binding protein 43 gene (TARDBP or TDP-43) may occur in 3-4% of FALS cases, and less frequently, in FTLD. Recently, mutations in the fused in sarcoma/translation in liposarcoma gene (FUS/TLS) were identified as causing about 4-5% of FALS, SALS, and FTLD cases, but not SOD-1 ALS cases, indicating a pathogenic role of FUS, together with TDP-43, in possibly all types of ALS, except for SOD-1 linked ALS. TDP-43 and FUS have striking structural and functional similarities, most likely implicating altered RNA processing as a major event in ALS pathogenesis. Thus, TARDBP and FUS/TLS mutations define a novel class of neurodegenerative diseases called TDP-43- and FUS-proteinopathies, in which both misfolded proteins are novel targets for the development of therapeutics in this spectrum of diseases. However, SOD-1 linked ALS may have a pathogenic pathway distinct from other types of ALS.
Collapse
Affiliation(s)
- Rajka M Liscic
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | | |
Collapse
|
1802
|
Kryndushkin D, Wickner RB, Shewmaker F. FUS/TLS forms cytoplasmic aggregates, inhibits cell growth and interacts with TDP-43 in a yeast model of amyotrophic lateral sclerosis. Protein Cell 2011; 2:223-36. [PMID: 21452073 DOI: 10.1007/s13238-011-1525-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 03/06/2011] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease characterized by the premature loss of motor neurons. While the underlying cellular mechanisms of neuron degeneration are unknown, the cytoplasmic aggregation of several proteins is associated with sporadic and familial forms of the disease. Both wild-type and mutant forms of the RNA-binding proteins FUS and TDP-43 accumulate in cytoplasmic inclusions in the neurons of ALS patients. It is not known if these so-called proteinopathies are due to a loss of function or a gain of toxicity resulting from the formation of cytoplasmic aggregates. Here we present a model of FUS toxicity using the yeast Saccharomyces cerevisiae in which toxicity is associated with greater expression and accumulation of FUS in cytoplasmic aggregates. We find that FUS and TDP-43 have a high propensity for co-aggregation, unlike the aggregation patterns of several other aggregation-prone proteins. Moreover, the biophysical properties of FUS aggregates in yeast are distinctly different from many amyloidogenic proteins, suggesting they are not composed of amyloid.
Collapse
Affiliation(s)
- Dmitry Kryndushkin
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
1803
|
Swarup V, Julien JP. ALS pathogenesis: recent insights from genetics and mouse models. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:363-9. [PMID: 20728492 DOI: 10.1016/j.pnpbp.2010.08.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 07/15/2010] [Accepted: 08/11/2010] [Indexed: 12/11/2022]
Abstract
For the vast majority of cases of amyotrophic lateral sclerosis (ALS) the etiology remains unknown. After the discovery of missense mutations in the gene coding for the Cu/Zn superoxide dismutase 1 (SOD1) in subsets of familial ALS, several transgenic mouse lines have been generated with various forms of SOD1 mutants overexpressed at different levels. Studies with these mice yielded complex results with multiple targets of damage in disease including mitochondria, proteasomes, and secretory pathways. Many unexpected discoveries were made. For instance, the toxicity of mutant SOD1 seems unrelated to copper-mediated catalysis but rather to formation of misfolded SOD1 species and aggregates. Transgenic studies revealed a potential role of wtSOD1 in exacerbating mutant SOD1-mediated disease. Another key finding came from chimeric mouse studies and from Cre-lox mediated gene deletion experiments which have highlighted the importance of non-neuronal cells in the disease progression. Involvement of cytoskeletal components in ALS pathogenesis is supported by several mouse models of motor neuron disease with neurofilament abnormalities and with genetic defects in microtubule-based transport. Recently, the generation of new animal models of ALS has been made possible with the discovery of ALS-linked mutations in other genes encoding for alsin, dynactin, senataxin, VAPB, TDP-43 and FUS. Following the discovery of mutations in the TARDBP gene linked to ALS, there have been some reports of transgenic mice with high level overexpression of WT or mutant forms of TDP-43 under strong gene promoters. However, these TDP-43 transgenic mice do not exhibit all pathological features the human ALS disease. Here, we will describe these new TDP-43 transgenic mice and discuss their validity as animal models of human ALS.
Collapse
Affiliation(s)
- Vivek Swarup
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Department of Psychiatry and Neuroscience of Laval University, Quebec, QC, Canada
| | | |
Collapse
|
1804
|
Estes PS, Boehringer A, Zwick R, Tang JE, Grigsby B, Zarnescu DC. Wild-type and A315T mutant TDP-43 exert differential neurotoxicity in a Drosophila model of ALS. Hum Mol Genet 2011; 20:2308-21. [PMID: 21441568 DOI: 10.1093/hmg/ddr124] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The RNA-binding protein TDP-43 has been linked to amyotrophic lateral sclerosis (ALS) both as a causative locus and as a marker of pathology. With several missense mutations being identified within TDP-43, efforts have been directed towards generating animal models of ALS in mouse, zebrafish, Drosophila and worms. Previous loss of function and overexpression studies have shown that alterations in TDP-43 dosage recapitulate hallmark features of ALS pathology, including neuronal loss and locomotor dysfunction. Here we report a direct in vivo comparison between wild-type and A315T mutant TDP-43 overexpression in Drosophila neurons. We found that when expressed at comparable levels, wild-type TDP-43 exerts more severe effects on neuromuscular junction architecture, viability and motor neuron loss compared with the A315T allele. A subset of these differences can be compensated by higher levels of A315T expression, indicating a direct correlation between dosage and neurotoxic phenotypes. Interestingly, larval locomotion is the sole parameter that is more affected by the A315T allele than wild-type TDP-43. RNA interference and genetic interaction experiments indicate that TDP-43 overexpression mimics a loss-of-function phenotype and suggest a dominant-negative effect. Furthermore, we show that neuronal apoptosis does not require the cytoplasmic localization of TDP-43 and that its neurotoxicity is modulated by the proteasome, the HSP70 chaperone and the apoptosis pathway. Taken together, our findings provide novel insights into the phenotypic consequences of the A315T TDP-43 missense mutation and suggest that studies of individual mutations are critical for elucidating the molecular mechanisms of ALS and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Patricia S Estes
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | | | | | | | | | | |
Collapse
|
1805
|
Eschbach J, Dupuis L. Cytoplasmic dynein in neurodegeneration. Pharmacol Ther 2011; 130:348-63. [PMID: 21420428 DOI: 10.1016/j.pharmthera.2011.03.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 03/01/2011] [Indexed: 12/11/2022]
Abstract
Cytoplasmic dynein 1 (later referred to as dynein) is the major molecular motor moving cargoes such as mitochondria, organelles and proteins towards the minus end of microtubules. Dynein is involved in multiple basic cellular functions, such as mitosis, autophagy and structure of endoplasmic reticulum and Golgi, but also in neuron specific functions in particular retrograde axonal transport. Dynein is regulated by a number of protein complexes, notably by dynactin. Several studies have supported indirectly the involvement of dynein in neurodegeneration associated with Alzheimer's disease, Parkinson's disease, Huntington's disease and motor neuron diseases. First, axonal transport disruption represents a common feature occurring in neurodegenerative diseases. Second, a number of dynein-dependent processes, including autophagy or clearance of aggregation-prone proteins, are found defective in most of these diseases. Third, a number of mutant genes in various neurodegenerative diseases are involved in the regulation of dynein transport. This includes notably mutations in the P150Glued subunit of dynactin that are found in Perry syndrome and motor neuron diseases. Interestingly, gene products that are mutant in Huntington's disease, Parkinson's disease, motor neuron disease or spino-cerebellar ataxia are also involved in the regulation of dynein motor activity or of cargo binding. Despite a constellation of indirect evidence, direct links between the motor itself and neurodegeneration are few, and this might be due to the requirement of fully active dynein for development. Here, we critically review the evidence of dynein involvement in different neurodegenerative diseases and discuss potential underlying mechanisms.
Collapse
Affiliation(s)
- Judith Eschbach
- Inserm U692, Laboratoire de Signalisations Moléculaires et Neurodégénérescence, Strasbourg, F-67085, France
| | | |
Collapse
|
1806
|
Kao AW, Eisenhut RJ, Martens LH, Nakamura A, Huang A, Bagley JA, Zhou P, de Luis A, Neukomm LJ, Cabello J, Farese RV, Kenyon C. A neurodegenerative disease mutation that accelerates the clearance of apoptotic cells. Proc Natl Acad Sci U S A 2011; 108:4441-6. [PMID: 21368173 PMCID: PMC3060230 DOI: 10.1073/pnas.1100650108] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Frontotemporal lobar degeneration is a progressive neurodegenerative syndrome that is the second most common cause of early-onset dementia. Mutations in the progranulin gene are a major cause of familial frontotemporal lobar degeneration [Baker M, et al. (2006) Nature 442:916-919 and Cruts M, et al. (2006) Nature 442:920-924]. Although progranulin is involved in wound healing, inflammation, and tumor growth, its role in the nervous system and the mechanism by which insufficient levels result in neurodegeneration are poorly understood [Eriksen and Mackenzie (2008) J Neurochem 104:287-297]. We have characterized the normal function of progranulin in the nematode Caenorhabditis elegans. We found that mutants lacking pgrn-1 appear grossly normal, but exhibit fewer apoptotic cell corpses during development. This reduction in corpse number is not caused by reduced apoptosis, but instead by more rapid clearance of dying cells. Likewise, we found that macrophages cultured from progranulin KO mice displayed enhanced rates of apoptotic-cell phagocytosis. Although most neurodegenerative diseases are thought to be caused by the toxic effects of aggregated proteins, our findings suggest that susceptibility to neurodegeneration may be increased by a change in the kinetics of programmed cell death. We propose that cells that might otherwise recover from damage or injury are destroyed in progranulin mutants, which in turn facilitates disease progression.
Collapse
Affiliation(s)
- Aimee W. Kao
- Departments of Biochemistry and Biophysics and
- Neurology, University of California, San Francisco, CA 94158
| | | | | | | | - Anne Huang
- Departments of Biochemistry and Biophysics and
| | | | - Ping Zhou
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
| | - Alberto de Luis
- Center for Biomedical Research of La Rioja, 26006 Logrono, Spain
| | - Lukas J. Neukomm
- Institute of Molecular Life Sciences, University of Zurich, CH8057 Zurich, Switzerland; and
| | - Juan Cabello
- Center for Biomedical Research of La Rioja, 26006 Logrono, Spain
| | - Robert V. Farese
- Departments of Biochemistry and Biophysics and
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158
- Department of Internal Medicine, University of California, San Francisco, CA 94143
| | | |
Collapse
|
1807
|
Abstract
Amyotrophic lateral sclerosis (ALS) is an idiopathic, fatal neurodegenerative disease of the human motor system. In this Seminar, we summarise current concepts about the origin of the disease, what predisposes patients to develop the disorder, and discuss why all cases of ALS are not the same. In the 150 years since Charcot originally described ALS, painfully slow progress has been made towards answering these questions. We focus on what is known about ALS and where research is heading-from the small steps of extending longevity, improving therapies, undertaking clinical trials, and compiling population registries to the overarching goals of establishing the measures that guard against onset and finding the triggers for this neurodegenerative disorder.
Collapse
Affiliation(s)
- Matthew C Kiernan
- Neuroscience Research Australia and Prince of Wales Clinical School, University of New South Wales, Sydney, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
1808
|
Lunn JS, Sakowski SA, Federici T, Glass JD, Boulis NM, Feldman EL. Stem cell technology for the study and treatment of motor neuron diseases. Regen Med 2011; 6:201-13. [PMID: 21391854 PMCID: PMC3154698 DOI: 10.2217/rme.11.6] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis and spinal muscular atrophy are devastating neurodegenerative diseases that lead to the specific loss of motor neurons. Recently, stem cell technologies have been developed for the investigation and treatment of both diseases. Here we discuss the different stem cells currently being studied for mechanistic discovery and therapeutic development, including embryonic, adult and induced pluripotent stem cells. We also present supporting evidence for the utilization of stem cell technology in the treatment of amyotrophic lateral sclerosis and spinal muscular atrophy, and describe key issues that must be considered for the transition of stem cell therapies for motor neuron diseases from bench to bedside. Finally, we discuss the first-in-human Phase I trial currently underway examining the safety and feasibility of intraspinal stem cell injections in amyotrophic lateral sclerosis patients as a foundation for translating stem cell therapies for various neurological diseases.
Collapse
Affiliation(s)
- J Simon Lunn
- University of Michigan Department of Neurology, 109 Zina Pitcher Place, 5017 BSRB, Ann Arbor, MI 48109, USA
| | - Stacey A Sakowski
- University of Michigan Department of Neurology, 109 Zina Pitcher Place, 5017 BSRB, Ann Arbor, MI 48109, USA
| | - Thais Federici
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
| | | | | | - Eva L Feldman
- University of Michigan Department of Neurology, 109 Zina Pitcher Place, 5017 BSRB, Ann Arbor, MI 48109, USA
| |
Collapse
|
1809
|
Chiò A, Calvo A, Moglia C, Ossola I, Brunetti M, Sbaiz L, Lai SL, Abramzon Y, Traynor BJ, Restagno G. A de novo missense mutation of the FUS gene in a "true" sporadic ALS case. Neurobiol Aging 2011; 32:553.e23-6. [PMID: 20598774 PMCID: PMC2972379 DOI: 10.1016/j.neurobiolaging.2010.05.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 04/30/2010] [Accepted: 05/16/2010] [Indexed: 12/12/2022]
Abstract
Mutations in the Cu/Zn superoxide dismutase (SOD1), transactive response (TAR)-DNA binding protein (TARDBP) and fused in sarcoma (FUS) genes account for approximately 1 third of familial amyotrophic lateral sclerosis (ALS) cases. Mutations in these genes have been found in 1% to 2% of apparently sporadic cases. We present the first case of an ALS patient carrying a de novo missense mutation of the FUS gene (c.1561C>T, p.R521C). This report highlights the importance of screening ALS patients, both familial and sporadic, for FUS mutations and also suggests that de novo mutations is a relevant mechanism underlying sporadic neurodegenerative disease.
Collapse
Affiliation(s)
- Adriano Chiò
- ALS Center, Department of Neuroscience, University of Torino, Torino, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1810
|
Huang C, Zhou H, Tong J, Chen H, Liu YJ, Wang D, Wei X, Xia XG. FUS transgenic rats develop the phenotypes of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. PLoS Genet 2011; 7:e1002011. [PMID: 21408206 PMCID: PMC3048370 DOI: 10.1371/journal.pgen.1002011] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 01/03/2011] [Indexed: 12/12/2022] Open
Abstract
Fused in Sarcoma (FUS) proteinopathy is a feature of frontotemporal lobar dementia (FTLD), and mutation of the fus gene segregates with FTLD and amyotrophic lateral sclerosis (ALS). To study the consequences of mutation in the fus gene, we created transgenic rats expressing the human fus gene with or without mutation. Overexpression of a mutant (R521C substitution), but not normal, human FUS induced progressive paralysis resembling ALS. Mutant FUS transgenic rats developed progressive paralysis secondary to degeneration of motor axons and displayed a substantial loss of neurons in the cortex and hippocampus. This neuronal loss was accompanied by ubiquitin aggregation and glial reaction. While transgenic rats that overexpressed the wild-type human FUS were asymptomatic at young ages, they showed a deficit in spatial learning and memory and a significant loss of cortical and hippocampal neurons at advanced ages. These results suggest that mutant FUS is more toxic to neurons than normal FUS and that increased expression of normal FUS is sufficient to induce neuron death. Our FUS transgenic rats reproduced some phenotypes of ALS and FTLD and will provide a useful model for mechanistic studies of FUS-related diseases.
Collapse
Affiliation(s)
- Cao Huang
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Hongxia Zhou
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Jianbin Tong
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Han Chen
- Center for Biotechnology, University of Nebraska–Lincoln, Lincoln, Nebraska, United States of America
| | - Yong-Jian Liu
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Dian Wang
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Xiaotao Wei
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Xu-Gang Xia
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
1811
|
Lai SL, Abramzon Y, Schymick JC, Stephan DA, Dunckley T, Dillman A, Cookson M, Calvo A, Battistini S, Giannini F, Caponnetto C, Mancardi GL, Spataro R, Monsurro MR, Tedeschi G, Marinou K, Sabatelli M, Conte A, Mandrioli J, Sola P, Salvi F, Bartolomei I, Lombardo F, the ITALSGEN consortium, Mora G, Restagno G, Chiò A, Traynor BJ. FUS mutations in sporadic amyotrophic lateral sclerosis. Neurobiol Aging 2011; 32:550.e1-4. [PMID: 20138404 PMCID: PMC2891336 DOI: 10.1016/j.neurobiolaging.2009.12.020] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 12/08/2009] [Accepted: 12/24/2009] [Indexed: 12/13/2022]
Abstract
Mutations in the FUS gene have recently been described as a cause of familial amyotrophic lateral sclerosis (ALS), but their role in the pathogenesis of sporadic ALS is unclear. We undertook mutational screening of all coding exons of FUS in 228 sporadic ALS cases, and, as previous reports suggest that exon 15 represents a mutational hotspot, we sequenced this exon in an additional 1295 sporadic cases. Six variants in six different cases were found, indicating that FUS mutations can underlie apparently sporadic ALS, but account for less than 1% of this form of disease.
Collapse
Affiliation(s)
- Shiao-Lin Lai
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
- Department of Molecular Neuroscience and Reta Lila Weston Institute of Neurological Studies, Institute of Neurology, Queen Square, London WC1 3BG, UK
- Chang Gung University College of Medicine, Taiwan
| | - Yevgeniya Abramzon
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Jennifer C. Schymick
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
- Department of Clinical Medicine, University of Oxford, Oxford OX1 3QX, UK
| | - Dietrich A. Stephan
- Navigenics, Foster City, CA 94404, USA
- IGNITE Institute for Individualized Health, Fairfax, VA 22042, USA
| | - Travis Dunckley
- Neurogenomics Division, Translational Genomics Institute (TGEN), Phoenix, AZ 85004, USA
| | - Allissa Dillman
- Cell Biology and Gene Expression Unit, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Mark Cookson
- Cell Biology and Gene Expression Unit, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| | - Andrea Calvo
- Department of Neuroscience, University of Turin, Via Cherasco 15, 10126 Turin, Italy
| | - Stefania Battistini
- Department of Neuroscience, Neurology Section, University of Siena, Siena, Italy
| | - Fabio Giannini
- Department of Neuroscience, Neurology Section, University of Siena, Siena, Italy
| | - Claudia Caponnetto
- Department of Neuroscience, Ophthalmology and Genetics, University of Genoa, Genoa, Italy
| | | | - Rossella Spataro
- Department of Clinical Neurosciences, University of Palermo, Italy
| | - Maria Rosaria Monsurro
- Department of Neuroscience, Second University of Naples, Naples, Italy
- Capodimonte-Hermitage Institute, Naples, Italy
| | - Gioacchino Tedeschi
- Department of Neuroscience, Second University of Naples, Naples, Italy
- Capodimonte-Hermitage Institute, Naples, Italy
| | | | - Mario Sabatelli
- Neurological Institute, Catholic University and I.CO.M.M. Association for ALS Research, Rome, Italy
| | - Amelia Conte
- Neurological Institute, Catholic University and I.CO.M.M. Association for ALS Research, Rome, Italy
| | - Jessica Mandrioli
- Department of Neuroscience, S. Agostino- Estense Hospital and University of Modena, Modena, Italy
| | - Patrizia Sola
- Department of Neuroscience, S. Agostino- Estense Hospital and University of Modena, Modena, Italy
| | - Fabrizio Salvi
- Department of Neurology, Center for Diagnosis and Cure of Rare Diseases, Bellaria Hospital, Bologna, Italy
| | - Ilaria Bartolomei
- Department of Neurology, Center for Diagnosis and Cure of Rare Diseases, Bellaria Hospital, Bologna, Italy
| | - Federica Lombardo
- Molecular Genetics Unit, Department of Clinical Pathology, A.S.O. O.I.R.M.-S.Anna, Turin, Italy
| | | | | | - Gabriella Restagno
- Molecular Genetics Unit, Department of Clinical Pathology, A.S.O. O.I.R.M.-S.Anna, Turin, Italy
| | - Adriano Chiò
- Department of Neuroscience, University of Turin, Via Cherasco 15, 10126 Turin, Italy
| | - Bryan J. Traynor
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA
| |
Collapse
Collaborators
Claudia Ricci, Cristina Moglia, Stefania Cammarosano, Roberto Mutani, Maura Brunetti, Irene Ossola, Laura Papetti, Marco Luigetti, Vincenzo La Bella, Piera Paladino, Gabriele Siciliano,
Collapse
|
1812
|
Cohen NR, Hammans SR, Macpherson J, Nicoll JAR. New neuropathological findings in Unverricht-Lundborg disease: neuronal intranuclear and cytoplasmic inclusions. Acta Neuropathol 2011; 121:421-7. [PMID: 20721566 DOI: 10.1007/s00401-010-0738-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 08/09/2010] [Accepted: 08/11/2010] [Indexed: 11/25/2022]
Abstract
Unverricht-Lundborg disease (EPM1A), also known as Baltic myoclonus, is the most common form of progressive myoclonic epilepsy. It is inherited as an autosomal recessive trait, due to mutations in the Cystatin-B gene promoter region. Although there is much work on rodent models of this disease, there is very little published neuropathology in patients with EPM1A. Here, we present the neuropathology of a patient with genetically confirmed EPM1A, who died at the age of 76. There was atrophy and gliosis affecting predominantly the cerebellum, frontotemporal cortex, hippocampus and thalamus. We have identified neuronal cytoplasmic inclusions containing the lysosomal proteins, Cathepsin-B and CD68. These inclusions also showed immunopositivity to both TDP-43 and FUS, in some cases associated with an absence of normal neuronal nuclear TDP-43 staining. There were also occasional ubiquitinylated neuronal intranuclear inclusions, some of which were FUS immunopositive. This finding is consistent with neurodegeneration in EPM1A as at least a partial consequence of lysosomal damage to neurons, which have reduced Cystatin-B-related neuroprotection. It also reveals a genetically defined neurodegenerative disease with both FUS and TDP-43 related pathology.
Collapse
Affiliation(s)
- Nicola R Cohen
- Cellular Pathology, Southampton General Hospital, Southampton, UK.
| | | | | | | |
Collapse
|
1813
|
Casula M, Iyer AM, Spliet WGM, Anink JJ, Steentjes K, Sta M, Troost D, Aronica E. Toll-like receptor signaling in amyotrophic lateral sclerosis spinal cord tissue. Neuroscience 2011; 179:233-43. [PMID: 21303685 DOI: 10.1016/j.neuroscience.2011.02.001] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 01/31/2011] [Accepted: 02/01/2011] [Indexed: 02/07/2023]
Abstract
Increasing evidence indicates that inflammatory responses could play a critical role in the pathogenesis of motor neuron injury in amyotrophic lateral sclerosis (ALS). Recent findings have underlined the role of Toll-like receptors (TLRs) and the receptor for advanced glycation endproducts (RAGE) in the regulation of both innate and adaptive immunity in different pathologies associated with neuroinflammation. In the present study we investigated the expression and cellular distribution of TLR2, TLR4, RAGE and their endogenous ligand high mobility group box 1 (HMGB1) in the spinal cord of control (n=6) and sporadic ALS (n=12) patients. The immunohistochemical analysis of TLR2, TLR4 and RAGE showed increased expression in reactive glial cells in both gray (ventral horn) and white matter of ALS spinal cord. TLR2 was predominantly detected in cells of the microglia/macrophage lineage, whereas the TLR4 and RAGE was strongly expressed in astrocytes. Real-time quantitative PCR analysis confirmed the increased expression of both TLR2 and TLR4 and HMGB1 mRNA level in ALS patients. In ALS spinal cord, HMGB1 signal is increased in the cytoplasm of reactive glia, indicating a possible release of this molecule from glial cells. Our findings show increased expression of TLR2, TLR4, RAGE and HMGB1 in reactive glia in human ALS spinal cord, suggesting activation of the TLR/RAGE signaling pathways. The activation of these pathways may contribute to the progression of inflammation, resulting in motor neuron injury. In this context, future studies, using animal models, will be important to achieve a better understanding of these signaling pathways in ALS in view of the development of new therapeutic strategies.
Collapse
Affiliation(s)
- M Casula
- Department of (Neuro) Pathology, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
1814
|
Wilde IB, Brack M, Winget JM, Mayor T. Proteomic characterization of aggregating proteins after the inhibition of the ubiquitin proteasome system. J Proteome Res 2011; 10:1062-72. [PMID: 21204586 DOI: 10.1021/pr1008543] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Protein aggregation, which is associated with the impairment of the ubiquitin proteasome system, is a hallmark of many neurodegenerative diseases. To better understand the contribution of proteasome inhibition in aggregation, we analyzed which proteins may potentially localize in chemically induced aggregates in human neuroblastoma tissue culture cells. We enriched for proteins in high-density structures by using a sucrose gradient in combination with stable isotope labeling with amino acids in cell culture (SILAC). The quantitative analysis allowed us to distinguish which proteins were specifically affected by the proteasome inhibition. We identified 642 potentially aggregating proteins, including the p62/sequestosome 1 and NBR1 ubiquitin-binding proteins involved in aggregation. We also identified the ubiquitin-associated protein 2 like (UBAP2L). We verified that it cofractionated with ubiquitin in the high-density fraction and that it was colocalized in the ubiquitin-containing aggregates after proteasome inhibition. In addition, we identified several chaperone proteins and used data from protein interaction networks to show that they potentially interact with distinct subgroups of proteins within the aggregating structures. Several other proteins associated with neurodegenerative diseases, like UCHL1, were identified, further underlining the potential of our analysis to better understand the aggregation process and proteotoxic stress caused by proteasome inhibition.
Collapse
Affiliation(s)
- Inga B Wilde
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia, 2125 East Mall, Vancouver, British Columbia, V6T 1Z4, Canada
| | | | | | | |
Collapse
|
1815
|
Lee T, Li YR, Ingre C, Weber M, Grehl T, Gredal O, de Carvalho M, Meyer T, Tysnes OB, Auburger G, Gispert S, Bonini NM, Andersen PM, Gitler AD. Ataxin-2 intermediate-length polyglutamine expansions in European ALS patients. Hum Mol Genet 2011; 20:1697-700. [PMID: 21292779 DOI: 10.1093/hmg/ddr045] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disease primarily affecting motor neurons. We recently identified intermediate-length polyglutamine (polyQ) expansions (27-33 Qs) in ataxin 2 as a genetic risk factor for sporadic ALS in North American ALS patients. To extend these findings, we assessed the ataxin 2 polyQ repeat length in 1294 European ALS patients and 679 matched healthy controls. We observed a significant association between polyQ expansions and ALS (>30 Qs; P= 6.2 × 10(-3)). Thus, intermediate-length ataxin 2 polyQ repeat expansions are associated with increased risk for ALS also in the European cohort. The specific polyQ length cutoff, however, appears to vary between different populations, with longer repeat lengths showing a clear association. Our findings support the hypothesis that ataxin 2 plays an important role in predisposing to ALS and that polyQ expansions in ataxin 2 are a significant risk factor for the disease.
Collapse
Affiliation(s)
- Teresa Lee
- Department of Cell and Developmental Biology, the University of Pennsylvania School of Medicine, 1109 BRB II/III, 421 Curie Blvd., Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1816
|
Nizzardo M, Nardini M, Ronchi D, Salani S, Donadoni C, Fortunato F, Colciago G, Falcone M, Simone C, Riboldi G, Govoni A, Bresolin N, Comi GP, Corti S. Beta-lactam antibiotic offers neuroprotection in a spinal muscular atrophy model by multiple mechanisms. Exp Neurol 2011; 229:214-25. [PMID: 21295027 DOI: 10.1016/j.expneurol.2011.01.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 01/21/2011] [Accepted: 01/24/2011] [Indexed: 10/18/2022]
Abstract
Spinal muscular atrophy (SMA) is a devastating genetic motoneuron disease leading to infant death. No effective therapy is currently available. It has been suggested that β-lactam antibiotics such as ceftriaxone may offer neuroprotection in motoneuron diseases. Here, we investigate the therapeutic effect of ceftriaxone in a murine model of SMA. Treated animals present a modest, but significant ameliorated neuromuscular phenotype and increased survival, which correlate with protection of neuromuscular units. Whole gene expression profiling in treated mice demonstrates modifications in several genes including those involved in RNA metabolism toward wild-type. The neuroprotective effect seems to be mediated by multiple mechanisms that encompass the increase of the glutamate transporter Glt1, the transcription factor Nrf2, as well as SMN protein. This study provides the first evidence of a potential positive effect of this class of molecules in SMA. Further investigation of analogs with increased and more specific therapeutic effects warrants the development of useful therapies for SMA.
Collapse
Affiliation(s)
- Monica Nizzardo
- Dino Ferrari Centre, Department of Neurological Sciences, University of Milan, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1817
|
Sau D, Rusmini P, Crippa V, Onesto E, Bolzoni E, Ratti A, Poletti A. Dysregulation of axonal transport and motorneuron diseases. Biol Cell 2011; 103:87-107. [PMID: 21250942 DOI: 10.1042/bc20100093] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
MNDs (motorneuron diseases) are neurodegenerative disorders in which motorneurons located in the motor cortex, in the brainstem and in the spinal cord are affected. These diseases in their inherited or sporadic forms are mainly characterized by motor dysfunctions, occasionally associated with cognitive and behavioural alterations. Although these diseases show high variability in onset, progression and clinical symptoms, they share common pathological features, and motorneuronal loss invariably leads to muscle weakness and atrophy. One of the most relevant aspect of these disorders is the occurrence of defects in axonal transport, which have been postulated to be either a direct cause, or a consequence, of motorneuron degeneration. In fact, due to their peculiar morphology and high energetic metabolism, motorneurons deeply rely on efficient axonal transport processes. Dysfunction of axonal transport is known to adversely affect motorneuronal metabolism, inducing progressive degeneration and cell death. In this regard, the understanding of the fine mechanisms at the basis of the axonal transport process and of their possible alterations may help shed light on MND pathological processes. In the present review, we will summarize what is currently known about the alterations of axonal transport found to be either causative or a consequence of MNDs.
Collapse
Affiliation(s)
- Daniela Sau
- Dipartimento di Endocrinologia, Fisiopatologia e Biologia Applicata, and Centre of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
1818
|
Mackenzie IRA, Munoz DG, Kusaka H, Yokota O, Ishihara K, Roeber S, Kretzschmar HA, Cairns NJ, Neumann M. Distinct pathological subtypes of FTLD-FUS. Acta Neuropathol 2011; 121:207-18. [PMID: 21052700 DOI: 10.1007/s00401-010-0764-0] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/18/2010] [Accepted: 10/19/2010] [Indexed: 11/27/2022]
Abstract
Most cases of frontotemporal lobar degeneration (FTLD) are characterized by abnormal intracellular accumulation of either tau or TDP-43 protein. However, in ~10% of cases, composed of a heterogenous collection of uncommon disorders, the molecular basis remains to be uncertain. We recently discovered that the pathological changes in several tau/TDP-43-negative FTLD subtypes are immunoreactive (ir) for the fused in sarcoma (FUS) protein. In this study, we directly compared the pattern of FUS-ir pathology in cases of atypical FTLD-U (aFTLD-U, N = 10), neuronal intermediate filament inclusion disease (NIFID, N = 5) and basophilic inclusion body disease (BIBD, N = 8), to determine whether these are discrete entities or represent a pathological continuum. All cases had FUS-ir pathology in the cerebral neocortex, hippocampus and a similar wide range of subcortical regions. Although there was significant overlap, each group showed specific differences that distinguished them from the others. Cases of aFTLD-U consistently had less pathology in subcortical regions. In addition, the neuronal inclusions in aFTLD-U usually had a uniform, round shape, whereas NIFID and BIBD were characterized by a variety of inclusion morphologies. In all cases of aFTLD-U and NIFID, vermiform neuronal intranuclear inclusions (NII) were readily identified in the hippocampus and neocortex. In contrast, only two cases of BIBD had very rare NII in a single subcortical region. These findings support aFTLD-U, NIFID and BIBD as representing closely related, but distinct entities that share a common molecular pathogenesis. Although cases with overlapping pathology may exist, we recommend retaining the terms aFTLD-U, NIFID and BIBD for specific FTLD-FUS subtypes.
Collapse
Affiliation(s)
- Ian R A Mackenzie
- Department of Pathology, Vancouver General Hospital, University of British Columbia, BC, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
1819
|
Armstrong RA, Gearing M, Bigio EH, Cruz-Sanchez FF, Duyckaerts C, Mackenzie IRA, Perry RH, Skullerud K, Yokoo H, Cairns NJ. The spectrum and severity of FUS-immunoreactive inclusions in the frontal and temporal lobes of ten cases of neuronal intermediate filament inclusion disease. Acta Neuropathol 2011; 121:219-28. [PMID: 20886222 PMCID: PMC3035044 DOI: 10.1007/s00401-010-0753-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 09/24/2010] [Accepted: 09/25/2010] [Indexed: 12/13/2022]
Abstract
Neuronal intermediate filament inclusion disease (NIFID), a rare form of frontotemporal lobar degeneration (FTLD), is characterized neuropathologically by focal atrophy of the frontal and temporal lobes, neuronal loss, gliosis, and neuronal cytoplasmic inclusions (NCI) containing epitopes of ubiquitin and neuronal intermediate filament proteins. Recently, the 'fused in sarcoma' (FUS) protein (encoded by the FUS gene) has been shown to be a component of the inclusions of familial amyotrophic lateral sclerosis with FUS mutation, NIFID, basophilic inclusion body disease, and atypical FTLD with ubiquitin-immunoreactive inclusions (aFTLD-U). To further characterize FUS proteinopathy in NIFID, and to determine whether the pathology revealed by FUS immunohistochemistry (IHC) is more extensive than α-internexin, we have undertaken a quantitative assessment of ten clinically and neuropathologically well-characterized cases using FUS IHC. The densities of NCI were greatest in the dentate gyrus (DG) and in sectors CA1/2 of the hippocampus. Anti-FUS antibodies also labeled glial inclusions (GI), neuronal intranuclear inclusions (NII), and dystrophic neurites (DN). Vacuolation was extensive across upper and lower cortical layers. Significantly greater densities of abnormally enlarged neurons and glial cell nuclei were present in the lower compared with the upper cortical laminae. FUS IHC revealed significantly greater numbers of NCI in all brain regions especially the DG. Our data suggest: (1) significant densities of FUS-immunoreactive NCI in NIFID especially in the DG and CA1/2; (2) infrequent FUS-immunoreactive GI, NII, and DN; (3) widely distributed vacuolation across the cortex, and (4) significantly more NCI revealed by FUS than α-internexin IHC.
Collapse
|
1820
|
Fushimi K, Long C, Jayaram N, Chen X, Li L, Wu JY. Expression of human FUS/TLS in yeast leads to protein aggregation and cytotoxicity, recapitulating key features of FUS proteinopathy. Protein Cell 2011; 2:141-9. [PMID: 21327870 PMCID: PMC3093303 DOI: 10.1007/s13238-011-1014-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 01/28/2011] [Indexed: 10/18/2022] Open
Abstract
Mutations in the fused in sarcoma/translocated in liposarcoma (FUS/TLS) gene have been associated with amyotrophic lateral sclerosis (ALS). FUS-positive neuropathology is reported in a range of neurodegenerative diseases, including ALS and fronto-temporal lobar degeneration with ubiquitin-positive pathology (FTLDU). To examine protein aggregation and cytotoxicity, we expressed human FUS protein in yeast. Expression of either wild type or ALS-associated R524S or P525L mutant FUS in yeast cells led to formation of aggregates and cytotoxicity, with the two ALS mutants showing increased cytotoxicity. Therefore, yeast cells expressing human FUS protein recapitulate key features of FUS-positive neurodegenerative diseases. Interestingly, a significant fraction of FUS expressing yeast cells stained by propidium iodide were without detectable protein aggregates, suggesting that membrane impairment and cellular damage caused by FUS expression may occur before protein aggregates become microscopically detectable and that aggregate formation might protect cells from FUS-mediated cytotoxicity. The N-terminus of FUS, containing the QGSY and G rich regions, is sufficient for the formation of aggregates but not cytotoxicity. The C-terminal domain, which contains a cluster of mutations, did not show aggregation or cytotoxicity. Similar to TDP-43 when expressed in yeast, FUS protein has the intrinsic property of forming aggregates in the absence of other human proteins. On the other hand, the aggregates formed by FUS are thioflavin T-positive and resistant to 0.5% sarkosyl, unlike TDP-43 when expressed in yeast cells. Furthermore, TDP-43 and FUS display distinct domain requirements in aggregate formation and cytotoxicity.
Collapse
Affiliation(s)
- Kazuo Fushimi
- Department of Neurology, Center for Genetic Medicine, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL 60611 USA
| | - Charles Long
- Department of Neurology, Center for Genetic Medicine, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL 60611 USA
| | - Neha Jayaram
- Department of Neurology, Center for Genetic Medicine, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL 60611 USA
| | - Xiaoping Chen
- Department of Neurology, Center for Genetic Medicine, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL 60611 USA
| | - Liming Li
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL 60611 USA
| | - Jane Y. Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL 60611 USA
| |
Collapse
|
1821
|
Webster YW, Dow ER, Koehler J, Gudivada RC, Palakal MJ. Leveraging health social networking communities in translational research. J Biomed Inform 2011; 44:536-44. [PMID: 21284958 DOI: 10.1016/j.jbi.2011.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 12/17/2010] [Accepted: 01/25/2011] [Indexed: 01/19/2023]
Abstract
Health social networking communities are emerging resources for translational research. We have designed and implemented a framework called HyGen, which combines Semantic Web technologies, graph algorithms and user profiling to discover and prioritize novel associations across disciplines. This manuscript focuses on the key strategies developed to overcome the challenges in handling patient-generated content in Health social networking communities. Heuristic and quantitative evaluations were carried out in colorectal cancer. The results demonstrate the potential of our approach to bridge silos and to identify hidden links among clinical observations, drugs, genes and diseases. In Amyotrophic Lateral Sclerosis case studies, HyGen has identified 15 of the 20 published disease genes. Additionally, HyGen has highlighted new candidates for future investigations, as well as a scientifically meaningful connection between riluzole and alcohol abuse.
Collapse
Affiliation(s)
- Yue W Webster
- School of Informatics, Indiana University Purdue University, IN, USA.
| | | | | | | | | |
Collapse
|
1822
|
Matus S, Glimcher LH, Hetz C. Protein folding stress in neurodegenerative diseases: a glimpse into the ER. Curr Opin Cell Biol 2011; 23:239-52. [PMID: 21288706 DOI: 10.1016/j.ceb.2011.01.003] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 01/05/2011] [Accepted: 01/11/2011] [Indexed: 01/19/2023]
Abstract
Several neurodegenerative diseases share common neuropathology, primarily featuring the presence in the brain of abnormal protein inclusions containing specific misfolded proteins. Recent evidence indicates that alteration in organelle function is a common pathological feature of protein misfolding disorders, highlighting perturbations in the homeostasis of the endoplasmic reticulum (ER). Signs of ER stress have been detected in most experimental models of neurological disorders and more recently in brain samples from human patients with neurodegenerative disease. To cope with ER stress, cells activate an integrated signaling response termed the unfolded protein response (UPR), which aims to reestablish homeostasis in part through regulation of genes involved in protein folding, quality control and degradation pathways. Here we discuss the particular mechanisms currently proposed to be involved in the generation of protein folding stress in different neurodegenerative conditions and speculate about possible therapeutic interventions.
Collapse
Affiliation(s)
- Soledad Matus
- Center for Molecular Studies of Cell, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | |
Collapse
|
1823
|
Sleegers K, Cruts M, Van Broeckhoven C. Molecular pathways of frontotemporal lobar degeneration. Annu Rev Neurosci 2011; 33:71-88. [PMID: 20415586 DOI: 10.1146/annurev-neuro-060909-153144] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) is a neurodegenerative condition that predominantly affects behavior, social awareness, and language. It is characterized by extensive heterogeneity at the clinical, pathological, and genetic levels. Recognition of these levels of heterogeneity is important for proper disease management. The identification of progranulin and TDP-43 as key proteins in a significant proportion of FTLD patients has provided the impetus for a wealth of studies probing their role in neurodegeneration. This review highlights the most recent developments and future directions in this field and puts them in perspective of the novel insights into the neurodegenerative process, which have been gained from related disorders, e.g., the role of FUS in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Kristel Sleegers
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Universiteitsplein 1, B-2610 Antwerpen, Belgium.
| | | | | |
Collapse
|
1824
|
Association between divalent metal transport 1 encoding gene (SLC11A2) and disease duration in amyotrophic lateral sclerosis. J Neurol Sci 2011; 303:124-7. [PMID: 21276595 DOI: 10.1016/j.jns.2010.12.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 12/17/2010] [Indexed: 12/12/2022]
Abstract
BACKGROUND Dysregulation of iron homeostasis is one possible pathophysiological mechanism involved in motor neuron degeneration in amyotrophic lateral sclerosis (ALS). SLC11A2 gene encodes the divalent metal transport 1 (DMT1) mediating iron transport in cerebral endosomal compartments. The objective of the study was to analyze DMT1 as a possible risk or modulating factor in sporadic ALS (SALS). METHODS We performed a case-control association study on an intronic polymorphism (rs407135) previously analyzed in another neurodegenerative disease, Alzheimer's disease. This polymorphism was studied by DNA sequencing in 579 French patients with SALS and 517 healthy matched individuals. The clinical characteristics of patients were analyzed in relation to their genotypes. RESULTS We observed that the C allele of rs407135 in SLC11A2 was associated with a shorter disease duration in SALS patients with onset in the legs [Hazard ratio: 1.5 [1.1-2.1] (p=0.02)]. These results are in line with previous observations suggesting that bulbar and spinal motor neurons have different metabolic regulation and gene expression profiles. CONCLUSIONS Our findings support an implication for iron metabolism in ALS and suggest that the genotype of the SLC11A2 gene could modulate the duration of the disease in French SALS patients.
Collapse
|
1825
|
Belzil VV, Daoud H, St-Onge J, Desjarlais A, Bouchard JP, Dupre N, Lacomblez L, Salachas F, Pradat PF, Meininger V, Camu W, Dion PA, Rouleau GA. Identification of novel FUS mutations in sporadic cases of amyotrophic lateral sclerosis. ACTA ACUST UNITED AC 2011; 12:113-7. [PMID: 21261515 DOI: 10.3109/17482968.2010.536840] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mutations in the FUS gene have been recently associated with amyotrophic lateral sclerosis (ALS). While most of the variants have been identified in patients with a family history of the disease, a few mutations were also found in sporadic patients. Considering this, we wanted to evaluate the frequency of mutations in the coding region of the FUS gene in a sporadic ALS (SALS) cohort compared to a control population. We tested 475 SALS cases of European origin and 475 matched controls for coding variations in the 15 exons of the FUS gene. Rare novel variants were identified in a total of five SALS patients: one missense, one deletion, one frameshift, and one nonsense substitution. Two of the four variants are located in the carboxy terminal of the protein where the previously reported variants were mostly clustered. In conclusion, FUS gene mutations are rare in SALS, with four new FUS variants identified in five different SALS cases. These findings will help evaluate the proportion of FUS variations in the SALS population, and to better understand its contributing role to ALS pathology.
Collapse
Affiliation(s)
- Veronique V Belzil
- Centre of Excellence in Neuromics of Université de Montréal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1826
|
Nguyen CD, Mansfield RE, Leung W, Vaz PM, Loughlin FE, Grant RP, Mackay JP. Characterization of a family of RanBP2-type zinc fingers that can recognize single-stranded RNA. J Mol Biol 2011; 407:273-83. [PMID: 21256132 DOI: 10.1016/j.jmb.2010.12.041] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 12/24/2010] [Accepted: 12/28/2010] [Indexed: 11/17/2022]
Abstract
The recognition of single-stranded RNA (ssRNA) is an important aspect of gene regulation, and a number of different classes of protein domains that recognize ssRNA in a sequence-specific manner have been identified. Recently, we demonstrated that the RanBP2-type zinc finger (ZnF) domains from the human splicing factor ZnF Ran binding domain-containing protein 2 (ZRANB2) can bind to a sequence containing the consensus AGGUAA. Six other human proteins, namely, Ewing's sarcoma (EWS), translocated in liposarcoma (TLS)/FUS, RNA-binding protein 56 (RBP56), RNA-binding motif 5 (RBM5), RNA-binding motif 10 (RBM10) and testis-expressed sequence 13A (TEX13A), each contains a single ZnF with homology to the ZRANB2 ZnFs, and several of these proteins have been implicated in the regulation of mRNA processing. Here, we show that all of these ZnFs are able to bind with micromolar affinities to ssRNA containing a GGU motif. NMR titration data reveal that binding is mediated by the corresponding surfaces on each ZnF, and we also show that sequence selectivity is largely limited to the GGU core motif and that substitution of the three flanking adenines that were selected in our original selection experiment has a minimal effect on binding affinity. These data establish a subset of RanBP2-type ZnFs as a new family of ssRNA-binding motifs.
Collapse
Affiliation(s)
- Cuong D Nguyen
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | | | | |
Collapse
|
1827
|
Praline J, Blasco H, Vourc'h P, Garrigue MA, Gordon PH, Camu W, Corcia P, Andres CR. APOE ε4 allele is associated with an increased risk of bulbar-onset amyotrophic lateral sclerosis in men. Eur J Neurol 2011; 18:1046-52. [PMID: 21251163 DOI: 10.1111/j.1468-1331.2010.03330.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Several association studies have identified possible susceptibility factors for sporadic amyotrophic lateral sclerosis (SALS). Studies on the APOE gene provided conflicting results, especially about the effect on bulbar onset. We assessed the possible role of APOE gene in a large cohort of patients with ALS and matched controls. METHODS The APOE alleles were determined in 1482 patients with SALS and 955 controls and analysed by univariate and multivariate statistics, taking into account gender, site-of-onset and age-at-onset. RESULTS Patients with bulbar onset were more likely to be women [odds ratio (OR)=2.17; 95% CI: 1.74-2.72] and to be older (OR=3.47; 95% CI: 2.58-4.67). The ε4-carriers were more frequent in the bulbar-onset group than in the limb-onset group (OR=1.39 bulbar onset versus limb onset; 95% CI: 1.08-1.80) but this association was observed amongst men (OR=1.78; 95% CI: 1.25-2.53) and not women (OR=1.09; 95% CI: 0.75-1.59). CONCLUSION Our study provides evidence for a contribution of the ε4 allele in the occurrence of bulbar-onset ALS amongst men. We propose that men are normally protected by androgens against bulbar onset and that the ε4 allele inhibits this protection, perhaps by interfering with the androgen pathway.
Collapse
Affiliation(s)
- J Praline
- UMR INSERM U930, Université François-Rabelais, Tours Centre SLA, Service de Neurologie et Neurophysiologie Clinique, CHRU de Tours, Tours Cedex 2, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
1828
|
Mizuno Y, Fujita Y, Takatama M, Okamoto K. Peripherin partially localizes in Bunina bodies in amyotrophic lateral sclerosis. J Neurol Sci 2011; 302:14-8. [PMID: 21241994 DOI: 10.1016/j.jns.2010.12.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Revised: 12/22/2010] [Accepted: 12/23/2010] [Indexed: 12/13/2022]
Abstract
Peripherin is a type III intermediate filament protein expressed with low levels in spinal motor neurons. Amyotrophic lateral sclerosis (ALS) is characterized by the presence of Bunina bodies, skein-like inclusions, and Lewy body-like inclusions (LBLIs) in the remaining anterior horn cells, where the first and third structures are detected by Hematoxylin-Eosin (H & E) staining. We examined paraffin sections of lumbar spinal cords from six ALS patients, using H & E staining and immunostaining for human peripherin. The results demonstrated that there were a total of 73 anterior horn cells containing one or more Bunina bodies, and that twelve of these cells (approximately 16.4%) demonstrated peripherin-positive Bunina bodies. In fact, some part of chain-like Bunina bodies showed peripherin-positive reaction, although there were a much higher number of non-immunoreacitive Bunina bodies in each neuron. LBLIs were clearly immunostained for peripherin corresponding to the core, while some of them showed different types of immunoreactivities due to oblique cutting of inclusions. Our findings suggest that although the mechanisms underlying peripherin co-localization in Bunina bodies are unknown, peripherin could be involved in forming these inclusions. Furthermore, following cystatin C and transferrin, peripherin is the third most prevalent protein that partially localizes in Bunina bodies.
Collapse
Affiliation(s)
- Yuji Mizuno
- Department of Neurology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| | | | | | | |
Collapse
|
1829
|
Genetic rodent models of amyotrophic lateral sclerosis. J Biomed Biotechnol 2011; 2011:348765. [PMID: 21274268 PMCID: PMC3022221 DOI: 10.1155/2011/348765] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 11/29/2010] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the selective death of motor neurons in the motor cortex, brainstem, and spinal cord. A large number of rodent models are available that show motor neuron death and a progressive motor phenotype that is more or less reminiscent of what occurs in patients. These rodent models contain genes with spontaneous or induced mutations or (over) express different (mutant) genes. Some of these models have been of great value to delineate potential pathogenic mechanisms that cause and/or modulate selective motor neuron degeneration. In addition, these genetic rodent models play a crucial role in testing and selecting potential therapeutics that can be used to treat ALS and/or other motor neuron disorders. In this paper, we give a systematic overview of the most important genetic rodent models that show motor neuron degeneration and/or develop a motor phenotype. In addition, we discuss the value and limitations of the different models and conclude that it remains a challenge to find more and better rodent models based on mutations in new genes causing ALS.
Collapse
|
1830
|
Chen T, Benmohamed R, Arvanites AC, Ranaivo HR, Morimoto RI, Ferrante RJ, Watterson DM, Kirsch DR, Silverman RB. Arylsulfanyl pyrazolones block mutant SOD1-G93A aggregation. Potential application for the treatment of amyotrophic lateral sclerosis. Bioorg Med Chem 2011; 19:613-22. [PMID: 21095130 PMCID: PMC3014451 DOI: 10.1016/j.bmc.2010.10.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 10/21/2010] [Accepted: 10/25/2010] [Indexed: 10/18/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an orphan neurodegenerative disease currently without a cure. Mutations in copper/zinc superoxide dismutase 1 (SOD1) have been implicated in the pathophysiology of this disease. Using a high-throughput screening assay expressing mutant G93A SOD1, two bioactive chemical hit compounds (1 and 2), identified as arylsulfanyl pyrazolones, were identified. The structural optimization of this scaffold led to the generation of a more potent analogue (19) with an EC(50) of 170nM. To determine the suitability of this class of compounds for further optimization, 1 was subjected to a battery of pharmacokinetic assays; most of the properties of 1 were good for a screening hit, except it had a relatively rapid clearance and short microsomal half-life stability. Compound 2 was found to be blood-brain barrier penetrating with a brain/plasma ratio=0.19. The optimization of this class of compounds could produce novel therapeutic candidates for ALS patients.
Collapse
Affiliation(s)
- Tian Chen
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113 USA
| | | | | | - Hantamalala Ralay Ranaivo
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208-3113
| | - Richard I. Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208-3500 USA
| | - Robert J. Ferrante
- Geriatric Research Education and Clinical Center, Bedford Veterans Administration Medical Center, Bedford, Massachusetts 01730, USA and the Department of Neurology, Laboratory Medicine and Pathology, and Psychiatry, Boston University School of Medicine, Boston, Massachusetts 02118 USA
| | - D. Martin Watterson
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208-3113
| | | | - Richard B. Silverman
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113 USA
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208-3113 USA
| |
Collapse
|
1831
|
Brady OA, Meng P, Zheng Y, Mao Y, Hu F. Regulation of TDP-43 aggregation by phosphorylation and p62/SQSTM1. J Neurochem 2011; 116:248-59. [PMID: 21062285 DOI: 10.1111/j.1471-4159.2010.07098.x] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
TAR DNA-binding protein-43 (TDP-43) proteinopathy has been linked to several neurodegenerative diseases, such as frontotemporal lobar degeneration with ubiquitin-positive inclusions and amyotrophic lateral sclerosis. Phosphorylated and ubiquitinated TDP-43 C-terminal fragments have been found in cytoplasmic inclusions in frontotemporal lobar degeneration with ubiquitin-positive inclusions and amyotrophic lateral sclerosis patients. However, the factors and pathways that regulate TDP-43 aggregation are still not clear. We found that the C-terminal 15 kDa fragment of TDP-43 is sufficient to induce aggregation but the aggregation phenotype is modified by additional sequences. Aggregation is accompanied by phosphorylation at serine residues 409/410. Mutation of 409/410 to phosphomimetic aspartic acid residues significantly reduces aggregation. Inhibition of either proteasome or autophagy dramatically increases TDP-43 aggregation. Furthermore, TDP-43 aggregates colocalize with markers of autophagy and the adaptor protein p62/SQSTM1. Over-expression of p62/SQSTM1 reduces TDP-43 aggregation in an autophagy and proteasome-dependent manner. These studies suggest that aggregation of TDP-43 C-terminal fragments is regulated by phosphorylation events and both the autophagy and proteasome-mediated degradation pathways.
Collapse
Affiliation(s)
- Owen A Brady
- Weill Institute for Cell and Molecular Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | | | | | | | | |
Collapse
|
1832
|
Trancikova A, Ramonet D, Moore DJ. Genetic Mouse Models of Neurodegenerative Diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 100:419-82. [DOI: 10.1016/b978-0-12-384878-9.00012-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
1833
|
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are clinically overlapping neurodegenerative disorders whose pathophysiology remains incompletely understood. ALS initiates in a discrete location, and typically progresses in a pattern consistent with spread of the degenerative process to involve neighboring regions of the motor system, although the basis of the apparent "spread" remains elusive. Recently mutations in two RNA binding proteins, TDP-43 and FUS, were identified in patients with familial ALS. In addition to being involved in numerous events related to RNA metabolism, each forms aggregates in neurons in ALS and FTLD. Recent evidence also indicates that both TDP-43 and FUS contain prion-related domains rich in glutamine (Q) and asparagine (N) residues, and in the case of TDP-43 this is the location of most disease causing mutations. This review discusses the potential relevance of the prion-related domains in TDP-43 and FUS in normal physiology, pathologic aggregation, and disease progression in ALS and FTLD.
Collapse
Affiliation(s)
- Maria Udan
- Department of Neurology, Neuromuscular Division, Washington University, Saint Louis, MO, USA
| | | |
Collapse
|
1834
|
Kurokawa R. Promoter-associated long noncoding RNAs repress transcription through a RNA binding protein TLS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 722:196-208. [PMID: 21915790 DOI: 10.1007/978-1-4614-0332-6_12] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The majority of the human genome is found to be transcribed and generates mostly noncoding (nc) RNAs that do not possess protein information. MicroRNAs are one of the well-identified small ncRNAs, but occupy merely a fraction of ncRNAs. Long (large) ncRNAs are emerging as a novel class of ncRNAs, but knowledge of these ncRNAs is far less accumulated. Long ncRNAs are tentatively classified as an ncRNA species containing more than 200 nucleotides. Recently, a long promoter-associated ncRNA (pncRNA) has been identified to be transcribed from the cyclin D1 promoter upon induction by genotoxic factors like ionizing-irradiation. The cyclin D1 pncRNA is specifically bound with an RNA-binding protein TLS (Translocated in liposarcoma) and exerts transcriptional repression through histone acetyltransferase (HAT) inhibitory activity. Analysis of TLS and the pncRNAs could provide a model for elucidating their roles inregulation of mammalian transcriptional programs. The pncRNA binding to TLS turns out to be an essential event for the HAT inhibitory activity. A key consensus sequence of the pncRNA is composed of GGUG, while not every RNA sequence bearing GGUG is targeted by TLS, suggesting that a secondary structure of the GGUG-bearing RNAs is also involved in recognition by TLS. Taken together, TLS is a unique mediator between signals of the long ncRNAs and transcription, suggesting that RNA networking functions in living cells.(1-3).
Collapse
Affiliation(s)
- Riki Kurokawa
- Saitama Medical University, Yamane, Hidaka-shi, Saitama-Ken, Japan.
| |
Collapse
|
1835
|
Guest WC, Silverman JM, Pokrishevsky E, O'Neill MA, Grad LI, Cashman NR. Generalization of the prion hypothesis to other neurodegenerative diseases: an imperfect fit. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2011; 74:1433-1459. [PMID: 22043906 DOI: 10.1080/15287394.2011.618967] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Protein misfolding diseases have been classically understood as diffuse errors in protein folding, with misfolded protein arising autonomously throughout a tissue due to a pathologic stressor. The field of prion science has provided an alternative mechanism whereby a seed of pathologically misfolded protein, arising exogenously or through a rare endogenous structural fluctuation, yields a template to catalyze misfolding of the native protein. The misfolded protein may then spread intercellularly to communicate the misfold to adjacent areas and ultimately infect a whole tissue. Mounting evidence implicates a prion-like process in the propagation of several neurodegenerative diseases, including Alzheimer's, Parkinson's, Huntington's, amyotrophic lateral sclerosis, and the tauopathies. However, the parallels between the events observed in these conditions and those in prion disease are often incomplete. The aim of this review was to examine the current state of knowledge concerning the mechanisms of protein misfolding and aggregation for neurodegeneration-associated proteins. In addition, possible methods of intercellular spread are described that focus on the hypothesis that released microvesicles function as misfolded protein delivery vehicles, and the therapeutic options enabled by viewing these diseases from the prion perspective.
Collapse
Affiliation(s)
- Will C Guest
- Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | |
Collapse
|
1836
|
Du K, Arai S, Kawamura T, Matsushita A, Kurokawa R. TLS and PRMT1 synergistically coactivate transcription at the survivin promoter through TLS arginine methylation. Biochem Biophys Res Commun 2010; 404:991-6. [PMID: 21187067 DOI: 10.1016/j.bbrc.2010.12.097] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 12/10/2010] [Indexed: 10/18/2022]
Abstract
TLS (Translocated in LipoSarcoma), also termed FUS, is a multifunctional protein implicated in diverse cellular events such as maintaining genome integrity and regulating gene expression. We have focused on the role of TLS as a coregulator in transcriptional regulation. In the process of investigating TLS-binding proteins, we found that PRMT1 (protein arginine methyltransferase 1) was in complex with TLS. We analyzed the methylation status of endogenous TLS and demonstrated that TLS was arginine-methylated by PRMT1. Using mass spectrometry, we identified that four arginine residues within TLS (R216, R218, R242 and R394) were consistently dimethylated. We performed luciferase reporter assays to assess the functional consequence of TLS arginine methylation in transcriptional regulation and, interestingly, observed that TLS and PRMT1 synergistically coactivated transcription at the survivin promoter. Further analysis using a catalytic-dead PRMT1 or methylation inhibitor both showed that the synergistic transcriptional activation was mediated by TLS arginine-methylation. These results revealed a cooperative role of TLS and PRMT1 in transcriptional regulation.
Collapse
Affiliation(s)
- Kun Du
- Division of Gene Structure and Function, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama-ken 350-1241, Japan
| | | | | | | | | |
Collapse
|
1837
|
New frontiers in the treatment of liposarcoma, a therapeutically resistant malignant cohort. Drug Resist Updat 2010; 14:52-66. [PMID: 21169051 DOI: 10.1016/j.drup.2010.11.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 11/23/2010] [Indexed: 01/10/2023]
Abstract
The adipogenic origin-derived liposarcoma (LPS) family is the most common soft tissue sarcoma histological subtype. This group is composed of three categories as per the 2002 WHO guidelines: (1) well-differentiated and dedifferentiated liposarcoma (WDLPS/DDLPS); (2) myxoid and round cell liposarcoma (MLS and RCL); and (3) pleomorphic liposarcoma (PLS). While clustered together, these histological subtypes are widely diverse in their clinical, pathological, and molecular characteristics. In general, surgery still remains the mainstay of LPS therapy and the only approach offering the potential of cure. Effective therapeutic strategies for locally advanced and metastatic disease are currently lacking and are crucially needed. With the current gradually increasing knowledge of LPS genetic- and epigenetic-associated deregulations, the ultimate goal is to develop drugs that can specifically eliminate LPS cells while sparing normal tissues. This tumor-tailored target-orientated approach will hopefully result in a significant improvement in the outcome of patients suffering from these poor prognosis malignancies.
Collapse
|
1838
|
Identification of a FUS splicing mutation in a large family with amyotrophic lateral sclerosis. J Hum Genet 2010; 56:247-9. [PMID: 21160488 DOI: 10.1038/jhg.2010.162] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease characterized by the degeneration of upper and lower motor neurons. Genetic studies have led, thus far, to the identification of 12 loci and 9 genes for familial ALS (FALS). Although the distribution and impact of superoxide dismutase 1 mutations has been extensively examined for over a decade, the recently identified FALS-associated FUS gene has been less studied. Therefore, we set out to screen our collection of FALS cases for FUS mutations. All 15 exons of FUS were amplified and sequenced in 154 unrelated FALS cases and 475 ethnically matched healthy individuals. One substitution located in the acceptor splice site of intron 14 was identified in all affected members of a large family, causing the skipping of the last 13 amino acids of the protein and the translation of 7 novel amino acids, resulting from the new translation of a part of the 3' untranslated region. Our study identified a new splicing mutation in the highly conserved C-terminal of the FUS protein. Thus far most FUS mutations are missenses, and our findings, combined with those of others, confirm the importance of the C-terminal portion of the protein, adding additional support for FUS mutations having a critical role in ALS.
Collapse
|
1839
|
Nassif M, Matus S, Castillo K, Hetz C. Amyotrophic lateral sclerosis pathogenesis: a journey through the secretory pathway. Antioxid Redox Signal 2010; 13:1955-89. [PMID: 20560784 DOI: 10.1089/ars.2009.2991] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common adult-onset motoneuron degenerative disease characterized by the selective loss of motoneurons in the spinal ventral horn, most brainstem nuclei, and the cerebral cortex. Although approximately 90% of ALS cases are sporadic (sALS), analyses of familial ALS (fALS)-causative genes have generated relevant insight into molecular events involved in the pathology. Here we overview an emerging concept indicating the occurrence of secretory pathway stress in the disease process. These alterations include a failure in the protein folding machinery at the endoplasmic reticulum (ER), engagement of the unfolded protein response (UPR), modifications of the Golgi apparatus network, impaired vesicular trafficking, inhibition of protein quality control mechanisms, oxidative damage to ER proteins, and sustained activation of degradative pathways such as autophagy. A common feature predicted for most of these alterations is abnormal protein homeostasis associated with the accumulation of misfolded proteins at the ER, possibly leading to chronic ER stress and neuronal dysfunction. Signs of ER stress are observed even during presymptomatic stages in fALS mouse models, and pharmacological strategies to alleviate protein misfolding slow disease progression. Because the secretory pathway stress occurs in both sALS and several forms of fALS, it may offer a unique common target for possible therapeutic strategies to treat this devastating disease.
Collapse
Affiliation(s)
- Melissa Nassif
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences , Faculty of Medicine, NEMO Millennium Nucleus, Santiago, Chile
| | | | | | | |
Collapse
|
1840
|
Piguet O, Hornberger M, Mioshi E, Hodges JR. Behavioural-variant frontotemporal dementia: diagnosis, clinical staging, and management. Lancet Neurol 2010; 10:162-72. [PMID: 21147039 DOI: 10.1016/s1474-4422(10)70299-4] [Citation(s) in RCA: 356] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Patients with behavioural-variant frontotemporal dementia (bvFTD) present with insidious changes in personality and interpersonal conduct that indicate progressive disintegration of the neural circuits involved in social cognition, emotion regulation, motivation, and decision making. The underlying pathological changes are heterogeneous and are characterised by various intraneuronal inclusions. Biomarkers to detect these histopathological changes in life are becoming increasingly important with the development of disease-modifying drugs. Gene mutations have been found that collectively account for around 10-20% of cases. Recently, criteria proposed for bvFTD define three levels of diagnostic certainty: possible, probable, and definite. Detailed history taking from family members to elicit behavioural features underpins the diagnostic process, with support from neuropsychological testing designed to detect impairment in decision making, emotion processing, and social cognition. Brain imaging is important for increasing the level of diagnostic certainty. A recently developed staging instrument shows much promise for monitoring patients and evaluating therapies, which at present are aimed at symptom amelioration. Carer education and support remain of paramount importance.
Collapse
Affiliation(s)
- Olivier Piguet
- Neuroscience Research Australia, Randwick, NSW, Australia
| | | | | | | |
Collapse
|
1841
|
Barmada SJ, Finkbeiner S. Pathogenic TARDBP mutations in amyotrophic lateral sclerosis and frontotemporal dementia: disease-associated pathways. Rev Neurosci 2010; 21:251-72. [PMID: 21086759 DOI: 10.1515/revneuro.2010.21.4.251] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are late-onset neurodegenerative disorders that are associated with mutations in the TARDBP gene. The product of this gene, TDP-43, has also been identified as the main component of the intracellular inclusions typical of most cases of ALS and FTD. Recent evidence suggests that TDP-43 is essential for proper development and involved in several fundamental cellular processes, including gene transcription, RNA processing, and the spatial regulation of mRNA translation. Pathogenic TARDBP mutations that impair TDP-43 function could therefore be related to neuronal degeneration in ALS and FTD. Conversely, cellular and animal studies have shown that pathogenic TARDBP mutations induce neuronal toxicity through mislocalization or elevated concentrations of TDP-43, consistent with a gain-of-function mechanism. In this review, we focus on the physiologic functions of TDP-43 within the central nervous system and discuss how these functions may be perturbed or pathologically altered by disease-associated mutations.
Collapse
Affiliation(s)
- Sami J Barmada
- Gladstone Institute of Neurological Disease, University of California, San Francisco 94158, USA
| | | |
Collapse
|
1842
|
Dr. Jekyll and Mr. Hyde: The Two Faces of the FUS/EWS/TAF15 Protein Family. Sarcoma 2010; 2011:837474. [PMID: 21197473 PMCID: PMC3005952 DOI: 10.1155/2011/837474] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 10/20/2010] [Accepted: 11/01/2010] [Indexed: 12/13/2022] Open
Abstract
FUS, EWS, and TAF15 form the FET family of RNA-binding proteins whose genes are found rearranged with various transcription factor genes predominantly in sarcomas and in rare hematopoietic and epithelial cancers. The resulting fusion gene products have attracted considerable interest as diagnostic and promising therapeutic targets. So far, oncogenic FET fusion proteins have been regarded as strong transcription factors that aberrantly activate or repress target genes of their DNA-binding fusion partners. However, the role of the transactivating domain in the context of the normal FET proteins is poorly defined, and, therefore, our knowledge on how FET aberrations impact on tumor biology is incomplete. Since we believe that a full understanding of aberrant FET protein function can only arise from looking at both sides of the coin, the good and the evil, this paper summarizes evidence for the central function of FET proteins in bridging RNA transcription, processing, transport, and DNA repair.
Collapse
|
1843
|
Johnson JO, Mandrioli J, Benatar M, Abramzon Y, Van Deerlin VM, Trojanowski JQ, Gibbs JR, Brunetti M, Gronka S, Wuu J, Ding J, McCluskey L, Martinez-Lage M, Falcone D, Hernandez DG, Arepalli S, Chong S, Schymick JC, Rothstein J, Landi F, Wang M, Calvo A, Mora G, Sabatelli M, Monsurrò MR, Battistini S, Salvi F, Spataro R, Sola P, Borghero G, ITALSGEN, Galassi G, Scholz SW, Taylor JP, Restagno G, Chiò A, Traynor BJ. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 2010; 68:857-64. [PMID: 21145000 PMCID: PMC3032425 DOI: 10.1016/j.neuron.2010.11.036] [Citation(s) in RCA: 976] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2010] [Indexed: 12/12/2022]
Abstract
Using exome sequencing, we identified a p.R191Q amino acid change in the valosin-containing protein (VCP) gene in an Italian family with autosomal dominantly inherited amyotrophic lateral sclerosis (ALS). Mutations in VCP have previously been identified in families with Inclusion Body Myopathy, Paget disease, and Frontotemporal Dementia (IBMPFD). Screening of VCP in a cohort of 210 familial ALS cases and 78 autopsy-proven ALS cases identified four additional mutations including a p.R155H mutation in a pathologically proven case of ALS. VCP protein is essential for maturation of ubiquitin-containing autophagosomes, and mutant VCP toxicity is partially mediated through its effect on TDP-43 protein, a major constituent of ubiquitin inclusions that neuropathologically characterize ALS. Our data broaden the phenotype of IBMPFD to include motor neuron degeneration, suggest that VCP mutations may account for ∼1%-2% of familial ALS, and provide evidence directly implicating defects in the ubiquitination/protein degradation pathway in motor neuron degeneration.
Collapse
Affiliation(s)
- Janel O. Johnson
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
| | - Jessica Mandrioli
- Department of Neuroscience, S. Agostino-Estense Hospital and University of Modena, Via Pietro Giardini 1355, 41126 Modena, Italy
| | - Michael Benatar
- Department of Neurology, Emory University School of Medicine, Woodruff Memorial Research Building, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Yevgeniya Abramzon
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
| | - Vivianna M. Van Deerlin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA 19104, USA
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA 19104, USA
| | - J Raphael Gibbs
- Computational Biology Core, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
- Department of Molecular Neuroscience and Reta Lila Weston Laboratories, Institute of Neurology, University College London, Queen Square House, 9 Queen Square, London WC1 3BG, United Kingdom
| | - Maura Brunetti
- Molecular Genetics Unit, Department of Clinical Pathology, A.S.O. O.I.R.M.-S. Anna, Piazza Polonia 94, 10126 Turin, Italy
| | - Susan Gronka
- Department of Neurology, Emory University School of Medicine, Woodruff Memorial Research Building, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Joanne Wuu
- Department of Neurology, Emory University School of Medicine, Woodruff Memorial Research Building, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Jinhui Ding
- Computational Biology Core, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
| | - Leo McCluskey
- Department of Neurology, University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA 19104, USA
| | - Maria Martinez-Lage
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA 19104, USA
| | - Dana Falcone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 3600 Spruce Street, Philadelphia, PA 19104, USA
| | - Dena G. Hernandez
- Department of Molecular Neuroscience and Reta Lila Weston Laboratories, Institute of Neurology, University College London, Queen Square House, 9 Queen Square, London WC1 3BG, United Kingdom
- Molecular Genetics Unit, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
| | - Sampath Arepalli
- Molecular Genetics Unit, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
| | - Sean Chong
- Molecular Genetics Unit, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
| | - Jennifer C. Schymick
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
| | - Jeffrey Rothstein
- Department of Neurology, Johns Hopkins Hospital, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Francesco Landi
- Department of Gerontology, Geriatrics, and Rehabilitative Medicine, Catholic University of Sacred Heart, Rome, Italy
| | - Michael Wang
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Andrea Calvo
- Department of Neuroscience, University of Turin, Via Cherasco 15, 10126 Turin, Italy
| | - Gabriele Mora
- ALS Center, Salvatore Maugeri Foundation, Via Camaldoli 64, 20100 Milan, Italy
| | - Mario Sabatelli
- Neurological Institute, Catholic University and I.CO.M.M. Association for ALS Research, Largo Agostino Gemelli 8, 10100 Rome, Italy
| | - Maria Rosaria Monsurrò
- Department of Neurological Sciences, Second University of Naples, Piazza Miraglia 2, 80138 Naples, Italy
| | - Stefania Battistini
- Department of Neuroscience, Neurology Section, University of Siena, Viale Bracci 2, 53100 Siena, Italy
| | - Fabrizio Salvi
- Center for Diagnosis and Cure of Rare Diseases, Department of Neurology, Bellaria Hospital, Via Altura 3, 40100 Bologna, Italy
| | - Rossella Spataro
- Department of Clinical Neurosciences, University of Palermo, Via G. La Loggia 1, 90129 Palermo, Italy
| | - Patrizia Sola
- Department of Neuroscience, S. Agostino-Estense Hospital and University of Modena, Via Pietro Giardini 1355, 41126 Modena, Italy
| | - Giuseppe Borghero
- Neurological Clinic, Azienda Hospital, University of Cagliari, S.S. 554 Bivio Sestu, 09042 Monserrato-Cagliari, Italy
| | | | - Giuliana Galassi
- Department of Neuroscience, S. Agostino-Estense Hospital and University of Modena, Via Pietro Giardini 1355, 41126 Modena, Italy
| | - Sonja W. Scholz
- Molecular Genetics Unit, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
- Department of Neuroscience, Georgetown University, 3970 Reservoir Road NW, Washington DC 20057, USA
| | - J. Paul Taylor
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Gabriella Restagno
- Molecular Genetics Unit, Department of Clinical Pathology, A.S.O. O.I.R.M.-S. Anna, Piazza Polonia 94, 10126 Turin, Italy
| | - Adriano Chiò
- Department of Neuroscience, University of Turin, Via Cherasco 15, 10126 Turin, Italy
| | - Bryan J. Traynor
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, NIA, NIH, Porter Neuroscience Building, 35 Convent Drive, Bethesda, MD 20892, USA
- Department of Neurology, Johns Hopkins Hospital, 600 North Wolfe Street, Baltimore, MD 21287, USA
| |
Collapse
Collaborators
Fabio Giannini, Claudia Ricci, Cristina Moglia, Irene Ossola, Antonio Canosa, Sara Gallo, Gioacchino Tedeschi, Patrizia Sola, Ilaria Bartolomei, Kalliopi Marinou, Laura Papetti, Amelia Conte, Marco Luigetti, Vincenzo La Bella, Piera Paladino, Claudia Caponnetto, Paolo Volanti, Maria Giovanna Marrosu, Maria Rita Murru,
Collapse
|
1844
|
Abstract
TDP-43 mislocalization and aggregation are implicated in the pathogenesis of ALS and FTLD-U. Valosin containing protein (VCP) mutations also lead to TDP-43 deposition, resulting in Inclusion Body Myopathy, Paget disease, and Frontotemporal Dementia (IBMPFD). In this issue of Neuron, Johnson et al. used whole-exome capture to identify VCP mutations in familial ALS. This extends the VCP phenotype to include motor neuron degeneration and provides another molecular tool to explore neurodegeneration disease mechanisms underlying the TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Christopher E Shaw
- King's College London, MRC Centre for Neurodegeneration Research, Department of Clinical Neuroscience, Institute of Psychiatry, London SE5 8AF, UK.
| |
Collapse
|
1845
|
Ito D, Seki M, Tsunoda Y, Uchiyama H, Suzuki N. Nuclear transport impairment of amyotrophic lateral sclerosis-linked mutations in FUS/TLS. Ann Neurol 2010; 69:152-62. [PMID: 21280085 DOI: 10.1002/ana.22246] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 07/24/2010] [Accepted: 08/27/2010] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The fused in sarcoma/translated in liposarcoma (FUS/TLS) protein was recently identified as a cause of familial amyotrophic lateral sclerosis (ALS), as well as a major component of the inclusion bodies found in subtypes of frontotemporal lobar degeneration (FTLD). These diseases now are collectively known as the novel clinical spectrum, FUS proteinopathy. ALS-linked mutations of FUS are clustered in the C-terminal region; however, the molecular properties of mutant FUS remain unclear. To gain insight into the pathogenesis of FUS proteinopathy, we examined the biochemical and cellular characteristics of mutant FUS in expressing cells. METHODS AND RESULTS Expression of ALS-linked FUS mutations resulted in their assembly into cytoplasmic stress granules (SGs), cellular structures that package mRNA and RNA-binding proteins during cell stress. A deletion mutant series revealed that the C-terminal region in FUS is critical for nuclear retention via Ran guanosine triphosphatase-dependent transport machinery. A parallel study of subcellular distribution revealed that ALS-linked mutants additively disturb the function of the C-terminus for nuclear traffic, resulting in cytoplasmic accumulation and the formation of SGs. INTERPRETATION This study demonstrates that mutant FUS, which is missing the nuclear traffic activity of the C-terminus, is dislocated to cytoplasm and assembled into SGs, indicating that disruption of translational regulation and metabolism of mRNA via inappropriate/excessive SGs may be crucial for FUS proteinopathies. Our findings provide new biological and pathological insights into the FUS protein that should help our understanding of the pathogenesis of ALS/FTLD.
Collapse
Affiliation(s)
- Daisuke Ito
- Department of Neurology, School of Medicine, Keio University, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
1846
|
Syriani E, Morales M, Gamez J. FUS/TLSgene mutations are the second most frequent cause of familial ALS in the Spanish population. ACTA ACUST UNITED AC 2010; 12:118-23. [DOI: 10.3109/17482968.2010.539235] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
1847
|
Wegorzewska I, Baloh RH. TDP-43-based animal models of neurodegeneration: new insights into ALS pathology and pathophysiology. NEURODEGENER DIS 2010; 8:262-74. [PMID: 21124004 DOI: 10.1159/000321547] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 09/08/2010] [Indexed: 12/12/2022] Open
Abstract
The clinical and pathological overlap between amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) suggests these diseases share common underlying mechanisms, a suggestion underscored by the discovery that TDP-43 inclusions are a key pathologic feature in both ALS and FTLD. This finding, combined with the identification of TDP-43 mutations in ALS, directly implicates this DNA/RNA binding protein in disease pathogenesis in ALS and FTLD. However, many key questions remain, including what is the normal function of TDP-43, and whether disease-associated mutations produce toxicity in the nucleus, cytoplasm or both. Furthermore, although pathologic TDP-43 inclusions are clearly associated with many forms of neurodegeneration, whether TDP-43 aggregation is a key step in the pathogenesis in ALS, FTLD and other disorders remains to be proven. This review will compare the features of numerous recently developed animal models of TDP-43-related neurodegeneration, and discuss how they contribute to our understanding of the pathogenesis of human ALS and FTLD.
Collapse
Affiliation(s)
- Iga Wegorzewska
- Neuromuscular Division, Department of Neurology, Washington University, Saint Louis, MO 63110, USA
| | | |
Collapse
|
1848
|
Higashi S, Tsuchiya Y, Araki T, Wada K, Kabuta T. TDP-43 physically interacts with amyotrophic lateral sclerosis-linked mutant CuZn superoxide dismutase. Neurochem Int 2010; 57:906-13. [DOI: 10.1016/j.neuint.2010.09.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 09/10/2010] [Accepted: 09/22/2010] [Indexed: 10/19/2022]
|
1849
|
Penco S, Lunetta C, Mosca L, Maestri E, Avemaria F, Tarlarini C, Patrosso MC, Marocchi A, Corbo M. Phenotypic heterogeneity in a SOD1 G93D Italian ALS family: an example of human model to study a complex disease. J Mol Neurosci 2010; 44:25-30. [PMID: 21120636 DOI: 10.1007/s12031-010-9480-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 11/21/2010] [Indexed: 12/12/2022]
Abstract
We report different clinical expression in seven members of a large family with amyotrophic lateral sclerosis (ALS) and the G93D mutation in exon 4 of the Cu/Zn superoxide dismutase (SOD1) gene. The ALS clinical course in the proband showed an unusually fast progression of the disease compared to the paucisymptomatic presentation associated to this mutation in the two previously Italian families described. The remaining mutation carriers did not show the aggressive clinical course displayed by the proband. We selected few genes known to be ALS modifiers searching for genetic variants that could explain the wide phenotypic diversity within the family. Exclusion of causative genes such as TDP43, FUS, PGRN and VAPB was performed too. We believe that this kind of family with contrasting phenotypes of ALS may be considered an excellent human model to study the relationship between a wider genetic profile, including modifier genes, and the clinical expression of the disease. Therefore, the novelty of our approach is also represented by the study of a single family to reproduce a composite structure in which search for possible modifier genes/genetic variants linked to SOD1 mutated.
Collapse
Affiliation(s)
- Silvana Penco
- Department of Laboratory Medicine, Medical Genetics, Niguarda Ca' Granda Hospital, Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
1850
|
Cairns NJ, Perrin RJ, Schmidt RE, Gru A, Green KG, Carter D, Taylor-Reinwald L, Morris JC, Gitcho MA, Baloh RH. TDP-43 proteinopathy in familial motor neurone disease with TARDBP A315T mutation: a case report. Neuropathol Appl Neurobiol 2010; 36:673-9. [PMID: 20819167 PMCID: PMC2978282 DOI: 10.1111/j.1365-2990.2010.01121.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- N. J. Cairns
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - R. J. Perrin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - R. E. Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - A. Gru
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - K. G. Green
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - D. Carter
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - L. Taylor-Reinwald
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - J. C. Morris
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - M. A. Gitcho
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - R. H. Baloh
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|