151
|
Blakemore LJ, Trombley PQ. Zinc as a Neuromodulator in the Central Nervous System with a Focus on the Olfactory Bulb. Front Cell Neurosci 2017; 11:297. [PMID: 29033788 PMCID: PMC5627021 DOI: 10.3389/fncel.2017.00297] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/06/2017] [Indexed: 12/19/2022] Open
Abstract
The olfactory bulb (OB) is central to the sense of smell, as it is the site of the first synaptic relay involved in the processing of odor information. Odor sensations are first transduced by olfactory sensory neurons (OSNs) before being transmitted, by way of the OB, to higher olfactory centers that mediate olfactory discrimination and perception. Zinc is a common trace element, and it is highly concentrated in the synaptic vesicles of subsets of glutamatergic neurons in some brain regions including the hippocampus and OB. In addition, zinc is contained in the synaptic vesicles of some glycinergic and GABAergic neurons. Thus, zinc released from synaptic vesicles is available to modulate synaptic transmission mediated by excitatory (e.g., N-methyl-D aspartate (NMDA), alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)) and inhibitory (e.g., gamma-aminobutyric acid (GABA), glycine) amino acid receptors. Furthermore, extracellular zinc can alter the excitability of neurons through effects on a variety of voltage-gated ion channels. Consistent with the notion that zinc acts as a regulator of neuronal activity, we and others have shown zinc modulation (inhibition and/or potentiation) of amino acid receptors and voltage-gated ion channels expressed by OB neurons. This review summarizes the locations and release of vesicular zinc in the central nervous system (CNS), including in the OB. It also summarizes the effects of zinc on various amino acid receptors and ion channels involved in regulating synaptic transmission and neuronal excitability, with a special emphasis on the actions of zinc as a neuromodulator in the OB. An understanding of how neuroactive substances such as zinc modulate receptors and ion channels expressed by OB neurons will increase our understanding of the roles that synaptic circuits in the OB play in odor information processing and transmission.
Collapse
Affiliation(s)
- Laura J Blakemore
- Program in Neuroscience, Florida State UniversityTallahassee, FL, United States.,Department of Biological Science, Florida State UniversityTallahassee, FL, United States
| | - Paul Q Trombley
- Program in Neuroscience, Florida State UniversityTallahassee, FL, United States.,Department of Biological Science, Florida State UniversityTallahassee, FL, United States
| |
Collapse
|
152
|
Lin HC, Lin CH, Chen PL, Cheng SJ, Chen PH. Intrafamilial phenotypic heterogeneity in a Taiwanese family with a MAPT p.R5H mutation: a case report and literature review. BMC Neurol 2017; 17:186. [PMID: 28923025 PMCID: PMC5604294 DOI: 10.1186/s12883-017-0966-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/13/2017] [Indexed: 12/13/2022] Open
Abstract
Background Frontotemporal degeneration (FTD) is a clinically and genetically heterogeneous neurodegenerative disorder characterized by deficits in executive function that frequently overlaps with parkinsonism and motor neuron disorders. Several genes have been identified to cause autosomal dominant forms of FTD, including the gene coding for the protein associated with microtubule tau (MAPT). While most reported pathogenic mutations in MAPT occur in exons 9–13, few families have been reported with mutations outside of this region. Herein, we report a first Taiwanese family having the exon 1 p.Arg5His mutation in MAPT with intrafamilial phenotype heterogeneity. Case presentation A 63-year-old man presented with progressive non-fluent speech and impaired memory for 3 years. He then developed apraxia, myoclonus and parkinsonism feature at his right hand. Extensive neurologic and neurocognitive examination lead to a diagnosis of FTD mixed with corticobasal syndrome. Magnetic resonance imaging revealed asymmetric atrophy in the left frontal and temporal lobes and single-photon emission computed tomography indicated decreased metabolism in the same areas as well as the left basal ganglia. The patient’s mother had been diagnosed with amyotrophic lateral sclerosis (ALS) at the age of 60 and was deceased 10 years later due to respiratory failure. The patient’s younger sister had persistent depressive disorder in her early forties and did not have any prominent cognitive or motor dysfunctions. We performed genetic analysis applying a targeted next generation sequencing (NGS) panel covering MAPT, GRN, VCP, FUS, CHMP2B, and TARDBP on the proband, followed by Sanger sequencing of candidate genes in eight family members. Hexanucleotide repeat expansion of C9Orf72 was determined by repeat-primed PCR. We identified a missense mutation in exon 1 of MAPT gene, c.14G > A (p.R5H), which was previously reported in only two Japanese patients in a literature review. This substitution co-segregated with the disease phenotypes in the family. Conclusions This is the first report of the occurrence of the MAPT p.R5H mutation in the Taiwanese population. Our findings extend the current knowledge of phenotypic heterogeneity among family members carrying the MAPT p.R5H mutation.
Collapse
Affiliation(s)
- Hui-Chi Lin
- Department of Neurology, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Zhongshan Dist, Taipei City, 10449, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Pei-Lung Chen
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, No. 7, Chung-Shan South Road, Taipei, Taiwan
| | - Shih-Jung Cheng
- Department of Neurology, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Zhongshan Dist, Taipei City, 10449, Taiwan
| | - Pei-Hao Chen
- Department of Neurology, MacKay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Zhongshan Dist, Taipei City, 10449, Taiwan. .,Department of Medicine, Mackay Medical College, New Taipei, Taiwan. .,Graduate Institute of Mechanical and Electrical Engineering, National Taipei University of Technology, Taipei, Taiwan.
| |
Collapse
|
153
|
Chang MC, Srinivasan K, Friedman BA, Suto E, Modrusan Z, Lee WP, Kaminker JS, Hansen DV, Sheng M. Progranulin deficiency causes impairment of autophagy and TDP-43 accumulation. J Exp Med 2017; 214:2611-2628. [PMID: 28778989 PMCID: PMC5584112 DOI: 10.1084/jem.20160999] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 04/11/2017] [Accepted: 07/06/2017] [Indexed: 11/05/2022] Open
Abstract
It is unclear how progranulin deficiency causes frontotemporal dementia, a neurodegenerative disease characterized by TDP-43 inclusions. Chang et al. show that loss of progranulin causes impairment of autophagy and autophagy signaling, which leads to accumulation of pathological TDP-43 in neurons. Loss-of-function mutations in GRN cause frontotemporal dementia (FTD) with transactive response DNA-binding protein of 43 kD (TDP-43)–positive inclusions and neuronal ceroid lipofuscinosis (NCL). There are no disease-modifying therapies for either FTD or NCL, in part because of a poor understanding of how mutations in genes such as GRN contribute to disease pathogenesis and neurodegeneration. By studying mice lacking progranulin (PGRN), the protein encoded by GRN, we discovered multiple lines of evidence that PGRN deficiency results in impairment of autophagy, a key cellular degradation pathway. PGRN-deficient mice are sensitive to Listeria monocytogenes because of deficits in xenophagy, a specialized form of autophagy that mediates clearance of intracellular pathogens. Cells lacking PGRN display reduced autophagic flux, and pathological forms of TDP-43 typically cleared by autophagy accumulate more rapidly in PGRN-deficient neurons. Our findings implicate autophagy as a novel therapeutic target for GRN-associated NCL and FTD and highlight the emerging theme of defective autophagy in the broader FTD/amyotrophic lateral sclerosis spectrum of neurodegenerative disease.
Collapse
Affiliation(s)
- Michael C Chang
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA
| | | | - Brad A Friedman
- Department of Bioinformatics and Computational Biology, Genentech, Inc., South San Francisco, CA
| | - Eric Suto
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA
| | - Zora Modrusan
- Department of Molecular Biology, Genentech, Inc., South San Francisco, CA
| | - Wyne P Lee
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA
| | - Joshua S Kaminker
- Department of Bioinformatics and Computational Biology, Genentech, Inc., South San Francisco, CA
| | - David V Hansen
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA
| | - Morgan Sheng
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA
| |
Collapse
|
154
|
She A, Kurtser I, Reis SA, Hennig K, Lai J, Lang A, Zhao WN, Mazitschek R, Dickerson BC, Herz J, Haggarty SJ. Selectivity and Kinetic Requirements of HDAC Inhibitors as Progranulin Enhancers for Treating Frontotemporal Dementia. Cell Chem Biol 2017; 24:892-906.e5. [PMID: 28712747 DOI: 10.1016/j.chembiol.2017.06.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 02/06/2017] [Accepted: 06/19/2017] [Indexed: 11/18/2022]
Abstract
Frontotemporal dementia (FTD) arises from neurodegeneration in the frontal, insular, and anterior temporal lobes. Autosomal dominant causes of FTD include heterozygous mutations in the GRN gene causing haploinsufficiency of progranulin (PGRN) protein. Recently, histone deacetylase (HDAC) inhibitors have been identified as enhancers of PGRN expression, although the mechanisms through which GRN is epigenetically regulated remain poorly understood. Using a chemogenomic toolkit, including optoepigenetic probes, we show that inhibition of class I HDACs is sufficient to upregulate PGRN in human neurons, and only inhibitors with apparent fast binding to their target HDAC complexes are capable of enhancing PGRN expression. Moreover, we identify regions in the GRN promoter in which elevated H3K27 acetylation and transcription factor EB (TFEB) occupancy correlate with HDAC-inhibitor-mediated upregulation of PGRN. These findings have implications for epigenetic and cis-regulatory mechanisms controlling human GRN expression and may advance translational efforts to develop targeted therapeutics for treating PGRN-deficient FTD.
Collapse
Affiliation(s)
- Angela She
- Chemical Neurobiology Laboratory, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Iren Kurtser
- Chemical Neurobiology Laboratory, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Surya A Reis
- Chemical Neurobiology Laboratory, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Krista Hennig
- Chemical Neurobiology Laboratory, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Jenny Lai
- Chemical Neurobiology Laboratory, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Audrey Lang
- Chemical Neurobiology Laboratory, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Wen-Ning Zhao
- Chemical Neurobiology Laboratory, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Ralph Mazitschek
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Bradford C Dickerson
- MGH Frontotemporal Disorders Unit, Gerontology Research Unit, Alzheimer's Disease Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9046, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
155
|
Pottier C, Ravenscroft TA, Sanchez-Contreras M, Rademakers R. Genetics of FTLD: overview and what else we can expect from genetic studies. J Neurochem 2017; 138 Suppl 1:32-53. [PMID: 27009575 DOI: 10.1111/jnc.13622] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/26/2016] [Accepted: 03/18/2016] [Indexed: 12/11/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) comprises a highly heterogeneous group of disorders clinically associated with behavioral and personality changes, language impairment, and deficits in executive functioning, and pathologically associated with degeneration of frontal and temporal lobes. Some patients present with motor symptoms including amyotrophic lateral sclerosis. Genetic research over the past two decades in FTLD families led to the identification of three common FTLD genes (microtubule-associated protein tau, progranulin, and chromosome 9 open reading frame 72) and a small number of rare FTLD genes, explaining the disease in almost all autosomal dominant FTLD families but only a minority of apparently sporadic patients or patients in whom the family history is less clear. Identification of additional FTLD (risk) genes is therefore highly anticipated, especially with the emerging use of next-generation sequencing. Common variants in the transmembrane protein 106 B were identified as a genetic risk factor of FTLD and disease modifier in patients with known mutations. This review summarizes for each FTLD gene what we know about the type and frequency of mutations, their associated clinical and pathological features, and potential disease mechanisms. We also provide an overview of emerging disease pathways encompassing multiple FTLD genes. We further discuss how FTLD specific issues, such as disease heterogeneity, the presence of an unclear family history and the possible role of an oligogenic basis of FTLD, can pose challenges for future FTLD gene identification and risk assessment of specific variants. Finally, we highlight emerging clinical, genetic, and translational research opportunities that lie ahead. Genetic research led to the identification of three common FTLD genes with rare variants (MAPT, GRN, and C9orf72) and a small number of rare genes. Efforts are now ongoing, which aimed at the identification of rare variants with high risk and/or low frequency variants with intermediate effect. Common risk variants have also been identified, such as TMEM106B. This review discusses the current knowledge on FTLD genes and the emerging disease pathways encompassing multiple FTLD genes.
Collapse
Affiliation(s)
- Cyril Pottier
- Mayo Clinic Jacksonville, Department of Neuroscience, Jacksonville, FL, USA
| | | | | | - Rosa Rademakers
- Mayo Clinic Jacksonville, Department of Neuroscience, Jacksonville, FL, USA
| |
Collapse
|
156
|
Luzzi S, Colleoni L, Corbetta P, Baldinelli S, Fiori C, Girelli F, Silvestrini M, Caroppo P, Giaccone G, Tagliavini F, Rossi G. Missense mutation in GRN gene affecting RNA splicing and plasma progranulin level in a family affected by frontotemporal lobar degeneration. Neurobiol Aging 2017; 54:214.e1-214.e6. [DOI: 10.1016/j.neurobiolaging.2017.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 12/18/2022]
|
157
|
Fletcher EV, Simon CM, Pagiazitis JG, Chalif JI, Vukojicic A, Drobac E, Wang X, Mentis GZ. Reduced sensory synaptic excitation impairs motor neuron function via Kv2.1 in spinal muscular atrophy. Nat Neurosci 2017; 20:905-916. [PMID: 28504671 PMCID: PMC5487291 DOI: 10.1038/nn.4561] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 04/04/2017] [Indexed: 12/30/2022]
Abstract
Behavioral deficits in neurodegenerative diseases are often attributed to the selective dysfunction of vulnerable neurons via cell-autonomous mechanisms. Although vulnerable neurons are embedded in neuronal circuits, the contribution of their synaptic partners to the disease process is largely unknown. Here, we show that in a mouse model of spinal muscular atrophy (SMA), a reduction in proprioceptive synaptic drive leads to motor neuron dysfunction and motor behavior impairments. In SMA mice or after the blockade of proprioceptive synaptic transmission we observed a decrease in the motor neuron firing which could be explained by the reduction in the expression of the potassium channel Kv2.1 at the surface of motor neurons. Increasing neuronal activity pharmacologically by chronic exposure in vivo led to a normalization of Kv2.1 expression and an improvement in motor function. Our results demonstrate a key role of excitatory synaptic drive in shaping the function of motor neurons during development and the contribution of its disruption to a neurodegenerative disease.
Collapse
Affiliation(s)
- Emily V Fletcher
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Christian M Simon
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - John G Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Joshua I Chalif
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Aleksandra Vukojicic
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Estelle Drobac
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Xiaojian Wang
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - George Z Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA.,Department of Neurology, Columbia University, New York, New York, USA
| |
Collapse
|
158
|
Morriss GR, Cooper TA. Protein sequestration as a normal function of long noncoding RNAs and a pathogenic mechanism of RNAs containing nucleotide repeat expansions. Hum Genet 2017; 136:1247-1263. [PMID: 28484853 DOI: 10.1007/s00439-017-1807-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/28/2017] [Indexed: 12/12/2022]
Abstract
An emerging class of long noncoding RNAs (lncRNAs) function as decoy molecules that bind and sequester proteins thereby inhibiting their normal functions. Titration of proteins by lncRNAs has wide-ranging effects affecting nearly all steps in gene expression. While decoy lncRNAs play a role in normal physiology, RNAs expressed from alleles containing nucleotide repeat expansions can be pathogenic due to protein sequestration resulting in disruption of normal functions. This review focuses on commonalities between decoy lncRNAs that regulate gene expression by competitive inhibition of protein function through sequestration and specific examples of nucleotide repeat expansion disorders mediated by toxic RNA that sequesters RNA-binding proteins and impedes their normal functions. Understanding how noncoding RNAs compete with various RNA and DNA molecules for binding of regulatory proteins will provide insight into how similar mechanisms contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Ginny R Morriss
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Thomas A Cooper
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
159
|
Korhonen VE, Solje E, Suhonen NM, Rauramaa T, Vanninen R, Remes AM, Leinonen V. Frontotemporal dementia as a comorbidity to idiopathic normal pressure hydrocephalus (iNPH): a short review of literature and an unusual case. Fluids Barriers CNS 2017; 14:10. [PMID: 28420385 PMCID: PMC5395836 DOI: 10.1186/s12987-017-0060-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/05/2017] [Indexed: 12/12/2022] Open
Abstract
Behavioural variant frontotemporal dementia (bvFTD) and idiopathic normal pressure hydrocephalus (iNPH) are neurodegenerative diseases that can present with similar symptoms. These include decline in executive functions, psychomotor slowness, and behavioural and personality changes. Ventricular enlargement is a key radiological finding in iNPH that may also be present in bvFTD caused by the C9ORF72 expansion mutation. Due to this, bvFTD has been hypothesized as a potential comorbidity to iNPH but bvFTD patients have never been identified in studies focusing in clinical comorbidities with iNPH. Here we describe a patient with the C9ORF72 expansion-associated bvFTD who also showed enlarged ventricles on brain imaging. The main clinical symptoms were severe gait disturbances and psychiatric problems with mild cognitive decline. Cerebrospinal fluid removal increased the patient's walking speed, so a ventriculoperitoneal shunt was placed. After insertion of the shunt, there was a significant improvement in walking speed as well as mild improvement in cognitive function but not in neuropsychiatric symptoms relating to bvFTD. Comorbid iNPH should be considered in bvFTD patients who have enlarged ventricles and severely impaired gait.
Collapse
Affiliation(s)
- V. E. Korhonen
- Department of Neurosurgery, Kuopio University Hospital, P.O. Box 100, 70029 KYS Kuopio, Finland
- University of Eastern Finland, P.O. Box 100, 70029 KYS Kuopio, Finland
| | - E. Solje
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - N. M. Suhonen
- Medical Research Center, Oulu University Hospital, P.O. Box 20, 90029 Oulu, Finland
- Unit of Clinical Neuroscience, Neurology, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
| | - T. Rauramaa
- Institute of Clinical Medicine-Pathology, School of Medicine, University of Eastern, Kuopio, Finland
- Department of Pathology, Kuopio University Hospital, P.O. Box 162, 70211 Kuopio, Finland
| | - R. Vanninen
- Department of Radiology, Kuopio University Hospital, P.O. Box 100, 70029 KYS Kuopio, Finland
| | - A. M. Remes
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
- Medical Research Center, Oulu University Hospital, P.O. Box 20, 90029 Oulu, Finland
- Unit of Clinical Neuroscience, Neurology, University of Oulu, P.O. Box 5000, 90014 Oulu, Finland
| | - V. Leinonen
- Department of Neurosurgery, Kuopio University Hospital, P.O. Box 100, 70029 KYS Kuopio, Finland
- University of Eastern Finland, P.O. Box 100, 70029 KYS Kuopio, Finland
| |
Collapse
|
160
|
Hinz FI, Geschwind DH. Molecular Genetics of Neurodegenerative Dementias. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a023705. [PMID: 27940516 DOI: 10.1101/cshperspect.a023705] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative dementias are clinically heterogeneous, progressive diseases with frequently overlapping symptoms, such as cognitive impairments and behavior and movement deficits. Although a majority of cases appear to be sporadic, there is a large genetic component that has yet to be fully explained. Here, we review the recent genetic and genomic findings pertaining to Alzheimer's disease, frontotemporal dementia, Lewy body dementia, and prion dementia. In this review, we describe causal and susceptibility genes identified for these dementias and discuss recent research pertaining to the molecular function of these genes. Of particular interest, there is a large overlap in clinical phenotypes, genes, and/or aggregating protein products involved in these diseases, as well as frequent comorbid presentation, indicating that these dementias may represent a continuum of syndromes rather than individual diseases.
Collapse
Affiliation(s)
- Flora I Hinz
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095.,Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California 90024
| |
Collapse
|
161
|
Chitramuthu BP, Kay DG, Bateman A, Bennett HPJ. Neurotrophic effects of progranulin in vivo in reversing motor neuron defects caused by over or under expression of TDP-43 or FUS. PLoS One 2017; 12:e0174784. [PMID: 28358904 PMCID: PMC5373598 DOI: 10.1371/journal.pone.0174784] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/15/2017] [Indexed: 12/12/2022] Open
Abstract
Progranulin (PGRN) is a glycoprotein with multiple roles in normal and disease states. Mutations within the GRN gene cause frontotemporal lobar degeneration (FTLD). The affected neurons display distinctive TAR DNA binding protein 43 (TDP-43) inclusions. How partial loss of PGRN causes TDP-43 neuropathology is poorly understood. TDP-43 inclusions are also found in affected neurons of patients with other neurodegenerative diseases including amyotrophic lateral sclerosis (ALS) and Alzheimer's disease. In ALS, TDP-43 inclusions are typically also immunoreactive for fused in sarcoma (FUS). Mutations within TDP-43 or FUS are themselves neuropathogenic in ALS and some cases of FTLD. We used the outgrowth of caudal primary motor neurons (MNs) in zebrafish embryos to investigate the interaction of PGRN with TDP-43 and FUS in vivo. As reported previously, depletion of zebrafish PGRN-A (zfPGRN-A) is associated with truncated primary MNs and impaired motor function. Here we found that depletion of zfPGRN-A results in primary MNs outgrowth stalling at the horizontal myoseptum, a line of demarcation separating the myotome into dorsal and ventral compartments that is where the final destination of primary motor is assigned. Successful axonal outgrowth beyond the horizontal myoseptum depends in part upon formation of acetylcholine receptor clusters and this was found to be disorganized upon depletion of zfPGRN-A. PGRN reversed the effects of zfPGRN-A knockdown, but a related gene, zfPGRN-1, was without effect. Both knockdown of TDP-43 or FUS, as well as expression of humanTDP-43 and FUS mutants results in MN abnormalities that are reversed by co-expression of hPGRN mRNA. Neither TDP-43 nor FUS reversed MN phenotypes caused by the depletion of PGRN. Thus TDP-43 and FUS lie upstream of PGRN in a gene complementation pathway. The ability of PGRN to override TDP-43 and FUS neurotoxicity due to partial loss of function or mutation in the corresponding genes may have therapeutic relevance.
Collapse
Affiliation(s)
- Babykumari P. Chitramuthu
- Endocrine Research Laboratory, Royal Victoria Hospital, McGill University Health Centre Research Institute, Montreal, Québec, Canada
- Neurodyn Inc., Charlottetown, Prince Edward Island, Canada
- * E-mail: (BPC); (HPJB)
| | - Denis G. Kay
- Neurodyn Inc., Charlottetown, Prince Edward Island, Canada
| | - Andrew Bateman
- Endocrine Research Laboratory, Royal Victoria Hospital, McGill University Health Centre Research Institute, Montreal, Québec, Canada
| | - Hugh P. J. Bennett
- Endocrine Research Laboratory, Royal Victoria Hospital, McGill University Health Centre Research Institute, Montreal, Québec, Canada
- * E-mail: (BPC); (HPJB)
| |
Collapse
|
162
|
Gatto EM, Allegri RF, Da Prat G, Chrem Mendez P, Hanna DS, Dorschner MO, Surace EI, Zabetian CP, Mata IF. Intrafamilial variable phenotype including corticobasal syndrome in a family with p.P301L mutation in the MAPT gene: first report in South America. Neurobiol Aging 2017; 53:195.e11-195.e17. [PMID: 28268100 DOI: 10.1016/j.neurobiolaging.2017.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/17/2017] [Accepted: 02/02/2017] [Indexed: 12/14/2022]
Abstract
Frontotemporal lobar degeneration is a neuropathological disorder that causes a variety of clinical syndromes including frontotemporal dementia (FTD), progressive supranuclear palsy, and corticobasal syndrome (CBS). FTD associated with parkinsonism occurs frequently as a result of mutations in the C9orf72 gene and also in the genes coding for the protein associated with microtubule tau (MAPT) and progranulin (GRN) on chromosome 17 (FTDP-17). Herein, we report an Argentinean family, of Basque ancestry, with an extensive family history of behavioral variant of FTD. Twenty-one members over 6 generations composed the pedigree. An extensive neurologic and neurocognitive examination was performed on 2 symptomatic individuals and 3 nonsymptomatic individuals. Two different phenotypes were identified among affected members, CBS in the proband and FTD in his brother. DNA was extracted from blood for these 5 individuals and whole-exome sequencing was performed on 3 of them followed by Sanger sequencing of candidate genes on the other 2. In both affected individuals, a missense mutation (p.P301L; rs63751273) in exon 10 of the MAPT gene (chr17q21.3) was identified. Among MAPT mutations, p.P301L is the most frequently associated to different phenotypes: (1) aggressive, symmetrical, and early-onset Parkinsonism; (2) late parkinsonism associated with FTD; and (3) progressive supranuclear palsy but only exceptionally it is reported associated to CBS. This is the first report of the occurrence of the p.P301L-MAPT mutation in South America and supports the marked phenotypic heterogeneity among members of the same family as previously reported.
Collapse
Affiliation(s)
- Emilia M Gatto
- Department of Movement Disorders, Fundación INEBA, Buenos Aires, Argentina; Department of Neurology, Sanatorio de la Trinidad Mitre, Buenos Aires, Argentina
| | - Ricardo F Allegri
- Department of Cognitive Neurology, Memory and Aging Center, Instituto de Investigaciones Neurológicas Dr. Raúl Carrea (FLENI), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Investigator at CONICET the Argentine Research Council (CONICET), Cdad. Autónoma de Buenos Aires, Argentina; Universidad de la Costa (CUC), Barranquilla, Colombia
| | - Gustavo Da Prat
- Department of Neurology, Sanatorio de la Trinidad Mitre, Buenos Aires, Argentina
| | - Patricio Chrem Mendez
- Department of Cognitive Neurology, Memory and Aging Center, Instituto de Investigaciones Neurológicas Dr. Raúl Carrea (FLENI), Buenos Aires, Argentina
| | - David S Hanna
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Michael O Dorschner
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Ezequiel I Surace
- Department of Neuropathology, Laboratorio de Biología Molecular, Instituto de Investigaciones Neurológicas Dr. Raúl Carrea (FLENI), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Investigator at CONICET the Argentine Research Council (CONICET), Cdad. Autónoma de Buenos Aires, Argentina
| | - Cyrus P Zabetian
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Seattle, WA, USA; Department of Neurology, University of Washington, Seattle, Seattle, WA, USA
| | - Ignacio F Mata
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Seattle, WA, USA; Department of Neurology, University of Washington, Seattle, Seattle, WA, USA.
| |
Collapse
|
163
|
Abstract
PURPOSE OF REVIEW This article reviews the common behavioral and cognitive features of frontotemporal dementia (FTD) and related disorders as well as the distinguishing clinical, genetic, and pathologic features of the most common subtypes. RECENT FINDINGS Advances in clinical phenotyping, genetics, and biomarkers have enabled improved predictions of the specific underlying molecular pathology associated with different presentations of FTD. Evaluation of large international cohorts has led to recent refinements in diagnostic criteria for several of the FTD subtypes. SUMMARY The FTDs are a group of neurodegenerative disorders featuring progressive deterioration of behavior or language and associated pathology in the frontal or temporal lobes. Based on anatomic, genetic, and neuropathologic categorizations, the six clinical subtypes of FTD or related disorders are: (1) behavioral variant of FTD, (2) semantic variant primary progressive aphasia, (3) nonfluent agrammatic variant primary progressive aphasia, (4) corticobasal syndrome, (5) progressive supranuclear palsy, and (6) FTD associated with motor neuron disease. Recognition and accurate diagnoses of FTD subtypes will aid the neurologist in the management of patients with FTD.
Collapse
|
164
|
Metzger FG, Schopp B, Haeussinger FB, Dehnen K, Synofzik M, Fallgatter AJ, Ehlis AC. Brain activation in frontotemporal and Alzheimer's dementia: a functional near-infrared spectroscopy study. ALZHEIMERS RESEARCH & THERAPY 2016; 8:56. [PMID: 27931245 PMCID: PMC5146884 DOI: 10.1186/s13195-016-0224-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/16/2016] [Indexed: 12/12/2022]
Abstract
Background Frontotemporal dementia is an increasingly studied disease, the underlying functional impairments on a neurobiological level of which have not been fully understood. Patients with the behavioral-subtype frontotemporal dementia (bvFTD) are particularly challenging for clinical measurements such as functional imaging due to their behavioral symptoms. Here, an alternative imaging method, functional near-infrared spectroscopy (fNIRS), is introduced to measure task-related cortical brain activation based on blood oxygenation. The current study investigated differences in cortical activation patterns of patients with bvFTD, Alzheimer’s dementia (AD), and healthy elderly subjects measured by fNIRS. Method Eight probable bvFTD patients completed the semantic, phonological, and control conditions of a verbal fluency task. Eight AD patients and eight healthy controls were compared on the same task. Simultaneously, an fNIRS measurement was conducted and analyzed using a correction method based on the expected negative correlation between oxygenated and deoxygenated hemoglobin. Results Healthy controls show an increase in cortical activation measured in frontoparietal areas such as the dorsolateral prefrontal cortex. The activation pattern of patients with AD is similar, but weaker. In contrast, bvFTD patients show a more frontopolar pattern, with activation of Broca’s area, instead of the dorsolateral prefrontal cortex and the superior temporal gyrus. The frontoparietal compensation mechanisms, seen in the healthy elderly, were missing in bvFTD patients. Conclusion Different frontoparietal cortical activation patterns may indicate a correlate of diverse pathophysiological mechanisms of AD and bvFTD during verbal fluency processing. The AD pattern is weaker and more similar to the healthy pattern, whereas the bvFTD pattern is qualitatively different, namely more frontopolar and without frontoparietal compensation activation. It adheres to a change of cortical activation during the course of the disease. Electronic supplementary material The online version of this article (doi:10.1186/s13195-016-0224-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Florian G Metzger
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Calwerstraße 14, 72076, Tuebingen, Germany. .,Geriatric Center at the University Hospital of Tuebingen, Calwerstraße 14, 72076, Tuebingen, Germany.
| | - Betti Schopp
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Calwerstraße 14, 72076, Tuebingen, Germany
| | - Florian B Haeussinger
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Calwerstraße 14, 72076, Tuebingen, Germany
| | - Katja Dehnen
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Calwerstraße 14, 72076, Tuebingen, Germany
| | - Matthis Synofzik
- Center of Neurology, Department of Neurodegeneration and Hertie Institute for Clinical Brain Research, University Hospital of Tuebingen, Hoppe-Seyler-Straße 3, 72076, Tuebingen, Germany.,German Center of Neurodegenerative Disorders (DZNE), University Hospital of Tuebingen, Otfried-Müller-Straße 23, 72076, Tuebingen, Germany
| | - Andreas J Fallgatter
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Calwerstraße 14, 72076, Tuebingen, Germany.,German Center of Neurodegenerative Disorders (DZNE), University Hospital of Tuebingen, Otfried-Müller-Straße 23, 72076, Tuebingen, Germany
| | - Ann-Christine Ehlis
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Calwerstraße 14, 72076, Tuebingen, Germany
| |
Collapse
|
165
|
Abstract
Amyloid plaques, along with neurofibrillary tangles, are a neuropathologic hallmark of Alzheimer disease (AD). Recently, amyloid PET radiotracers have been developed and approved for clinical use in the evaluation of suspected neurodegenerative disorders. In both research and clinical settings, amyloid PET imaging has provided important diagnostic and prognostic information for the management of patients with possible AD, mild cognitive impairment (MCI), and other challenging diagnostic presentations. Although the overall impact of amyloid imaging is still being evaluated, the Society of Nuclear Medicine and Molecular Imaging and Alzheimer's Association Amyloid Imaging Task Force have created appropriate use criteria for the standard clinical use of amyloid PET imaging. By the appropriate use criteria, amyloid imaging is appropriate for patients with (1) persistent or unexplained MCI, (2) AD as a possible but still uncertain diagnosis after expert evaluation and (3) atypically early-age-onset progressive dementia. To better understand the clinical and economic effect of amyloid imaging, the Imaging Dementia-Evidence for Amyloid Scanning (IDEAS) study is an ongoing large multicenter study in the United States, which is evaluating how amyloid imaging affects diagnosis, management, and outcomes for cognitively impaired patients who cannot be completely evaluated by clinical assessment alone. Multiple other large-scale studies are evaluating the prognostic role of amyloid PET imaging for predicting MCI progression to AD in general and high-risk populations. At the same time, amyloid imaging is an important tool for evaluating potential disease-modifying therapies for AD. Overall, the increased use of amyloid PET imaging has led to a better understanding of the strengths and limitations of this imaging modality and how it may best be used with other clinical, molecular, and imaging assessment techniques for the diagnosis and management of neurodegenerative disorders.
Collapse
Affiliation(s)
- Atul Mallik
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT.
| | - Alex Drzezga
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| |
Collapse
|
166
|
Markopoulou K, Chase BA, Robowski P, Strongosky A, Narożańska E, Sitek EJ, Berdynski M, Barcikowska M, Baker MC, Rademakers R, Sławek J, Klein C, Hückelheim K, Kasten M, Wszolek ZK. Assessment of Olfactory Function in MAPT-Associated Neurodegenerative Disease Reveals Odor-Identification Irreproducibility as a Non-Disease-Specific, General Characteristic of Olfactory Dysfunction. PLoS One 2016; 11:e0165112. [PMID: 27855167 PMCID: PMC5113898 DOI: 10.1371/journal.pone.0165112] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 10/06/2016] [Indexed: 01/30/2023] Open
Abstract
Olfactory dysfunction is associated with normal aging, multiple neurodegenerative disorders, including Parkinson's disease, Lewy body disease and Alzheimer's disease, and other diseases such as diabetes, sleep apnea and the autoimmune disease myasthenia gravis. The wide spectrum of neurodegenerative disorders associated with olfactory dysfunction suggests different, potentially overlapping, underlying pathophysiologies. Studying olfactory dysfunction in presymptomatic carriers of mutations known to cause familial parkinsonism provides unique opportunities to understand the role of genetic factors, delineate the salient characteristics of the onset of olfactory dysfunction, and understand when it starts relative to motor and cognitive symptoms. We evaluated olfactory dysfunction in 28 carriers of two MAPT mutations (p.N279K, p.P301L), which cause frontotemporal dementia with parkinsonism, using the University of Pennsylvania Smell Identification Test. Olfactory dysfunction in carriers does not appear to be allele specific, but is strongly age-dependent and precedes symptomatic onset. Severe olfactory dysfunction, however, is not a fully penetrant trait at the time of symptom onset. Principal component analysis revealed that olfactory dysfunction is not odor-class specific, even though individual odor responses cluster kindred members according to genetic and disease status. Strikingly, carriers with incipient olfactory dysfunction show poor inter-test consistency among the sets of odors identified incorrectly in successive replicate tests, even before severe olfactory dysfunction appears. Furthermore, when 78 individuals without neurodegenerative disease and 14 individuals with sporadic Parkinson's disease were evaluated twice at a one-year interval using the Brief Smell Identification Test, the majority also showed inconsistency in the sets of odors they identified incorrectly, independent of age and cognitive status. While these findings may reflect the limitations of these tests used and the sample sizes, olfactory dysfunction appears to be associated with the inability to identify odors reliably and consistently, not with the loss of an ability to identify specific odors. Irreproducibility in odor identification appears to be a non-disease-specific, general feature of olfactory dysfunction that is accelerated or accentuated in neurodegenerative disease. It may reflect a fundamental organizational principle of the olfactory system, which is more "error-prone" than other sensory systems.
Collapse
Affiliation(s)
- Katerina Markopoulou
- NorthShore University Health System, Evanston, Illinois, United States of America
- * E-mail:
| | - Bruce A. Chase
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, United States of America
| | - Piotr Robowski
- Department of Neurological and Psychiatric Nursing, Medical University of Gdańsk, Gdańsk, Poland
- Department of Neurology, St. Adalbert Hospital, Copernicus PL Sp. z o.o, Gdańsk, Poland
| | - Audrey Strongosky
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, Florida, United States of America
| | - Ewa Narożańska
- Department of Neurological and Psychiatric Nursing, Medical University of Gdańsk, Gdańsk, Poland
- Department of Neurology, St. Adalbert Hospital, Copernicus PL Sp. z o.o, Gdańsk, Poland
| | - Emilia J. Sitek
- Department of Neurological and Psychiatric Nursing, Medical University of Gdańsk, Gdańsk, Poland
- Department of Neurology, St. Adalbert Hospital, Copernicus PL Sp. z o.o, Gdańsk, Poland
| | - Mariusz Berdynski
- Department of Neurodegenerative Disorders, Mossakowski Medical Research Center, Polish Academy of Sciences, Warsaw, Poland
| | - Maria Barcikowska
- Department of Neurodegenerative Disorders, Mossakowski Medical Research Center, Polish Academy of Sciences, Warsaw, Poland
| | - Matt C. Baker
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, Florida, United States of America
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, Florida, United States of America
| | - Jarosław Sławek
- Department of Neurological and Psychiatric Nursing, Medical University of Gdańsk, Gdańsk, Poland
- Department of Neurology, St. Adalbert Hospital, Copernicus PL Sp. z o.o, Gdańsk, Poland
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Katja Hückelheim
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Meike Kasten
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Zbigniew K. Wszolek
- Department of Neurology, Mayo Clinic Jacksonville, Jacksonville, Florida, United States of America
| |
Collapse
|
167
|
Altmann C, Vasic V, Hardt S, Heidler J, Häussler A, Wittig I, Schmidt MHH, Tegeder I. Progranulin promotes peripheral nerve regeneration and reinnervation: role of notch signaling. Mol Neurodegener 2016; 11:69. [PMID: 27770818 PMCID: PMC5075406 DOI: 10.1186/s13024-016-0132-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 09/28/2016] [Indexed: 01/11/2023] Open
Abstract
Background Peripheral nerve injury is a frequent cause of lasting motor deficits and chronic pain. Although peripheral nerves are capable of regrowth they often fail to re-innervate target tissues. Results Using newly generated transgenic mice with inducible neuronal progranulin overexpression we show that progranulin accelerates axonal regrowth, restoration of neuromuscular synapses and recovery of sensory and motor functions after injury of the sciatic nerve. Oppositely, progranulin deficient mice have long-lasting deficits in motor function tests after nerve injury due to enhanced losses of motor neurons and stronger microglia activation in the ventral horn of the spinal cord. Deep proteome and gene ontology (GO) enrichment analysis revealed that the proteins upregulated in progranulin overexpressing mice were involved in ‘regulation of transcription’ and ‘response to insulin’ (GO terms). Transcription factor prediction pointed to activation of Notch signaling and indeed, co-immunoprecipitation studies revealed that progranulin bound to the extracellular domain of Notch receptors, and this was functionally associated with higher expression of Notch target genes in the dorsal root ganglia of transgenic mice with neuronal progranulin overexpression. Functionally, these transgenic mice recovered normal gait and running, which was not achieved by controls and was stronger impaired in progranulin deficient mice. Conclusion We infer that progranulin activates Notch signaling pathways, enhancing thereby the regenerative capacity of partially injured neurons, which leads to improved motor function recovery. Graphical abstract ![]()
Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0132-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christine Altmann
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Verica Vasic
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stefanie Hardt
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Juliana Heidler
- Functional Proteomics, SFB815 Core Unit, Goethe-University, Frankfurt, Germany
| | - Annett Häussler
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Ilka Wittig
- Functional Proteomics, SFB815 Core Unit, Goethe-University, Frankfurt, Germany
| | - Mirko H H Schmidt
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany.
| |
Collapse
|
168
|
Coppola C, Saracino D, Puoti G, Lus G, Dato C, Le Ber I, Pariente J, Caroppo P, Piccoli E, Tagliavini F, Di Iorio G, Rossi G. A cluster of progranulin C157KfsX97 mutations in Southern Italy: clinical characterization and genetic correlations. Neurobiol Aging 2016; 49:219.e5-219.e13. [PMID: 27814992 DOI: 10.1016/j.neurobiolaging.2016.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/02/2016] [Indexed: 01/08/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is a group of neurodegenerative diseases displaying high clinical, pathologic, and genetic heterogeneity. Several autosomal dominant progranulin (GRN) mutations have been reported, accounting for 5%-10% of FTLD cases worldwide. In this study, we described the clinical characteristics of 7 Italian patients, 5 with a diagnosis of frontotemporal dementia behavioral variant and 2 of corticobasal syndrome (CBS), carrying the GRN deletion g.101349_101355delCTGCTGT, resulting in the C157KfsX97 null mutation, and hypothesized the existence of a founder effect by means of haplotype sharing analysis. We performed plasma progranulin dosage, GRN gene sequencing, and haplotype sharing study, analyzing 10 short tandem repeat markers, spanning a region of 11.08 Mb flanking GRN on chromosome 17q21. We observed shared alleles among 6 patients for 8 consecutive short tandem repeat markers spanning a 7.29 Mb region. Therefore, also with this particular mutation, the elevated clinical variability described among GRN-mutated FTLD cases is confirmed. Moreover, this is the first study reporting the likely existence of a founder effect for C157KfsX97 mutation in Southern Italy.
Collapse
Affiliation(s)
- Cinzia Coppola
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy.
| | - Dario Saracino
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Gianfranco Puoti
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Giacomo Lus
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Clemente Dato
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Isabelle Le Ber
- Institut du Cerveau et de la Moelle épinière (ICM), INSERM U1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UPMC-P6 UMR S 1127 Hôpital de la Pitié-Salpêtrière, Paris, France; AP-HP, Hôpital de la Pitié-Salpêtrière, Centre de Référence des Démences Rares & Fédération des maladies du système nerveux, Paris, France
| | | | - Paola Caroppo
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Piccoli
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fabrizio Tagliavini
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Di Iorio
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Giacomina Rossi
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
169
|
Ljubenkov PA, Miller BL. A Clinical Guide to Frontotemporal Dementias. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2016; 14:448-464. [PMID: 31975825 PMCID: PMC6519586 DOI: 10.1176/appi.focus.20160018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term frontotemporal dementia (FTD) describes a diverse group of clinical syndromes, including behavioral-variant FTD (bvFTD), nonfluent/agrammatic-variant primary progressive aphasia (nfvPPA), semantic-variant primary progressive aphasia (svPPA), FTD motor neuron disease (FTD-MND), progressive supranuclear palsy syndrome (PSP-S), and corticobasal syndrome (CBS). Although each of these syndromes may be distinguished by their respective disturbances in behavior, language, or motor function and characteristic imaging findings, they may present a diagnostic dilemma when encountered clinically. In this article, we review the clinical features, diagnostic criteria, pathology, genetics, and therapeutic interventions for FTD spectrum disorders.
Collapse
Affiliation(s)
- Peter A Ljubenkov
- Dr. Ljubenkov is a clinical fellow and Dr. Miller is professor of neurology in the Department of Neurology, University of California, San Francisco, School of Medicine (e-mail: )
| | - Bruce L Miller
- Dr. Ljubenkov is a clinical fellow and Dr. Miller is professor of neurology in the Department of Neurology, University of California, San Francisco, School of Medicine (e-mail: )
| |
Collapse
|
170
|
Wen X, Westergard T, Pasinelli P, Trotti D. Pathogenic determinants and mechanisms of ALS/FTD linked to hexanucleotide repeat expansions in the C9orf72 gene. Neurosci Lett 2016; 636:16-26. [PMID: 27619540 DOI: 10.1016/j.neulet.2016.09.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/12/2016] [Accepted: 09/06/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two apparently distinct neurodegenerative diseases, the former characterized by selective loss of motor neurons in the brain and spinal cord and the latter characterized by selective atrophy of frontal and temporal lobes. Over the years, however, growing evidence from clinical, pathological and genetic findings has suggested that ALS and FTD belong to the same clinic-pathological spectrum disorder. This concept has been further supported by the identification of the most common genetic cause for both diseases, an aberrantly expanded hexanucleotide repeat GGGGCC/ CCCCGG sequence located in a non-coding region of the gene C9orf72. Three hypotheses have been proposed to explain how this repeats expansion causes diseases: 1) C9orf72 haploinsufficiency-expanded repeats interfere with transcription or translation of the gene, leading to decreased expression of the C9orf72 protein; 2) RNA gain of function-RNA foci formed by sense and antisense transcripts of expanded repeats interact and sequester essential RNA binding proteins, causing neurotoxicity; 3) Repeat associated non-ATG initiated (RAN) translation of expanded sense GGGGCC and antisense CCCCGG repeats produces potential toxic dipeptide repeat protein (DPR). In this review, we assess current evidence supporting or arguing against each proposed mechanism in C9 ALS/FTD disease pathogenesis. Additionally, controversial findings are also discussed. Lastly, we discuss the possibility that the three pathogenic mechanisms are not mutually exclusive and all three might be involved in disease.
Collapse
Affiliation(s)
- Xinmei Wen
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Thomas Westergard
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Piera Pasinelli
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
171
|
Abstract
Today, frontotemporal dementia (FTD) remains one of the most common forms of early-onset dementia, that is, before the age of 65, thus posing several diagnostic challenges to clinicians since symptoms are often mistaken for psychiatric or neurological diseases causing a delay in correct diagnosis, and the majority of patients with FTD present with symptoms at ages between 50 and 60. Genetic components are established risk factors for FTD, but the influence of lifestyle, comorbidity, and environmental factors on the risk of FTD is still unclear. Approximately 40% of individuals with FTD have a family history of dementia but less than 10% have a clear autosomal dominant pattern of inheritance. Lack of insight is often an early clue to FTD. A tailored treatment option at an early phase can mitigate suffering and improve patients' and caregivers' quality of life.
Collapse
Affiliation(s)
| | - Knut Engedal
- Norwegian National Advisory Unit on Ageing and Health, Tønsberg, Norway
| | - Zeina Chemali
- Department of Neurology and Psychiatry, Neuropsychiatry Clinics, Massachusetts General Hospital, Boston, MA, USA Department of Psychiatry, Neuropsychiatry Clinics, Massachusetts General Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| |
Collapse
|
172
|
Stoica R, Paillusson S, Gomez-Suaga P, Mitchell JC, Lau DH, Gray EH, Sancho RM, Vizcay-Barrena G, De Vos KJ, Shaw CE, Hanger DP, Noble W, Miller CC. ALS/FTD-associated FUS activates GSK-3β to disrupt the VAPB-PTPIP51 interaction and ER-mitochondria associations. EMBO Rep 2016; 17:1326-42. [PMID: 27418313 PMCID: PMC5007559 DOI: 10.15252/embr.201541726] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 05/06/2016] [Accepted: 06/13/2016] [Indexed: 12/12/2022] Open
Abstract
Defective FUS metabolism is strongly associated with amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), but the mechanisms linking FUS to disease are not properly understood. However, many of the functions disrupted in ALS/FTD are regulated by signalling between the endoplasmic reticulum (ER) and mitochondria. This signalling is facilitated by close physical associations between the two organelles that are mediated by binding of the integral ER protein VAPB to the outer mitochondrial membrane protein PTPIP51, which act as molecular scaffolds to tether the two organelles. Here, we show that FUS disrupts the VAPB-PTPIP51 interaction and ER-mitochondria associations. These disruptions are accompanied by perturbation of Ca(2+) uptake by mitochondria following its release from ER stores, which is a physiological read-out of ER-mitochondria contacts. We also demonstrate that mitochondrial ATP production is impaired in FUS-expressing cells; mitochondrial ATP production is linked to Ca(2+) levels. Finally, we demonstrate that the FUS-induced reductions to ER-mitochondria associations and are linked to activation of glycogen synthase kinase-3β (GSK-3β), a kinase already strongly associated with ALS/FTD.
Collapse
Affiliation(s)
- Radu Stoica
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Sébastien Paillusson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Patricia Gomez-Suaga
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Jacqueline C Mitchell
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Dawn Hw Lau
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Emma H Gray
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Rosa M Sancho
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | | | - Kurt J De Vos
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Christopher E Shaw
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Diane P Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Christopher Cj Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| |
Collapse
|
173
|
Yang M, Liang C, Swaminathan K, Herrlinger S, Lai F, Shiekhattar R, Chen JF. A C9ORF72/SMCR8-containing complex regulates ULK1 and plays a dual role in autophagy. SCIENCE ADVANCES 2016; 2:e1601167. [PMID: 27617292 PMCID: PMC5010369 DOI: 10.1126/sciadv.1601167] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/30/2016] [Indexed: 05/10/2023]
Abstract
The intronic GGGGCC hexanucleotide repeat expansion in chromosome 9 open reading frame 72 (C9ORF72) is a prevalent genetic abnormality identified in both frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Smith-Magenis syndrome chromosomal region candidate gene 8 (SMCR8) is a protein with unclear functions. We report that C9ORF72 is a component of a multiprotein complex containing SMCR8, WDR41, and ATG101 (an important regulator of autophagy). The C9ORF72 complex displays guanosine triphosphatase (GTPase) activity and acts as a guanosine diphosphate-guanosine 5'-triphosphate (GDP-GTP) exchange factor (GEF) for RAB39B. We created Smcr8 knockout mice and found that Smcr8 mutant cells exhibit impaired autophagy induction, which is similarly observed in C9orf72 knockdown cells. Mechanistically, SMCR8/C9ORF72 interacts with the key autophagy initiation ULK1 complex and regulates expression and activity of ULK1. The complex has an additional role in regulating later stages of autophagy. Whereas autophagic flux is enhanced in C9orf72 knockdown cells, depletion of Smcr8 results in a reduced flux with an abnormal expression of lysosomal enzymes. Thus, C9ORF72 and SMCR8 have similar functions in modulating autophagy induction by regulating ULK1 and play distinct roles in regulating autophagic flux.
Collapse
Affiliation(s)
- Mei Yang
- Department of Genetics and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Chen Liang
- Department of Genetics and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | | | - Stephanie Herrlinger
- Department of Genetics and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Fan Lai
- Department of Human Genetics, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ramin Shiekhattar
- Department of Human Genetics, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Corresponding author. (J.-F.C.); (R.S.); (F.L.)
| | - Jian-Fu Chen
- Department of Genetics and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
- Corresponding author. (J.-F.C.); (R.S.); (F.L.)
| |
Collapse
|
174
|
Suzuki H, Matsuoka M. The Lysosomal Trafficking Transmembrane Protein 106B Is Linked to Cell Death. J Biol Chem 2016; 291:21448-21460. [PMID: 27563066 DOI: 10.1074/jbc.m116.737171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 08/24/2016] [Indexed: 12/12/2022] Open
Abstract
A common genetic variation in the transmembrane protein 106B (TMEM106B) gene has been suggested to be a risk factor for frontotemporal lobar degeneration (FTLD) with inclusions of transactive response DNA-binding protein-43 (TDP-43) (FTLD-TDP), the most common pathological subtype in FTLD. Furthermore, previous studies have shown that TMEM106B levels are up-regulated in the brains of FTLD-TDP patients, although the significance of this finding remains unknown. In this study, we show that the overexpression of TMEM106B and its N-terminal fragments induces cell death, enhances oxidative stress-induced cytotoxicity, and causes the cleavage of TDP-43, which represents TDP-43 pathology, using cell-based models. TMEM106B-induced death is mediated by the caspase-dependent mitochondrial cell death pathways and possibly by the lysosomal cell death pathway. These findings suggest that the up-regulation of TMEM106B may increase the risk of FTLD by directly causing neurotoxicity and a pathological phenotype linked to FTLD-TDP.
Collapse
Affiliation(s)
| | - Masaaki Matsuoka
- From the Departments of Pharmacology and .,Dermatological Neuroscience, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| |
Collapse
|
175
|
Papegaey A, Eddarkaoui S, Deramecourt V, Fernandez-Gomez FJ, Pantano P, Obriot H, Machala C, Anquetil V, Camuzat A, Brice A, Maurage CA, Le Ber I, Duyckaerts C, Buée L, Sergeant N, Buée-Scherrer V. Reduced Tau protein expression is associated with frontotemporal degeneration with progranulin mutation. Acta Neuropathol Commun 2016; 4:74. [PMID: 27435172 PMCID: PMC4952067 DOI: 10.1186/s40478-016-0345-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/10/2016] [Indexed: 12/12/2022] Open
Abstract
Reduction of Tau protein expression was described in 2003 by Zhukareva et al. in a variant of frontotemporal lobar degeneration (FTLD) referred to as diagnosis of dementia lacking distinctive histopathology, then re-classified as FTLD with ubiquitin inclusions. However, the analysis of Tau expression in FTLD has not been reconsidered since then. Knowledge of the molecular basis of protein aggregates and genes that are mutated in the FTLD spectrum would enable to determine whether the “Tau-less” is a separate pathological entity or if it belongs to an existing subclass of FTLD. To address this question, we have analyzed Tau expression in the frontal brain areas from control, Alzheimer’s disease and FTLD cases, including FTLD- Tau (MAPT), FTLD-TDP (sporadic, FTLD-TDP-GRN, FTLD-TDP-C9ORF72) and sporadic FTLD-FUS, using western blot and 2D-DIGE (Two-Dimensional fluorescence Difference Gel Electrophoresis) approaches. Surprisingly, we found that most of the FTLD-TDP-GRN brains are characterized by a huge reduction of Tau protein expression without any decrease in Tau mRNA levels. Interestingly, only cases affected by point mutations, rather than cases with total deletion of one GRN allele, seem to be affected by this reduction of Tau protein expression. Moreover, proteomic analysis highlighted correlations between reduced Tau protein level, synaptic impairment and massive reactive astrogliosis in these FTLD-GRN cases. Consistent with a recent study, our data also bring new insights regarding the role of progranulin in neurodegeneration by suggesting its involvement in lysosome and synaptic regulation. Together, our results demonstrate a strong association between progranulin deficiency and reduction of Tau protein expression that could lead to severe neuronal and glial dysfunctions. Our study also indicates that this FTLD-TDP-GRN subgroup could be part as a distinct entity of FTLD classification.
Collapse
|
176
|
Abstract
Neurodegenerative disorders such as Alzheimer disease (AD), frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), Parkinson disease (PD), Huntington's disease (HD), and multiple sclerosis (MS) affect different neuronal cells, and have a variable age of onset, clinical symptoms, and pathological features. Despite the great progress in understanding the etiology of these disorders, the underlying mechanisms remain largely unclear. Among the processes affected in neurodegenerative diseases, alteration in RNA metabolism is emerging as a crucial player. RNA-binding proteins (RBPs) are involved at all stages of RNA metabolism and display a broad range of functions, including modulation of mRNA transcription, splicing, editing, export, stability, translation and localization and miRNA biogenesis, thus enormously impacting regulation of gene expression. On the other hand, aberrant regulation of RBP expression or activity can contribute to disease onset and progression. Recent reports identified mutations causative of neurological disorders in the genes encoding a family of RBPs named FET (FUS/TLS, EWS and TAF15). This review summarizes recent works documenting the involvement of FET proteins in the pathology of ALS, FTLD, essential tremor (ET) and other neurodegenerative diseases. Moreover, clinical implications of recent advances in FET research are critically discussed.
Collapse
Affiliation(s)
- Francesca Svetoni
- a University of Rome "Foro Italico," , Rome , Italy.,b Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia , Rome , Italy
| | - Paola Frisone
- b Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia , Rome , Italy
| | - Maria Paola Paronetto
- a University of Rome "Foro Italico," , Rome , Italy.,b Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia , Rome , Italy
| |
Collapse
|
177
|
Chen H, Liu S, Zheng M, Ji L, Wu T, Huang G, Ji Y. Factors Associated with Frontotemporal Dementia in China: A Cross-Sectional Study. Arch Med Res 2016; 47:388-393. [DOI: 10.1016/j.arcmed.2016.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022]
|
178
|
Nakamagoe K, Kadono K, Koganezawa T, Takiguchi M, Terada M, Yamamoto F, Moriyama T, Yanagiha K, Nohara S, Tozaka N, Miyake Z, Aizawa S, Furusho K, Tamaoka A. Vestibular Impairment in Frontotemporal Dementia Syndrome. Dement Geriatr Cogn Dis Extra 2016; 6:194-204. [PMID: 27350780 PMCID: PMC4913767 DOI: 10.1159/000445870] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Background No studies to date have attempted to evaluate frontotemporal lobar degeneration from the perspective of the vestibular system. Objective The present study examined vestibular function in patients with frontotemporal dementia (FTD) clinical syndrome and evaluated whether vestibular disorders are involved in the clinical symptoms due to FTD. Methods Fourteen patients with FTD syndrome, as well as healthy elderly controls without dementia, were included in the present study. All subjects underwent vestibular function tests using electronystagmography, such as caloric tests and visual suppression (VS) tests, in which the induced caloric nystagmus was suppressed by visual stimuli. The association between clinical symptoms and vestibular function in the FTD syndrome group was further examined. Results In the FTD syndrome group, caloric nystagmus was not necessarily suppressed during VS tests. Furthermore, VS was observed to be significantly impaired in FTD syndrome patients with gait disturbance as compared to those without such disturbance. Conclusion The present study revealed that impairment of VS in patients with FTD results in an inability to regulate vestibular function by means of visual perception, regardless of multiple presumed neuropathological backgrounds. This could also be associated with gait disturbance in patients with FTD syndrome.
Collapse
Affiliation(s)
- Kiyotaka Nakamagoe
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kotarou Kadono
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tadachika Koganezawa
- Department of Physiology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Mao Takiguchi
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Makoto Terada
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Fumiko Yamamoto
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tetsuya Moriyama
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kumi Yanagiha
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Seitaro Nohara
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoki Tozaka
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Zenshi Miyake
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoshi Aizawa
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kentaro Furusho
- Department of Neurology, Ryugasaki Saiseikai Hospital, Ryugasaki, Japan
| | - Akira Tamaoka
- Department of Neurology, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
179
|
The inhibition of TDP-43 mitochondrial localization blocks its neuronal toxicity. Nat Med 2016; 22:869-78. [PMID: 27348499 PMCID: PMC4974139 DOI: 10.1038/nm.4130] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 05/23/2016] [Indexed: 12/11/2022]
Abstract
Genetic mutations in TAR DNA-binding protein 43 (TARDBP, also known as TDP-43) cause amyotrophic lateral sclerosis (ALS), and an increase in the presence of TDP-43 (encoded by TARDBP) in the cytoplasm is a prominent histopathological feature of degenerating neurons in various neurodegenerative diseases. However, the molecular mechanisms by which TDP-43 contributes to ALS pathophysiology remain elusive. Here we have found that TDP-43 accumulates in the mitochondria of neurons in subjects with ALS or frontotemporal dementia (FTD). Disease-associated mutations increase TDP-43 mitochondrial localization. In mitochondria, wild-type (WT) and mutant TDP-43 preferentially bind mitochondria-transcribed messenger RNAs (mRNAs) encoding respiratory complex I subunits ND3 and ND6, impair their expression and specifically cause complex I disassembly. The suppression of TDP-43 mitochondrial localization abolishes WT and mutant TDP-43-induced mitochondrial dysfunction and neuronal loss, and improves phenotypes of transgenic mutant TDP-43 mice. Thus, our studies link TDP-43 toxicity directly to mitochondrial bioenergetics and propose the targeting of TDP-43 mitochondrial localization as a promising therapeutic approach for neurodegeneration.
Collapse
|
180
|
Mackenzie IRA, Neumann M. Molecular neuropathology of frontotemporal dementia: insights into disease mechanisms from postmortem studies. J Neurochem 2016; 138 Suppl 1:54-70. [PMID: 27306735 DOI: 10.1111/jnc.13588] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/12/2016] [Accepted: 02/16/2016] [Indexed: 12/13/2022]
Abstract
Frontotemporal dementia (FTD) is a clinical syndrome with a heterogeneous molecular basis. The past decade has seen the discovery of several new FTD-causing genetic mutations and the identification of many of the relevant pathological proteins. The current neuropathological classification is based on the predominant protein abnormality and allows most cases of FTD to be placed into one of three broad molecular subgroups; frontotemporal lobar degeneration with tau, TDP-43 or FET protein accumulation. This review will describe our current understanding of the molecular basis of FTD, focusing on insights gained from the study of human postmortem tissue, as well as some of the current controversies. Most cases of FTD can be subclassified into one of three broad molecular subgroups based on the predominant protein that accumulates as pathological cellular inclusions. Understanding the associated pathogenic mechanisms and recognizing these FTD molecular subtypes in vivo will likely be crucial for the development and use of targeted therapies. This article is part of the Frontotemporal Dementia special issue.
Collapse
Affiliation(s)
- Ian R A Mackenzie
- Department of Pathology, University of British Columbia and Vancouver General Hospital, Vancouver, Canada
| | - Manuela Neumann
- Department of Neuropathology, University of Tübingen and German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| |
Collapse
|
181
|
Patel AN, Sampson JB. Cognitive Profile of C9orf72 in Frontotemporal Dementia and Amyotrophic Lateral Sclerosis. Curr Neurol Neurosci Rep 2016. [PMID: 26198888 DOI: 10.1007/s11910-015-0582-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This review article focuses on the cognitive profile associated with the C9orf72 gene with GGGGCC (G4C2) hexanucleotide repeat expansions that is commonly found in both familial and sporadic forms of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) in order to aid clinicians in the screening process. In this growing clinical continuum between FTD and ALS, understanding and recognizing a neurocognitive profile is important for diagnosis. Key features of this profile include executive dysfunction with memory impairment and language deficits as the disease progresses. Behaviorally, patients are prone to disinhibition, apathy, and psychosis. With the discovery of this mutation, studies have begun to characterize the different phenotypes associated with this mutation in terms of epidemiology, clinical presentation, imaging, and pathology. Greater awareness and increased surveillance for this mutation will benefit patients and their families in terms of access to genetic counseling, research studies, and improved understanding of the disease process.
Collapse
Affiliation(s)
- Anjali N Patel
- Division of Aging and Dementia, Columbia University Medical Center, 622 West 168th Street, PH-19, Room 121, New York, NY, 10032, USA,
| | | |
Collapse
|
182
|
Sasaguri H, Chew J, Xu YF, Gendron TF, Garrett A, Lee CW, Jansen-West K, Bauer PO, Perkerson EA, Tong J, Stetler C, Zhang YJ. The extreme N-terminus of TDP-43 mediates the cytoplasmic aggregation of TDP-43 and associated toxicity in vivo. Brain Res 2016; 1647:57-64. [PMID: 27155453 DOI: 10.1016/j.brainres.2016.04.069] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 12/13/2022]
Abstract
Inclusions of Tar DNA- binding protein 43 (TDP-43) are a pathological hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with TDP-43-positive inclusions (FTLD-TDP). Pathological TDP-43 exhibits the disease-specific biochemical signatures, which include its ubiquitination, phosphorylation and truncation. Recently, we demonstrated that the extreme N-terminus of TDP-43 regulates formation of abnormal cytoplasmic TDP-43 aggregation in cultured cells and primary neurons. However, it remained unclear whether this N-terminal domain mediates TDP-43 aggregation and the associated toxicity in vivo. To investigate this, we expressed a GFP-tagged TDP-43 with a nuclear localization signal mutation (GFP-TDP-43NLSm) and a truncated form without the extreme N-terminus (GFP-TDP-4310-414-NLSm) by adeno-associated viral (AAV) vectors in mouse primary cortical neurons and murine central nervous system. Compared to neurons containing GFP alone, expression of GFP-TDP-43NLSm resulted in the formation of ubiquitin-positive cytoplasmic inclusions and activation of caspase-3, an indicator of cell death. Moreover, mice expressing GFP-TDP-43NLSm proteins show reactive gliosis and develop neurological abnormalities. However, by deletion of TDP-43's extreme N-terminus, these pathological alterations can be abrogated. Together, our study provides further evidence confirming the critical role of the extreme N-terminus of TDP-43 in regulating protein structure as well as mediating toxicity associated with its aggregation. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
Affiliation(s)
- Hiroki Sasaguri
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Jeannie Chew
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA; Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Ya-Fei Xu
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Aliesha Garrett
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Chris W Lee
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Karen Jansen-West
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Peter O Bauer
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Emilie A Perkerson
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Jimei Tong
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Caroline Stetler
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| |
Collapse
|
183
|
Gasca-Salas C, Masellis M, Khoo E, Shah BB, Fisman D, Lang AE, Kleiner-Fisman G. Characterization of Movement Disorder Phenomenology in Genetically Proven, Familial Frontotemporal Lobar Degeneration: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0153852. [PMID: 27100392 PMCID: PMC4839564 DOI: 10.1371/journal.pone.0153852] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/05/2016] [Indexed: 12/11/2022] Open
Abstract
Background Mutations in granulin (PGRN) and tau (MAPT), and hexanucleotide repeat expansions near the C9orf72 genes are the most prevalent genetic causes of frontotemporal lobar degeneration. Although behavior, language and movement presentations are common, the relationship between genetic subgroup and movement disorder phenomenology is unclear. Objective We conducted a systematic review and meta-analysis of the literature characterizing the spectrum and prevalence of movement disorders in genetic frontotemporal lobar degeneration. Methods Electronic databases were searched using terms related to frontotemporal lobar degeneration and movement disorders. Articles were included when cases had a proven genetic cause. Study-specific prevalence estimates for clinical features were transformed using Freeman-Tukey arcsine transformation, allowing for pooled estimates of prevalence to be generated using random-effects models. Results The mean age at onset was earlier in those with MAPT mutations compared to PGRN (p<0.001) and C9orf72 (p = 0.024). 66.5% of subjects had an initial non-movement presentation that was most likely a behavioral syndrome (35.7%). At any point during the disease, parkinsonism was the most common movement syndrome reported in 79.8% followed by progressive supranuclear palsy (PSPS) and corticobasal (CBS) syndromes in 12.2% and 10.7%, respectively. The prevalence of movement disorder as initial presentation was higher in MAPT subjects (35.8%) compared to PGRN subjects (10.1). In those with a non-movement presentation, language disorder was more common in PGRN subjects (18.7%) compared to MAPT subjects (5.4%). Summary This represents the first systematic review and meta-analysis of the occurrence of movement disorder phenomenology in genetic frontotemporal lobar degeneration. Standardized prospective collection of clinical information in conjunction with genetic characterization will be crucial for accurate clinico-genetic correlation.
Collapse
Affiliation(s)
- Carmen Gasca-Salas
- The Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson's Disease, TWH, Toronto, Canada
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, Canada
- Centro integral en Neurociencias A.C. (CINAC)/HM Hospitales- Puerta del Sur, CEU-San Pablo University, Madrid, Spain
- * E-mail:
| | - Mario Masellis
- Centro integral en Neurociencias A.C. (CINAC)/HM Hospitales- Puerta del Sur, CEU-San Pablo University, Madrid, Spain
- Cognitive & Movement Disorders Clinic, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Edwin Khoo
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Binit B. Shah
- Department of Neurology, University of Virginia, Charlottesville, Virginia, United States of America
| | - David Fisman
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Anthony E. Lang
- The Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson's Disease, TWH, Toronto, Canada
| | - Galit Kleiner-Fisman
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, Canada
- Jeff and Diane Ross Movement Disorders Clinic, Baycrest Center for Geriatric Health, Toronto, Canada
| |
Collapse
|
184
|
Kawakami I, Kobayashi Z, Arai T, Yokota O, Nonaka T, Aoki N, Niizato K, Oshima K, Higashi S, Katsuse O, Hosokawa M, Hasegawa M, Akiyama H. Chorea as a clinical feature of the basophilic inclusion body disease subtype of fused-in-sarcoma-associated frontotemporal lobar degeneration. Acta Neuropathol Commun 2016; 4:36. [PMID: 27044537 PMCID: PMC4820861 DOI: 10.1186/s40478-016-0304-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/18/2016] [Indexed: 12/14/2022] Open
Abstract
Choreoathetoid involuntary movements are rarely reported in patients with frontotemporal lobar degeneration (FTLD), suggesting their exclusion as a supportive feature in clinical diagnostic criteria for FTLD. Here, we identified three cases of the behavioral variant of frontotemporal dementia (bvFTD) that display chorea with fused in sarcoma (FUS)-positive inclusions (FTLD-FUS) and the basophilic inclusion body disease (BIBD) subtype. We determined the behavioral and cognitive features in this group that were distinct from other FTLD-FUS cases. We also reviewed the clinical records of 72 FTLD cases, and clarified additional clinical features that are predictive of the BIBD pathology. Symptom onset in the three patients with chorea was at 44.0 years of age (±12.0 years), and occurred in the absence of a family history of dementia. The cases were consistent with a clinical form of FTD known as bvFTD, as well as reduced neurological muscle tone in addition to chorea. The three patients showed no or mild parkinsonism, which by contrast, increased substantially in the other FTLD cases until a later stage of disease. The three patients exhibited severe caudate atrophy, which has previously been reported as a histological feature distinguishing FTLD-FUS from FTLD-tau or FTLD-TAR DNA-binding protein 43. Thus, our findings suggest that the clinical feature of choreoathetosis in bvFTD might be associated with FTLD-FUS, and in particular, with the BIBD subtype.
Collapse
Affiliation(s)
- Ito Kawakami
- />Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
- />Department of Psychiatry, Tokyo Metropolitan Matsuzawa Hospital (TMMH), Tokyo, Japan
| | - Zen Kobayashi
- />Department of Neurology, JA Toride Medical Center, Ibaraki, Japan
| | - Tetsuaki Arai
- />Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
- />Division of Clinical Medicine, Department of Neuropsychiatry, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Osamu Yokota
- />Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takashi Nonaka
- />Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Naoya Aoki
- />Department of Psychiatry, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Kazuhiro Niizato
- />Department of Psychiatry, Tokyo Metropolitan Matsuzawa Hospital (TMMH), Tokyo, Japan
| | - Kenichi Oshima
- />Department of Psychiatry, Tokyo Metropolitan Matsuzawa Hospital (TMMH), Tokyo, Japan
| | - Shinji Higashi
- />Division of Clinical Medicine, Department of Neuropsychiatry, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Omi Katsuse
- />Department of Psychiatry, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Masato Hosokawa
- />Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Masato Hasegawa
- />Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| | - Haruhiko Akiyama
- />Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506 Japan
| |
Collapse
|
185
|
|
186
|
Benussi L, Ciani M, Tonoli E, Morbin M, Palamara L, Albani D, Fusco F, Forloni G, Glionna M, Baco M, Paterlini A, Fostinelli S, Santini B, Galbiati E, Gagni P, Cretich M, Binetti G, Tagliavini F, Prosperi D, Chiari M, Ghidoni R. Loss of exosomes in progranulin-associated frontotemporal dementia. Neurobiol Aging 2016; 40:41-49. [DOI: 10.1016/j.neurobiolaging.2016.01.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 12/04/2015] [Accepted: 01/02/2016] [Indexed: 02/04/2023]
|
187
|
Impulsivity, decreased social exploration, and executive dysfunction in a mouse model of frontotemporal dementia. Neurobiol Learn Mem 2016; 130:34-43. [DOI: 10.1016/j.nlm.2016.01.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 01/07/2016] [Accepted: 01/16/2016] [Indexed: 12/12/2022]
|
188
|
Abstract
Although psychotic features have long been recognized in association with frontotemporal dementia (FTD), recent genetic discoveries enabling further subtyping of FTD have revealed that psychotic symptoms are frequent in some forms of FTD. Hallucinations and delusions can even precede onset of other cognitive or behavioural symptoms in patients with FTD. In this review, we explore the frequency and types of psychotic symptoms reported in patients with FTD, as well as in other neuropsychiatric disorders, to aid practitioners' consideration of these features in the diagnosis of FTD and related disorders.
Collapse
|
189
|
Krasniak CS, Ahmad ST. The role of CHMP2B Intron5 in autophagy and frontotemporal dementia. Brain Res 2016; 1649:151-157. [PMID: 26972529 DOI: 10.1016/j.brainres.2016.02.051] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 02/05/2016] [Accepted: 02/10/2016] [Indexed: 12/12/2022]
Abstract
Charged multivesicular body protein 2B (CHMP2B) - a component of the endosomal complex required for transport-III (ESCRT-III) - is responsible for the vital membrane deformation functions in autophagy and endolysosomal trafficking. A dominant mutation in CHMP2B (CHMP2BIntron5) is associated with a subset of heritable frontotemporal dementia - frontotemporal dementia linked to chromosome 3 (FTD-3). ESCRT-III recruits Vps4, an AAA-ATPase that abscises the membrane during various cellular processes including autophagy and intraluminal vesicle formation. CHMP2BIntron5 results in a C-terminus truncation removing an important Vps4 binding site as well as eliminating the normal autoinhibitory resting state of CHMP2B. CHMP2B is expressed in most cell types but seems to be especially vital for proper neuronal function. CHMP2BIntron5-mediated phenotypes include misregulation of transmembrane receptors, accumulation of multilamellar structures, abnormal lysosomal morphology, down regulation of a brain-specific micro RNA (miRNA-124), abnormal dendritic spine morphology, decrease in dendritic arborization, and cell death. Currently, transgenic-fly,-mouse, and -human cell lines are being used to better understand the diverse phenotypes and develop therapeutic approaches for the CHMP2BIntron5-induced FTD-3. This article is part of a Special Issue entitled SI:Autophagy.
Collapse
Affiliation(s)
| | - S Tariq Ahmad
- Department of Biology, Colby College, 5720 Mayflower Hill, Waterville, ME 04901, USA.
| |
Collapse
|
190
|
Premi E, Cauda F, Costa T, Diano M, Gazzina S, Gualeni V, Alberici A, Archetti S, Magoni M, Gasparotti R, Padovani A, Borroni B. Looking for Neuroimaging Markers in Frontotemporal Lobar Degeneration Clinical Trials: A Multi-Voxel Pattern Analysis Study in Granulin Disease. J Alzheimers Dis 2016; 51:249-62. [DOI: 10.3233/jad-150340] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Enrico Premi
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Stroke Unit, Azienda Ospedaliera “Spedali Civili”, “Spedali Civili” Hospital, Brescia, Italy
| | - Franco Cauda
- GCS fMRI Koelliker Hospital, Turin, Italy
- Department of Psychology, University of Turin, Turin, Italy
| | - Tommaso Costa
- GCS fMRI Koelliker Hospital, Turin, Italy
- Department of Psychology, University of Turin, Turin, Italy
| | - Matteo Diano
- GCS fMRI Koelliker Hospital, Turin, Italy
- Department of Psychology, University of Turin, Turin, Italy
| | - Stefano Gazzina
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Vera Gualeni
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Antonella Alberici
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvana Archetti
- III Laboratory of Analyses, Azienda Ospedaliera “Spedali Civili”, “Spedali Civili” Hospital, Brescia, Italy
| | - Mauro Magoni
- Stroke Unit, Azienda Ospedaliera “Spedali Civili”, “Spedali Civili” Hospital, Brescia, Italy
| | - Roberto Gasparotti
- Neuroradiology Unit, Department of Surgery, Radiology and Public Health, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Barbara Borroni
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
191
|
An amyloid-like cascade hypothesis for C9orf72 ALS/FTD. Curr Opin Neurobiol 2016; 36:99-106. [DOI: 10.1016/j.conb.2015.10.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/08/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
|
192
|
Karch CM, Ezerskiy L, Redaelli V, Giovagnoli AR, Tiraboschi P, Pelliccioni G, Pelliccioni P, Kapetis D, D'Amato I, Piccoli E, Ferretti MG, Tagliavini F, Rossi G. Missense mutations in progranulin gene associated with frontotemporal lobar degeneration: study of pathogenetic features. Neurobiol Aging 2016; 38:215.e1-215.e12. [PMID: 26652843 PMCID: PMC4738142 DOI: 10.1016/j.neurobiolaging.2015.10.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/18/2015] [Accepted: 10/28/2015] [Indexed: 10/22/2022]
Abstract
GRN, the gene coding for the progranulin (PGRN) protein, was recognized as a gene linked to frontotemporal lobar degeneration (FTLD). The first mutations identified were null mutations giving rise to haploinsufficiency. Missense mutations were subsequently detected, but only a small subset has been functionally investigated. We identified missense mutations (C105Y, A199V, and R298H) in FTLD cases with family history and/or with low plasma PGRN levels. The aim of this study was to determine their pathogenicity. We performed functional studies, analyzing PGRN expression, secretion, and cleavage by elastase. GRN C105Y affected both secretion and elastase cleavage, likely representing a pathogenic mutation. GRN A199V did not alter the physiological properties of PGRN and GRN R298H produced only moderate effects on PGRN secretion, indicating that their pathogenicity is uncertain. In the absence of strong segregation data and neuropathological examinations, genetic, biomarker, and functional studies can be applied to an algorithm to assess the likelihood of pathogenicity for a mutation. This information can improve our understanding of the complex mechanisms by which GRN mutations lead to FTLD.
Collapse
Affiliation(s)
- Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA; Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St Louis, MO, USA
| | - Lubov Ezerskiy
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Veronica Redaelli
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Anna Rita Giovagnoli
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Pietro Tiraboschi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | - Paolo Pelliccioni
- Division of Neurology, Geriatric Hospital, INRCA-IRCCS, Ancona, Italy
| | - Dimos Kapetis
- Bioinformatics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Ilaria D'Amato
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Elena Piccoli
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Maria Giulia Ferretti
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Fabrizio Tagliavini
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giacomina Rossi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy.
| |
Collapse
|
193
|
C9orf72 repeat expansions that cause frontotemporal dementia are detectable among patients with psychosis. Psychiatry Res 2016; 235:200-2. [PMID: 26723138 PMCID: PMC4724461 DOI: 10.1016/j.psychres.2015.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/19/2015] [Accepted: 12/07/2015] [Indexed: 12/12/2022]
Abstract
A pathologic hexanucleotide repeat expansion in C9orf72 causes frontotemporal dementia (FTD) or amyotrophic lateral sclerosis (ALS). Behavioral abnormalities can also occur among mutation carriers with FTD, but it is uncertain whether such mutations occur among persons with psychoses per se. Among participants in a genetic study of psychoses (N=739), two pairs of related individuals had C9orf72 expansions, of whom three were diagnosed with schizophrenia (SZ) / schizoaffective disorder (SZA), but their clinical features did not suggest dementia or ALS. A few patients with SZ/SZA carry C9orf72 repeat expansions; such individuals are highly likely to develop FTD/ALS.
Collapse
|
194
|
Kim S, Kim MK, Oh D, Lee SH, Kim B. Induced Pluripotent Stem Cells as a Novel Tool in Psychiatric Research. Psychiatry Investig 2016; 13:8-17. [PMID: 26766942 PMCID: PMC4701689 DOI: 10.4306/pi.2016.13.1.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/11/2015] [Accepted: 06/26/2015] [Indexed: 12/19/2022] Open
Abstract
Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) provides a valuable opportunity to study neurodevelopmental and neurodegenerative psychiatric diseases by offering an unlimited source for patient-specific neuronal and glial cells. The present review focuses on the recent advancements in modeling psychiatric disorders such as Phelan-McDermid syndrome, Timothy syndrome, Rett syndrome, schizophrenia, bipolar disorder, and dementia. The treatment effects identified in studies on iPSCs using known therapeutic compounds are also summarized in this review. Here we discuss validation of cellular models and explore iPSCs as a novel drug screening tool. Although there are several limitations associated with the current methods used to study mental disorders, using iPSCs as a model system provides the advantage of rewinding and reviewing the development and degeneration of human neural cells.
Collapse
Affiliation(s)
- Sewoong Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Min-Kyoung Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Daeyoung Oh
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang-Hyuk Lee
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Borah Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
195
|
Coppola C, Saracino D, Califano F, Barbarulo AM, Di Fede G, Piccoli E, Tagliavini F, Di Iorio G, Rossi G. A case of progressive frontal lobe syndrome in a sporadic form of Cerebral Amyloid Angiopathy: A singular overlap with fronto-temporal dementia? J Neurol Sci 2015; 359:247-9. [PMID: 26671122 DOI: 10.1016/j.jns.2015.08.1533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 08/19/2015] [Accepted: 08/25/2015] [Indexed: 11/25/2022]
Affiliation(s)
- Cinzia Coppola
- Second Division of Neurology of the Second University of Naples, Isola 8-Edificio 10 Policlinico "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Dario Saracino
- Second Division of Neurology of the Second University of Naples, Isola 8-Edificio 10 Policlinico "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Francesca Califano
- Second Division of Neurology of the Second University of Naples, Isola 8-Edificio 10 Policlinico "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Anna Maria Barbarulo
- Second Division of Neurology of the Second University of Naples, Isola 8-Edificio 10 Policlinico "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Giuseppe Di Fede
- Division of Neurology V and Neuropathology, Neurological Institute "Carlo Besta" Foundation IRCCS - Milan, Italy
| | - Elena Piccoli
- Division of Neurology V and Neuropathology, Neurological Institute "Carlo Besta" Foundation IRCCS - Milan, Italy
| | - Fabrizio Tagliavini
- Division of Neurology V and Neuropathology, Neurological Institute "Carlo Besta" Foundation IRCCS - Milan, Italy
| | - Giuseppe Di Iorio
- Second Division of Neurology of the Second University of Naples, Isola 8-Edificio 10 Policlinico "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Giacomina Rossi
- Division of Neurology V and Neuropathology, Neurological Institute "Carlo Besta" Foundation IRCCS - Milan, Italy
| |
Collapse
|
196
|
Jun MH, Han JH, Lee YK, Jang DJ, Kaang BK, Lee JA. TMEM106B, a frontotemporal lobar dementia (FTLD) modifier, associates with FTD-3-linked CHMP2B, a complex of ESCRT-III. Mol Brain 2015; 8:85. [PMID: 26651479 PMCID: PMC4676093 DOI: 10.1186/s13041-015-0177-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/04/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Transmembrane protein 106B (TMEM106B) has been identified as a risk factor for frontotemporal lobar degeneration, which is the second most common form of progressive dementia in people under 65 years of age. Mutations in charged multivesicular body protein 2B (CHMP2B), which is involved in endosomal protein trafficking, have been found in chromosome 3-linked frontotemporal dementia. Despite the number of studies on both CHMP2B and TMEM106B in the endolysosomal pathway, little is known about the relationship between CHMP2B and TMEM106B in the endosomal/autophagy pathway. RESULTS This study found that endogenous TMEM106B was partially sequestered in CHMP2B-positive structures, suggesting its possible involvement in endosomal sorting complexes required for transport (ESCRT)-associated pathways. The role of single nucleotide polymorphisms of TMEM106B (T185, S185, or S134N) in the ESCRT-associated pathways were characterized. The T185 and S185 variants were more localized to Rab5-/Rab7-positive endosomes compared with S134N, while all of the variants were more localized to Rab7-positive endosomes compared to Rab5-positive endosomes. T185 was more associated with CHMP2B compared to S185. Autophagic flux was slightly reduced in the T185-expressing cells compared to the control or S185-expressing cells. Moreover, T185 slightly enhanced the accumulation of EGFR, impairments in autophagic flux, and neurotoxicity that were caused by CHMP2B(Intron5) compared to S185-expressing cells. CONCLUSIONS These findings suggest that the T185 variant functions as a risk factor in neurodegeneration with endolysosomal defects. This study provides a better understanding of pathogenic functions of TMEM106B, which is a risk factor for the progression of neurodegenerative diseases that are associated with endosomal defects in the aged brain.
Collapse
Affiliation(s)
- Mi-Hee Jun
- Department of Biotechnology and Biological Sciences, Hannam University, 1646 Yuseongdaero, Yu-seong-gu, Daejeon, 305-811, South Korea.
| | - Jeong-Ho Han
- Department of Biotechnology and Biological Sciences, Hannam University, 1646 Yuseongdaero, Yu-seong-gu, Daejeon, 305-811, South Korea.
| | - Yu-Kyung Lee
- Department of Biotechnology and Biological Sciences, Hannam University, 1646 Yuseongdaero, Yu-seong-gu, Daejeon, 305-811, South Korea.
| | - Deok-Jin Jang
- Applied Biology, College of Ecological Environment, Kyungpook National University, 386, Gajang-dong, Sang-Ju, South Korea.
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 1 Gwanangno, Gwanak-gu, Seoul, 08826, South Korea.
| | - Jin-A Lee
- Department of Biotechnology and Biological Sciences, Hannam University, 1646 Yuseongdaero, Yu-seong-gu, Daejeon, 305-811, South Korea.
| |
Collapse
|
197
|
Paternicò D, Premi E, Gazzina S, Cosseddu M, Alberici A, Archetti S, Cotelli MS, Micheli A, Turla M, Gasparotti R, Padovani A, Borroni B. White matter hyperintensities characterize monogenic frontotemporal dementia with granulin mutations. Neurobiol Aging 2015; 38:176-180. [PMID: 26827655 DOI: 10.1016/j.neurobiolaging.2015.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/02/2015] [Accepted: 11/19/2015] [Indexed: 11/29/2022]
Abstract
No study but one has suggested the presence of white matter hyperintensities (WMHs) in frontotemporal dementia (FTD), limited to 4 cases carrying pathogenic Granulin (GRN) gene mutations. We investigated the presence of WMHs in a cohort of 14 FTD patients with pathogenic GRN mutations (GRN+), 28 patients without GRN mutations (GRN-) and 18 healthy controls (HC). We further considered 11 asymptomatic GRN+ subjects and 11 young age-matched healthy controls (yHC). The WMH burden was automatically computed and a voxelwise-based analysis was carried out to explore the differences in WMH brain spatial distribution. FTD-GRN+ patients had increased total WMH burden than both HC (p < 0.001) and FTD-GRN-(p = 0.01) groups. WMHs were mainly localized in the right middle frontal and superior temporal gyri, in the left superior frontal in the left parietal gyri. No significant differences of WMH burden between asymptomatic GRN+ and yHC were observed. The presence of WMHs in cases of FTD may suggest a novel mechanism of GRN disease-related neurodegeneration, may be of help in the differential diagnosis, and in guiding genetic screening.
Collapse
Affiliation(s)
- Donata Paternicò
- Centre of Brain Aging, Neurology Unit, Department of Biomedical Sciences and Translational Medicine, University of Brescia, Brescia, Italy
| | - Enrico Premi
- Centre of Brain Aging, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Gazzina
- Centre of Brain Aging, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Maura Cosseddu
- Centre of Brain Aging, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Antonella Alberici
- Centre of Brain Aging, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvana Archetti
- The III Laboratory, Biotechnology, Spedali Civili Hospital, Brescia, Italy
| | | | | | | | | | - Alessandro Padovani
- Centre of Brain Aging, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Barbara Borroni
- Centre of Brain Aging, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
| |
Collapse
|
198
|
Filippi M, Agosta F, Ferraro PM. Charting Frontotemporal Dementia: From Genes to Networks. J Neuroimaging 2015; 26:16-27. [PMID: 26617288 DOI: 10.1111/jon.12316] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia (FTD) is a genetically and clinically heterogeneous syndrome that is characterized by overlapping clinical symptoms involving behavior, personality, language and/or motor functions and degeneration of the frontal and temporal lobes. The term frontotemporal lobar degeneration (FTLD) is used to describe the proteinopathies associated with clinical FTD. Emerging evidence from network-based neuroimaging studies, such as resting state functional MRI and diffusion tensor MRI studies, have implicated specific large-scale brain networks in the pathogenesis of FTD syndromes, suggesting a new paradigm for explaining the distributed and heterogeneous spreading patterns of pathological proteins in FTLD. In this review, we overview recent research on the study of FTD syndromes as connectivity disorders in symptomatic patients as well as genotype-specific changes in asymptomatic FTD-related gene mutation carriers. Characterizing brain network breakdown in these subjects using neuroimaging may help anticipate the diagnosis and perhaps prevent the devastating impact of FTD.
Collapse
Affiliation(s)
- Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Pilar M Ferraro
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
199
|
Zhou B, Liu C, Geng Y, Zhu G. Topology of a G-quadruplex DNA formed by C9orf72 hexanucleotide repeats associated with ALS and FTD. Sci Rep 2015; 5:16673. [PMID: 26564809 PMCID: PMC4643247 DOI: 10.1038/srep16673] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/07/2015] [Indexed: 12/13/2022] Open
Abstract
Abnormal expansions of an intronic hexanucleotide GGGGCC (G4C2) repeat of the C9orf72 gene are the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Previous studies suggested that the C9orf72 hexanucleotide repeat expansion (HRE), either as DNA or the transcribed RNA, can fold into G-quadruplexes with distinct structures. These structural polymorphisms lead to abortive transcripts and contribute to the pathogenesis of ALS and FTD. Using circular dichroism (CD) and nuclear magnetic resonance (NMR) spectroscopy, we analyzed the structures of C9orf72 HRE DNA with various G4C2 repeats. They exhibited diverse G-quadruplex folds in potassium ions. Furthermore, we determined the topology of a G-quadruplex formed by d(G4C2)4. It favors a monomeric fold and forms a chair-type G-quadruplex with a four-layer antiparallel G-tetra core and three edgewise loops, which is distinct from known structures of chair-type G-quadruplexes. Our findings highlight the conformational heterogeneity of C9orf72 HRE DNA, and may lay the necessary structural basis for designing small molecules for the modulation of ALS/FTD pathogenesis.
Collapse
Affiliation(s)
- Bo Zhou
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| | - Changdong Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| | - Yanyan Geng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| | - Guang Zhu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| |
Collapse
|
200
|
Murakami T, Qamar S, Lin JQ, Schierle GSK, Rees E, Miyashita A, Costa AR, Dodd RB, Chan FTS, Michel CH, Kronenberg-Versteeg D, Li Y, Yang SP, Wakutani Y, Meadows W, Ferry RR, Dong L, Tartaglia GG, Favrin G, Lin WL, Dickson DW, Zhen M, Ron D, Schmitt-Ulms G, Fraser PE, Shneider NA, Holt C, Vendruscolo M, Kaminski CF, St George-Hyslop P. ALS/FTD Mutation-Induced Phase Transition of FUS Liquid Droplets and Reversible Hydrogels into Irreversible Hydrogels Impairs RNP Granule Function. Neuron 2015; 88:678-90. [PMID: 26526393 PMCID: PMC4660210 DOI: 10.1016/j.neuron.2015.10.030] [Citation(s) in RCA: 648] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/18/2015] [Accepted: 10/15/2015] [Indexed: 12/14/2022]
Abstract
The mechanisms by which mutations in FUS and other RNA binding proteins cause ALS and FTD remain controversial. We propose a model in which low-complexity (LC) domains of FUS drive its physiologically reversible assembly into membrane-free, liquid droplet and hydrogel-like structures. ALS/FTD mutations in LC or non-LC domains induce further phase transition into poorly soluble fibrillar hydrogels distinct from conventional amyloids. These assemblies are necessary and sufficient for neurotoxicity in a C. elegans model of FUS-dependent neurodegeneration. They trap other ribonucleoprotein (RNP) granule components and disrupt RNP granule function. One consequence is impairment of new protein synthesis by cytoplasmic RNP granules in axon terminals, where RNP granules regulate local RNA metabolism and translation. Nuclear FUS granules may be similarly affected. Inhibiting formation of these fibrillar hydrogel assemblies mitigates neurotoxicity and suggests a potential therapeutic strategy that may also be applicable to ALS/FTD associated with mutations in other RNA binding proteins. FUS phase transitions between monomer, liquid droplet, and hydrogel states FUS mutants induce further phase transition into irreversible fibrillar hydrogels Irreversible hydrogels sequester RNP cargo and impair RNP granule function Formation of non-amyloid fibrillar hydrogels provides a compelling causative mechanism for neurodegeneration
Collapse
Affiliation(s)
- Tetsuro Murakami
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Seema Qamar
- Cambridge Institute for Medical Research, Cambridge National Institute for Health Research - Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK
| | - Julie Qiaojin Lin
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | | | - Eric Rees
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 3RA, UK
| | - Akinori Miyashita
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Ana R Costa
- Cambridge Institute for Medical Research, Cambridge National Institute for Health Research - Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK
| | - Roger B Dodd
- Cambridge Institute for Medical Research, Cambridge National Institute for Health Research - Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK
| | - Fiona T S Chan
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 3RA, UK
| | - Claire H Michel
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 3RA, UK
| | - Deborah Kronenberg-Versteeg
- Cambridge Institute for Medical Research, Cambridge National Institute for Health Research - Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK
| | - Yi Li
- Cambridge Institute for Medical Research, Cambridge National Institute for Health Research - Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK
| | - Seung-Pil Yang
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Yosuke Wakutani
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - William Meadows
- Cambridge Institute for Medical Research, Cambridge National Institute for Health Research - Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK
| | - Rodylyn Rose Ferry
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Liang Dong
- Cambridge Institute for Medical Research, Cambridge National Institute for Health Research - Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK
| | - Gian Gaetano Tartaglia
- Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK; Centre for Genomic Regulation and University Pompeu Fabra, Dr. Aiguader St. 88, and Universitat Pompeu Fabra, 08003, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 23 Passeig Lluís Companys, 08010 Barcelona, Spain
| | - Giorgio Favrin
- Cambridge Systems Biology Center & Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - Wen-Lang Lin
- Department of Research, Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Research, Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Mei Zhen
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - David Ron
- Department of Clinical Biochemistry, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Neil A Shneider
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University Medical Center, New York, NY 10032, USA
| | - Christine Holt
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | | | - Clemens F Kaminski
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB2 3RA, UK
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada; Cambridge Institute for Medical Research, Cambridge National Institute for Health Research - Biomedical Research Unit in Dementia, University of Cambridge, Cambridge CB2 0XY, UK.
| |
Collapse
|