1
|
Chen Y, Deng H, Zhang N. Autophagy-targeting modulation to promote peripheral nerve regeneration. Neural Regen Res 2025; 20:1864-1882. [PMID: 39254547 PMCID: PMC11691477 DOI: 10.4103/nrr.nrr-d-23-01948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/22/2024] [Accepted: 03/29/2024] [Indexed: 09/11/2024] Open
Abstract
Nerve regeneration following traumatic peripheral nerve injuries and neuropathies is a complex process modulated by diverse factors and intricate molecular mechanisms. Past studies have focused on factors that stimulate axonal outgrowth and myelin regeneration. However, recent studies have highlighted the pivotal role of autophagy in peripheral nerve regeneration, particularly in the context of traumatic injuries. Consequently, autophagy-targeting modulation has emerged as a promising therapeutic approach to enhancing peripheral nerve regeneration. Our current understanding suggests that activating autophagy facilitates the rapid clearance of damaged axons and myelin sheaths, thereby enhancing neuronal survival and mitigating injury-induced oxidative stress and inflammation. These actions collectively contribute to creating a favorable microenvironment for structural and functional nerve regeneration. A range of autophagy-inducing drugs and interventions have demonstrated beneficial effects in alleviating peripheral neuropathy and promoting nerve regeneration in preclinical models of traumatic peripheral nerve injuries. This review delves into the regulation of autophagy in cell types involved in peripheral nerve regeneration, summarizing the potential drugs and interventions that can be harnessed to promote this process. We hope that our review will offer novel insights and perspectives on the exploitation of autophagy pathways in the treatment of peripheral nerve injuries and neuropathies.
Collapse
Affiliation(s)
- Yan Chen
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Reproductive Endocrinology and Reproductive Regulation, Sichuan University, Chengdu, Sichuan Province, China
| | - Hongxia Deng
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Reproductive Endocrinology and Reproductive Regulation, Sichuan University, Chengdu, Sichuan Province, China
| | - Nannan Zhang
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Kamińska J, Kochański A. A Role of Inflammation in Charcot-Marie-Tooth Disorders-In a Perspective of Treatment? Int J Mol Sci 2024; 26:15. [PMID: 39795872 PMCID: PMC11720021 DOI: 10.3390/ijms26010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Despite the fact that there are published case reports and model work providing evidence of inflammation in Charcot-Marie-Tooth disorders (CMTs), in clinical practice, CMT and inflammatory neuropathies are always classified as two separate groups of disorders. This sharp separation of chronic neuropathies into two groups has serious clinical implications. As a consequence, the patients harboring CMT mutations are practically excluded from pharmacological anti-inflammatory treatments. In this review, we present that neuropathological studies of peripheral nerves taken from some patients representing familial aggregation of CMTs revealed the presence of inflammation within the nerves. This shows that neurodegeneration resulting from germline mutations and the inflammatory process are not mutually exclusive. We also point to reports demonstrating that, at the clinical level, a positive response to anti-inflammatory therapy was observed in some patients diagnosed with CMTs, confirming the role of the inflammatory component in CMT. We narrowed a group of more than 100 genes whose mutations were found in CMT-affected patients to the seven most common (MPZ, PMP22, GJB1, SEPT9, LITAF, FIG4, and GDAP1) as being linked to the coexistence of hereditary and inflammatory neuropathy. We listed studies of mouse models supporting the idea of the presence of an inflammatory process in some CMTs and studies demonstrating at the cellular level the presence of an inflammatory response. In the following, we discuss the possible molecular basis of some neuropathies involving neurodegenerative and inflammatory processes at both the clinical and morphological levels. Finally, we discuss the prospect of a therapeutic approach using immunomodulation in some patients affected by CMTs.
Collapse
Affiliation(s)
- Joanna Kamińska
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Andrzej Kochański
- Neuromuscular Unit, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warsaw, Poland
| |
Collapse
|
3
|
Gambarotto L, Russo L, Bresolin S, Persano L, D'Amore R, Ronchi G, Zen F, Muratori L, Cani A, Negro S, Megighian A, Calabrò S, Braghetta P, Bizzotto D, Cescon M. Schwann Cell-Specific Ablation of Beclin 1 Impairs Myelination and Leads to Motor and Sensory Neuropathy in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2308965. [PMID: 39680476 DOI: 10.1002/advs.202308965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/24/2024] [Indexed: 12/18/2024]
Abstract
The core component of the class III phosphatidylinositol 3-kinase complex, Beclin 1, takes part in different protein networks, thus switching its role from inducing autophagy to regulating autophagosomal maturation and endosomal trafficking. While assessed in neurons, astrocytes, and microglia, its role is far less investigated in myelinating glia, including Schwann cells (SCs), responsible for peripheral nerve myelination. Remarkably, the dysregulation in endosomal trafficking is emerging as a pathophysiological mechanism underlying peripheral neuropathies, such as demyelinating Charcot-Marie-Tooth (CMT) diseases. By knocking out Beclin 1 in SCs here a novel mouse model (Becn1 cKO) is generated, developing a severe and progressive neuropathy, accompanied by involuntary tremors, body weight loss, and premature death. Ultrastructural analysis revealed abated myelination and SCs displaying enlarged cytoplasm with progressive accumulation of intracellular vesicles. Transcriptomic and histological analysis from sciatic nerves of 10-day and 2-month-old mice revealed pro-mitotic gene deregulation and increased SCs proliferation at both stages with axonal loss and increased immune infiltration in adults, well reflecting the progressive motor and sensory functional impairment that characterizes Becn1 cKO mice, compared to controls. The study establishes a further step in understanding key mechanisms in SC development and points to Beclin 1 and its regulated pathways as targets for demyelinating CMT forms.
Collapse
Affiliation(s)
- Lisa Gambarotto
- Department of Molecular Medicine, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
- Department of Biology, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
| | - Loris Russo
- Department of Molecular Medicine, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
| | - Silvia Bresolin
- Department of Women and Children's Health, University of Padova, via Giustiniani 3, Padova, 35127, Italy
- Istituto di Ricerca Pediatrica - Città della Speranza, Corso Stati Uniti 4, Padova, 35128, Italy
| | - Luca Persano
- Department of Women and Children's Health, University of Padova, via Giustiniani 3, Padova, 35127, Italy
- Istituto di Ricerca Pediatrica - Città della Speranza, Corso Stati Uniti 4, Padova, 35128, Italy
| | - Rachele D'Amore
- Department of Molecular Medicine, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, Torino, 10043, Italy
| | - Federica Zen
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, Torino, 10043, Italy
| | - Luisa Muratori
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, Torino, 10043, Italy
| | - Alice Cani
- Department of Women and Children's Health, University of Padova, via Giustiniani 3, Padova, 35127, Italy
| | - Samuele Negro
- U.O.C. Clinica Neurologica, Azienda Ospedale-Università Padova, Via Giustiniani 5, Padova, 35128, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
- Padova Neuroscience Center, University of Padova, Via G. Orus, 2, Padova, 35131, Italy
| | - Sonia Calabrò
- Department of Molecular Medicine, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
- Department of Biology, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
| | - Dario Bizzotto
- Department of Molecular Medicine, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, Via U. Bassi 58/B, Padova, 35131, Italy
| |
Collapse
|
4
|
Scherer SS, Svaren J. Peripheral Nervous System (PNS) Myelin Diseases. Cold Spring Harb Perspect Biol 2024; 16:a041376. [PMID: 38253417 PMCID: PMC11065170 DOI: 10.1101/cshperspect.a041376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
This is a review of inherited and acquired causes of human demyelinating neuropathies and a subset of disorders that affect axon-Schwann cell interactions. Nearly all inherited demyelinating neuropathies are caused by mutations in genes that are expressed by myelinating Schwann cells, affecting diverse functions in a cell-autonomous manner. The most common acquired demyelinating neuropathies are Guillain-Barré syndrome and chronic, inflammatory demyelinating polyneuropathy, both of which are immune-mediated. An additional group of inherited and acquired disorders affect axon-Schwann cell interactions in the nodal region. Overall, these disorders affect the formation of myelin and its maintenance, with superimposed axonal loss that is clinically important.
Collapse
Affiliation(s)
- Steven S Scherer
- Department of Neurology, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - John Svaren
- Department of Comparative Biosciences, Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| |
Collapse
|
5
|
Li D, Sun F, Yang Y, Tu H, Cai H. Gradients of PI(4,5)P2 and PI(3,5)P2 Jointly Participate in Shaping the Back State of Dictyostelium Cells. Front Cell Dev Biol 2022; 10:835185. [PMID: 35186938 PMCID: PMC8855053 DOI: 10.3389/fcell.2022.835185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Polarity, which refers to the molecular or structural asymmetry in cells, is essential for diverse cellular functions. Dictyostelium has proven to be a valuable system for dissecting the molecular mechanisms of cell polarity. Previous studies in Dictyostelium have revealed a range of signaling and cytoskeletal proteins that function at the leading edge to promote pseudopod extension and migration. In contrast, how proteins are localized to the trailing edge is not well understood. By screening for asymmetrically localized proteins, we identified a novel trailing-edge protein we named Teep1. We show that a charged surface formed by two pleckstrin homology (PH) domains in Teep1 is necessary and sufficient for targeting it to the rear of cells. Combining biochemical and imaging analyses, we demonstrate that Teep1 interacts preferentially with PI(4,5)P2 and PI(3,5)P2in vitro and simultaneous elimination of these lipid species in cells blocks the membrane association of Teep1. Furthermore, a leading-edge localized myotubularin phosphatase likely mediates the removal of PI(3,5)P2 from the front, as well as the formation of a back-to-front gradient of PI(3,5)P2. Together our data indicate that PI(4,5)P2 and PI(3,5)P2 on the plasma membrane jointly participate in shaping the back state of Dictyostelium cells.
Collapse
Affiliation(s)
- Dong Li
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Feifei Sun
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yihong Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hui Tu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Huaqing Cai
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Huaqing Cai,
| |
Collapse
|
6
|
Shalev I, Somekh J, Eran A. Multimodal bioinformatic analyses of the neurodegenerative disease-associated TECPR2 gene reveal its diverse roles. J Med Genet 2021; 59:1002-1009. [PMID: 34933910 DOI: 10.1136/jmedgenet-2021-108193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/01/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND Loss of tectonin β-propeller repeat-containing 2 (TECPR2) function has been implicated in an array of neurodegenerative disorders, yet its physiological function remains largely unknown. Understanding TECPR2 function is essential for developing much needed precision therapeutics for TECPR2-related diseases. METHODS We leveraged considerable amounts of functional data to obtain a comprehensive perspective of the role of TECPR2 in health and disease. We integrated expression patterns, population variation, phylogenetic profiling, protein-protein interactions and regulatory network data for a minimally biased multimodal functional analysis. Genes and proteins linked to TECPR2 via multiple lines of evidence were subject to functional enrichment analyses to identify molecular mechanisms involving TECPR2. RESULTS TECPR2 was found to be part of a tight neurodevelopmental gene expression programme that includes KIF1A, ATXN1, TOM1L2 and FA2H, all implicated in neurological diseases. Functional enrichment analyses of TECPR2-related genes converged on a role in late autophagy and ribosomal processes. Large-scale population variation data demonstrated that this role is non-redundant. CONCLUSIONS TECPR2 might serve as an indicator for the energy balance between protein synthesis and autophagy, and a marker for diseases associated with their imbalance, such as Alzheimer's disease and Huntington's disease. Specifically, we speculate that TECPR2 plays an important role as a proteostasis regulator during synaptogenesis, highlighting its importance in developing neurons. By advancing our understanding of TECPR2 function, this work provides an essential stepping stone towards the development of precision diagnostics and targeted treatment options for TECPR2-related disorders.
Collapse
Affiliation(s)
- Ido Shalev
- Department of Psychology, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Zlotowsky Center for Brain Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Judith Somekh
- Department of Information Systems, University of Haifa, Haifa, Israel
| | - Alal Eran
- Zlotowsky Center for Brain Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel .,Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Computational Health Informatics Program, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Mammel AE, Delgado KC, Chin AL, Condon AF, Hill JQ, Aicher SA, Wang Y, Fedorov LM, Robinson FL. Distinct roles for the Charcot-Marie-tooth disease-causing endosomal regulators Mtmr5 and Mtmr13 in axon radial sorting and Schwann cell myelination. Hum Mol Genet 2021; 31:1216-1229. [PMID: 34718573 DOI: 10.1093/hmg/ddab311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 11/12/2022] Open
Abstract
The form of Charcot-Marie-Tooth type 4B (CMT4B) disease caused by mutations in myotubularin-related 5 (MTMR5; also called SET Binding Factor 1; SBF1) shows a spectrum of axonal and demyelinating nerve phenotypes. This contrasts with the CMT4B subtypes caused by MTMR2 or MTMR13 (SBF2) mutations, which are characterized by myelin outfoldings and classic demyelination. Thus, it is unclear whether MTMR5 plays an analogous or distinct role from that of its homolog, MTMR13, in the peripheral nervous system (PNS). MTMR5 and MTMR13 are pseudophosphatases predicted to regulate endosomal trafficking by activating Rab GTPases and binding to the phosphoinositide 3-phosphatase MTMR2. In the mouse PNS, Mtmr2 was required to maintain wild type levels of Mtmr5 and Mtmr13, suggesting that these factors function in discrete protein complexes. Genetic elimination of both Mtmr5 and Mtmr13 in mice led to perinatal lethality, indicating that the two proteins have partially redundant functions during embryogenesis. Loss of Mtmr5 in mice did not cause CMT4B-like myelin outfoldings. However, adult Mtmr5-/- mouse nerves contained fewer myelinated axons than control nerves, likely as a result of axon radial sorting defects. Consistently, Mtmr5 levels were highest during axon radial sorting and fell sharply after postnatal day seven. Our findings suggest that Mtmr5 and Mtmr13 ensure proper axon radial sorting and Schwann cell myelination, respectively, perhaps through their direct interactions with Mtmr2. This study enhances our understanding of the non-redundant roles of the endosomal regulators MTMR5 and MTMR13 during normal peripheral nerve development and disease.
Collapse
Affiliation(s)
- Anna E Mammel
- The Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.,Cell, Developmental & Cancer Biology Graduate Program, Oregon Health & Science University
| | - Katherine C Delgado
- The Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Andrea L Chin
- The Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Alec F Condon
- The Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.,Neuroscience Graduate Program, Oregon Health & Science University
| | - Jo Q Hill
- Department of Physiology and Pharmacology, Oregon Health & Science University
| | - Sue A Aicher
- Department of Physiology and Pharmacology, Oregon Health & Science University
| | - Yingming Wang
- OHSU Transgenic Mouse Models Shared Resource, Knight Cancer Institute, Oregon Health & Science University
| | - Lev M Fedorov
- OHSU Transgenic Mouse Models Shared Resource, Knight Cancer Institute, Oregon Health & Science University
| | - Fred L Robinson
- The Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.,Vollum Institute, Oregon Health & Science University
| |
Collapse
|
8
|
Abstract
Myelin is a key evolutionary specialization and adaptation of vertebrates formed by the plasma membrane of glial cells, which insulate axons in the nervous system. Myelination not only allows rapid and efficient transmission of electric impulses in the axon by decreasing capacitance and increasing resistance but also influences axonal metabolism and the plasticity of neural circuits. In this review, we will focus on Schwann cells, the glial cells which form myelin in the peripheral nervous system. Here, we will describe the main extrinsic and intrinsic signals inducing Schwann cell differentiation and myelination and how myelin biogenesis is achieved. Finally, we will also discuss how the study of human disorders in which molecules and pathways relevant for myelination are altered has enormously contributed to the current knowledge on myelin biology.
Collapse
Affiliation(s)
- Alessandra Bolino
- Human Inherited Neuropathies Unit, Institute of Experimental Neurology INSPE, Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
9
|
Abstract
Demyelinating forms of Charcot-Marie-Tooth disease (CMT) are genetically and phenotypically heterogeneous and result from highly diverse biological mechanisms including gain of function (including dominant negative effects) and loss of function. While no definitive treatment is currently available, rapid advances in defining the pathomechanisms of demyelinating CMT have led to promising pre-clinical studies, as well as emerging clinical trials. Especially promising are the recently completed pre-clinical genetic therapy studies in PMP-22, GJB1, and SH3TC2-associated neuropathies, particularly given the success of similar approaches in humans with spinal muscular atrophy and transthyretin familial polyneuropathy. This article focuses on neuropathies related to mutations in PMP-22, MPZ, and GJB1, which together comprise the most common forms of demyelinating CMT, as well as on select rarer forms for which promising treatment targets have been identified. Clinical characteristics and pathomechanisms are reviewed in detail, with emphasis on therapeutically targetable biological pathways. Also discussed are the challenges facing the CMT research community in its efforts to advance the rapidly evolving biological insights to effective clinical trials. These considerations include the limitations of currently available animal models, the need for personalized medicine approaches/allele-specific interventions for select forms of demyelinating CMT, and the increasing demand for optimal clinical outcome assessments and objective biomarkers.
Collapse
Affiliation(s)
- Vera Fridman
- Department of Neurology, University of Colorado Anschutz Medical Campus, 12631 E 17th Avenue, Mailstop B185, Room 5113C, Aurora, CO, 80045, USA.
| | - Mario A Saporta
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
10
|
Bosco L, Falzone YM, Previtali SC. Animal Models as a Tool to Design Therapeutical Strategies for CMT-like Hereditary Neuropathies. Brain Sci 2021; 11:1237. [PMID: 34573256 PMCID: PMC8465478 DOI: 10.3390/brainsci11091237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
Since ancient times, animal models have provided fundamental information in medical knowledge. This also applies for discoveries in the field of inherited peripheral neuropathies (IPNs), where they have been instrumental for our understanding of nerve development, pathogenesis of neuropathy, molecules and pathways involved and to design potential therapies. In this review, we briefly describe how animal models have been used in ancient medicine until the use of rodents as the prevalent model in present times. We then travel along different examples of how rodents have been used to improve our understanding of IPNs. We do not intend to describe all discoveries and animal models developed for IPNs, but just to touch on a few arbitrary and paradigmatic examples, taken from our direct experience or from literature. The idea is to show how strategies have been developed to finally arrive to possible treatments for IPNs.
Collapse
Affiliation(s)
| | | | - Stefano Carlo Previtali
- Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.B.); (Y.M.F.)
| |
Collapse
|
11
|
Markworth R, Bähr M, Burk K. Held Up in Traffic-Defects in the Trafficking Machinery in Charcot-Marie-Tooth Disease. Front Mol Neurosci 2021; 14:695294. [PMID: 34483837 PMCID: PMC8415527 DOI: 10.3389/fnmol.2021.695294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Charcot-Marie-Tooth disease (CMT), also known as motor and sensory neuropathy, describes a clinically and genetically heterogenous group of disorders affecting the peripheral nervous system. CMT typically arises in early adulthood and is manifested by progressive loss of motor and sensory functions; however, the mechanisms leading to the pathogenesis are not fully understood. In this review, we discuss disrupted intracellular transport as a common denominator in the pathogenesis of different CMT subtypes. Intracellular transport via the endosomal system is essential for the delivery of lipids, proteins, and organelles bidirectionally to synapses and the soma. As neurons of the peripheral nervous system are amongst the longest neurons in the human body, they are particularly susceptible to damage of the intracellular transport system, leading to a loss in axonal integrity and neuronal death. Interestingly, defects in intracellular transport, both in neurons and Schwann cells, have been found to provoke disease. This review explains the mechanisms of trafficking and subsequently summarizes and discusses the latest findings on how defects in trafficking lead to CMT. A deeper understanding of intracellular trafficking defects in CMT will expand our understanding of CMT pathogenesis and will provide novel approaches for therapeutic treatments.
Collapse
Affiliation(s)
- Ronja Markworth
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Katja Burk
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| |
Collapse
|
12
|
Dysregulation of myelin synthesis and actomyosin function underlies aberrant myelin in CMT4B1 neuropathy. Proc Natl Acad Sci U S A 2021; 118:2009469118. [PMID: 33653949 DOI: 10.1073/pnas.2009469118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Charcot-Marie-Tooth type 4B1 (CMT4B1) is a severe autosomal recessive demyelinating neuropathy with childhood onset, caused by loss-of-function mutations in the myotubularin-related 2 (MTMR2) gene. MTMR2 is a ubiquitously expressed catalytically active 3-phosphatase, which in vitro dephosphorylates the 3-phosphoinositides PtdIns3P and PtdIns(3,5)P 2, with a preference for PtdIns(3,5)P 2 A hallmark of CMT4B1 neuropathy are redundant loops of myelin in the nerve termed myelin outfoldings, which can be considered the consequence of altered growth of myelinated fibers during postnatal development. How MTMR2 loss and the resulting imbalance of 3'-phosphoinositides cause CMT4B1 is unknown. Here we show that MTMR2 by regulating PtdIns(3,5)P 2 levels coordinates mTORC1-dependent myelin synthesis and RhoA/myosin II-dependent cytoskeletal dynamics to promote myelin membrane expansion and longitudinal myelin growth. Consistent with this, pharmacological inhibition of PtdIns(3,5)P 2 synthesis or mTORC1/RhoA signaling ameliorates CMT4B1 phenotypes. Our data reveal a crucial role for MTMR2-regulated lipid turnover to titrate mTORC1 and RhoA signaling thereby controlling myelin growth.
Collapse
|
13
|
Mattei AM, Smailys JD, Hepworth EMW, Hinton SD. The Roles of Pseudophosphatases in Disease. Int J Mol Sci 2021; 22:ijms22136924. [PMID: 34203203 PMCID: PMC8269279 DOI: 10.3390/ijms22136924] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 01/07/2023] Open
Abstract
The pseudophosphatases, atypical members of the protein tyrosine phosphatase family, have emerged as bona fide signaling regulators within the past two decades. Their roles as regulators have led to a renaissance of the pseudophosphatase and pseudoenyme fields, catapulting interest from a mere curiosity to intriguing and relevant proteins to investigate. Pseudophosphatases make up approximately fourteen percent of the phosphatase family, and are conserved throughout evolution. Pseudophosphatases, along with pseudokinases, are important players in physiology and pathophysiology. These atypical members of the protein tyrosine phosphatase and protein tyrosine kinase superfamily, respectively, are rendered catalytically inactive through mutations within their catalytic active signature motif and/or other important domains required for catalysis. This new interest in the pursuit of the relevant functions of these proteins has resulted in an elucidation of their roles in signaling cascades and diseases. There is a rapid accumulation of knowledge of diseases linked to their dysregulation, such as neuropathies and various cancers. This review analyzes the involvement of pseudophosphatases in diseases, highlighting the function of various role(s) of pseudophosphatases involvement in pathologies, and thus providing a platform to strongly consider them as key therapeutic drug targets.
Collapse
|
14
|
Svaren J. Adeno-associated virus gene therapy to the rescue for Charcot-Marie-Tooth disease type 4J. J Clin Invest 2021; 131:e149492. [PMID: 34060476 DOI: 10.1172/jci149492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The genetic peripheral neuropathy known as Charcot-Marie-Tooth disease type 4J (CMT4J) is caused by recessive mutations in the FIG4 gene. The transformational success of adeno-associated virus (AAV) gene therapy for spinal muscular atrophy has generated substantial interest in using this approach to create similar treatments for CMT. In this issue of the JCI, Presa et al. provide a preclinical demonstration of efficacy using AAV-directed gene therapy for CMT4J. The study showed a dramatic improvement in both survival and neuropathy symptoms in a severe mouse model of CMT4J after administration of AAV gene therapy at several time points. The authors' approach advances the technique for delivering treatments to individuals with CMT, for which FDA-approved therapies have not yet come to the clinic.
Collapse
|
15
|
Chen Z, Malia PC, Hatakeyama R, Nicastro R, Hu Z, Péli-Gulli MP, Gao J, Nishimura T, Eskes E, Stefan CJ, Winderickx J, Dengjel J, De Virgilio C, Ungermann C. TORC1 Determines Fab1 Lipid Kinase Function at Signaling Endosomes and Vacuoles. Curr Biol 2020; 31:297-309.e8. [PMID: 33157024 DOI: 10.1016/j.cub.2020.10.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/25/2020] [Accepted: 10/08/2020] [Indexed: 01/17/2023]
Abstract
Organelles of the endomembrane system maintain their identity and integrity during growth or stress conditions by homeostatic mechanisms that regulate membrane flux and biogenesis. At lysosomes and endosomes, the Fab1 lipid kinase complex and the nutrient-regulated target of rapamycin complex 1 (TORC1) control the integrity of the endolysosomal homeostasis and cellular metabolism. Both complexes are functionally connected as Fab1-dependent generation of PI(3,5)P2 supports TORC1 activity. Here, we identify Fab1 as a target of TORC1 on signaling endosomes, which are distinct from multivesicular bodies, and provide mechanistic insight into their crosstalk. Accordingly, TORC1 can phosphorylate Fab1 proximal to its PI3P-interacting FYVE domain, which causes Fab1 to shift to signaling endosomes, where it generates PI(3,5)P2. This, in turn, regulates (1) vacuole morphology, (2) recruitment of TORC1 and the TORC1-regulatory Rag GTPase-containing EGO complex to signaling endosomes, and (3) TORC1 activity. Thus, our study unravels a regulatory feedback loop between TORC1 and the Fab1 complex that controls signaling at endolysosomes.
Collapse
Affiliation(s)
- Zilei Chen
- Department of Biology/Chemistry, Biochemistry Section, University of Osnabrück, Barbarastrasse 13, 49076 Osnabrück, Germany
| | - Pedro Carpio Malia
- Department of Biology/Chemistry, Biochemistry Section, University of Osnabrück, Barbarastrasse 13, 49076 Osnabrück, Germany
| | - Riko Hatakeyama
- Department of Biology, University of Fribourg, Chemin du Musée, CH-1700 Fribourg, Switzerland
| | - Raffaele Nicastro
- Department of Biology, University of Fribourg, Chemin du Musée, CH-1700 Fribourg, Switzerland
| | - Zehan Hu
- Department of Biology, University of Fribourg, Chemin du Musée, CH-1700 Fribourg, Switzerland
| | - Marie-Pierre Péli-Gulli
- Department of Biology, University of Fribourg, Chemin du Musée, CH-1700 Fribourg, Switzerland
| | - Jieqiong Gao
- Department of Biology/Chemistry, Biochemistry Section, University of Osnabrück, Barbarastrasse 13, 49076 Osnabrück, Germany
| | - Taki Nishimura
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Elja Eskes
- Functional Biology, KU Leuven, Kasteelpark Arensberg 31, 3000 Leuven, Belgium
| | - Christopher J Stefan
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Joris Winderickx
- Functional Biology, KU Leuven, Kasteelpark Arensberg 31, 3000 Leuven, Belgium
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Chemin du Musée, CH-1700 Fribourg, Switzerland
| | - Claudio De Virgilio
- Department of Biology, University of Fribourg, Chemin du Musée, CH-1700 Fribourg, Switzerland.
| | - Christian Ungermann
- Department of Biology/Chemistry, Biochemistry Section, University of Osnabrück, Barbarastrasse 13, 49076 Osnabrück, Germany; Center of Cellular Nanoanalytics Osnabrück (CellNanOs), University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany.
| |
Collapse
|
16
|
Edgar JR, Ho AK, Laurá M, Horvath R, Reilly MM, Luzio JP, Roberts RC. A dysfunctional endolysosomal pathway common to two sub-types of demyelinating Charcot-Marie-Tooth disease. Acta Neuropathol Commun 2020; 8:165. [PMID: 33059769 PMCID: PMC7559459 DOI: 10.1186/s40478-020-01043-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/24/2020] [Indexed: 12/17/2022] Open
Abstract
Autosomal dominant mutations in LITAF are responsible for the rare demyelinating peripheral neuropathy, Charcot-Marie-Tooth disease type 1C (CMT1C). The LITAF protein is expressed in many human cell types and we have investigated the consequences of two different LITAF mutations in primary fibroblasts from CMT1C patients using confocal and electron microscopy. We observed the appearance of vacuolation/enlargement of late endocytic compartments (late endosomes and lysosomes). This vacuolation was also observed after knocking out LITAF from either control human fibroblasts or from the CMT1C patient-derived cells, consistent with it being the result of loss-of-function mutations in the CMT1C fibroblasts. The vacuolation was similar to that previously observed in fibroblasts from CMT4J patients, which have autosomal recessive mutations in FIG4. The FIG4 protein is a component of a phosphoinositide kinase complex that synthesises phosphatidylinositol 3,5-bisphosphate on the limiting membrane of late endosomes. Phosphatidylinositol 3,5-bisphosphate activates the release of lysosomal Ca2+ through the cation channel TRPML1, which is required to maintain the homeostasis of endosomes and lysosomes in mammalian cells. We observed that a small molecule activator of TRPML1, ML-SA1, was able to rescue the vacuolation phenotype of LITAF knockout, FIG4 knockout and CMT1C patient fibroblasts. Our data describe the first cellular phenotype common to two different subtypes of demyelinating CMT and are consistent with LITAF and FIG4 functioning on a common endolysosomal pathway that is required to maintain the homeostasis of late endosomes and lysosomes. Although our experiments were on human fibroblasts, they have implications for our understanding of the molecular pathogenesis and approaches to therapy in two subtypes of demyelinating Charcot-Marie-Tooth disease.
Collapse
Affiliation(s)
- James R Edgar
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| | - Anita K Ho
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
- Department of Biology, University of York, York, YO10 5DD, UK
| | - Matilde Laurá
- Centre for Neuromuscular Diseases, UCL, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Mary M Reilly
- Centre for Neuromuscular Diseases, UCL, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - J Paul Luzio
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Rhys C Roberts
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK.
| |
Collapse
|
17
|
Visigalli D, Capodivento G, Basit A, Fernández R, Hamid Z, Pencová B, Gemelli C, Marubbi D, Pastorino C, Luoma AM, Riekel C, Kirschner DA, Schenone A, Fernández JA, Armirotti A, Nobbio L. Exploiting Sphingo- and Glycerophospholipid Impairment to Select Effective Drugs and Biomarkers for CMT1A. Front Neurol 2020; 11:903. [PMID: 32982928 PMCID: PMC7477391 DOI: 10.3389/fneur.2020.00903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/14/2020] [Indexed: 01/12/2023] Open
Abstract
In Charcot-Marie-Tooth type 1A (CMT1A), Schwann cells exhibit a preponderant transcriptional deficiency of genes involved in lipid biosynthesis. This perturbed lipid metabolism affects the peripheral nerve physiology and the structure of peripheral myelin. Nevertheless, the identification and functional characterization of the lipid species mainly responsible for CMT1A myelin impairment currently lack. This is critical in the pathogenesis of the neuropathy since lipids are many and complex molecules which play essential roles in the cell, including the structural components of cellular membranes, cell signaling, and membrane trafficking. Moreover, lipids themselves are able to modify gene transcription, thereby affecting the genotype-phenotype correlation of well-defined inherited diseases, including CMT1A. Here we report for the first time a comprehensive lipid profiling in experimental and human CMT1A, demonstrating a previously unknown specific alteration of sphingolipid (SP) and glycerophospholipid (GP) metabolism. Notably, SP, and GP changes even emerge in biological fluids of CMT1A rat and human patients, implying a systemic metabolic dysfunction for these specific lipid classes. Actually, SP and GP are not merely reduced; their expression is instead aberrant, contributing to the ultrastructural abnormalities that we detailed by X-ray diffraction in rat and human internode myelin. The modulation of SP and GP pathways in myelinating dorsal root ganglia cultures clearly sustains this issue. In fact, just selected molecules interacting with these pathways are able to modify the altered geometric parameters of CMT1A myelinated fibers. Overall, we propose to exploit the present SP and GP metabolism impairment to select effective drugs and validate a set of reliable biomarkers, which remain a challenge in CMT1A neuropathy.
Collapse
Affiliation(s)
- Davide Visigalli
- DINOGMI, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico S. Martino, UO Clinica Neurologica, Genoa, Italy
| | - Giovanna Capodivento
- DINOGMI, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico S. Martino, UO Clinica Neurologica, Genoa, Italy
| | - Abdul Basit
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Roberto Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country, Leioa, Spain
| | - Zeeshan Hamid
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Barbora Pencová
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country, Leioa, Spain
| | - Chiara Gemelli
- DINOGMI, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico S. Martino, UO Clinica Neurologica, Genoa, Italy
| | - Daniela Marubbi
- DIMES, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico S. Martino, UO Oncologia Cellulare Genoa, Genoa, Italy
| | - Cecilia Pastorino
- DINOGMI, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico S. Martino, UO Clinica Neurologica, Genoa, Italy
| | - Adrienne M Luoma
- Department of Biology, Boston College, Boston, MA, United States
| | | | | | - Angelo Schenone
- DINOGMI, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico S. Martino, UO Clinica Neurologica, Genoa, Italy
| | - José A Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country, Leioa, Spain
| | - Andrea Armirotti
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Lucilla Nobbio
- DINOGMI, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico S. Martino, UO Clinica Neurologica, Genoa, Italy
| |
Collapse
|
18
|
Guimarães-Costa R, Villar-Quiles RN, Latour P, Sole G, Husson I, Lacour A, Leonard-Louis S, Stojkovic T. Confounding clinical presentation and different disease progression in CMT4B1. Neuromuscul Disord 2020; 30:576-582. [DOI: 10.1016/j.nmd.2020.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/30/2020] [Accepted: 05/07/2020] [Indexed: 11/27/2022]
|
19
|
Rzepnikowska W, Kaminska J, Kabzińska D, Binięda K, Kochański A. A Yeast-Based Model for Hereditary Motor and Sensory Neuropathies: A Simple System for Complex, Heterogeneous Diseases. Int J Mol Sci 2020; 21:ijms21124277. [PMID: 32560077 PMCID: PMC7352270 DOI: 10.3390/ijms21124277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Charcot–Marie–Tooth (CMT) disease encompasses a group of rare disorders that are characterized by similar clinical manifestations and a high genetic heterogeneity. Such excessive diversity presents many problems. Firstly, it makes a proper genetic diagnosis much more difficult and, even when using the most advanced tools, does not guarantee that the cause of the disease will be revealed. Secondly, the molecular mechanisms underlying the observed symptoms are extremely diverse and are probably different for most of the disease subtypes. Finally, there is no possibility of finding one efficient cure for all, or even the majority of CMT diseases. Every subtype of CMT needs an individual approach backed up by its own research field. Thus, it is little surprise that our knowledge of CMT disease as a whole is selective and therapeutic approaches are limited. There is an urgent need to develop new CMT models to fill the gaps. In this review, we discuss the advantages and disadvantages of yeast as a model system in which to study CMT diseases. We show how this single-cell organism may be used to discriminate between pathogenic variants, to uncover the mechanism of pathogenesis, and to discover new therapies for CMT disease.
Collapse
Affiliation(s)
- Weronika Rzepnikowska
- Neuromuscular Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.R.); (D.K.); (K.B.)
| | - Joanna Kaminska
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Dagmara Kabzińska
- Neuromuscular Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.R.); (D.K.); (K.B.)
| | - Katarzyna Binięda
- Neuromuscular Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.R.); (D.K.); (K.B.)
| | - Andrzej Kochański
- Neuromuscular Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.R.); (D.K.); (K.B.)
- Correspondence:
| |
Collapse
|
20
|
Sawade L, Grandi F, Mignanelli M, Patiño-López G, Klinkert K, Langa-Vives F, Di Guardo R, Echard A, Bolino A, Haucke V. Rab35-regulated lipid turnover by myotubularins represses mTORC1 activity and controls myelin growth. Nat Commun 2020; 11:2835. [PMID: 32503983 PMCID: PMC7275063 DOI: 10.1038/s41467-020-16696-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 05/18/2020] [Indexed: 01/21/2023] Open
Abstract
Inherited peripheral neuropathies (IPNs) represent a broad group of disorders including Charcot-Marie-Tooth (CMT) neuropathies characterized by defects primarily arising in myelin, axons, or both. The molecular mechanisms by which mutations in nearly 100 identified IPN/CMT genes lead to neuropathies are poorly understood. Here we show that the Ras-related GTPase Rab35 controls myelin growth via complex formation with the myotubularin-related phosphatidylinositol (PI) 3-phosphatases MTMR13 and MTMR2, encoded by genes responsible for CMT-types 4B2 and B1 in humans, and found that it downregulates lipid-mediated mTORC1 activation, a pathway known to crucially regulate myelin biogenesis. Targeted disruption of Rab35 leads to hyperactivation of mTORC1 signaling caused by elevated levels of PI 3-phosphates and to focal hypermyelination in vivo. Pharmacological inhibition of phosphatidylinositol 3,5-bisphosphate synthesis or mTORC1 signaling ameliorates this phenotype. These findings reveal a crucial role for Rab35-regulated lipid turnover by myotubularins to repress mTORC1 activity and to control myelin growth. Charcot-Marie-Tooth (CMT) is an inherited peripheral neuropathy. Here, the authors show that Rab35 forms a complex with genes implicated in CMT, MTMR13 and MTMR2, which regulates myelin growth by controlling mTORC1 signaling through lipid turnover.
Collapse
Affiliation(s)
- Linda Sawade
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Federica Grandi
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Marianna Mignanelli
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.,San Raffaele Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy
| | - Genaro Patiño-López
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México, Federico Gómez. C.P, 06720, Ciudad de México, México
| | - Kerstin Klinkert
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, CNRS, 25-28 rue du Dr Roux, F-75015, Paris, France.,Sorbonne Université, Collège doctoral, F-75005, Paris, France
| | - Francina Langa-Vives
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Roberta Di Guardo
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Arnaud Echard
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, CNRS, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Alessandra Bolino
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125, Berlin, Germany. .,Freie Universität Berlin, Faculty of Biology, Chemistry and Pharmacy, 14195, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, NeuroCure Cluster of Excellence, 10117, Berlin, Germany.
| |
Collapse
|
21
|
Halperin D, Sapir A, Wormser O, Drabkin M, Yogev Y, Dolgin V, Flusser H, Birk OS. Novel MTMR2 mutation causing severe Charcot-Marie-Tooth type 4B1 disease: a case report. Neurogenetics 2020; 21:301-304. [PMID: 32488727 DOI: 10.1007/s10048-020-00617-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/16/2020] [Indexed: 11/25/2022]
Abstract
Mutations in myotubularin-related protein 2 (MTMR2) were shown to underlie Charcot-Marie-Tooth type 4B1 (CMT4B1) disease, a rare autosomal recessive demyelinating neuropathy, characterized by severe early-onset motor and sensory neuropathy. We describe three siblings of consanguineous kindred presenting with hypotonia, reduced muscle tone, action tremor, dysmetria, areflexia, and skeletal deformities, consistent with a diagnosis of CMT. Whole-exome sequencing identified a novel homozygous c.336_337 insertion mutation in MTMR2, resulting in a frameshift and putative truncated protein. In this concise report, we discuss the clinical presentation of this rare disease and support the limited number of observations regarding the pathogenesis of MTMR2-related neuropathies.
Collapse
Affiliation(s)
- Daniel Halperin
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Aviad Sapir
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ohad Wormser
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Max Drabkin
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yuval Yogev
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Vadim Dolgin
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Hagit Flusser
- Zusman Child Development Center, Division of Pediatrics, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ohad S Birk
- The Morris Kahn Laboratory of Human Genetics, National Institute for Biotechnology in the Negev and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- Genetics Institute, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
22
|
Severe Consequences of SAC3/FIG4 Phosphatase Deficiency to Phosphoinositides in Patients with Charcot-Marie-Tooth Disease Type-4J. Mol Neurobiol 2019; 56:8656-8667. [PMID: 31313076 DOI: 10.1007/s12035-019-01693-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/01/2019] [Indexed: 01/26/2023]
Abstract
Charcot-Marie-Tooth disease type-4J (CMT4J), an autosomal recessively inherited peripheral neuropathy characterized by neuronal degeneration, segmental demyelination, and limb muscle weakness, is caused by compound heterozygous mutations in the SAC3/FIG4 gene, resulting in SAC3/FIG4 protein deficiency. SAC3/FIG4 is a phosphatase that not only turns over PtdIns(3,5)P2 to PtdIns3P but also promotes PtdIns(3,5)P2 synthesis by activating the PIKFYVE kinase that also makes PtdIns5P. Whether CMT4J patients have alterations in PtdIns(3,5)P2, PtdIns5P or in other phosphoinositides (PIs), and if yes, in what direction these changes might be, has never been examined. We performed PI profiling in primary fibroblasts from a cohort of CMT4J patients. Subsequent to myo-[2-3H]inositol cell labeling to equilibrium, steady-state levels of PIs were quantified by HPLC under conditions concurrently detecting PtdIns5P, PtdIns(3,5)P2, and the other PIs. Immunoblotting verified SAC3/FIG4 depletion in CMT4J fibroblasts. Compared to normal human controls (n = 9), both PtdIns(3,5)P2 and PtdIns5P levels were significantly decreased in CMT4J fibroblasts (n = 13) by 36.4 ± 3.6% and 43.1 ± 4.4%, respectively (p < 0.0001). These reductions were independent of patients' gender or disease onset. Although mean values for PtdIns3P in the CMT4J cohort remained unchanged, there were high variations in PtdIns3P among individual patients. Aberrant endolysosomal vacuoles, typically seen under PtdIns(3,5)P2 reduction, were apparent but not in fibroblasts from all patients. The subset of patients without aberrant vacuoles exhibited especially low PtdIns3P levels. Concomitant decreases in PtdIns5P and PtdIns(3,5)P2 and the link between PtdIns3P levels and cellular vacuolization are novel insights shedding further light into the molecular determinants in CMT4J polyneuropathy.
Collapse
|
23
|
Wang H, Kaçar Bayram A, Sprute R, Ozdemir O, Cooper E, Pergande M, Efthymiou S, Nedic I, Mazaheri N, Stumpfe K, Azizi Malamiri R, Shariati G, Zeighami J, Bayram N, Naghibzadeh SK, Tajik M, Yaşar M, Sami Güven A, Bibi F, Sultan T, Salpietro V, Houlden H, Per H, Galehdari H, Shalbafan B, Jamshidi Y, Cirak S. Genotype-Phenotype Correlations in Charcot-Marie-Tooth Disease Due to MTMR2 Mutations and Implications in Membrane Trafficking. Front Neurosci 2019; 13:974. [PMID: 31680794 PMCID: PMC6807680 DOI: 10.3389/fnins.2019.00974] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/30/2019] [Indexed: 11/13/2022] Open
Abstract
Charcot-Marie-Tooth type 4 (CMT4) is an autosomal recessive severe form of neuropathy with genetic heterogeneity. CMT4B1 is caused by mutations in the myotubularin-related 2 (MTMR2) gene and as a member of the myotubularin family, the MTMR2 protein is crucial for the modulation of membrane trafficking. To enable future clinical trials, we performed a detailed review of the published cases with MTMR2 mutations and describe four novel cases identified through whole-exome sequencing (WES). The four unrelated families harbor novel homozygous mutations in MTMR2 (NM_016156, Family 1: c.1490dupC; p.Phe498IlefsTer2; Family 2: c.1479+1G>A; Family 3: c.1090C>T; p.Arg364Ter; Family 4: c.883C>T; p.Arg295Ter) and present with CMT4B1-related severe early-onset motor and sensory neuropathy, generalized muscle atrophy, facial and bulbar weakness, and pes cavus deformity. The clinical description of the new mutations reported here overlap with previously reported CMT4B1 phenotypes caused by mutations in the phosphatase domain of MTMR2, suggesting that nonsense MTMR2 mutations, which are predicted to result in loss or disruption of the phosphatase domain, are associated with a severe phenotype and loss of independent ambulation by the early twenties. Whereas the few reported missense mutations and also those truncating mutations occurring at the C-terminus after the phosphatase domain cause a rather mild phenotype and patients were still ambulatory above the age 30 years. Charcot-Marie-Tooth neuropathy and Centronuclear Myopathy causing mutations have been shown to occur in proteins involved in membrane remodeling and trafficking pathway mediated by phosphoinositides. Earlier studies have showing the rescue of MTM1 myopathy by MTMR2 overexpression, emphasize the importance of maintaining the phosphoinositides equilibrium and highlight a potential compensatory mechanism amongst members of this pathway. This proved that the regulation of expression of these proteins involved in the membrane remodeling pathway may compensate each other's loss- or gain-of-function mutations by restoring the phosphoinositides equilibrium. This provides a potential therapeutic strategy for neuromuscular diseases resulting from mutations in the membrane remodeling pathway.
Collapse
Affiliation(s)
- Haicui Wang
- Department of Pediatrics, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Ayşe Kaçar Bayram
- Department of Pediatric Neurology, University of Health Sciences, Kayseri City Hospital, Kayseri, Turkey
| | - Rosanne Sprute
- Department of Pediatrics, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Ozkan Ozdemir
- Department of Pediatrics, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Emily Cooper
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St. George's, University of London, London, United Kingdom
| | - Matthias Pergande
- Department of Pediatrics, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Stephanie Efthymiou
- Department of Neuromuscular Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - Ivana Nedic
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St. George's, University of London, London, United Kingdom
| | - Neda Mazaheri
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran.,Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Katharina Stumpfe
- Department of Pediatrics, University Hospital Cologne, Cologne, Germany
| | - Reza Azizi Malamiri
- Paediatric Neurology, Department of Paediatric Neurology, Golestan Medical, Educational, and Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Gholamreza Shariati
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran.,Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Jawaher Zeighami
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Nurettin Bayram
- Department of Ophthalmology, University of Health Sciences, Kayseri City Hospital, Kayseri, Turkey
| | | | - Mohamad Tajik
- Department of Neurology, Firoozgar General Hospital, University of Medical Sciences, Tehran, Iran
| | - Mehmet Yaşar
- Department of Ear Nose and Throat, University of Health Sciences, Kayseri City Hospital, Kayseri, Turkey
| | - Ahmet Sami Güven
- Department of Pediatric Neurology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Farah Bibi
- Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture University, Rawalpindi, Pakistan
| | - Tipu Sultan
- Department of Pediatric Neurology, Institute of Child Health, The Children's Hospital Lahore, Lahore, Pakistan
| | - Vincenzo Salpietro
- Department of Neuromuscular Disorders, Institute of Neurology, University College London, London, United Kingdom.,Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Henry Houlden
- Department of Neuromuscular Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - Hüseyin Per
- Department of Pediatric Neurology, Erciyes University Medical School, Kayseri, Turkey
| | - Hamid Galehdari
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Bita Shalbafan
- Iran Social Security Organization, Labafinejad Hospital, Tehran, Iran
| | - Yalda Jamshidi
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St. George's, University of London, London, United Kingdom
| | - Sebahattin Cirak
- Department of Pediatrics, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
24
|
Raghu P, Joseph A, Krishnan H, Singh P, Saha S. Phosphoinositides: Regulators of Nervous System Function in Health and Disease. Front Mol Neurosci 2019; 12:208. [PMID: 31507376 PMCID: PMC6716428 DOI: 10.3389/fnmol.2019.00208] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
Phosphoinositides, the seven phosphorylated derivatives of phosphatidylinositol have emerged as regulators of key sub-cellular processes such as membrane transport, cytoskeletal function and plasma membrane signaling in eukaryotic cells. All of these processes are also present in the cells that constitute the nervous system of animals and in this setting too, these are likely to tune key aspects of cell biology in relation to the unique structure and function of neurons. Phosphoinositides metabolism and function are mediated by enzymes and proteins that are conserved in evolution, and analysis of knockouts of these in animal models implicate this signaling system in neural function. Most recently, with the advent of human genome analysis, mutations in genes encoding components of the phosphoinositide signaling pathway have been implicated in human diseases although the cell biological basis of disease phenotypes in many cases remains unclear. In this review we evaluate existing evidence for the involvement of phosphoinositide signaling in human nervous system diseases and discuss ways of enhancing our understanding of the role of this pathway in the human nervous system's function in health and disease.
Collapse
Affiliation(s)
- Padinjat Raghu
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| | | | | | | | | |
Collapse
|
25
|
Volpatti JR, Al-Maawali A, Smith L, Al-Hashim A, Brill JA, Dowling JJ. The expanding spectrum of neurological disorders of phosphoinositide metabolism. Dis Model Mech 2019; 12:12/8/dmm038174. [PMID: 31413155 PMCID: PMC6737944 DOI: 10.1242/dmm.038174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Phosphoinositides (PIPs) are a ubiquitous group of seven low-abundance phospholipids that play a crucial role in defining localized membrane properties and that regulate myriad cellular processes, including cytoskeletal remodeling, cell signaling cascades, ion channel activity and membrane traffic. PIP homeostasis is tightly regulated by numerous inositol kinases and phosphatases, which phosphorylate and dephosphorylate distinct PIP species. The importance of these phospholipids, and of the enzymes that regulate them, is increasingly being recognized, with the identification of human neurological disorders that are caused by mutations in PIP-modulating enzymes. Genetic disorders of PIP metabolism include forms of epilepsy, neurodegenerative disease, brain malformation syndromes, peripheral neuropathy and congenital myopathy. In this Review, we provide an overview of PIP function and regulation, delineate the disorders associated with mutations in genes that modulate or utilize PIPs, and discuss what is understood about gene function and disease pathogenesis as established through animal models of these diseases. Summary: This Review highlights the intersection between phosphoinositides and the enzymes that regulate their metabolism, which together are crucial regulators of myriad cellular processes and neurological disorders.
Collapse
Affiliation(s)
- Jonathan R Volpatti
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Almundher Al-Maawali
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Lindsay Smith
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Aqeela Al-Hashim
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Neuroscience, King Fahad Medical City, Riyadh 11525, Saudi Arabia
| | - Julie A Brill
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.,Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - James J Dowling
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
26
|
Pareyson D, Stojkovic T, Reilly MM, Leonard-Louis S, Laurà M, Blake J, Parman Y, Battaloglu E, Tazir M, Bellatache M, Bonello-Palot N, Lévy N, Sacconi S, Guimarães-Costa R, Attarian S, Latour P, Solé G, Megarbane A, Horvath R, Ricci G, Choi BO, Schenone A, Gemelli C, Geroldi A, Sabatelli M, Luigetti M, Santoro L, Manganelli F, Quattrone A, Valentino P, Murakami T, Scherer SS, Dankwa L, Shy ME, Bacon CJ, Herrmann DN, Zambon A, Tramacere I, Pisciotta C, Magri S, Previtali SC, Bolino A. A multicenter retrospective study of charcot-marie-tooth disease type 4B (CMT4B) associated with mutations in myotubularin-related proteins (MTMRs). Ann Neurol 2019; 86:55-67. [PMID: 31070812 DOI: 10.1002/ana.25500] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 04/23/2019] [Accepted: 05/05/2019] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Charcot-Marie-Tooth (CMT) disease 4B1 and 4B2 (CMT4B1/B2) are characterized by recessive inheritance, early onset, severe course, slowed nerve conduction, and myelin outfoldings. CMT4B3 shows a more heterogeneous phenotype. All are associated with myotubularin-related protein (MTMR) mutations. We conducted a multicenter, retrospective study to better characterize CMT4B. METHODS We collected clinical and genetic data from CMT4B subjects in 18 centers using a predefined minimal data set including Medical Research Council (MRC) scores of nine muscle pairs and CMT Neuropathy Score. RESULTS There were 50 patients, 21 of whom never reported before, carrying 44 mutations, of which 21 were novel and six representing novel disease associations of known rare variants. CMT4B1 patients had significantly more-severe disease than CMT4B2, with earlier onset, more-frequent motor milestones delay, wheelchair use, and respiratory involvement as well as worse MRC scores and motor CMT Examination Score components despite younger age at examination. Vocal cord involvement was common in both subtypes, whereas glaucoma occurred in CMT4B2 only. Nerve conduction velocities were similarly slowed in both subtypes. Regression analyses showed that disease severity is significantly associated with age in CMT4B1. Slopes are steeper for CMT4B1, indicating faster disease progression. Almost none of the mutations in the MTMR2 and MTMR13 genes, responsible for CMT4B1 and B2, respectively, influence the correlation between disease severity and age, in agreement with the hypothesis of a complete loss of function of MTMR2/13 proteins for such mutations. INTERPRETATION This is the largest CMT4B series ever reported, demonstrating that CMT4B1 is significantly more severe than CMT4B2, and allowing an estimate of prognosis. ANN NEUROL 2019.
Collapse
Affiliation(s)
- Davide Pareyson
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Tanya Stojkovic
- Hôpital Pitié-Salpêtrière, AP-HP, Centre de référence des maladies neuromusculaires Nord/Est/Ile de France, Paris, France
| | - Mary M Reilly
- MRC Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Sarah Leonard-Louis
- Hôpital Pitié-Salpêtrière, AP-HP, Centre de référence des maladies neuromusculaires Nord/Est/Ile de France, Paris, France
| | - Matilde Laurà
- MRC Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Julian Blake
- MRC Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom.,Department of Clinical Neurophysiology, Norfolk and Norwich University Hospital, Norfolk, United Kingdom
| | - Yesim Parman
- Istanbul University, Istanbul Faculty of Medicine, Neurology Dep. Istanbul, Turkey
| | - Esra Battaloglu
- Bogazici University, Department of Molecular Biology and Genetics, Istanbul, Turkey
| | - Meriem Tazir
- Laboratoire de Recherche en Neurosciences Service de Neurologie, CHU, Alger, Algeria
| | - Mounia Bellatache
- Laboratoire de Recherche en Neurosciences Service de Neurologie, CHU, Alger, Algeria
| | - Nathalie Bonello-Palot
- Department of Medical Genetics, Timone Hospital, Marseille, France.2, Aix-Marseille University, INSERM, MMG, U1251, Marseille, France
| | - Nicolas Lévy
- Department of Medical Genetics, Timone Hospital, Marseille, France.2, Aix-Marseille University, INSERM, MMG, U1251, Marseille, France
| | - Sabrina Sacconi
- Université Côte d'Azur, Service Système Nerveux Périphérique, Muscle et SLA, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Raquel Guimarães-Costa
- Hôpital Pitié-Salpêtrière, AP-HP, Centre de référence des maladies neuromusculaires Nord/Est/Ile de France, Paris, France
| | - Sharham Attarian
- Reference center for neuromuscular disorders and ALS, CHU La Timone, Aix-Marseille University, Marseille, France
| | - Philippe Latour
- Center of Biology and Pathology Laboratory of Molecular Neurogenetics, Hospices Civils, Lyon, France
| | - Guilhem Solé
- Reference center for neuromuscular disorders AOC (Atlantique Occitanie Caraibes), CHU de Bordeaux, Bordeaux, France
| | - André Megarbane
- Institut Jérôme Lejeune, Paris, France.,INOVIE, Beirut, Lebanon
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.,Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | - Giulia Ricci
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Byung-Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Angelo Schenone
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetic and MATERNAL Infantile Sciences, University of Genoa, and IRCCS Policlinico San Martino, Genoa, Italy
| | - Chiara Gemelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetic and MATERNAL Infantile Sciences, University of Genoa, and IRCCS Policlinico San Martino, Genoa, Italy
| | - Alessandro Geroldi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetic and MATERNAL Infantile Sciences, University of Genoa, and IRCCS Policlinico San Martino, Genoa, Italy
| | - Mario Sabatelli
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS. Centro Clinico Nemo Adulti Rome, Rome, Italy.,Università Cattolica del Sacro Cuore. Sede di Roma, Rome, Italy
| | - Marco Luigetti
- Università Cattolica del Sacro Cuore. Sede di Roma, Rome, Italy.,UOC Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Lucio Santoro
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University Federico II of Naples, Naples, Italy
| | - Fiore Manganelli
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University Federico II of Naples, Naples, Italy
| | - Aldo Quattrone
- Department of Neurology, Università Magna Graecia di Catanzaro, Catanzaro, Italy
| | - Paola Valentino
- Department of Neurology, Università Magna Graecia di Catanzaro, Catanzaro, Italy
| | | | - Steven S Scherer
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Lois Dankwa
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael E Shy
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa, IA
| | - Chelsea J Bacon
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa, IA
| | | | - Alberto Zambon
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Irene Tramacere
- Department of Research and Clinical Development, Scientific Directorate, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Pisciotta
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefania Magri
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefano C Previtali
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandra Bolino
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
27
|
Phosphatidylinositol 5 Phosphate (PI5P): From Behind the Scenes to the Front (Nuclear) Stage. Int J Mol Sci 2019; 20:ijms20092080. [PMID: 31035587 PMCID: PMC6539119 DOI: 10.3390/ijms20092080] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/20/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Phosphatidylinositol (PI)-related signaling plays a pivotal role in many cellular aspects, including survival, cell proliferation, differentiation, DNA damage, and trafficking. PI is the core of a network of proteins represented by kinases, phosphatases, and lipases which are able to add, remove or hydrolyze PI, leading to different phosphoinositide products. Among the seven known phosphoinositides, phosphatidylinositol 5 phosphate (PI5P) was the last to be discovered. PI5P presence in cells is very low compared to other PIs. However, much evidence collected throughout the years has described the role of this mono-phosphoinositide in cell cycles, stress response, T-cell activation, and chromatin remodeling. Interestingly, PI5P has been found in different cellular compartments, including the nucleus. Here, we will review the nuclear role of PI5P, describing how it is synthesized and regulated, and how changes in the levels of this rare phosphoinositide can lead to different nuclear outputs.
Collapse
|
28
|
Wang H, Schänzer A, Kampschulte B, Daimagüler HS, Logeswaran T, Schlierbach H, Petzinger J, Ehrhardt H, Hahn A, Cirak S. A novel SPEG mutation causes non-compaction cardiomyopathy and neuropathy in a floppy infant with centronuclear myopathy. Acta Neuropathol Commun 2018; 6:83. [PMID: 30157964 PMCID: PMC6114030 DOI: 10.1186/s40478-018-0589-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 08/21/2018] [Indexed: 01/02/2023] Open
|
29
|
Tied up: Does altering phosphoinositide-mediated membrane trafficking influence neurodegenerative disease phenotypes? J Genet 2018. [DOI: 10.1007/s12041-018-0961-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
30
|
Nadiminti SSP, Kamak M, Koushika SP. Tied up: Does altering phosphoinositide-mediated membrane trafficking influence neurodegenerative disease phenotypes? J Genet 2018; 97:753-771. [PMID: 30027907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Phosphoinositides are a class of membrane lipids that are found on several intracellular compartments and play diverse roles inside cells, such as vesicle formation, protein trafficking, endocytosis etc. Intracellular distribution and levels of phosphoinositides are regulated by enzymes that generate and breakdown these lipids as well as other proteins that associate with phosphoinositides. These events lead to differing levels of specific phosphoinositides on different intracellular compartments. At these intracellular locations, phosphoinositides and their associated proteins, such as Rab GTPases, dynamin and BAR domain-containing proteins, regulate a variety of membrane trafficking pathways. Neurodegenerative phenotypes in disorders such as Parkinson's disease (PD) can arise as a consequence of altered or hampered intracellular trafficking. Altered trafficking can cause proteins such as α-synuclein to aggregate intracellularly. Several trafficking pathways are regulated bymaster regulators such as LRRK2,which is known to regulate the activity of phosphoinositide effector proteins. Perturbing either the levels of phosphoinositides or their interactions with different proteins disrupts intracellular trafficking pathways, contributing to phenotypes often observed in disorders such as Alzheimer's or PDs. Thus, studying phosphoinositide regulation and its role in trafficking can give us a deeper understanding of the contribution of disrupted trafficking to neurodegenerative phenotypes.
Collapse
Affiliation(s)
- Sravanthi S P Nadiminti
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400 005, India.
| | | | | |
Collapse
|
31
|
Narayanan P, Hütte M, Kudryasheva G, Taberner FJ, Lechner SG, Rehfeldt F, Gomez-Varela D, Schmidt M. Myotubularin related protein-2 and its phospholipid substrate PIP 2 control Piezo2-mediated mechanotransduction in peripheral sensory neurons. eLife 2018. [PMID: 29521261 PMCID: PMC5898911 DOI: 10.7554/elife.32346] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Piezo2 ion channels are critical determinants of the sense of light touch in vertebrates. Yet, their regulation is only incompletely understood. We recently identified myotubularin related protein-2 (Mtmr2), a phosphoinositide (PI) phosphatase, in the native Piezo2 interactome of murine dorsal root ganglia (DRG). Here, we demonstrate that Mtmr2 attenuates Piezo2-mediated rapidly adapting mechanically activated (RA-MA) currents. Interestingly, heterologous Piezo1 and other known MA current subtypes in DRG appeared largely unaffected by Mtmr2. Experiments with catalytically inactive Mtmr2, pharmacological blockers of PI(3,5)P2 synthesis, and osmotic stress suggest that Mtmr2-dependent Piezo2 inhibition involves depletion of PI(3,5)P2. Further, we identified a PI(3,5)P2 binding region in Piezo2, but not Piezo1, that confers sensitivity to Mtmr2 as indicated by functional analysis of a domain-swapped Piezo2 mutant. Altogether, our results propose local PI(3,5)P2 modulation via Mtmr2 in the vicinity of Piezo2 as a novel mechanism to dynamically control Piezo2-dependent mechanotransduction in peripheral sensory neurons. We often take our sense of touch for granted. Yet, our every-day life greatly depends on the ability to perceive our environment to alert us of danger or to further social interactions, such as mother-child bonding. Our sense of touch relies on the conversion of mechanical stimuli to electrical signals (this is known as mechanotransduction), which then travel to brain to be processed. This task is fulfilled by specific ion channels called Piezo2, which are activated when cells are exposed to pressure and other mechanical forces. These channels can be found in sensory nerves and specialized structures in the skin, where they help to detect physical contact, roughness of surfaces and the position of our body parts. It is still not clear how Piezo2 channels are regulated but previous research by several laboratories suggests that they work in conjunction with other proteins. One of these proteins is the myotubularin related protein-2, or Mtmr2 for short. Now, Narayanan et al. – including some of the researchers involved in the previous research – set out to advance our understanding of the molecular basis of touch and looked more closely at Mtmr2. To test if Mtmr2 played a role in mechanotransduction, Narayanan et al. both increased and reduced the levels of this protein in sensory neurons of mice grown in the laboratory. When Mtmr2 levels were low, the activity of Piezo2 channels increased. However, when the protein levels were high, Piezo2 channels were inhibited. These results suggest that Mtmr2 can control the activity of Piezo2. Further experiments, in which Mtmr2 was genetically modified or sensory neurons were treated with chemicals, revealed that Mtmr2 reduces a specific fatty acid in the membrane of nerve cells, which in turn attenuates the activity of Piezo2. This study identified Mtmr2 and distinct fatty acids in the cell membrane as new components of the complex setup required for the sense of touch. A next step will be to test if these molecules also influence the activity of Piezo2 when the skin has become injured or upon inflammation.
Collapse
Affiliation(s)
- Pratibha Narayanan
- Emmy Noether-Group Somatosensory Signaling and Systems Biology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| | - Meike Hütte
- Emmy Noether-Group Somatosensory Signaling and Systems Biology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| | - Galina Kudryasheva
- Third Institute of Physics - Biophysics, University of Goettingen, Goettingen, Germany
| | | | | | - Florian Rehfeldt
- Third Institute of Physics - Biophysics, University of Goettingen, Goettingen, Germany
| | - David Gomez-Varela
- Emmy Noether-Group Somatosensory Signaling and Systems Biology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| | - Manuela Schmidt
- Emmy Noether-Group Somatosensory Signaling and Systems Biology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| |
Collapse
|
32
|
Abstract
Our understanding of the processes controlling peripheral nervous system myelination have been significantly benefited by the development of an in vitro myelinating culture system in which primary Schwann cells are cocultured together with primary sensory neurons. In this chapter, we describe the protocol currently used in our laboratories to establish Schwann cells neuronal myelinating cocultures. We also include a detailed description of the various substrates that can be used to establish it.
Collapse
Affiliation(s)
- Carla Taveggia
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, Milan, Italy.
| | - Alessandra Bolino
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
33
|
Yamaguchi M, Takashima H. Drosophila Charcot-Marie-Tooth Disease Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1076:97-117. [PMID: 29951817 DOI: 10.1007/978-981-13-0529-0_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Charcot-Marie-Tooth disease (CMT) was initially described in 1886. It is characterized by defects in the peripheral nervous system, including sensory and motor neurons. Although more than 80 CMT-causing genes have been identified to date, an effective therapy has not yet been developed for this disease. Since Drosophila does not have axons surrounded by myelin sheaths or Schwann cells, the establishment of a demyelinating CMT model is not appropriate. In this chapter, after overviewing CMT, examples of Drosophila CMT models with axonal neuropathy and other animal CMT models are described.
Collapse
Affiliation(s)
| | - Hiroshi Takashima
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
34
|
Robinson DC, Mammel AE, Logan AM, Larson AA, Schmidt EJ, Condon AF, Robinson FL. An In Vitro Model of Charcot-Marie-Tooth Disease Type 4B2 Provides Insight Into the Roles of MTMR13 and MTMR2 in Schwann Cell Myelination. ASN Neuro 2018; 10:1759091418803282. [PMID: 30419760 PMCID: PMC6236487 DOI: 10.1177/1759091418803282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 01/16/2023] Open
Abstract
Charcot-Marie-Tooth Disorder Type 4B (CMT4B) is a demyelinating peripheral neuropathy caused by mutations in myotubularin-related (MTMR) proteins 2, 13, or 5 (CMT4B1/2/3), which regulate phosphoinositide turnover and endosomal trafficking. Although mouse models of CMT4B2 exist, an in vitro model would make possible pharmacological and reverse genetic experiments needed to clarify the role of MTMR13 in myelination. We have generated such a model using Schwann cell-dorsal root ganglion (SC-DRG) explants from Mtmr13-/- mice. Myelin sheaths in mutant cultures contain outfoldings highly reminiscent of those observed in the nerves of Mtmr13-/- mice and CMT4B2 patients. Mtmr13-/- SC-DRG explants also contain reduced Mtmr2, further supporting a role of Mtmr13 in stabilizing Mtmr2. Elevated PI(3,5)P2 has been implicated as a cause of myelin outfoldings in Mtmr2-/- models. In contrast, the role of elevated PI3P or PI(3,5)P2 in promoting outfoldings in Mtmr13-/- models is unclear. We found that over-expression of MTMR2 in Mtmr13-/- SC-DRGs moderately reduced the prevalence of myelin outfoldings. Thus, a manipulation predicted to lower PI3P and PI(3,5)P2 partially suppressed the phenotype caused by Mtmr13 deficiency. We also explored the relationship between CMT4B2-like myelin outfoldings and kinases that produce PI3P and PI(3,5)P2 by analyzing nerve pathology in mice lacking both Mtmr13 and one of two specific PI 3-kinases. Intriguingly, the loss of vacuolar protein sorting 34 or PI3K-C2β in Mtmr13-/- mice had no impact on the prevalence of myelin outfoldings. In aggregate, our findings suggest that the MTMR13 scaffold protein likely has critical functions other than stabilizing MTMR2 to achieve an adequate level of PI 3-phosphatase activity.
Collapse
Affiliation(s)
- Danielle C. Robinson
- Department of Neurology, Jungers Center for
Neurosciences Research, Oregon Health & Science University,
Portland, OR, USA
- Neuroscience Graduate Program, Oregon Health &
Science University, Portland, OR, USA
| | - Anna E. Mammel
- Department of Neurology, Jungers Center for
Neurosciences Research, Oregon Health & Science University,
Portland, OR, USA
- Cell, Developmental & Cancer Biology Graduate
Program, Oregon Health & Science University, Portland, OR,
USA
| | - Anne M. Logan
- Department of Neurology, Jungers Center for
Neurosciences Research, Oregon Health & Science University,
Portland, OR, USA
- Neuroscience Graduate Program, Oregon Health &
Science University, Portland, OR, USA
| | - Aubree A. Larson
- Department of Neurology, Jungers Center for
Neurosciences Research, Oregon Health & Science University,
Portland, OR, USA
| | - Eric J. Schmidt
- Department of Neurology, Jungers Center for
Neurosciences Research, Oregon Health & Science University,
Portland, OR, USA
| | - Alec F. Condon
- Department of Neurology, Jungers Center for
Neurosciences Research, Oregon Health & Science University,
Portland, OR, USA
- Neuroscience Graduate Program, Oregon Health &
Science University, Portland, OR, USA
| | - Fred L. Robinson
- Department of Neurology, Jungers Center for
Neurosciences Research, Oregon Health & Science University,
Portland, OR, USA
- Vollum Institute, Oregon Health & Science
University, Portland, OR, USA
| |
Collapse
|
35
|
Sphingomyelin as a myelin biomarker in CSF of acquired demyelinating neuropathies. Sci Rep 2017; 7:7831. [PMID: 28798317 PMCID: PMC5552737 DOI: 10.1038/s41598-017-08314-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/07/2017] [Indexed: 01/06/2023] Open
Abstract
Fast, accurate and reliable methods to quantify the amount of myelin still lack, both in humans and experimental models. The overall objective of the present study was to demonstrate that sphingomyelin (SM) in the cerebrospinal fluid (CSF) of patients affected by demyelinating neuropathies is a myelin biomarker. We found that SM levels mirror both peripheral myelination during development and small myelin rearrangements in experimental models. As in acquired demyelinating peripheral neuropathies myelin breakdown occurs, SM amount in the CSF of these patients might detect the myelin loss. Indeed, quantification of SM in 262 neurological patients showed a significant increase in patients with peripheral demyelination (p = 3.81 * 10 − 8) compared to subjects affected by non-demyelinating disorders. Interestingly, SM alone was able to distinguish demyelinating from axonal neuropathies and differs from the principal CSF indexes, confirming the novelty of this potential CSF index. In conclusion, SM is a specific and sensitive biomarker to monitor myelin pathology in the CSF of peripheral neuropathies. Most importantly, SM assay is simple, fast, inexpensive, and promising to be used in clinical practice and drug development.
Collapse
|
36
|
Logan AM, Mammel AE, Robinson DC, Chin AL, Condon AF, Robinson FL. Schwann cell-specific deletion of the endosomal PI 3-kinase Vps34 leads to delayed radial sorting of axons, arrested myelination, and abnormal ErbB2-ErbB3 tyrosine kinase signaling. Glia 2017; 65:1452-1470. [PMID: 28617998 DOI: 10.1002/glia.23173] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 05/04/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022]
Abstract
The PI 3-kinase Vps34 (Pik3c3) synthesizes phosphatidylinositol 3-phosphate (PI3P), a lipid critical for both endosomal membrane traffic and macroautophagy. Human genetics have implicated PI3P dysregulation, and endosomal trafficking in general, as a recurring cause of demyelinating Charcot-Marie-Tooth (CMT) peripheral neuropathy. Here, we investigated the role of Vps34, and PI3P, in mouse Schwann cells by selectively deleting Vps34 in this cell type. Vps34-Schwann cell knockout (Vps34SCKO ) mice show severe hypomyelination in peripheral nerves. Vps34-/- Schwann cells interact abnormally with axons, and there is a delay in radial sorting, a process by which large axons are selected for myelination. Upon reaching the promyelinating stage, Vps34-/- Schwann cells are significantly impaired in the elaboration of myelin. Nerves from Vps34SCKO mice contain elevated levels of the LC3 and p62 proteins, indicating impaired autophagy. However, in the light of recent demonstrations that autophagy is dispensable for myelination, it is unlikely that hypomyelination in Vps34SCKO mice is caused by impaired autophagy. Endosomal trafficking is also disturbed in Vps34-/- Schwann cells. We investigated the activation of the ErbB2/3 receptor tyrosine kinases in Vps34SCKO nerves, as these proteins, which play essential roles in Schwann cell myelination, are known to traffic through endosomes. In Vps34SCKO nerves, ErbB3 was hyperphosphorylated on a tyrosine known to be phosphorylated in response to neuregulin 1 exposure. ErbB2 protein levels were also decreased during myelination. Our findings suggest that the loss of Vps34 alters the trafficking of ErbB2/3 through endosomes. Abnormal ErbB2/3 signaling to downstream targets may contribute to the hypomyelination observed in Vps34SCKO mice.
Collapse
Affiliation(s)
- Anne M Logan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Neuroscience Graduate Program, Oregon Health and Science University, Portland, Oregon, 97239
| | - Anna E Mammel
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Cell, Developmental and Cancer Biology Graduate Program, Oregon Health and Science University, Portland, Oregon, 97239
| | - Danielle C Robinson
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Neuroscience Graduate Program, Oregon Health and Science University, Portland, Oregon, 97239
| | - Andrea L Chin
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239
| | - Alec F Condon
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Neuroscience Graduate Program, Oregon Health and Science University, Portland, Oregon, 97239
| | - Fred L Robinson
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Vollum Institute, Oregon Health and Science University, Portland, Oregon, 97239
| |
Collapse
|
37
|
De Craene JO, Bertazzi DL, Bär S, Friant S. Phosphoinositides, Major Actors in Membrane Trafficking and Lipid Signaling Pathways. Int J Mol Sci 2017; 18:ijms18030634. [PMID: 28294977 PMCID: PMC5372647 DOI: 10.3390/ijms18030634] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/02/2017] [Accepted: 03/06/2017] [Indexed: 11/30/2022] Open
Abstract
Phosphoinositides are lipids involved in the vesicular transport of proteins and lipids between the different compartments of eukaryotic cells. They act by recruiting and/or activating effector proteins and thus are involved in regulating various cellular functions, such as vesicular budding, membrane fusion and cytoskeleton dynamics. Although detected in small concentrations in membranes, their role is essential to cell function, since imbalance in their concentrations is a hallmark of many cancers. Their synthesis involves phosphorylating/dephosphorylating positions D3, D4 and/or D5 of their inositol ring by specific lipid kinases and phosphatases. This process is tightly regulated and specific to the different intracellular membranes. Most enzymes involved in phosphoinositide synthesis are conserved between yeast and human, and their loss of function leads to severe diseases (cancer, myopathy, neuropathy and ciliopathy).
Collapse
Affiliation(s)
- Johan-Owen De Craene
- Department of Molecular and Cellular Genetics, Université de Strasbourg, CNRS, GMGM UMR 7156, F-67000 Strasbourg, France.
| | - Dimitri L Bertazzi
- Department of Molecular and Cellular Genetics, Université de Strasbourg, CNRS, GMGM UMR 7156, F-67000 Strasbourg, France.
| | - Séverine Bär
- Department of Molecular and Cellular Genetics, Université de Strasbourg, CNRS, GMGM UMR 7156, F-67000 Strasbourg, France.
| | - Sylvie Friant
- Department of Molecular and Cellular Genetics, Université de Strasbourg, CNRS, GMGM UMR 7156, F-67000 Strasbourg, France.
| |
Collapse
|
38
|
Hasegawa J, Strunk BS, Weisman LS. PI5P and PI(3,5)P 2: Minor, but Essential Phosphoinositides. Cell Struct Funct 2017; 42:49-60. [PMID: 28302928 DOI: 10.1247/csf.17003] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In most eukaryotes, phosphoinositides (PIs) have crucial roles in multiple cellular functions. Although the cellular levels of phosphatidylinositol 5-phosphate (PI5P) and phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) are extremely low relative to some other PIs, emerging evidence demonstrates that both lipids are crucial for the endocytic pathway, intracellular signaling, and adaptation to stress. Mutations that causes defects in the biosynthesis of PI5P and PI(3,5)P2 are linked to human diseases including neurodegenerative disorders. Here, we review recent findings on cellular roles of PI5P and PI(3,5)P2, as well as the pathophysiological importance of these lipids.Key words: Phosphoinositides, Membrane trafficking, Endocytosis, Vacuoles/Lysosomes, Fab1/PIKfyve.
Collapse
|
39
|
Mucke HA. Drug Repurposing Patent Applications July–September 2016. Assay Drug Dev Technol 2016; 14:577-582. [DOI: 10.1089/adt.2016.29053.pq3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
40
|
Bolino A, Piguet F, Alberizzi V, Pellegatta M, Rivellini C, Guerrero-Valero M, Noseda R, Brombin C, Nonis A, D'Adamo P, Taveggia C, Previtali SC. Niacin-mediated Tace activation ameliorates CMT neuropathies with focal hypermyelination. EMBO Mol Med 2016; 8:1438-1454. [PMID: 27799291 PMCID: PMC5167133 DOI: 10.15252/emmm.201606349] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Charcot–Marie–Tooth (CMT) neuropathies are highly heterogeneous disorders caused by mutations in more than 70 genes, with no available treatment. Thus, it is difficult to envisage a single suitable treatment for all pathogenetic mechanisms. Axonal Neuregulin 1 (Nrg1) type III drives Schwann cell myelination and determines myelin thickness by ErbB2/B3‐PI3K–Akt signaling pathway activation. Nrg1 type III is inhibited by the α‐secretase Tace, which negatively regulates PNS myelination. We hypothesized that modulation of Nrg1 levels and/or secretase activity may constitute a unifying treatment strategy for CMT neuropathies with focal hypermyelination as it could restore normal levels of myelination. Here we show that in vivo delivery of Niaspan, a FDA‐approved drug known to enhance TACE activity, efficiently rescues myelination in the Mtmr2−/− mouse, a model of CMT4B1 with myelin outfoldings, and in the Pmp22+/− mouse, which reproduces HNPP (hereditary neuropathy with liability to pressure palsies) with tomacula. Importantly, we also found that Niaspan reduces hypermyelination of Vim (vimentin)−/− mice, characterized by increased Nrg1 type III and Akt activation, thus corroborating the hypothesis that Niaspan treatment downregulates Nrg1 type III signaling.
Collapse
Affiliation(s)
- Alessandra Bolino
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy .,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Françoise Piguet
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Alberizzi
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Marta Pellegatta
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Rivellini
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Marta Guerrero-Valero
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Noseda
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Brombin
- University Centre of Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Nonis
- University Centre of Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Patrizia D'Adamo
- Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Carla Taveggia
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Carlo Previtali
- INSPE-Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy.,Department of Neurology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
41
|
Phosphatidylinositol 3,5-bisphosphate: regulation of cellular events in space and time. Biochem Soc Trans 2016; 44:177-84. [PMID: 26862203 DOI: 10.1042/bst20150174] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Phosphorylated phosphatidylinositol lipids are crucial for most eukaryotes and have diverse cellular functions. The low-abundance signalling lipid phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] is critical for cellular homoeostasis and adaptation to stimuli. A large complex of proteins that includes the lipid kinase Fab1-PIKfyve, dynamically regulates the levels of PI(3,5)P2. Deficiencies in PI(3,5)P2 are linked to some human diseases, especially those of the nervous system. Future studies will probably determine new, undiscovered regulatory roles of PI(3,5)P2, as well as uncover mechanistic insights into how PI(3,5)P2 contributes to normal human physiology.
Collapse
|
42
|
Sabha N, Volpatti JR, Gonorazky H, Reifler A, Davidson AE, Li X, Eltayeb NM, Dall'Armi C, Di Paolo G, Brooks SV, Buj-Bello A, Feldman EL, Dowling JJ. PIK3C2B inhibition improves function and prolongs survival in myotubular myopathy animal models. J Clin Invest 2016; 126:3613-25. [PMID: 27548528 DOI: 10.1172/jci86841] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/19/2016] [Indexed: 11/17/2022] Open
Abstract
Myotubular myopathy (MTM) is a devastating pediatric neuromuscular disorder of phosphoinositide (PIP) metabolism resulting from mutations of the PIP phosphatase MTM1 for which there are no treatments. We have previously shown phosphatidylinositol-3-phosphate (PI3P) accumulation in animal models of MTM. Here, we tested the hypothesis that lowering PI3P levels may prevent or reverse the MTM disease process. To test this, we targeted class II and III PI3 kinases (PI3Ks) in an MTM1-deficient mouse model. Muscle-specific ablation of Pik3c2b, but not Pik3c3, resulted in complete prevention of the MTM phenotype, and postsymptomatic targeting promoted a striking rescue of disease. We confirmed this genetic interaction in zebrafish, and additionally showed that certain PI3K inhibitors prevented development of the zebrafish mtm phenotype. Finally, the PI3K inhibitor wortmannin improved motor function and prolonged lifespan of the Mtm1-deficient mice. In all, we have identified Pik3c2b as a genetic modifier of Mtm1 mutation and demonstrated that PIK3C2B inhibition is a potential treatment strategy for MTM. In addition, we set the groundwork for similar reciprocal inhibition approaches for treating other PIP metabolic disorders and highlight the importance of modifier gene pathways as therapeutic targets.
Collapse
|
43
|
Fogarty EA, Brewer MH, Rodriguez-Molina JF, Law WD, Ma KH, Steinberg NM, Svaren J, Antonellis A. SOX10 regulates an alternative promoter at the Charcot-Marie-Tooth disease locus MTMR2. Hum Mol Genet 2016; 25:3925-3936. [PMID: 27466180 DOI: 10.1093/hmg/ddw233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/21/2016] [Accepted: 07/11/2016] [Indexed: 11/13/2022] Open
Abstract
Schwann cells are the myelinating glia of the peripheral nervous system and dysfunction of these cells causes motor and sensory peripheral neuropathy. The transcription factor SOX10 is critical for Schwann cell development and maintenance, and many SOX10 target genes encode proteins required for Schwann cell function. Loss-of-function mutations in the gene encoding myotubularin-related protein 2 (MTMR2) cause Charcot-Marie-Tooth disease type 4B1 (CMT4B1), a severe demyelinating peripheral neuropathy characterized by myelin outfoldings along peripheral nerves. Previous reports indicate that MTMR2 is ubiquitously expressed making it unclear how loss of this gene causes a Schwann cell-specific phenotype. To address this, we performed computational and functional analyses at MTMR2 to identify transcriptional regulatory elements important for Schwann cell expression. Through these efforts, we identified an alternative, SOX10-responsive promoter at MTMR2 that displays strong regulatory activity in immortalized rat Schwann (S16) cells. This promoter directs transcription of a previously unidentified MTMR2 transcript that is enriched in mouse Schwann cells compared to immortalized mouse motor neurons (MN-1), and is predicted to encode an N-terminally truncated protein isoform. The expression of the endogenous transcript is induced in a heterologous cell line by ectopically expressing SOX10, and is nearly ablated in Schwann cells by impairing SOX10 function. Intriguingly, overexpressing the two MTMR2 protein isoforms in HeLa cells revealed that both localize to nuclear puncta and the shorter isoform displays higher nuclear localization compared to the longer isoform. Combined, our data warrant further investigation of the truncated MTMR2 protein isoform in Schwann cells and in CMT4B1 pathogenesis.
Collapse
Affiliation(s)
| | - Megan H Brewer
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - William D Law
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ki H Ma
- Cellular and Molecular Pathology (CMP) Program
| | - Noah M Steinberg
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - John Svaren
- Waisman Center.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Anthony Antonellis
- Neuroscience Graduate Program .,Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.,Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Charcot-Marie-Tooth disease (CMT) is the common terminology used to describe the hereditary neuropathies. This update reviews advances in the past year in our understanding of these diseases, including some important earlier references. RECENT FINDINGS In the past year, advances in next-generation sequencing continued to increase the number of genes associated with CMT. The connection between genotype and phenotype has become more complicated. New insights into the pathogenesis of the diseases are reviewed. Treatment and clinical trial updates coming from these new insights, as well as use of high-throughput screening to match potential treatments with targets, are moving the field forward. There is a discussion of potential next steps, including the use of patient-derived induced pluripotent stem cells, to enhance our understanding of individual genotypes and phenotypes. SUMMARY The use of high-throughput screens, and techniques such as RNAi and induced pluripotent stem cell continue to push forward other therapies for specific genetic forms of CMT and are potentially more generalizable to peripheral neuropathies. These developments, along with the development of improved outcome measures and longitudinal natural history data, advance CMT, making the future for finding treatments and/or cures closer than it has ever been.
Collapse
|
45
|
Dysregulation of ErbB Receptor Trafficking and Signaling in Demyelinating Charcot-Marie-Tooth Disease. Mol Neurobiol 2016; 54:87-100. [PMID: 26732592 DOI: 10.1007/s12035-015-9668-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 12/17/2015] [Indexed: 12/12/2022]
Abstract
Charcot-Marie-Tooth (CMT) disease is the most common inherited peripheral neuropathy with the majority of cases involving demyelination of peripheral nerves. The pathogenic mechanisms of demyelinating CMT remain unclear, and no effective therapy currently exists for this disease. The discovery that mutations in different genes can cause a similar phenotype of demyelinating peripheral neuropathy raises the possibility that there may be convergent mechanisms leading to demyelinating CMT pathogenesis. Increasing evidence indicates that ErbB receptor-mediated signaling plays a major role in the control of Schwann cell-axon communication and myelination in the peripheral nervous system. Recent studies reveal that several demyelinating CMT-linked proteins are novel regulators of endocytic trafficking and/or phosphoinositide metabolism that may affect ErbB receptor signaling. Emerging data have begun to suggest that dysregulation of ErbB receptor trafficking and signaling in Schwann cells may represent a common pathogenic mechanism in multiple subtypes of demyelinating CMT. In this review, we focus on the roles of ErbB receptor trafficking and signaling in regulation of peripheral nerve myelination and discuss the emerging evidence supporting the potential involvement of altered ErbB receptor trafficking and signaling in demyelinating CMT pathogenesis and the possibility of modulating these trafficking and signaling processes for treating demyelinating peripheral neuropathy.
Collapse
|
46
|
Ikonomov OC, Sbrissa D, Compton LM, Kumar R, Tisdale EJ, Chen X, Shisheva A. The Protein Complex of Neurodegeneration-related Phosphoinositide Phosphatase Sac3 and ArPIKfyve Binds the Lewy Body-associated Synphilin-1, Preventing Its Aggregation. J Biol Chem 2015; 290:28515-28529. [PMID: 26405034 DOI: 10.1074/jbc.m115.669929] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Indexed: 12/14/2022] Open
Abstract
The 5-phosphoinositide phosphatase Sac3, in which loss-of-function mutations are linked to neurodegenerative disorders, forms a stable cytosolic complex with the scaffolding protein ArPIKfyve. The ArPIKfyve-Sac3 heterodimer interacts with the phosphoinositide 5-kinase PIKfyve in a ubiquitous ternary complex that couples PtdIns(3,5)P2 synthesis with turnover at endosomal membranes, thereby regulating the housekeeping endocytic transport in eukaryotes. Neuron-specific associations of the ArPIKfyve-Sac3 heterodimer, which may shed light on the neuropathological mechanisms triggered by Sac3 dysfunction, are unknown. Here we conducted mass spectrometry analysis for brain-derived interactors of ArPIKfyve-Sac3 and unraveled the α-synuclein-interacting protein Synphilin-1 (Sph1) as a new component of the ArPIKfyve-Sac3 complex. Sph1, a predominantly neuronal protein that facilitates aggregation of α-synuclein, is a major component of Lewy body inclusions in neurodegenerative α-synucleinopathies. Modulations in ArPIKfyve/Sac3 protein levels by RNA silencing or overexpression in several mammalian cell lines, including human neuronal SH-SY5Y or primary mouse cortical neurons, revealed that the ArPIKfyve-Sac3 complex specifically altered the aggregation properties of Sph1-GFP. This effect required an active Sac3 phosphatase and proceeded through mechanisms that involved increased Sph1-GFP partitioning into the cytosol and removal of Sph1-GFP aggregates by basal autophagy but not by the proteasomal system. If uncoupled from ArPIKfyve elevation, overexpressed Sac3 readily aggregated, markedly enhancing the aggregation potential of Sph1-GFP. These data identify a novel role of the ArPIKfyve-Sac3 complex in the mechanisms controlling aggregate formation of Sph1 and suggest that Sac3 protein deficiency or overproduction may facilitate aggregation of aggregation-prone proteins, thereby precipitating the onset of multiple neuronal disorders.
Collapse
Affiliation(s)
- Ognian C Ikonomov
- Departments of Physiology, Wayne State School of Medicine, Detroit, Michigan 48201
| | - Diego Sbrissa
- Departments of Physiology, Wayne State School of Medicine, Detroit, Michigan 48201
| | - Lauren M Compton
- Departments of Physiology, Wayne State School of Medicine, Detroit, Michigan 48201
| | - Rita Kumar
- Departments of Physiology, Wayne State School of Medicine, Detroit, Michigan 48201; Departments of Emergency Medicine, Wayne State School of Medicine, Detroit, Michigan 48201
| | - Ellen J Tisdale
- Departments of Pharmacology, Wayne State School of Medicine, Detroit, Michigan 48201
| | - Xuequn Chen
- Departments of Physiology, Wayne State School of Medicine, Detroit, Michigan 48201
| | - Assia Shisheva
- Departments of Physiology, Wayne State School of Medicine, Detroit, Michigan 48201.
| |
Collapse
|
47
|
Reactivation of Lysosomal Ca2+ Efflux Rescues Abnormal Lysosomal Storage in FIG4-Deficient Cells. J Neurosci 2015; 35:6801-12. [PMID: 25926456 DOI: 10.1523/jneurosci.4442-14.2015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Loss of function of FIG4 leads to Charcot-Marie-Tooth disease Type 4J, Yunis-Varon syndrome, or an epilepsy syndrome. FIG4 is a phosphatase with its catalytic specificity toward 5'-phosphate of phosphatidylinositol-3,5-diphosphate (PI3,5P2). However, the loss of FIG4 decreases PI3,5P2 levels likely due to FIG4's dominant effect in scaffolding a PI3,5P2 synthetic protein complex. At the cellular level, all these diseases share similar pathology with abnormal lysosomal storage and neuronal degeneration. Mice with no FIG4 expression (Fig4(-/-)) recapitulate the pathology in humans with FIG4 deficiency. Using a flow cytometry technique that rapidly quantifies lysosome sizes, we detected an impaired lysosomal fission, but normal fusion, in Fig4(-/-) cells. The fission defect was associated with a robust increase of intralysosomal Ca(2+) in Fig4(-/-) cells, including FIG4-deficient neurons. This finding was consistent with a suppressed Ca(2+) efflux of lysosomes because the endogenous ligand of lysosomal Ca(2+) channel TRPML1 is PI3,5P2 that is deficient in Fig4(-/-) cells. We reactivated the TRPML1 channels by application of TRPML1 synthetic ligand, ML-SA1. This treatment reduced the intralysosomal Ca(2+) level and rescued abnormal lysosomal storage in Fig4(-/-) culture cells and ex vivo DRGs. Furthermore, we found that the suppressed Ca(2+) efflux in Fig4(-/-) culture cells and Fig4(-/-) mouse brains profoundly downregulated the expression/activity of dynamin-1, a GTPase known to scissor organelle membranes during fission. This downregulation made dynamin-1 unavailable for lysosomal fission. Together, our study revealed a novel mechanism explaining abnormal lysosomal storage in FIG4 deficiency. Synthetic ligands of the TRPML1 may become a potential therapy against diseases with FIG4 deficiency.
Collapse
|
48
|
Waugh MG. PIPs in neurological diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1066-82. [PMID: 25680866 DOI: 10.1016/j.bbalip.2015.02.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/29/2015] [Accepted: 02/01/2015] [Indexed: 12/19/2022]
Abstract
Phosphoinositide (PIP) lipids regulate many aspects of cell function in the nervous system including receptor signalling, secretion, endocytosis, migration and survival. Levels of PIPs such as PI4P, PI(4,5)P2 and PI(3,4,5)P3 are normally tightly regulated by phosphoinositide kinases and phosphatases. Deregulation of these biochemical pathways leads to lipid imbalances, usually on intracellular endosomal membranes, and these changes have been linked to a number of major neurological diseases including Alzheimer's, Parkinson's, epilepsy, stroke, cancer and a range of rarer inherited disorders including brain overgrowth syndromes, Charcot-Marie-Tooth neuropathies and neurodevelopmental conditions such as Lowe's syndrome. This article analyses recent progress in this area and explains how PIP lipids are involved, to varying degrees, in almost every class of neurological disease. This article is part of a Special Issue entitled Brain Lipids.
Collapse
Affiliation(s)
- Mark G Waugh
- Lipid and Membrane Biology Group, Institute for Liver and Digestive Health, UCL, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom.
| |
Collapse
|
49
|
Idevall-Hagren O, De Camilli P. Detection and manipulation of phosphoinositides. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:736-45. [PMID: 25514766 DOI: 10.1016/j.bbalip.2014.12.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/27/2014] [Accepted: 12/09/2014] [Indexed: 12/23/2022]
Abstract
Phosphoinositides (PIs) are minor components of cell membranes, but play key roles in cell function. Recent refinements in techniques for their detection, together with imaging methods to study their distribution and changes, have greatly facilitated the study of these lipids. Such methods have been complemented by the parallel development of techniques for the acute manipulation of their levels, which in turn allow bypassing the long-term adaptive changes implicit in genetic perturbations. Collectively, these advancements have helped elucidate the role of PIs in physiology and the impact of the dysfunction of their metabolism in disease. Combining methods for detection and manipulation enables the identification of specific roles played by each of the PIs and may eventually lead to the complete deconstruction of the PI signaling network. Here, we review current techniques used for the study and manipulation of cellular PIs and also discuss advantages and disadvantages associated with the various methods. This article is part of a Special Issue entitled Phosphoinositides.
Collapse
Affiliation(s)
- Olof Idevall-Hagren
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, 75123 Uppsala, Sweden.
| | - Pietro De Camilli
- Department of Cell Biology, Howard Hughes Medical Institute and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
50
|
Mendelian disorders of PI metabolizing enzymes. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:867-81. [PMID: 25510381 DOI: 10.1016/j.bbalip.2014.12.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/18/2014] [Accepted: 12/01/2014] [Indexed: 12/18/2022]
Abstract
More than twenty different genetic diseases have been described that are caused by mutations in phosphoinositide metabolizing enzymes, mostly in phosphoinositide phosphatases. Although generally ubiquitously expressed, mutations in these enzymes, which are mainly loss-of-function, result in tissue-restricted clinical manifestations through mechanisms that are not completely understood. Here we analyze selected disorders of phosphoinositide metabolism grouped according to the principle tissue affected: the nervous system, muscle, kidney, the osteoskeletal system, the eye, and the immune system. We will highlight what has been learnt so far from the study of these disorders about not only the cellular and molecular pathways that are involved or are governed by phosphoinositides, but also the many gaps that remain to be filled to gain a full understanding of the pathophysiological mechanisms underlying the clinical manifestations of this steadily growing class of diseases, most of which still remain orphan in terms of treatment. This article is part of a Special Issue entitled Phosphoinositides.
Collapse
|