UB Vice President for Research and Economic Development markup
M Laura Feltri UB Profile
Biochemistry
Professor of Biochemistry and Neurology
Specialty/Research Focus: Neurology; Cytoskeleton and cell motility; Molecular and Cellular Biology; Molecular Basis of Disease; Molecular genetics; Neurobiology; Signal Transduction; Inherited Metabolic Disorders; Transgenic organisms
701 Ellicott Street
Hunter James Kelly Research Institute
NYS Center of Excellence in Bioinformatics & Life Sciences, B4-322
Buffalo NY 14203
716-881-8969
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Jordan J S VerPlank, Sudarsanareddy Lokireddy, M Laura Feltri, Alfred L Goldberg, Lawrence Wrabetz
Keywords: Schwann cell deubiquitinase myelin protein quality control
Abstract: In several neurodegenerative diseases in which misfolded proteins accumulate there is impairment of the ubiquitin proteasome system (UPS). We tested if a similar disruption of proteostasis occurs in hereditary peripheral neuropathies. In sciatic nerves from mouse models of two human neuropathies, Myelin Protein Zero mutation (S63del) and increased copy number (P0 overexpression), polyubiquitinated proteins accumulated, and the overall rates of protein degradation were decreased. 26S proteasomes affinity-purified from sciatic nerves of S63del mice were defective in degradation of peptides and a ubiquitinated protein, unlike proteasomes from P0 overexpression, which appeared normal. Nevertheless, cellular levels of 26S proteasomes were increased in both, through the proteolytic-activation of the transcription factor Nrf1, as occurs in response to proteasome inhibitors. In S63del, increased amounts of the deubiquitinating enzymes USP14, UCH37, and USP5 were associated with proteasomes, the first time this has been reported in a human disease model. Inhibitors of USP14 increased the rate of protein degradation in S63del sciatic nerves and unexpectedly increased the phosphorylation of eIF2 by Perk. Thus, proteasome content, composition and activity are altered in these diseases and USP14 inhibitors have therapeutic potential in S63del neuropathy.
PubMedId: 29076578
Journal-article: Glia, vol. 66, 2017
Authors: Anna Bianchi-Smiraglia, Mitra S Rana, Colleen E Foley, Leslie M Paul, Brittany C Lipchick, Sudha Moparthy, Kalyana Moparthy, Emily E Fink, Archis Bagati, Edward Hurley, Hayley C Affronti, Andrei V Bakin, Eugene S Kandel, Dominic J Smiraglia, Maria Laura Feltri, Rui Sousa, Mikhail A Nikiforov
Abstract: GTP is a major regulator of multiple cellular processes, but tools for quantitative evaluation of GTP levels in live cells have not been available. We report the development and characterization of genetically encoded GTP sensors, which we constructed by inserting a circularly permuted yellow fluorescent protein (cpYFP) into a region of the bacterial G protein FeoB that undergoes a GTP-driven conformational change. GTP binding to these sensors results in a ratiometric change in their fluorescence, thereby providing an internally normalized response to changes in GTP levels while minimally perturbing those levels. Mutations introduced into FeoB to alter its affinity for GTP created a series of sensors with a wide dynamic range. Critically, in mammalian cells the sensors showed consistent changes in ratiometric signal upon depletion or restoration of GTP pools. We show that these GTP evaluators (GEVALs) are suitable for detection of spatiotemporal changes in GTP levels in living cells and for high-throughput screening of molecules that modulate GTP levels.
PubMedId: 28869758
Journal-article: Nature methods, vol. 14, 2017
Authors: Gustavo Della-Flora Nunes, Lauren Mueller, Nicholas Silvestri, Mulchand S Patel, Lawrence Wrabetz, M Laura Feltri, Yannick Poitelon
Keywords: PDC Schwann cell myelin oligodendrocyte pyruvate dehydrogenase complex deficiency
Abstract: Oligodendrocytes and Schwann cells not only form myelin in the central and peripheral nervous system, but also provide metabolic and trophic support to the axons they ensheathe. Acetyl-CoA is potentially a key molecule in Schwann cells and oligodendrocytes because it is at the crossroads of cellular lipid biosynthesis and energy generation. The main route for acetyl-CoA production is the oxidation of pyruvate by the pyruvate dehydrogenase complex (PDC). PDC deficiency in humans results in neurodegeneration and developmental impairments in both white and gray matter structures. To address the importance of PDC in myelinating glia, we deleted Pdha1 gene specifically in oligodendrocytes and Schwann cells. Surprisingly, sciatic and optic nerve morphology and the motor performance of Pdha1f/Y; CnpCre/+ mice are undistinguishable from those of controls at 1 month of age. In addition, myelin is stably maintained for at least 10 months. However, Pdha1f/Y; CnpCre/+ mice showed reduced fiber density and signs of axonal degeneration in both sciatic and optic nerves from 6 months of age. In contrast, 10 month-old mice bearing a floxed Pdha1 gene with either P0-Cre (expressed only by Schwann cells) or NG2-CreER (expressed in oligodendrocyte precursor cells) do not show any sign of axonal pathology or alterations in myelin structure or thickness. This indicates that the axonopathy is specific to the Pdha1f/Y; CnpCre/+ mice. Taken together, these results suggest that acetyl-CoA derived from pyruvate is not necessary for myelin maintenance and, thus, myelin-forming cells are not likely to contribute to the pathophysiology of PDC deficiency.
PubMedId: 28657129
Journal-article: Glia, vol. 65, 2017
Authors: Monica Ghidinelli, Yannick Poitelon, Yoon Kyoung Shin, Dominique Ameroso, Courtney Williamson, Cinzia Ferri, Marta Pellegatta, Kevin Espino, Amit Mogha, Kelly Monk, Paola Podini, Carla Taveggia, Klaus-Armin Nave, Lawrence Wrabetz, Hwan Tae Park, Maria Laura Feltri
Abstract: Myelin is required for proper nervous system function. Schwann cells in developing nerves depend on extrinsic signals from the axon and from the extracellular matrix to first sort and ensheathe a single axon and then myelinate it. Neuregulin 1 type III (Nrg1III) and laminin 211 (Lm211) are the key axonal and matrix signals, respectively, but how their signaling is integrated and if each molecule controls both axonal sorting and myelination is unclear. Here, we use a series of epistasis experiments to show that Lm211 modulates neuregulin signaling to ensure the correct timing and amount of myelination. Lm211 can inhibit Nrg1III by limiting protein kinase A (PKA) activation, which is required to initiate myelination. We provide evidence that excessive PKA activation amplifies promyelinating signals downstream of neuregulin, including direct activation of the neuregulin receptor ErbB2 and its effector Grb2-Associated Binder-1 (Gab1), thereby elevating the expression of the key transcription factors Oct6 and early growth response protein 2 (Egr2). The inhibitory effect of Lm211 is seen only in fibers of small caliber. These data may explain why hereditary neuropathies associated with decreased laminin function are characterized by focally thick and redundant myelin.
PubMedId: 28636612
Journal-article: PLoS biology, vol. 15, 2016
Authors: Yannick Poitelon, Gustavo Della-Flora Nunes, M Laura Feltri
Keywords: Neuroscience Schwann cell TAZ YAP
PubMedId: 28031544
Journal-article: Oncotarget, vol. 8, 2016
Authors: Nadav I Weinstock, Lawrence Wrabetz, M Laura Feltri, Daesung Shin
Keywords: Krabbe disease biomarker globoid cell leukodystrophy glucose metabolism metabolomics mitochondrial dysfunction oxidative stress twitcher
Abstract: Krabbe disease (KD) is caused by mutations in the galactosylceramidase (GALC) gene, which encodes a lysosomal enzyme that degrades galactolipids, including galactosylceramide and galactosylsphingosine (psychosine). GALC deficiency results in progressive intracellular accumulation of psychosine, which is believed to be the main cause for the demyelinating neurodegeneration in KD pathology. Umbilical cord blood transplantation slows disease progression when performed presymptomatically but carries a significant risk of morbidity and mortality. Accurate presymptomatic diagnosis is therefore critical to facilitate the efficacy of existing transplant approaches and to avoid unnecessary treatment of children who will not develop KD. Unfortunately, current diagnostic criteria, including GALC activity, genetic analysis, and psychosine measurement, are insufficient for secure presymptomatic diagnosis. This study performs a global metabolomic analysis to identify pathogenetic metabolic pathways and novel biomarkers implicated in the authentic mouse model of KD known as twitcher. At a time point before onset of signs of disease, twitcher hindbrains had metabolic profiles similar to WT, with the exception of a decrease in metabolites related to glucose energy metabolism. Many metabolic pathways were altered after early signs of disease in the twitcher, including decreased phospholipid turnover, restricted mitochondrial metabolism of branched-chain amino acids, increased inflammation, and changes in neurotransmitter metabolism and osmolytes. Hypoxanthine, a purine derivative, is increased before signs of disease appear, suggesting its potential as a biomarker for early diagnosis of KD. Additionally, given the early changes in glucose metabolism in the pathogenesis of KD, diagnostic modalities that report metabolic function, such as positron emission tomography, may be useful in KD. 2016 Wiley Periodicals, Inc.
PubMedId: 27638595
Journal-article: Journal of neuroscience research, vol. 94, 2016
Authors: Camila Lopez-Anido, Yannick Poitelon, Chetna Gopinath, John J Moran, Ki Hwan Ma, William D Law, Anthony Antonellis, M Laura Feltri, John Svaren
Abstract: Schwann cells are myelinating glia in the peripheral nervous system that form the myelin sheath. A major cause of peripheral neuropathy is a copy number variant involving the Peripheral Myelin Protein 22 (PMP22) gene, which is located within a 1.4-Mb duplication on chromosome 17 associated with the most common form of Charcot-Marie-Tooth Disease (CMT1A). Rodent models of CMT1A have been used to show that reducing Pmp22 overexpression mitigates several aspects of a CMT1A-related phenotype. Mechanistic studies of Pmp22 regulation identified enhancers regulated by the Sox10 (SRY sex determining region Y-box 10) and Egr2/Krox20 (Early growth response protein 2) transcription factors in myelinated nerves. However, relatively little is known regarding how other transcription factors induce Pmp22 expression during Schwann cell development and myelination. Here, we examined Pmp22 enhancers as a function of cell type-specificity, nerve injury and development. While Pmp22 enhancers marked by active histone modifications were lost or remodeled after injury, we found that these enhancers were permissive in early development prior to Pmp22 upregulation. Pmp22 enhancers contain binding motifs for TEA domain (Tead) transcription factors of the Hippo signaling pathway. We discovered that Tead1 and co-activators Yap and Taz are required for Pmp22 expression, as well as for the expression of Egr2 Tead1 directly binds Pmp22 and Egr2 enhancers early in development and Tead1 binding is induced during myelination, correlating with Pmp22 expression. The data identify Tead1 as a novel regulator of Pmp22 expression during development in concert with Sox10 and Egr2.
PubMedId: 27288457
Journal-article: Human molecular genetics, vol. 25, 2016
Authors: Daesung Shin, M Laura Feltri, Lawrence Wrabetz
Keywords: Krabbe disease galactosylceramidase globoid cell leukodystrophy lysosomal processing lysosomal storage disorders protein trafficking
Abstract: Globoid cell leukodystrophy (GLD, Krabbe disease) is due to autosomal recessive mutations in the lysosomal enzyme galactosylceramidase (GALC). Many GLD patients develop infantile-onset of progressive neurologic deterioration and death by 2 years of age, whereas others have a later-onset, milder disease. Cord blood transplant slows disease progression much more effectively when performed presymptomatically, highlighting the importance of early diagnosis. Current diagnosis is based on reduced GALC activity, DNA sequence, and clinical examination. However, presymptomatic diagnosis is hampered by imperfect genotype-GALC activity-phenotype correlations. In addition, three polymorphisms in the GALC gene are variably associated with disease mutations and have unknown effects on GALC activity and disease outcome. Here, we study mutations that cause infantile or later-onset GLD, and show that GALC activity is significantly lower in infantile versus later-onset mutants when measured in the lysosomal fraction, but not in whole-cell lysates. In parallel, infantile-onset mutant GALCs showed reduced trafficking to lysosomes and processing than later-onset mutant GALCs. Finally, the cis-polymorphisms also affected trafficking to the lysosome and processing of GALC. These differences potentially explain why the activity of different mutations appears similar in whole-cell extracts from lymphocytes, and suggest that measure of GALC activity in lysosomes may better predict the onset and severity of disease for a given GLD genotype. Globoid cell leukodystrophy (GLD, Krabbe disease) is diagnosed by measuring galactosylceramidase (GALC) activity and DNA analysis. However, genotype and phenotype often do not correlate due to considerable clinical variability, even for the same mutation, for unknown reasons. We find that altered trafficking to the lysosome and processing of GALC correlates with GLD severity and is modulated by cis-polymorphisms. Current diagnosis of GLD is based on GALC activity of total cell lysates from blood, which does not discriminate whether the activity comes from the lysosome or other subcellular organelles. Measurement of GALC activity in lysosomes may predict which infants are at high risk for the infantile phenotype while distinguishing other children who will develop later-onset phenotypes without onset of symptoms for years.
PubMedId: 26865610
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 36, 2016
Authors: Yannick Poitelon, Camila Lopez-Anido, Kathleen Catignas, Caterina Berti, Marilena Palmisano, Courtney Williamson, Dominique Ameroso, Kansho Abiko, Yoonchan Hwang, Alex Gregorieff, Jeffrey L Wrana, Mohammadnabi Asmani, Ruogang Zhao, Fraser James Sim, Lawrence Wrabetz, John Svaren, Maria Laura Feltri
Abstract: Myelination is essential for nervous system function. Schwann cells interact with neurons and the basal lamina to myelinate axons using known receptors, signals and transcription factors. In contrast, the transcriptional control of axonal sorting and the role of mechanotransduction in myelination are largely unknown. Yap and Taz are effectors of the Hippo pathway that integrate chemical and mechanical signals in cells. We describe a previously unknown role for the Hippo pathway in myelination. Using conditional mutagenesis in mice, we show that Taz is required in Schwann cells for radial sorting and myelination and that Yap is redundant with Taz. Yap and Taz are activated in Schwann cells by mechanical stimuli and regulate Schwann cell proliferation and transcription of basal lamina receptor genes, both necessary for radial sorting of axons and subsequent myelination. These data link transcriptional effectors of the Hippo pathway and of mechanotransduction to myelin formation in Schwann cells.
PubMedId: 27273766
Journal-article: Nature neuroscience, vol. 19, 2015
Authors: M Laura Feltri, Yannick Poitelon, Stefano Carlo Previtali
Keywords: Schwann cells development human peripheral neuropathies myelin peripheral nervous system
Abstract: Peripheral nerves contain large myelinated and small unmyelinated (Remak) fibers that perform different functions. The choice to myelinate or not is dictated to Schwann cells by the axon itself, based on the amount of neuregulin I-type III exposed on its membrane. Peripheral axons are more important in determining the final myelination fate than central axons, and the implications for this difference in Schwann cells and oligodendrocytes are discussed. Interestingly, this choice is reversible during pathology, accounting for the remarkable plasticity of Schwann cells, and contributing to the regenerative potential of the peripheral nervous system. Radial sorting is the process by which Schwann cells choose larger axons to myelinate during development. This crucial morphogenetic step is a prerequisite for myelination and for differentiation of Remak fibers, and is arrested in human diseases due to mutations in genes coding for extracellular matrix and linkage molecules. In this review we will summarize progresses made in the last years by a flurry of reverse genetic experiments in mice and fish. This work revealed novel molecules that control radial sorting, and contributed unexpected ideas to our understanding of the cellular and molecular mechanisms that control radial sorting of axons.
PubMedId: 25686621
Journal-article: The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry, vol. 22, 2015
Authors: Roberta Noseda, Marta Guerrero-Valero, Valeria Alberizzi, Stefano C Previtali, Diane L Sherman, Marilena Palmisano, Richard L Huganir, Klaus-Armin Nave, Ana Cuenda, Maria Laura Feltri, Peter J Brophy, Alessandra Bolino
Abstract: Microtubule-based kinesin motors have many cellular functions, including the transport of a variety of cargos. However, unconventional roles have recently emerged, and kinesins have also been reported to act as scaffolding proteins and signaling molecules. In this work, we further extend the notion of unconventional functions for kinesin motor proteins, and we propose that Kif13b kinesin acts as a signaling molecule regulating peripheral nervous system (PNS) and central nervous system (CNS) myelination. In this process, positive and negative signals must be tightly coordinated in time and space to orchestrate myelin biogenesis. Here, we report that in Schwann cells Kif13b positively regulates myelination by promoting p38 mitogen-activated protein kinase (MAPK)-mediated phosphorylation and ubiquitination of Discs large 1 (Dlg1), a known brake on myelination, which downregulates the phosphatidylinositol 3-kinase (PI3K)/v-AKT murine thymoma viral oncogene homolog (AKT) pathway. Interestingly, Kif13b also negatively regulates Dlg1 stability in oligodendrocytes, in which Dlg1, in contrast to Schwann cells, enhances AKT activation and promotes myelination. Thus, our data indicate that Kif13b is a negative regulator of CNS myelination. In summary, we propose a novel function for the Kif13b kinesin in glial cells as a key component of the PI3K/AKT signaling pathway, which controls myelination in both PNS and CNS.
PubMedId: 27070899
Journal-article: PLoS biology, vol. 14, 2015
Authors: Y Poitelon, S Bogni, V Matafora, G Della-Flora Nunes, E Hurley, M Ghidinelli, B S Katzenellenbogen, C Taveggia, N Silvestri, A Bachi, A Sannino, L Wrabetz, M L Feltri
Abstract: Cell-cell interactions promote juxtacrine signals in specific subcellular domains, which are difficult to capture in the complexity of the nervous system. For example, contact between axons and Schwann cells triggers signals required for radial sorting and myelination. Failure in this interaction causes dysmyelination and axonal degeneration. Despite its importance, few molecules at the axo-glial surface are known. To identify novel molecules in axo-glial interactions, we modified the 'pseudopodia' sub-fractionation system and isolated the projections that glia extend when they receive juxtacrine signals from axons. By proteomics we identified the signalling networks present at the glial-leading edge, and novel proteins, including members of the Prohibitin family. Glial-specific deletion of Prohibitin-2 in mice impairs axo-glial interactions and myelination. We thus validate a novel method to model morphogenesis and juxtacrine signalling, provide insights into the molecular organization of the axo-glial contact, and identify a novel class of molecules in myelination.
PubMedId: 26383514
Journal-article: Nature communications, vol. 6, 2015
Authors: Kelly R Monk, M Laura Feltri, Carla Taveggia
Keywords: Remak Schwann cell Schwann cell Schwann cell precursor immature Schwann cell myelinating Schwann cell neural crest peripheral nervous system radial sorting
Abstract: In the peripheral nervous system, Schwann cells are glial cells that are in intimate contact with axons throughout development. Schwann cells generate the insulating myelin sheath and provide vital trophic support to the neurons that they ensheathe. Schwann cell precursors arise from neural crest progenitor cells, and a highly ordered developmental sequence controls the progression of these cells to become mature myelinating or nonmyelinating Schwann cells. Here, we discuss both seminal discoveries and recent advances in our understanding of the molecular mechanisms that drive Schwann cell development and myelination with a focus on cell-cell and cell-matrix signaling events.
PubMedId: 25921593
Journal-article: Glia, vol. 63, 2015
Authors: Bradley A Heller, Monica Ghidinelli, Jakob Voelkl, Steven Einheber, Ryan Smith, Ethan Grund, Grant Morahan, David Chandler, Luba Kalaydjieva, Filippo Giancotti, Rosalind H King, Aniko Naray Fejes-Toth, Gerard Fejes-Toth, Maria Laura Feltri, Florian Lang, James L Salzer
Abstract: The PI 3-kinase (PI 3-K) signaling pathway is essential for Schwann cell myelination. Here we have characterized PI 3-K effectors activated during myelination by probing myelinating cultures and developing nerves with an antibody that recognizes phosphorylated substrates for this pathway. We identified a discrete number of phospho-proteins including the S6 ribosomal protein (S6rp), which is down-regulated at the onset of myelination, and N-myc downstream-regulated gene-1 (NDRG1), which is up-regulated strikingly with myelination. We show that type III Neuregulin1 on the axon is the primary activator of S6rp, an effector of mTORC1. In contrast, laminin-2 in the extracellular matrix (ECM), signaling through the 64 integrin and Sgk1 (serum and glucocorticoid-induced kinase 1), drives phosphorylation of NDRG1 in the Cajal bands of the abaxonal compartment. Unexpectedly, mice deficient in 64 integrin signaling or Sgk1 exhibit hypermyelination during development. These results identify functionally and spatially distinct PI 3-K pathways: an early, pro-myelinating pathway driven by axonal Neuregulin1 and a later-acting, laminin-integrin-dependent pathway that negatively regulates myelination.
PubMedId: 24687281
Journal-article: The Journal of cell biology, vol. 204, 2014
Authors: Xinghua Yin, Sumiko Kiryu-Seo, Grahame J Kidd, M Laura Feltri, Lawrence Wrabetz, Bruce D Trapp
Keywords: Schwann cell axon hypomyelination myelin compaction myelin evolution
Abstract: The central nervous system (CNS) of terrestrial vertebrates underwent a prominent molecular change when proteolipid protein (PLP) replaced P0 protein as the most abundant protein of CNS myelin. However, PLP did not replace P0 in peripheral nervous system (PNS) myelin. To investigate the possible consequences of a PLP to P0 shift in PNS myelin, we engineered mice to express PLP instead of P0 in PNS myelin (PLP-PNS mice). PLP-PNS mice had severe neurological disabilities and died between 3 and 6 months of age. Schwann cells in sciatic nerves from PLP-PNS mice sorted axons into one-to-one relationships but failed to form myelin internodes. Mice with equal amounts of P0 and PLP had normal PNS myelination and lifespans similar to wild-type (WT) mice. When PLP was overexpressed with one copy of the P0 gene, sciatic nerves were hypomyelinated; mice displayed motor deficits, but had normal lifespans. These data support the hypothesis that while PLP can co-exist with P0 in PNS myelin, PLP cannot replace P0 as the major structural protein of PNS myelin.
PubMedId: 25066805
Journal-article: Glia, vol. 63, 2014
Authors: Peiwen Chen, Matilde Cescon, Gaia Zuccolotto, Lucilla Nobbio, Cristina Colombelli, Monica Filaferro, Giovanni Vitale, M Laura Feltri, Paolo Bonaldo
Abstract: Macrophages contribute to peripheral nerve regeneration and produce collagen VI, an extracellular matrix protein involved in nerve function. Here, we show that collagen VI is critical for macrophage migration and polarization during peripheral nerve regeneration. Nerve injury induces a robust upregulation of collagen VI, whereas lack of collagen VI in Col6a1(-/-) mice delays peripheral nerve regeneration. In vitro studies demonstrated that collagen VI promotes macrophage migration and polarization via AKT and PKA pathways. Col6a1(-/-) macrophages exhibit impaired migration abilities and reduced antiinflammatory (M2) phenotype polarization, but are prone to skewing toward the proinflammatory (M1) phenotype. In vivo, macrophage recruitment and M2 polarization are impaired in Col6a1(-/-) mice after nerve injury. The delayed nerve regeneration of Col6a1(-/-) mice is induced by macrophage deficits and rejuvenated by transplantation of wild-type bone marrow cells. These results identify collagen VI as a novel regulator for peripheral nerve regeneration by modulating macrophage function.
PubMedId: 25421425
Journal-article: Acta neuropathologica, vol. 129, 2014
Authors: Emanuela Porrello, Cristina Rivellini, Giorgia Dina, Daniela Triolo, Ubaldo Del Carro, Daniela Ungaro, Martina Panattoni, Maria Laura Feltri, Lawrence Wrabetz, Ruggero Pardi, Angelo Quattrini, Stefano Carlo Previtali
Abstract: Axonal sorting is a crucial event in nerve formation and requires proper Schwann cell proliferation, differentiation, and contact with axons. Any defect in axonal sorting results in dysmyelinating peripheral neuropathies. Evidence from mouse models shows that axonal sorting is regulated by laminin211- and, possibly, neuregulin 1 (Nrg1)-derived signals. However, how these signals are integrated in Schwann cells is largely unknown. We now report that the nuclear Jun activation domain-binding protein 1 (Jab1) may transduce laminin211 signals to regulate Schwann cell number and differentiation during axonal sorting. Mice with inactivation of Jab1 in Schwann cells develop a dysmyelinating neuropathy with axonal sorting defects. Loss of Jab1 increases p27 levels in Schwann cells, which causes defective cell cycle progression and aberrant differentiation. Genetic down-regulation of p27 levels in Jab1-null mice restores Schwann cell number, differentiation, and axonal sorting and rescues the dysmyelinating neuropathy. Thus, Jab1 constitutes a regulatory molecule that integrates laminin211 signals in Schwann cells to govern cell cycle, cell number, and differentiation. Finally, Jab1 may constitute a key molecule in the pathogenesis of dysmyelinating neuropathies.
PubMedId: 24344238
Journal-article: The Journal of experimental medicine, vol. 211, 2013
Authors: Sumihisa Orita, Kenneth Henry, Elisabetta Mantuano, Kazuyo Yamauchi, Alice De Corato, Tetsuhiro Ishikawa, M Laura Feltri, Lawrence Wrabetz, Alban Gaultier, Melanie Pollack, Mark Ellisman, Kazuhisa Takahashi, Steven L Gonias, W Marie Campana
Abstract: Trophic support and myelination of axons by Schwann cells in the PNS are essential for normal nerve function. Herein, we show that deletion of the LDL receptor-related protein-1 (LRP1) gene in Schwann cells (scLRP1(-/-)) induces abnormalities in axon myelination and in ensheathment of axons by nonmyelinating Schwann cells in Remak bundles. These anatomical changes in the PNS were associated with mechanical allodynia, even in the absence of nerve injury. In response to crush injury, sciatic nerves in scLRP1(-/-) mice showed accelerated degeneration and Schwann cell death. Remyelinated axons were evident 20 d after crush injury in control mice, yet were largely absent in scLRP1(-/-) mice. In the partial nerve ligation model, scLRP1(-/-) mice demonstrated significantly increased and sustained mechanical allodynia and loss of motor function. Evidence for central sensitization in pain processing included increased p38MAPK activation and activation of microglia in the spinal cord. These studies identify LRP1 as an essential mediator of normal Schwann cell-axonal interactions and as a pivotal regulator of the Schwann cell response to PNS injury in vivo. Mice in which LRP1 is deficient in Schwann cells represent a model for studying how abnormalities in Schwann cell physiology may facilitate and sustain chronic pain.
PubMedId: 23536074
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 33, 2013
Authors: Robin D S Doddrell, Xin-Peng Dun, Aditya Shivane, M Laura Feltri, Lawrence Wrabetz, Michael Wegner, Elisabeth Sock, C Oliver Hanemann, David B Parkinson
Abstract: Loss of the Merlin tumour suppressor causes abnormal de-differentiation and proliferation of Schwann cells and formation of schwannoma tumours in patients with neurofibromatosis type 2. Within the mature peripheral nerve the normal development, differentiation and maintenance of myelinating and non-myelinating Schwann cells is regulated by a network of transcription factors that include SOX10, OCT6 (now known as POU3F1), NFATC4 and KROX20 (also known as Egr2). We have examined for the first time how their regulation of Schwann cell development is disrupted in primary human schwannoma cells. We find that induction of both KROX20 and OCT6 is impaired, whereas enforced expression of KROX20 drives both myelin gene expression and cell cycle arrest in Merlin-null cells. Importantly, we show that human schwannoma cells have reduced expression of SOX10 protein and messenger RNA. Analysis of mouse SOX10-null Schwann cells shows they display many of the characteristics of human schwannoma cells, including increased expression of platelet derived growth factor receptor beta (PDGFRB) messenger RNA and protein, enhanced proliferation, increased focal adhesions and schwannoma-like morphology. Correspondingly, reintroduction of SOX10 into human Merlin-null cells restores the ability of these cells to induce KROX20 and myelin protein zero (MPZ), localizes NFATC4 to the nucleus, reduces cell proliferation and suppresses PDGFRB expression. Thus, we propose that loss of the SOX10 protein, which is vital for normal Schwann cell development, is also key to the pathology of Merlin-null schwannoma tumours.
PubMedId: 23413263
Journal-article: Brain : a journal of neurology, vol. 136, 2013
Authors: Mario A C Saporta, Brian R Shy, Agnes Patzko, Yunhong Bai, Maria Pennuto, Cinzia Ferri, Elisa Tinelli, Paola Saveri, Dan Kirschner, Michelle Crowther, Cherie Southwood, Xingyao Wu, Alexander Gow, M Laura Feltri, Lawrence Wrabetz, Michael E Shy
Abstract: Mutations in myelin protein zero (MPZ) cause Charcot-Marie-Tooth disease type 1B. Many dominant MPZ mutations, including R98C, present as infantile onset dysmyelinating neuropathies. We have generated an R98C 'knock-in' mouse model of Charcot-Marie-Tooth type 1B, where a mutation encoding R98C was targeted to the mouse Mpz gene. Both heterozygous (R98C/+) and homozygous (R98C/R98C) mice develop weakness, abnormal nerve conduction velocities and morphologically abnormal myelin; R98C/R98C mice are more severely affected. MpzR98C is retained in the endoplasmic reticulum of Schwann cells and provokes a transitory, canonical unfolded protein response. Ablation of Chop, a mediator of the protein kinase RNA-like endoplasmic reticulum kinase unfolded protein response pathway restores compound muscle action potential amplitudes of R98C/+ mice but does not alter the reduced conduction velocities, reduced axonal diameters or clinical behaviour of these animals. R98C/R98C Schwann cells are developmentally arrested in the promyelinating stage, whereas development is delayed in R98C/+ mice. The proportion of cells expressing c-Jun, an inhibitor of myelination, is elevated in mutant nerves, whereas the proportion of cells expressing the promyelinating transcription factor Krox-20 is decreased, particularly in R98C/R98C mice. Our results provide a potential link between the accumulation of MpzR98C in the endoplasmic reticulum and a developmental delay in myelination. These mice provide a model by which we can begin to understand the early onset dysmyelination seen in patients with R98C and similar mutations.
PubMedId: 22689911
Journal-article: Brain : a journal of neurology, vol. 135, 2012
Authors: Rosa La Marca, Federica Cerri, Keisuke Horiuchi, Angela Bachi, M Laura Feltri, Lawrence Wrabetz, Carl P Blobel, Angelo Quattrini, James L Salzer, Carla Taveggia
Abstract: Tumor necrosis factor--converting enzyme (TACE; also known as ADAM17) is a proteolytic sheddase that is responsible for the cleavage of several membrane-bound molecules. We report that TACE cleaves neuregulin-1 (NRG1) type III in the epidermal growth factor domain, probably inactivating it (as assessed by deficient activation of the phosphatidylinositol-3-OH kinase pathway), and thereby negatively regulating peripheral nervous system (PNS) myelination. Lentivirus-mediated knockdown of TACE in vitro in dorsal root ganglia neurons accelerates the onset of myelination and results in hypermyelination. In agreement, motor neurons of conditional knockout mice lacking TACE specifically in these cells are significantly hypermyelinated, and small-caliber fibers are aberrantly myelinated. Further, reduced TACE activity rescues hypomyelination in NRG1 type III haploinsufficient mice in vivo. We also show that the inhibitory effect of TACE is neuron-autonomous, as Schwann cells lacking TACE elaborate myelin of normal thickness. Thus, TACE is a modulator of NRG1 type III activity and is a negative regulator of myelination in the PNS.
PubMedId: 21666671
Journal-article: Nature neuroscience, vol. 14, 2011
Authors: Hwan Tae Park, M Laura Feltri
Abstract: After peripheral nerve injuries, Wallerian degeneration starts with a stereotypic fragmentation of myelin sheath into myelin ovoids, which occur near Schmidt-Lantermann incisures (SLI). This demyelination process requires a dramatic change in cytoskeletal structures in Schwann cells. We have recently shown that actin polymerization around SLI is an important step for cleavage of the myelin sheath. We described that Rac1 GTP ase regulates actin polymerization in SLI after injury. It has been previously reported that Rac-dependent cytoskeletal reorganization also plays an important role in myelination during the development of peripheral nerves. Thus, our findings suggest that Rac-dependent actin polymerization controls both myelination and demyelination in the peripheral nerves. We further discuss our new findings in relation to Schwann cell dedifferentiation and segmental demyelination.
PubMedId: 21922040
Journal-article: Bioarchitecture, vol. 1, 2011
Authors: Fuzi Jin, Baoxia Dong, John Georgiou, Qiuhong Jiang, Jinyi Zhang, Arjun Bharioke, Frank Qiu, Silvia Lommel, M Laura Feltri, Lawrence Wrabetz, John C Roder, Joel Eyer, Xiequn Chen, Alan C Peterson, Katherine A Siminovitch
Abstract: Schwann cells elaborate myelin sheaths around axons by spirally wrapping and compacting their plasma membranes. Although actin remodeling plays a crucial role in this process, the effectors that modulate the Schwann cell cytoskeleton are poorly defined. Here, we show that the actin cytoskeletal regulator, neural Wiskott-Aldrich syndrome protein (N-WASp), is upregulated in myelinating Schwann cells coincident with myelin elaboration. When N-WASp is conditionally deleted in Schwann cells at the onset of myelination, the cells continue to ensheath axons but fail to extend processes circumferentially to elaborate myelin. Myelin-related gene expression is also severely reduced in the N-WASp-deficient cells and in vitro process and lamellipodia formation are disrupted. Although affected mice demonstrate obvious motor deficits these do not appear to progress, the mutant animals achieving normal body weights and living to advanced age. Our observations demonstrate that N-WASp plays an essential role in Schwann cell maturation and myelin formation.
PubMedId: 21385763
Journal-article: Development (Cambridge, England), vol. 138, 2011
Authors: Junyang Jung, Wenting Cai, Hyun Kyoung Lee, Marta Pellegatta, Yoon Kyung Shin, So Young Jang, Duk Joon Suh, Lawrence Wrabetz, M Laura Feltri, Hwan Tae Park
Abstract: The mechanisms that trigger Wallerian degeneration (WD) of peripheral nerves after injury are not well understood. During the early period of WD, fragmentation of myelin into ovoid structures occurs near the Schmidt-Lantermann incisures (SLI), a noncompact region of the myelin sheath containing autotypical adherens junction. In this study, we found that new filamentous actin polymerization occurs in the SLI of mouse sciatic nerves after injury and that its inhibition prevented not only the degradation of E-cadherin in the SLI but also myelin ovoid formation. However, the inhibition of actin polymerization could not block Schwann cell dedifferentiation. The activation of Rac GTPase was observed in the distal stump of the injured nerves, and a specific Rac inhibitor, a dominant-negative Rac, and Rac1-RNA interference blocked myelin ovoid formation. Together, these findings suggest that dynamic changes in actin in the SLI are essential for initiation of demyelination after peripheral nerve injury.
PubMedId: 21307239
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 31, 2011
Authors: Pietro Fratta, Paola Saveri, Desiree Zambroni, Cinzia Ferri, Elisa Tinelli, Albee Messing, Maurizio D'Antonio, Maria Laura Feltri, Lawrence Wrabetz
Abstract: More than 120 mutations in the Myelin Protein Zero gene (MPZ, P0) cause various forms of hereditary neuropathy. Two human mutations encoding either P0S63C or P0S63del have been shown to cause demyelination in mice through different gain of function pathomechanisms. P0S63del, for example, is retained in the endoplasmic reticulum (ER) and elicits a pathogenetic unfolded protein response (UPR). As P0 likely forms oligomers, another gain of abnormal function could include a dominant-negative interaction between P0S63del and normal P0 (P0wt). To test this idea, we generated a transgenic mouse that expressed a form of P0wt with a myc epitope tag at the C terminus (P0ct-myc). We show that P0ct-myc is trafficked and functions like P0wt, thus providing a new tool to study P0 in vivo. In mice that express both P0ct-myc and P0S63del, P0S63del specifically delays the transit of P0ct-myc through the ER and reduces the level of P0wt in the myelin sheath by half-a level previously shown to cause demyelination in mice and humans. Surprisingly, P0ct-myc does not co-immunoprecipitate with P0S63del, suggesting an indirect interaction. Thus, P0S63del causes not only a UPR-related toxic mechanism, but also a dominant-negative effect on P0wt that probably contributes to demyelinating neuropathy.
PubMedId: 21363884
Journal-article: Human molecular genetics, vol. 20, 2011
Authors: Carla Taveggia, Maria Laura Feltri, Lawrence Wrabetz
Abstract: The myelin sheath wraps large axons in both the CNS and the PNS, and is a key determinant of efficient axonal function and health. Myelin is targeted in a series of diseases, notably multiple sclerosis (MS). In MS, demyelination is associated with progressive axonal damage, which determines the level of patient disability. The few treatments that are available for combating myelin damage in MS and related disorders, which largely comprise anti-inflammatory drugs, only show limited efficacy in subsets of patients. More-effective treatment of myelin disorders will probably be accomplished by early intervention with combinatorial therapies that target inflammation and other processes-for example, signaling pathways that promote remyelination. Indeed, evidence suggests that such pathways might be impaired in pathology and, hence, contribute to the failure of remyelination in such diseases. In this article, we review the molecular basis of signaling pathways that regulate myelination in the CNS and PNS, with a focus on signals that affect differentiation of myelinating glia. We also discuss factors such as extracellular molecules that act as modulators of these pathways. Finally, we consider the few preclinical and clinical trials of agents that augment this signaling.
PubMedId: 20404842
Journal-article: Nature reviews. Neurology, vol. 6, 2010
Authors: Robin L Avila, Maurizio D'Antonio, Angela Bachi, Hideyo Inouye, M Laura Feltri, Lawrence Wrabetz, Daniel A Kirschner
Abstract: P0 constitutes 50-60% of protein in peripheral nerve myelin and is essential for its structure and stability. Mutations within the P0 gene (MPZ) underlie a variety of hereditary neuropathies. MpzS63C transgenic mice encode a P0 with a serine to cysteine substitution at position 34 in the extracellular domain of mature P0 (P0S34C), associated with the hypomyelinating Djrine-Sottas syndrome in human. S63C mice develop a dysmyelinating neuropathy, with packing defects in peripheral myelin. Here, we used x-ray diffraction to examine time-dependent packing defects in unfixed myelin. At 7 h post-dissection, WT and S63C(+/+) myelin showed native periods (175 ) with the latter developing at most a few percent swollen myelin, whereas up to 50% of S63C(+/-) (mutant P0 on heterozygous P0 null background) or P0(+/-) myelin swelled to periods of 205 . In the same time frame, S63C(-/-) myelin was stable, remaining swollen at 210 . Surprisingly, treatment of whole S63C(-/-) nerves with a reducing agent completely reverted swollen arrays to native spacing and also normalized the swollen arrays that had formed in S63C(+/-) myelin, the genotype most closely related to the human disorder. Western blot revealed P0-positive bands at 27 and 50 kDa, and MALDI-TOF mass spectrometry showed these bands consisted of Ser(34)-containing peptides or P0 dimers having oxidized Cys(34) residues. We propose that P0S34C forms ectopic disulfide bonds in trans between apposed Cys(34) side chains that retard wrapping during myelin formation causing hypomyelination. Moreover, the new bonds create a packing defect by stabilizing swollen membrane arrays that leads to demyelination.
PubMedId: 20937820
Journal-article: The Journal of biological chemistry, vol. 285, 2010
Authors: Patrizia Dacci, Giorgia Dina, Federica Cerri, Stefano Carlo Previtali, Ignazio Diego Lopez, Giuseppe Lauria, Maria Laura Feltri, Alessandra Bolino, Giancarlo Comi, Lawrence Wrabetz, Angelo Quattrini
Abstract: Numerous transgenic and knockout mouse models of human hereditary neuropathies have become available over the past decade. We describe a simple, reproducible, and safe biopsy of mouse skin for histopathological evaluation of the peripheral nervous system (PNS) in models of hereditary neuropathies. We compared the diagnostic outcome between sciatic nerve and dermal nerves found in skin biopsy (SB) from the hind foot. A total of five animal models of different Charcot-Marie-Tooth neuropathies, and one model of congenital muscular dystrophy associated neuropathy were examined. In wild type mice, dermal nerve fibers were readily identified by immunohistochemistry, light, and electron microscopy and they appeared similar to myelinated fibers in sciatic nerve. In mutant mice, SB manifested myelin abnormalities similar to those observed in sciatic nerves, including hypomyelination, onion bulbs, myelin outfolding, redundant loops, and tomacula. In many strains, however, SB showed additional abnormalities--fiber loss, dense neurofilament packing with lower phosphorylation status, and axonal degeneration-undetected in sciatic nerve, possibly because SB samples distal nerves. SB, a reliable technique to investigate peripheral neuropathies in human beings, is also useful to investigate animal models of hereditary neuropathies. Our data indicate that SB may reveal distal axonal pathology in mouse models and permits sequential follow-up of the neuropathy in an individual mouse, thereby reducing the number of mice necessary to document pathology of the PNS.
PubMedId: 20878767
Journal-article: Glia, vol. 58, 2010
Authors: Beth Yun, Angela Anderegg, Daniela Menichella, Lawrence Wrabetz, M Laura Feltri, Rajeshwar Awatramani
Abstract: MicroRNAs, by modulating gene expression, have been implicated as regulators of various cellular and physiological processes, including differentiation, proliferation, and cancer. Here, we study the role of microRNAs in Schwann cell (SC) differentiation by conditional removal of the microRNA processing enzyme Dicer1. We reveal that both male and female mice lacking Dicer1 in SC (Dicer1 conditional knock-outs) display a severe neurological phenotype resembling congenital hypomyelination. Ultrastructural analyses show that many SC lacking Dicer1 are stalled in differentiation at the promyelinating state and fail to myelinate axons. Gene expression analyses reveal a failure to extinguish genes characteristic of the undifferentiated state such as Sox2, Jun, and Ccnd1. Sox2 and Jun are well characterized negative regulators of SC differentiation. Consistent with Sox2/Jun maintenance, Egr2, a master regulator of the myelinating program, is drastically downregulated and likely accounts for the myelination defect. We posit a model wherein microRNAs are critical for downregulation of antecedent programs of gene expression. In SC differentiation, this is particularly relevant in the key developmental transition from a promyelinating to myelinating SC.
PubMedId: 20519547
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 30, 2010
Authors: Karen B Carlson, Prabhjot Singh, Moses M Feaster, Anita Ramnarain, Constantine Pavlides, Zu-Lin Chen, Wei-Ming Yu, M Laura Feltri, Sidney Strickland
Abstract: Peripheral nerve function depends on a regulated process of axon and Schwann cell development. Schwann cells interact with peripheral neurons to sort and ensheath individual axons. Ablation of laminin 1 in the peripheral nervous system (PNS) arrests Schwann cell development prior to radial sorting of axons. Peripheral nerves of laminin-deficient animals are disorganized and hypomyelinated. In this study, sciatic nerves of laminin-deficient mice were treated with syngenic murine adipose-derived stem cells (ADSCs). ADSCs expressed laminin in vitro and in vivo following transplant into mutant sciatic nerves. ADSC-treatment of mutant nerves caused endogenous Schwann cells to differentiate past the point of developmental arrest to sort and myelinate axons. This was shown by (1) functional, (2) ultrastructural, and (3) immunohistochemical analysis. Treatment of laminin-deficient nerves with either soluble laminin or the immortalized laminin-expressing cell line 3T3/L1 did not overcome endogenous Schwann cell developmental arrest. In summary, these results indicate that (1) laminin-deficient Schwann cells can be rescued, (2) a cell-based approach is beneficial in comparison with soluble protein treatment, and (3) mesenchymal stem cells modify sciatic nerve function via trophic effects rather than transdifferentiation in this system.
PubMedId: 21125647
Journal-article: Glia, vol. 59, 2010
Authors: Barry M Bradford, Nadia L Tuzi, M Laura Feltri, Caroline McCorquodale, Enrico Cancellotti, Jean C Manson
Abstract: Expression of the prion protein (PrP(C)) is a requirement for host susceptibility to the transmissible spongiform encephalopathies (TSEs) and thought to be necessary for the replication and transport of the infectious agent. The mechanism of TSE neuroinvasion is not fully understood, although the routing of infection has been mapped through the peripheral nervous system (PNS) and Schwann cells have been implicated as a potential conduit for transport of the TSE infectious agent. To address whether Schwann cells are a requirement for spread of the TSE agent from the site of infection to the CNS, PrP(C) expression was selectively removed from Schwann cells in vivo. This dramatically reduced total PrP(C) within peripheral nerves by 90%, resulting in the selective loss of glycosylated PrP(C) species. Despite this, 139A and ME7 mouse-passaged scrapie agent strains were efficiently replicated and transported to the CNS following oral and intraperitoneal exposure. Thus, the myelinating glial cells within the PNS do not appear to play a significant role in TSE neuroinvasion.
PubMedId: 20007469
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 29, 2009
Authors: Maurizio D'Antonio, M Laura Feltri, Lawrence Wrabetz
Abstract: The capacity to fold proteins properly is fundamental for cell survival. Secreted and transmembrane proteins are synthesized in the endoplasmic reticulum (ER), an organelle that has the ability to discriminate between native and nonnative proteins, in a process called protein quality control. When folding is not properly achieved, misfolded proteins can accumulate. The terminally misfolded proteins are typically retrotranslocated into the cytoplasm for degradation by the proteasome, in a process known as endoplasmic reticulum-associated degradation. However, if the degradation is insufficient, accumulation of abnormal proteins in the ER activates the unfolded protein response (UPR), a complex set of new signals aimed to reduce further the load of abnormal protein in the ER. Massive synthesis of myelin lipids and proteins is necessary to support myelinogenesis. Not surprisingly, therefore, ER stress (including the UPR), the proteasome, and autophagy (lysosomes) have been implicated in myelin disorders, such as Pelizaeus-Merzbacher disease and vanishing white matter disease in the central nervous system and Charcot-Marie-Tooth neuropathies in the peripheral nervous system. Here we discuss recent evidence supporting an important role for ER stress in myelin disorders.
PubMedId: 19330777
Journal-article: Journal of neuroscience research, vol. 87, 2009
Authors: Christian S Lobsiger, Severine Boillee, Melissa McAlonis-Downes, Amir M Khan, M Laura Feltri, Koji Yamanaka, Don W Cleveland
Abstract: Neurodegeneration in an inherited form of ALS is non-cell-autonomous, with ALS-causing mutant SOD1 damage developed within multiple cell types. Selective inactivation within motor neurons of an ubiquitously expressed mutant SOD1 gene has demonstrated that mutant damage within motor neurons is a determinant of disease initiation, whereas mutant synthesis within neighboring astrocytes or microglia accelerates disease progression. We now report the surprising finding that diminished synthesis (by 70%) within Schwann cells of a fully dismutase active ALS-linked mutant (SOD1(G37R)) significantly accelerates disease progression, accompanied by reduction of insulin-like growth factor 1 (IGF-1) in nerves. Coupled with shorter disease duration in mouse models caused by dismutase inactive versus dismutase active SOD1 mutants, our findings implicate an oxidative cascade during disease progression that is triggered within axon ensheathing Schwann cells and that can be ameliorated by elevated dismutase activity. Thus, therapeutic down-regulation of dismutase active mutant SOD1 in familial forms of ALS should be targeted away from Schwann cells.
PubMedId: 19251638
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 106, 2009
Authors: Felipe A Court, Jane E Hewitt, Kay Davies, Bruce L Patton, Antonino Uncini, Lawrence Wrabetz, M Laura Feltri
Abstract: Animal and plant cells compartmentalize to perform morphogenetic functions. Compartmentalization of myelin-forming Schwann cells may favor elongation of myelin segments to the size required for efficient conduction of nerve impulses. Compartments in myelinated fibers were described by Ramn y Cajal and depend on periaxin, mutated in the hereditary neuropathy Charcot-Marie-Tooth disease type 4F (Charcot-Marie-Tooth 4F). Lack of periaxin in mice causes loss of compartments, formation of short myelin segments (internodes) and reduced nerve conduction velocity. How compartments are formed and maintained, and their relevance to human neuropathies is largely unknown. Here we show that formation of compartments around myelin is driven by the actin cytoskeleton, and maintained by actin and tubulin fences through linkage to the dystroglycan complex. Compartmentalization and establishment of correct internodal length requires the presence of glycosylated dystroglycan, utrophin and extracellular laminin-2/211. A neuropathic patient with reduced internodal length and nerve conduction velocity because of absence of laminin-2/211 (congenital muscular dystrophy 1A) also shows abnormal compartmentalization. These data link formation of compartments through a laminin2, dystroglycan, utrophin, actin axis to internodal length, and provide a common pathogenetic mechanism for two inherited human neuropathies. Other cell types may exploit dystroglycan complexes in similar fashions to create barriers and compartments.
PubMedId: 19321787
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 29, 2009
Authors: Maria Pennuto, Elisa Tinelli, Mariachiara Malaguti, Ubaldo Del Carro, Maurizio D'Antonio, David Ron, Angelo Quattrini, M Laura Feltri, Lawrence Wrabetz
Abstract: Deletion of serine 63 from P0 glycoprotein (P0S63del) causes Charcot-Marie-Tooth 1B neuropathy in humans, and P0S63del produces a similar demyelinating neuropathy in transgenic mice. P0S63del is retained in the endoplasmic reticulum and fails to be incorporated into myelin. Here we report that P0S63del is misfolded and Schwann cells mount a consequential canonical unfolded protein response (UPR), including expression of the transcription factor CHOP, previously associated with apoptosis in ER-stressed cells. UPR activation and CHOP expression respond dynamically to P0S63del levels and are reversible but are associated with only limited apoptosis of Schwann cells. Nonetheless, Chop ablation in S63del mice completely rescues their motor deficit and reduces active demyelination 2-fold. This indicates that signaling through the CHOP arm of the UPR provokes demyelination in inherited neuropathy. S63del mice also provide an opportunity to explore how cells can dysfunction yet survive in prolonged ER stress-important for neurodegeneration related to misfolded proteins.
PubMedId: 18255032
Journal-article: Neuron, vol. 57, 2007
Authors: Felipe A Court, Lawrence Wrabetz, M Laura Feltri
Abstract: Schwann cells form myelin in the peripheral nervous system. All Schwann cells are surrounded by a basal lamina. Extracellular matrix molecules in the basal lamina, such as laminin, regulate key aspects of Schwann cell development including the formation, architecture and function of myelin. Recent genetic and cell biological experiments suggest that Schwann cells regulate the basal lamina and its receptors in both time and space, resulting in differential functions. These findings have important implications for diseases resulting from laminin dysfunction, such as congenital muscular dystrophy 1A.
PubMedId: 16956757
Journal-article: Current opinion in neurobiology, vol. 16, 2006
Authors: Lawrence Wrabetz, Maurizio D'Antonio, Maria Pennuto, Gabriele Dati, Elisa Tinelli, Pietro Fratta, Stefano Previtali, Daniele Imperiale, Jurgen Zielasek, Klaus Toyka, Robin L Avila, Daniel A Kirschner, Albee Messing, M Laura Feltri, Angelo Quattrini
Abstract: Missense mutations in 22 genes account for one-quarter of Charcot-Marie-Tooth (CMT) hereditary neuropathies. Myelin Protein Zero (MPZ, P0) mutations produce phenotypes ranging from adult demyelinating (CMT1B) to early onset [Djrine-Sottas syndrome (DSS) or congenital hypomyelination] to predominantly axonal neuropathy, suggesting gain of function mechanisms. To test this directly, we produced mice in which either the MpzS63C (DSS) or MpzS63del (CMT1B) transgene was inserted randomly, so that the endogenous Mpz alleles could compensate for any loss of mutant P0 function. We show that either mutant allele produces demyelinating neuropathy that mimics the corresponding human disease. However, P0S63C creates a packing defect in the myelin sheath, whereas P0S63del does not arrive to the myelin sheath and is instead retained in the endoplasmic reticulum, where it elicits an unfolded protein response (UPR). This is the first evidence for UPR in association with neuropathy and provides a model to determine whether and how mutant proteins can provoke demyelination from outside of myelin.
PubMedId: 16495463
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 26, 2006
Authors: Maria Laura Feltri, Lawrence Wrabetz
Abstract: This review focuses on the influence of laminins, mediated through laminin receptors present on Schwann cells, on peripheral nerve development and pathology. Laminins influence multiple aspects of cell differentiation and tissue morphogenesis, including cell survival, proliferation, cytoskeletal rearrangements, and polarity. Peripheral nerves are no exception, as shown by the discovery that defective laminin signals contribute to the pathogenesis of diverse neuropathies such as merosin-deficient congenital muscular dystrophy and Charcot-Marie-Tooth 4F, neurofibromatosis, and leprosy. In the last 5 years, advanced molecular and cell biological techniques and conditional mutagenesis in mice began revealing the role of different laminins and receptors in developing nerves. In this way, we are starting to explain morphological and pathological observations beginning at the start of the last century. Here, we review these recent advances and show how the roles of laminins and their receptors are surprisingly varied in both time and place.
PubMedId: 15958125
Journal-article: Journal of the peripheral nervous system : JPNS, vol. 10, 2005
Authors: Holly Colognato, Charles ffrench-Constant, Maria Laura Feltri
Abstract: Extracellular matrix molecules such as laminins have a central role in regulating cell behaviour. However, our understanding of their functions in the mammalian nervous system is incomplete. It is important to establish these functions, both for an understanding of normal development and to devise strategies to enhance repair. Here, we review how insights gained from human diseases caused by genetic mutations in laminins or their receptors have revealed significant and sometimes unexpected roles for laminins in neural stem cells, migrating neurons and myelinating glia, in both the PNS and CNS.
PubMedId: 16043237
Journal-article: Trends in neurosciences, vol. 28, 2005
Authors: Annalisa Bolis, Silvia Coviello, Simona Bussini, Giorgia Dina, Celia Pardini, Stefano Carlo Previtali, Mariachiara Malaguti, Paolo Morana, Ubaldo Del Carro, Maria Laura Feltri, Angelo Quattrini, Lawrence Wrabetz, Alessandra Bolino
Abstract: Mutations in MTMR2, the myotubularin-related 2 gene, cause autosomal recessive Charcot-Marie-Tooth type 4B1 (CMT4B1). This disorder is characterized by childhood onset of weakness and sensory loss, severely decreased nerve conduction velocity, demyelination in the nerve with myelin outfoldings, and severe functional impairment of affected patients, mainly resulting from loss of myelinated fibers in the nerve. We recently generated Mtmr2-null(neo) mice, which show a dysmyelinating neuropathy with myelin outfoldings, thus reproducing human CMT4B1. Mtmr2 is detected in both Schwann cells and neurons, in which it interacts with discs large 1/synapse-associated protein 97 and neurofilament light chain, respectively. Here, we specifically ablated Mtmr2 in either Schwann cells or motor neurons. Disruption of Mtmr2 in Schwann cells produced a dysmyelinating phenotype very similar to that of the Mtmr2-null(neo) mouse. Disruption of Mtmr2 in motor neurons does not provoke myelin outfoldings nor axonal defects. We propose that loss of Mtmr2 in Schwann cells, but not in motor neurons, is both sufficient and necessary to cause CMT4B1 neuropathy. Thus, therapeutical approaches might be designed in the future to specifically deliver the Mtmr2 phospholipid phosphatase to Schwann cells in affected nerves.
PubMedId: 16162938
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 25, 2005
Authors: Alessandra Bolino, Annalisa Bolis, Stefano Carlo Previtali, Giorgia Dina, Simona Bussini, Gabriele Dati, Stefano Amadio, Ubaldo Del Carro, Dolores D Mruk, Maria Laura Feltri, C Yan Cheng, Angelo Quattrini, Lawrence Wrabetz
Abstract: Mutations in MTMR2, the myotubularin-related 2 gene, cause autosomal recessive Charcot-Marie-Tooth (CMT) type 4B1, a demyelinating neuropathy with myelin outfolding and azoospermia. MTMR2 encodes a ubiquitously expressed phosphatase whose preferred substrate is phosphatidylinositol (3,5)-biphosphate, a regulator of membrane homeostasis and vesicle transport. We generated Mtmr2-null mice, which develop progressive neuropathy characterized by myelin outfolding and recurrent loops, predominantly at paranodal myelin, and depletion of spermatids and spermatocytes from the seminiferous epithelium, which leads to azoospermia. Disruption of Mtmr2 in Schwann cells reproduces the myelin abnormalities. We also identified a novel physical interaction in Schwann cells, between Mtmr2 and discs large 1 (Dlg1)/synapse-associated protein 97, a scaffolding molecule that is enriched at the node/paranode region. Dlg1 homologues have been located in several types of cellular junctions and play roles in cell polarity and membrane addition. We propose that Schwann cell-autonomous loss of Mtmr2-Dlg1 interaction dysregulates membrane homeostasis in the paranodal region, thereby producing outfolding and recurrent loops of myelin.
PubMedId: 15557122
Journal-article: The Journal of cell biology, vol. 167, 2004
Authors: Carla Taveggia, Antonella Pizzagalli, Ernesta Fagiani, Albee Messing, M Laura Feltri, Lawrence Wrabetz
Abstract: Oligodendrocytes and Schwann cells use diverse regulatory elements to transcribe myelin basic protein (Mbp). For example, an enhancer 9.0 kb upstream of Mbp (MbpSCE1) activated either the proximal Mbp or hsp68 promoters only in Schwann cells in transgenic mice. Here, we analyze MbpSCE1 in vitro and in vivo and show that MbpSCE1 also activates another myelin gene, Mpz, specifically in Schwann cells in transgenic mice. Surprisingly, although MbpSCE1 behaves as an enhancer in Schwann cells, we show here for the first time that it also activates transcription robustly in cultured oligodendrocytes, but only from its original genomic position. Diverse nuclear proteins binding at distinct combinations of DNA elements in the two cell types may account for cell-specific MbpSCE1 function. A further surprise is that MbpSCE1 activation does not depend on consensus binding sites for the myelin-associated PPARbeta or Krox 20 transcription factors. Finally, chromatin context augments activation of MbpSCE1. Therefore, MbpSCE1 is active in both Schwann cells and oligodendrocytes, but by diverse mechanisms.
PubMedId: 15525335
Journal-article: Journal of neurochemistry, vol. 91, 2004
Authors: Xinghua Yin, Grahame J Kidd, Erik P Pioro, Jennifer McDonough, Ranjan Dutta, M Laura Feltri, Lawrence Wrabetz, Albee Messing, Ryan M Wyatt, Rita J Balice-Gordon, Bruce D Trapp
Abstract: Axonal degeneration is the major cause of permanent neurological disability in individuals with inherited diseases of myelin. Axonal and neuronal changes that precede axonal degeneration, however, are not well characterized. We show here that dysmyelinated lower motor neurons retract and regenerate dysfunctional presynaptic terminals, leading to severe neurological disability before axonal degeneration. In addition, dysmyelination led to a decreased synaptic quantal content, an indicator of synaptic dysfunction. The amplitude and rise time of miniature endplate potentials were also increased, but these changes were primarily consistent with an increase in the passive membrane properties of the transgenic muscle fibers. Maintenance of synaptic connections should be considered as a therapeutic target for slowing progression of neurological disability in primary diseases of myelin.
PubMedId: 15084670
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 24, 2004
Authors: M Laura Feltri, Diana Graus Porta, Stefano C Previtali, Alessandro Nodari, Barbara Migliavacca, Arianna Cassetti, Amanda Littlewood-Evans, Louis F Reichardt, Albee Messing, Angelo Quattrini, Ulrich Mueller, Lawrence Wrabetz
Abstract: In dystrophic mice, a model of merosin-deficient congenital muscular dystrophy, laminin-2 mutations produce peripheral nerve dysmyelination and render Schwann cells unable to sort bundles of axons. The laminin receptor and the mechanism through which dysmyelination and impaired sorting occur are unknown. We describe mice in which Schwann cell-specific disruption of beta1 integrin, a component of laminin receptors, causes a severe neuropathy with impaired radial sorting of axons. beta 1-null Schwann cells populate nerves, proliferate, and survive normally, but do not extend or maintain normal processes around axons. Interestingly, some Schwann cells surpass this problem to form normal myelin, possibly due to the presence of other laminin receptors such as dystroglycan and alpha 6 beta 4 integrin. These data suggest that beta 1 integrin links laminin in the basal lamina to the cytoskeleton in order for Schwann cells to ensheath axons, and alteration of this linkage contributes to the peripheral neuropathy of congenital muscular dystrophy.
PubMedId: 11777940
Journal-article: The Journal of cell biology, vol. 156, 2002
Authors: S Ferraresi, I Lorenzetti, R Nemni, J Kamholz, M L Feltri, L Wrabetz
Abstract: The molecular mechanisms necessary for remyelination by oligodendrocytes remain unexplored. We previously characterized a myelin basic protein promoter-lacZ (MBP-lacZ) transgene whose expression is regulated uniquely during development, and also in pathological situations, suggesting that it may be a useful reporter of molecular mechanisms during remyelination. As a first step toward creating a transgenic mouse model of remyelination, we cultured oligodendrocytes from these transgenic mice and showed that expression of MBP-lacZ appeared in parallel with a marker of oligodendrocyte maturation, galactocerebroside (GC). In addition, basic fibroblast growth factor blocked the expression of both MBP-lacZ and GC in these cells. Therefore, expression of MBP-lacZ reflects not only the developmental stage of oligodendrocytes, but also extrinsic influences on oligodendrocytes. These data suggest that MBP-lacZ may be a useful marker in transgenic mouse models of remyelination.
PubMedId: 9291158
Journal-article: Multiple sclerosis (Houndmills, Basingstoke, England), vol. 3, 1997
Authors: A Quattrini, S Previtali, M L Feltri, N Canal, R Nemni, L Wrabetz
Abstract: Schwann cell gene expression is dynamically regulated after peripheral nerve injury and during regeneration. We hypothesized that the changes in protein expression described after rat peripheral nerve injury could be used to identify single Schwann cell-axon units in human axonal neuropathy. Therefore, we performed immuno-fluorescence staining on sections of injured rat sciatic nerves compared with sections of neuropathic human sural nerves. We chose the markers beta 4 integrin, P0 glycoprotein, and glial fibrillary acidic protein (GFAP) to characterize Schwann cells, and neurofilament-heavy (NF-H) to recognize axons. Normal rat or human myelin-forming units demonstrated a sharp ring of beta 4 staining at their outer surface, P0 staining in the myelin sheath, and NF-H staining in the axon. Acutely denervated rat units transited from broken rings of beta 4 and P0 staining, to diffuse beta 4 and absent P0 and NF-H staining. Chronically denervated rat Schwann cells re-expressed beta 4 more highly, but in a diffuse, non-polarized pattern. In contrast, regenerating units re-expressed beta 4, P0, and NF-H; beta 4 staining was polarized to the outer surface of Schwann cells. Finally, GFAP staining increased progressively after injury and decreased during regeneration in the distal nerve stump. In neuropathic human sural nerves, we identified units exhibiting each of these beta 4, P0, and NF-H staining patterns; the proportion of each pattern correlated best with the extent and chronicity of axonal injury. Thus, synchronous injury of rat sciatic nerve predicts patterns of Schwann cell marker expression in human axonal neuropathy. In addition, the unique changes in the polarity of beta 4 integrin expression, in combination with changes in P0 and NF-H expression, may distinguish normal from denervated or reinnervated myelin-forming Schwann cells in human sural nerve biopsies.
PubMedId: 8856326
Journal-article: Glia, vol. 17, 1996
Authors: L Wrabetz, M L Feltri, H Kim, M Daston, J Kamholz, S S Scherer, N Ratner
Abstract: Loss of function mutations at the NF1 locus may act intrinsically in Schwann cells to cause the formation of benign Schwann cell tumors (neurofibromas) in patients with type 1 neurofibromatosis. To identify contexts in Schwann cells in which such mutations may play an important role, we measured the levels of NF1 mRNA and neurofibromin in rat sciatic nerve during development, after axotomy, and in cultured rat Schwann cells. NF1 mRNA was present in developing sciatic nerve throughout the period of active Schwann cell proliferation and myelination. After nerve transection, no alteration in NF1 message level was detected, but neurofibromin levels increased, as assessed by immunohistochemistry and Western blotting, suggesting that, in vivo, neurofibromin expression in Schwann cells is post-transcriptionally induced during Wallerian degeneration. Cultured rat Schwann cells constitutively expressed NF1 mRNA and neurofibromin. Schwann cell proliferation induced by exposure to serum and forskolin was not associated with changes in NF1 mRNA or neurofibromin expression, whereas Schwann cell proliferation induced by extracts of embryonic brain membranes was associated with increased NF1 message and neurofibromin expression. Thus, Schwann cells, both in vivo and in vitro, express NF1 mRNA constitutively; the expression of NF1 mRNA and neurofibromin is modulated by only some mitogenic stimuli in Schwann cells.
PubMedId: 8847098
Journal-article: Glia, vol. 15, 1995
Authors: S S Scherer, Y T Xu, D Roling, L Wrabetz, M L Feltri, J Kamholz
Abstract: We have examined the regulation of growth-associated protein 43 kD (GAP-43) in rat Schwann cells. In unlesioned adult nerves, GAP-43-immunoreactivity was restricted to non-myelinating Schwann cells and unmyelinated axons. When adult nerves were transected to cause permanent axotomy, previously myelinating Schwann cells expressed progressively more GAP-43-immunoreactivity over 3 weeks, and GAP-43 mRNA levels increased over a similar time course. The peak level of GAP-43 mRNA occurred at least 2 weeks later than that of nerve growth factor receptor, another marker of denervated Schwann cells. In contrast, after nerve-crush, which allows axonal regeneration, many fewer Schwann cells had GAP-43-immunoreactivity, and the amount of GAP-43 mRNA was markedly lower than in transected nerves. Forskolin, a drug that activates adenylate cyclase and mimics many effects of axon-Schwann cell interactions, markedly reduced GAP-43-immunoreactivity and mRNA expression in cultured Schwann cells, whereas interleukin-1 had no effect. These data demonstrate that axon-Schwann cell interactions inhibit the expression of GAP-43 in Schwann cells and that this effect is mimicked by forskolin.
PubMedId: 7815473
Journal-article: Journal of neuroscience research, vol. 38, 1994
Authors: L Wrabetz, S Shumas, J Grinspan, M L Feltri, D Bozyczko, F A McMorris, D Pleasure, J Kamholz
Abstract: Since the regulation of myelin basic protein expression depends primarily on the initiation of transcription, we analyzed the 5' flanking region of the human myelin basic protein gene in transient transfection studies in primary cultures of developing oligodendrocytes. We demonstrated that 149 base pairs 5' of the initiation of transcription was sufficient to direct oligodendrocyte-specific expression of myelin basic protein. The capsite of the fusion transcript was identical with that of the endogenous myelin basic protein transcript, and chloramphenicol acetyl transferase reporter gene expression was restricted to oligodendrocytes in these cultures. Within this 149 base pair region, one distal, negative cis-acting segment, containing a consensus nuclear factor I site, and one proximal, positive cis-acting segment were identified. The distal segment behaved more negatively in Cos-7 cells than in oligodendrocytes, reducing expression to background levels. Furthermore, these functionally important cis-acting segments bound oligodendrocyte nuclear proteins in a pattern differing from other cells, including Cos-7 cells. Interestingly, the distal segment increased heterologous SV40 promoter activity in oligodendrocytes but had no effect on the SV40 promoter in Cos-7 cells. We conclude that the functionally negative distal segment may mediate oligodendrocyte-specific expression of MBP by restricting its expression in other cells. These experiments strongly support using primary cultures of oligodendrocytes for analyzing the myelin-specific promoters.
PubMedId: 7505842
Journal-article: Journal of neuroscience research, vol. 36, 1993
Authors: R Nemni, M L Feltri, R Fazio, A Quattrini, I Lorenzetti, M Corbo, N Canal
Abstract: We report a 74-year-old woman with a slowly progressive sensory motor axonal neuropathy and a monoclonal IgG-kappa that bound to a 68-kd axonal protein identified as the low molecular weight neurofilament protein. The sera of control subjects and disease controls did not bind to neurofilament protein.
PubMedId: 2122795
Journal-article: Annals of neurology, vol. 28, 1990
Presented by CUBS © 2024