Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 16(3): 616-640, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383802

ABSTRACT

Haplo-insufficiency of the gene encoding the myelin protein PMP22 leads to focal myelin overgrowth in the peripheral nervous system and hereditary neuropathy with liability to pressure palsies (HNPP). Conversely, duplication of PMP22 causes Charcot-Marie-Tooth disease type 1A (CMT1A), characterized by hypomyelination of medium to large caliber axons. The molecular mechanisms of abnormal myelin growth regulation by PMP22 have remained obscure. Here, we show in rodent models of HNPP and CMT1A that the PI3K/Akt/mTOR-pathway inhibiting phosphatase PTEN is correlated in abundance with PMP22 in peripheral nerves, without evidence for direct protein interactions. Indeed, treating DRG neuron/Schwann cell co-cultures from HNPP mice with PI3K/Akt/mTOR pathway inhibitors reduced focal hypermyelination. When we treated HNPP mice in vivo with the mTOR inhibitor Rapamycin, motor functions were improved, compound muscle amplitudes were increased and pathological tomacula in sciatic nerves were reduced. In contrast, we found Schwann cell dedifferentiation in CMT1A uncoupled from PI3K/Akt/mTOR, leaving partial PTEN ablation insufficient for disease amelioration. For HNPP, the development of PI3K/Akt/mTOR pathway inhibitors may be considered as the first treatment option for pressure palsies.


Subject(s)
Arthrogryposis , Charcot-Marie-Tooth Disease , Hereditary Sensory and Motor Neuropathy , Phosphatidylinositol 3-Kinases , Mice , Animals , Proto-Oncogene Proteins c-akt , Rodentia/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Myelin Proteins/genetics , Myelin Proteins/metabolism , TOR Serine-Threonine Kinases
2.
Article in English | MEDLINE | ID: mdl-38199866

ABSTRACT

Peripheral nerves exist in a stable state in adulthood providing a rapid bidirectional signaling system to control tissue structure and function. However, following injury, peripheral nerves can regenerate much more effectively than those of the central nervous system (CNS). This multicellular process is coordinated by peripheral glia, in particular Schwann cells, which have multiple roles in stimulating and nurturing the regrowth of damaged axons back to their targets. Aside from the repair of damaged nerves themselves, nerve regenerative processes have been linked to the repair of other tissues and de novo innervation appears important in establishing an environment conducive for the development and spread of tumors. In contrast, defects in these processes are linked to neuropathies, aging, and pain. In this review, we focus on the role of peripheral glia, especially Schwann cells, in multiple aspects of nerve regeneration and discuss how these findings may be relevant for pathologies associated with these processes.


Subject(s)
Nerve Regeneration , Schwann Cells , Schwann Cells/physiology , Nerve Regeneration/physiology , Humans , Animals , Peripheral Nerves/physiology , Axons/physiology
3.
Cell Metab ; 35(12): 2136-2152.e9, 2023 12 05.
Article in English | MEDLINE | ID: mdl-37989315

ABSTRACT

The peripheral nervous system harbors a remarkable potential to regenerate after acute nerve trauma. Full functional recovery, however, is rare and critically depends on peripheral nerve Schwann cells that orchestrate breakdown and resynthesis of myelin and, at the same time, support axonal regrowth. How Schwann cells meet the high metabolic demand required for nerve repair remains poorly understood. We here report that nerve injury induces adipocyte to glial signaling and identify the adipokine leptin as an upstream regulator of glial metabolic adaptation in regeneration. Signal integration by leptin receptors in Schwann cells ensures efficient peripheral nerve repair by adjusting injury-specific catabolic processes in regenerating nerves, including myelin autophagy and mitochondrial respiration. Our findings propose a model according to which acute nerve injury triggers a therapeutically targetable intercellular crosstalk that modulates glial metabolism to provide sufficient energy for successful nerve repair.


Subject(s)
Myelin Sheath , Peripheral Nerves , Myelin Sheath/metabolism , Neuroglia , Schwann Cells/metabolism , Nerve Regeneration/physiology
4.
Nat Neurosci ; 26(7): 1218-1228, 2023 07.
Article in English | MEDLINE | ID: mdl-37386131

ABSTRACT

Axonal degeneration determines the clinical outcome of multiple sclerosis and is thought to result from exposure of denuded axons to immune-mediated damage. Therefore, myelin is widely considered to be a protective structure for axons in multiple sclerosis. Myelinated axons also depend on oligodendrocytes, which provide metabolic and structural support to the axonal compartment. Given that axonal pathology in multiple sclerosis is already visible at early disease stages, before overt demyelination, we reasoned that autoimmune inflammation may disrupt oligodendroglial support mechanisms and hence primarily affect axons insulated by myelin. Here, we studied axonal pathology as a function of myelination in human multiple sclerosis and mouse models of autoimmune encephalomyelitis with genetically altered myelination. We demonstrate that myelin ensheathment itself becomes detrimental for axonal survival and increases the risk of axons degenerating in an autoimmune environment. This challenges the view of myelin as a solely protective structure and suggests that axonal dependence on oligodendroglial support can become fatal when myelin is under inflammatory attack.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Humans , Myelin Sheath/metabolism , Axons/metabolism , Multiple Sclerosis/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Risk Factors
5.
Neurobiol Dis ; 176: 105952, 2023 01.
Article in English | MEDLINE | ID: mdl-36493976

ABSTRACT

The glial cell of the peripheral nervous system (PNS), the Schwann cell (SC), counts among the most multifaceted cells of the body. During development, SCs secure neuronal survival and participate in axonal path finding. Simultaneously, they orchestrate the architectural set up of the developing nerves, including the blood vessels and the endo-, peri- and epineurial layers. Perinatally, in rodents, SCs radially sort and subsequently myelinate individual axons larger than 1 µm in diameter, while small calibre axons become organised in non-myelinating Remak bundles. SCs have a vital role in maintaining axonal health throughout life and several specialized SC types perform essential functions at specific locations, such as terminal SC at the neuromuscular junction (NMJ) or SC within cutaneous sensory end organs. In addition, neural crest derived satellite glia maintain a tight communication with the soma of sensory, sympathetic, and parasympathetic neurons and neural crest derivatives are furthermore an indispensable part of the enteric nervous system. The remarkable plasticity of SCs becomes evident in the context of a nerve injury, where SC transdifferentiate into intriguing repair cells, which orchestrate a regenerative response that promotes nerve repair. Indeed, the multiple adaptations of SCs are captivating, but remain often ill-resolved on the molecular level. Here, we summarize and discuss the knowns and unknowns of the vast array of functions that this single cell type can cover in peripheral nervous system development, maintenance, and repair.


Subject(s)
Peripheral Nerve Injuries , Schwann Cells , Humans , Schwann Cells/metabolism , Peripheral Nerves/metabolism , Axons/metabolism , Neurons/metabolism , Peripheral Nervous System/metabolism , Nerve Regeneration/physiology , Peripheral Nerve Injuries/metabolism
6.
Dev Neurobiol ; 81(5): 490-506, 2021 07.
Article in English | MEDLINE | ID: mdl-32628805

ABSTRACT

Axons share a close relationship with Schwann cells, their glial partners in peripheral nerves. An intricate axo-glia network of signals and bioactive molecules regulates the major aspects of nerve development and normal functioning of the peripheral nervous system. Disruptions to these complex axo-glial interactions can have serious neurological consequences, as typically seen in injured nerves. Recent studies in inherited neuropathies have demonstrated that damage to one of the partners in this symbiotic unit ultimately leads to impairment of the other partner, emphasizing the bidirectional influence of axon to glia and glia to axon signaling in these diseases. After physical trauma to nerves, dramatic alterations in the architecture and signaling environment of peripheral nerves take place. Here, axons and Schwann cells respond adaptively to these perturbations and change the nature of their reciprocal interactions, thereby driving the remodeling and regeneration of peripheral nerves. In this review, we focus on the nature and importance of axon-glia interactions in injured nerves, both for the reshaping and repair of nerves after trauma, and in driving pathology in inherited peripheral neuropathies.


Subject(s)
Peripheral Nervous System Diseases , Axons/physiology , Humans , Nerve Regeneration , Neuroglia/physiology , Peripheral Nervous System , Schwann Cells/physiology
7.
Development ; 146(21)2019 11 12.
Article in English | MEDLINE | ID: mdl-31719044

ABSTRACT

During the development of the peripheral nervous system, axons and myelinating Schwann cells form a unique symbiotic unit, which is realized by a finely tuned network of molecular signals and reciprocal interactions. The importance of this complex interplay becomes evident after injury or in diseases in which aspects of axo-glial interaction are perturbed. This Review focuses on the specific interdependence of axons and Schwann cells in peripheral nerve development that enables axonal outgrowth, Schwann cell lineage progression, radial sorting and, finally, formation and maintenance of the myelin sheath.


Subject(s)
Axons/physiology , Gene Expression Regulation, Developmental , Myelin Sheath/physiology , Neuroglia/physiology , Peripheral Nerves/embryology , Schwann Cells/physiology , Animals , Cell Differentiation , Cell Lineage , Cell Separation , Mice , Nerve Regeneration , Peripheral Nerves/physiology , Peripheral Nervous System , Rats , Signal Transduction
8.
Nat Commun ; 10(1): 1467, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30931926

ABSTRACT

In contrast to acute peripheral nerve injury, the molecular response of Schwann cells in chronic neuropathies remains poorly understood. Onion bulb structures are a pathological hallmark of demyelinating neuropathies, but the nature of these formations is unknown. Here, we show that Schwann cells induce the expression of Neuregulin-1 type I (NRG1-I), a paracrine growth factor, in various chronic demyelinating diseases. Genetic disruption of Schwann cell-derived NRG1 signalling in a mouse model of Charcot-Marie-Tooth Disease 1A (CMT1A), suppresses hypermyelination and the formation of onion bulbs. Transgenic overexpression of NRG1-I in Schwann cells on a wildtype background is sufficient to mediate an interaction between Schwann cells via an ErbB2 receptor-MEK/ERK signaling axis, which causes onion bulb formations and results in a peripheral neuropathy reminiscent of CMT1A. We suggest that diseased Schwann cells mount a regeneration program that is beneficial in acute nerve injury, but that overstimulation of Schwann cells in chronic neuropathies is detrimental.


Subject(s)
Demyelinating Diseases/genetics , Neuregulin-1/genetics , Paracrine Communication , Schwann Cells/metabolism , Sural Nerve/metabolism , Animals , Animals, Genetically Modified , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Diabetes Mellitus, Type 1/complications , Diabetic Neuropathies/etiology , Diabetic Neuropathies/genetics , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Humans , MAP Kinase Signaling System , Mice , Mice, Transgenic , Microscopy, Electron , Motor Activity , Myelin Proteins/genetics , Neuregulin-1/metabolism , Neuritis, Autoimmune, Experimental/genetics , Neuritis, Autoimmune, Experimental/metabolism , Neuritis, Autoimmune, Experimental/pathology , Neuroglia/metabolism , Rats , Receptor, ErbB-2/metabolism , Schwann Cells/ultrastructure , Sciatic Nerve/injuries , Signal Transduction , Sural Nerve/ultrastructure , Tibial Nerve
9.
Nat Commun ; 10(1): 1840, 2019 Apr 16.
Article in English | MEDLINE | ID: mdl-30992451

ABSTRACT

Michael W. Sereda was incorrectly associated with the Department of Cellular Neurophysiology, Hanover Medical School, Carl-Neuberg-Str. 1, 30625 Hanover, Germany. The correct affiliations for Michael W. Sereda are Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany and Department of Clinical Neurophysiology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany.

10.
Front Neurosci ; 12: 467, 2018.
Article in English | MEDLINE | ID: mdl-30050403

ABSTRACT

Axons are electrically excitable, cable-like neuronal processes that relay information between neurons within the nervous system and between neurons and peripheral target tissues. In the central and peripheral nervous systems, most axons over a critical diameter are enwrapped by myelin, which reduces internodal membrane capacitance and facilitates rapid conduction of electrical impulses. The spirally wrapped myelin sheath, which is an evolutionary specialisation of vertebrates, is produced by oligodendrocytes and Schwann cells; in most mammals myelination occurs during postnatal development and after axons have established connection with their targets. Myelin covers the vast majority of the axonal surface, influencing the axon's physical shape, the localisation of molecules on its membrane and the composition of the extracellular fluid (in the periaxonal space) that immerses it. Moreover, myelinating cells play a fundamental role in axonal support, at least in part by providing metabolic substrates to the underlying axon to fuel its energy requirements. The unique architecture of the myelinated axon, which is crucial to its function as a conduit over long distances, renders it particularly susceptible to injury and confers specific survival and maintenance requirements. In this review we will describe the normal morphology, ultrastructure and function of myelinated axons, and discuss how these change following disease, injury or experimental perturbation, with a particular focus on the role the myelinating cell plays in shaping and supporting the axon.

11.
Ann Clin Transl Neurol ; 2(8): 787-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26339673

ABSTRACT

OBJECTIVE: Pelizaeus-Merzbacher disease (PMD) is a progressive and lethal leukodystrophy caused by mutations affecting the proteolipid protein (PLP1) gene. The most common cause of PMD is a duplication of PLP1 and at present there is no curative therapy available. METHODS: By using transgenic mice carrying additional copies of Plp1, we investigated whether curcumin diet ameliorates PMD symptoms. The diet of Plp1 transgenic mice was supplemented with curcumin for 10 consecutive weeks followed by phenotypical, histological and immunohistochemical analyses of the central nervous system. Plp1 transgenic and wild-type mice fed with normal chow served as controls. RESULTS: Curcumin improved the motor phenotype performance of Plp1 transgenic mice by 50% toward wild-type level and preserved myelinated axons by 35% when compared to Plp1 transgenic controls. Furthermore, curcumin reduced astrocytosis, microgliosis and lymphocyte infiltration in Plp1 transgenic mice. Curcumin diet did not affect the pathologically increased Plp1 mRNA abundance. However, high glutathione levels indicating an oxidative misbalance in the white matter of Plp1 transgenic mice were restored by curcumin treatment. INTERPRETATION: Curcumin may potentially serve as an antioxidant therapy of PMD caused by PLP1 gene duplication.

12.
Nat Med ; 20(9): 1055-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25150498

ABSTRACT

Duplication of the gene encoding the peripheral myelin protein of 22 kDa (PMP22) underlies the most common inherited neuropathy, Charcot-Marie-Tooth 1A (CMT1A), a disease without a known cure. Although demyelination represents a characteristic feature, the clinical phenotype of CMT1A is determined by the degree of axonal loss, and patients suffer from progressive muscle weakness and impaired sensation. CMT1A disease manifests within the first two decades of life, and walking disabilities, foot deformities and electrophysiological abnormalities are already present in childhood. Here, we show in Pmp22-transgenic rodent models of CMT1A that Schwann cells acquire a persistent differentiation defect during early postnatal development, caused by imbalanced activity of the PI3K-Akt and the Mek-Erk signaling pathways. We demonstrate that enhanced PI3K-Akt signaling by axonally overexpressed neuregulin-1 (NRG1) type I drives diseased Schwann cells toward differentiation and preserves peripheral nerve axons. Notably, in a preclinical experimental therapy using a CMT1A rat model, when treatment is restricted to early postnatal development, soluble NRG1 effectively overcomes impaired peripheral nerve development and restores axon survival into adulthood. Our findings suggest a model in which Schwann cell differentiation within a limited time window is crucial for the long-term maintenance of axonal support.


Subject(s)
Charcot-Marie-Tooth Disease/physiopathology , Disease Models, Animal , Neuregulin-1/physiology , Animals , Male , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Transgenic
13.
Am J Hum Genet ; 94(4): 533-46, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24680886

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is a severe hypomyelinating disease, characterized by ataxia, intellectual disability, epilepsy, and premature death. In the majority of cases, PMD is caused by duplication of PLP1 that is expressed in myelinating oligodendrocytes. Despite detailed knowledge of PLP1, there is presently no curative therapy for PMD. We used a Plp1 transgenic PMD mouse model to test the therapeutic effect of Lonaprisan, an antagonist of the nuclear progesterone receptor, in lowering Plp1 mRNA overexpression. We applied placebo-controlled Lonaprisan therapy to PMD mice for 10 weeks and performed the grid slip analysis to assess the clinical phenotype. Additionally, mRNA expression and protein accumulation as well as histological analysis of the central nervous system were performed. Although Plp1 mRNA levels are increased 1.8-fold in PMD mice compared to wild-type controls, daily Lonaprisan treatment reduced overexpression at the RNA level to about 1.5-fold, which was sufficient to significantly improve the poor motor phenotype. Electron microscopy confirmed a 25% increase in the number of myelinated axons in the corticospinal tract when compared to untreated PMD mice. Microarray analysis revealed the upregulation of proapoptotic genes in PMD mice that could be partially rescued by Lonaprisan treatment, which also reduced microgliosis, astrogliosis, and lymphocyte infiltration.


Subject(s)
Estrenes/therapeutic use , Hormone Antagonists/therapeutic use , Pelizaeus-Merzbacher Disease/drug therapy , Progesterone/antagonists & inhibitors , Animals , Disease Models, Animal , Estrenes/pharmacokinetics , Estrenes/pharmacology , Gene Expression Regulation/drug effects , Hormone Antagonists/pharmacokinetics , Hormone Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteolipid Protein/genetics , Phenotype , RNA, Messenger/genetics
14.
Nat Neurosci ; 16(1): 48-54, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23222914

ABSTRACT

After peripheral nerve injury, axons regenerate and become remyelinated by resident Schwann cells. However, myelin repair never results in the original myelin thickness, suggesting insufficient stimulation by neuronal growth factors. Upon testing this hypothesis, we found that axonal neuregulin-1 (NRG1) type III and, unexpectedly, also NRG1 type I restored normal myelination when overexpressed in transgenic mice. This led to the observation that Wallerian degeneration induced de novo NRG1 type I expression in Schwann cells themselves. Mutant mice lacking a functional Nrg1 gene in Schwann cells are fully myelinated but exhibit impaired remyelination in adult life. We suggest a model in which loss of axonal contact triggers denervated Schwann cells to transiently express NRG1 as an autocrine/paracrine signal that promotes Schwann cell differentiation and remyelination.


Subject(s)
Demyelinating Diseases/metabolism , Neuregulin-1/metabolism , Recovery of Function/genetics , Schwann Cells/metabolism , Sciatic Neuropathy/pathology , Animals , Animals, Newborn , Axons/drug effects , Axons/pathology , Axons/ultrastructure , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Demyelinating Diseases/etiology , Disease Models, Animal , Early Growth Response Protein 2/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/physiology , Ganglia, Spinal/cytology , Gene Expression Regulation/genetics , Hedgehog Proteins/genetics , Ki-67 Antigen/metabolism , Locomotion/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Myelin Basic Protein/metabolism , Myelin P0 Protein/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Neuregulin-1/genetics , Neurons/chemistry , Neurons/drug effects , Neurons/metabolism , Octamer Transcription Factor-6/metabolism , RNA, Messenger/metabolism , Rats , Recovery of Function/drug effects , S100 Proteins/metabolism , Schwann Cells/drug effects , Schwann Cells/ultrastructure , Sciatic Nerve/cytology , Sciatic Neuropathy/physiopathology , Signal Transduction/drug effects , Signal Transduction/genetics , Statistics, Nonparametric , Time Factors
15.
Br Med Bull ; 102: 89-113, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22551516

ABSTRACT

INTRODUCTION OR BACKGROUND: Charcot-Marie-Tooth (CMT) disease represents a broad group of inherited motor and sensory neuropathies which can originate from various genetic aberrations, e.g. mutations, deletions and duplications. SOURCES OF DATA: We performed a literature review on murine animal models of CMT disease with regard to experimental therapeutic approaches. Hereby, we focussed on the demyelinating subforms of CMT (CMT1). PubMed items were CMT, animal model, demyelination and therapy. AREAS OF AGREEMENT: Patients affected by CMT suffer from slowly progressive, distally pronounced muscle atrophy caused by an axonal loss. The disease severity is highly variable and impairments may result in wheelchair boundness. No therapy is available yet. AREAS OF CONTROVERSY: Numerous rodent models for the various CMT subtypes are available today. The selection of the correct animal model for the specific CMT subtype provides an important prerequisite for the successful translation of experimental findings in patients. GROWING POINTS: Despite more than 20 years of remarkable progress in CMT research, the disease is still left untreatable. There is a growing number of experimental therapeutic strategies that may be translated into future clinical trials in patients with CMT. AREAS TIMELY FOR DEVELOPING RESEARCH: The slow disease progression and insensitive outcome measures hamper clinical therapy trials in CMT. Biomarkers may provide powerful tools to monitor therapeutic efficacy. Recently, we have shown that transcriptional profiling can be utilized to assess and predict the disease severity in a transgenic rat model and in affected humans.


Subject(s)
Charcot-Marie-Tooth Disease/therapy , Disease Models, Animal , Animals , Animals, Genetically Modified , Charcot-Marie-Tooth Disease/genetics , Genetic Predisposition to Disease , Mice , Molecular Targeted Therapy/methods , Rats
16.
Brain ; 135(Pt 1): 72-87, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22189569

ABSTRACT

Charcot-Marie-Tooth disease is the most common inherited neuropathy and a duplication of the peripheral myelin protein 22 gene causes the most frequent subform Charcot-Marie-Tooth 1A. Patients develop a slowly progressive dysmyelinating and demyelinating peripheral neuropathy and distally pronounced muscle atrophy. The amount of axonal loss determines disease severity. Although patients share an identical monogenetic defect, the disease progression is strikingly variable and the impending disease course can not be predicted in individual patients. Despite promising experimental data, recent therapy trials have failed. Established clinical outcome measures are thought to be too insensitive to detect amelioration within trials. Surrogate biomarkers of disease severity in Charcot-Marie-Tooth 1A are thus urgently needed. Peripheral myelin protein 22 transgenic rats harbouring additional copies of the peripheral myelin protein 22 gene ('Charcot-Marie-Tooth rats'), which were kept on an outbred background mimic disease hallmarks and phenocopy the variable disease severity of patients with Charcot-Marie-Tooth 1A. Hence, we used the Charcot-Marie-Tooth rat to dissect prospective and surrogate markers of disease severity derived from sciatic nerve and skin tissue messenger RNA extracts. Gene set enrichment analysis of sciatic nerve transcriptomes revealed that dysregulation of lipid metabolism associated genes such as peroxisome proliferator-activated receptor gamma constitutes a modifier of present and future disease severity. Importantly, we directly validated disease severity markers from the Charcot-Marie-Tooth rats in 46 patients with Charcot-Marie-Tooth 1A. Our data suggest that the combination of age and cutaneous messenger RNA levels of glutathione S-transferase theta 2 and cathepsin A composes a strong indicator of disease severity in patients with Charcot-Marie-Tooth 1A, as quantified by the Charcot-Marie-Tooth Neuropathy Score. This translational approach, utilizing a transgenic animal model, demonstrates that transcriptional analysis of skin biopsy is suitable to identify biomarkers of Charcot-Marie-Tooth 1A.


Subject(s)
Axons/pathology , Charcot-Marie-Tooth Disease/pathology , Myelin Proteins/genetics , Sciatic Nerve/pathology , Animals , Axons/physiology , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Disease Models, Animal , Glutathione Transferase/genetics , Myelin P0 Protein/genetics , Neural Conduction/physiology , PPAR gamma/genetics , Pain Measurement , Phenotype , Rats , Rats, Transgenic , Sciatic Nerve/physiopathology , Severity of Illness Index
17.
Neurobiol Dis ; 42(1): 1-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21168501

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is the most common inherited neuropathy and a duplication of the peripheral myelin protein of 22 kDa (PMP22) gene causes the most frequent subform CMT1A. Clinical impairments are determined by the amount of axonal loss. Axons of the spontaneous mouse mutant Wallerian degeneration slow (Wlds) show markedly reduced degeneration following various types of injuries. Protection is conferred by a chimeric Wlds gene encoding an N-terminal part of ubiquitination factor Ube4b and full length nicotinamide mononucleotide adenylyl transferase 1 (Nmnat1). Nmnat1 enzyme generates nicotinamide adenine dinucleotide (NAD) from nicotinamide mononucleotide. Here, in a Pmp22 transgenic animal model of Charcot-Marie-Tooth disease type 1A (CMT rat), the Wlds transgene reduced axonal loss and clinical impairments without altering demyelination. Furthermore, nicotinamide - substrate precursor of the Nmnat1 enzyme - transiently delayed posttraumatic axonal degeneration in an in vivo model of acute peripheral nerve injury, but to a lower extent than Wlds. In contrast, 8 weeks of nicotinamide treatment did not influence axonal loss or clinical manifestations in the CMT rat. Therefore, nicotinamide can partially substitute for the protective Wlds effect in acute traumatic, but not in chronic secondary axonal injury. Future studies are needed to develop axon protective therapy in CMT1A which may be combined with therapeutic strategies aimed at downregulation of toxic PMP22 overexpression.


Subject(s)
Axons/pathology , Charcot-Marie-Tooth Disease/genetics , Nerve Tissue Proteins/genetics , Neuroprotective Agents/therapeutic use , Niacinamide/therapeutic use , Sciatic Neuropathy/genetics , Wallerian Degeneration/genetics , Wallerian Degeneration/prevention & control , Animals , Axons/metabolism , Charcot-Marie-Tooth Disease/drug therapy , Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Female , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Sciatic Neuropathy/complications , Sciatic Neuropathy/pathology , Wallerian Degeneration/pathology
18.
Neuron ; 59(4): 581-95, 2008 Aug 28.
Article in English | MEDLINE | ID: mdl-18760695

ABSTRACT

Understanding the control of myelin formation by oligodendrocytes is essential for treating demyelinating diseases. Neuregulin-1 (NRG1) type III, an EGF-like growth factor, is essential for myelination in the PNS. It is thus thought that NRG1/ErbB signaling also regulates CNS myelination, a view suggested by in vitro studies and the overexpression of dominant-negative ErbB receptors. To directly test this hypothesis, we generated a series of conditional null mutants that completely lack NRG1 beginning at different stages of neural development. Unexpectedly, these mice assemble normal amounts of myelin. In addition, double mutants lacking oligodendroglial ErbB3 and ErbB4 become myelinated in the absence of any stimulation by neuregulins. In contrast, a significant hypermyelination is achieved by transgenic overexpression of NRG1 type I or NRG1 type III. Thus, NRG1/ErbB signaling is markedly different between Schwann cells and oligodendrocytes that have evolved an NRG/ErbB-independent mechanism of myelination control.


Subject(s)
Nerve Fibers, Myelinated/metabolism , Neuregulin-1/metabolism , Oligodendroglia/metabolism , Receptor, ErbB-3/metabolism , Schwann Cells/metabolism , Animals , Central Nervous System/cytology , Central Nervous System/growth & development , Central Nervous System/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Dosage , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Mice, Mutant Strains , Neuregulin-1/genetics , Neurons/metabolism , Oligodendroglia/cytology , Peripheral Nervous System/cytology , Peripheral Nervous System/growth & development , Peripheral Nervous System/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-4 , Schwann Cells/cytology , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...