Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Mol Ther ; 31(11): 3290-3307, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37641403

ABSTRACT

Type 4C Charcot-Marie-Tooth (CMT4C) demyelinating neuropathy is caused by autosomal recessive SH3TC2 gene mutations. SH3TC2 is highly expressed in myelinating Schwann cells. CMT4C is a childhood-onset progressive disease without effective treatment. Here, we generated a gene therapy for CMT4C mediated by an adeno-associated viral 9 vector (AAV9) to deliver the human SH3TC2 gene in the Sh3tc2-/- mouse model of CMT4C. We used a minimal fragment of the myelin protein zero (Mpz) promoter (miniMpz), which was cloned and validated to achieve Schwann cell-targeted expression of SH3TC2. Following the demonstration of AAV9-miniMpz.SH3TC2myc vector efficacy to re-establish SH3TC2 expression in the peripheral nervous system, we performed an early as well as a delayed treatment trial in Sh3tc2-/- mice. We demonstrate both after early as well as following late treatment improvements in multiple motor performance tests and nerve conduction velocities. Moreover, treatment led to normalization of the organization of the nodes of Ranvier, which is typically deficient in CMT4C patients and Sh3tc2-/- mice, along with reduced ratios of demyelinated fibers, increased myelin thickness and reduced g-ratios at both time points of intervention. Taken together, our results provide a proof of concept for an effective and potentially translatable gene replacement therapy for CMT4C treatment.


Subject(s)
Charcot-Marie-Tooth Disease , Genetic Therapy , Intracellular Signaling Peptides and Proteins , Animals , Humans , Mice , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/therapy , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Schwann Cells/metabolism
2.
J Peripher Nerv Syst ; 28(3): 329-340, 2023 09.
Article in English | MEDLINE | ID: mdl-37220142

ABSTRACT

BACKGROUND AND AIMS: Mitofusin 1 (MFN1) and MFN2 are outer mitochondrial membrane fusogenic proteins regulating mitochondrial network morphology. MFN2 mutations cause Charcot-Marie-Tooth type 2A (CMT2A), an axonal neuropathy characterized by mitochondrial fusion defects, which in the case of a GTPase domain mutant, were rescued following wild-type MFN1/2 (MFN1/2WT ) overexpression. In this study, we compared the therapeutic efficiency between MFN1WT and MFN2WT overexpression in correcting mitochondrial defects induced by the novel MFN2K357T mutation located in the highly conserved R3 region. METHODS: Constructs expressing either MFN2K357T , MFN2WT , or MFN1WT under the ubiquitous chicken ß-actin hybrid (CBh) promoter were generated. Flag or myc tag was used for their detection. Differentiated SH-SY5Y cells were single transfected with MFN1WT , MFN2WT , or MFN2K357T , as well as double transfected with MFN2K357T /MFN2WT or MFN2K357T /MFN1WT . RESULTS: SH-SY5Y cells transfected with MFN2K357T exhibited severe perinuclear mitochondrial clustering with axon-like processes devoid of mitochondria. Single transfection with MFN1WT resulted in a more interconnected mitochondrial network than transfection with MFN2WT , accompanied by mitochondrial clusters. Double transfection of MFN2K357T with either MFN1WT or MFN2WT resolved the mutant-induced mitochondrial clusters and led to detectable mitochondria throughout the axon-like processes. MFN1WT showed higher efficacy than MFN2WT in rescuing these defects. INTERPRETATION: These results further demonstrate the higher potential of MFN1WT over MFN2WT overexpression to rescue CMT2A-induced mitochondrial network abnormalities due to mutations outside the GTPase domain. This higher phenotypic rescue conferred by MFN1WT , possibly due to its higher mitochondrial fusogenic ability, may be applied to different CMT2A cases regardless of the MFN2 mutation type.


Subject(s)
Charcot-Marie-Tooth Disease , Neuroblastoma , Humans , Mitochondrial Dynamics , Neuroblastoma/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mutation , GTP Phosphohydrolases/genetics , Mitochondrial Proteins/genetics , Charcot-Marie-Tooth Disease/genetics
3.
Brain ; 142(5): 1227-1241, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30907403

ABSTRACT

Charcot-Marie-Tooth disease type 4C is the most common recessively inherited demyelinating neuropathy that results from loss of function mutations in the SH3TC2 gene. Sh3tc2-/- mice represent a well characterized disease model developing early onset progressive peripheral neuropathy with hypo- and demyelination, slowing of nerve conduction velocities and disturbed nodal architecture. The aim of this project was to develop a gene replacement therapy for treating Charcot-Marie-Tooth disease type 4C to rescue the phenotype of the Sh3tc2-/- mouse model. We generated a lentiviral vector LV-Mpz.SH3TC2.myc to drive expression of the human SH3TC2 cDNA under the control of the Mpz promoter specifically in myelinating Schwann cells. The vector was delivered into 3-week-old Sh3tc2-/- mice by lumbar intrathecal injection and gene expression was assessed 4-8 weeks after injection. Immunofluorescence analysis showed presence of myc-tagged human SH3TC2 in sciatic nerves and lumbar roots in the perinuclear cytoplasm of a subset of Schwann cells, in a dotted pattern co-localizing with physiologically interacting protein Rab11. Quantitative PCR analysis confirmed SH3TC2 mRNA expression in different peripheral nervous system tissues. A treatment trial was initiated in 3 weeks old randomized Sh3tc2-/- littermate mice which received either the full or mock (LV-Mpz.Egfp) vector. Behavioural analysis 8 weeks after injection showed improved motor performance in rotarod and foot grip tests in treated Sh3tc2-/- mice compared to mock vector-treated animals. Moreover, motor nerve conduction velocities were increased in treated Sh3tc2-/- mice. On a structural level, morphological analysis revealed significant improvement in g-ratios, myelin thickness, and ratios of demyelinated fibres in lumbar roots and sciatic nerves of treated Sh3tc2-/- mice. Finally, treated mice also showed improved nodal molecular architecture and reduction of blood neurofilament light levels, a clinically relevant biomarker for axonal injury/degeneration. This study provides a proof of principle for viral gene replacement therapy targeted to Schwann cells to treat Charcot-Marie-Tooth disease type 4C and potentially other similar demyelinating inherited neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/therapy , Disease Models, Animal , Genetic Therapy/methods , Intracellular Signaling Peptides and Proteins/genetics , Animals , Charcot-Marie-Tooth Disease/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Mice, Knockout , Mice, Transgenic
4.
Proc Natl Acad Sci U S A ; 113(17): E2421-9, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27035961

ABSTRACT

Inherited demyelinating peripheral neuropathies are progressive incurable diseases without effective treatment. To develop a gene therapy approach targeting myelinating Schwann cells that can be translatable, we delivered a lentiviral vector using a single lumbar intrathecal injection and a myelin-specific promoter. The human gene of interest, GJB1, which is mutated in X-linked Charcot-Marie-Tooth Disease (CMT1X), was delivered intrathecally into adult Gjb1-null mice, a genetically authentic model of CMT1X that develops a demyelinating peripheral neuropathy. We obtained widespread, stable, and cell-specific expression of connexin32 in up to 50% of Schwann cells in multiple lumbar spinal roots and peripheral nerves. Behavioral and electrophysiological analysis revealed significantly improved motor performance, quadriceps muscle contractility, and sciatic nerve conduction velocities. Furthermore, treated mice exhibited reduced numbers of demyelinated and remyelinated fibers and fewer inflammatory cells in lumbar motor roots, as well as in the femoral motor and sciatic nerves. This study demonstrates that a single intrathecal lentiviral gene delivery can lead to Schwann cell-specific expression in spinal roots extending to multiple peripheral nerves. This clinically relevant approach improves the phenotype of an inherited neuropathy mouse model and provides proof of principle for treating inherited demyelinating neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Demyelinating Diseases/genetics , Animals , Genetic Therapy , Humans , Myelin Sheath/metabolism , Schwann Cells/metabolism
5.
Hum Mol Genet ; 24(7): 2049-64, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25524707

ABSTRACT

Oligodendrocytes are coupled by gap junctions (GJs) formed mainly by connexin47 (Cx47) and Cx32. Recessive GJC2/Cx47 mutations cause Pelizaeus-Merzbacher-like disease, a hypomyelinating leukodystrophy, while GJB1/Cx32 mutations cause neuropathy and chronic or acute-transient encephalopathy syndromes. Cx32/Cx47 double knockout (Cx32/Cx47dKO) mice develop severe CNS demyelination beginning at 1 month of age leading to death within weeks, offering a relevant model to study disease mechanisms. In order to clarify whether the loss of oligodendrocyte connexins has cell autonomous effects, we generated transgenic mice expressing the wild-type human Cx32 under the control of the mouse proteolipid protein promoter, obtaining exogenous hCx32 expression in oligodendrocytes. By crossing these mice with Cx32KO mice, we obtained expression of hCx32 on Cx32KO background. Immunohistochemical and immunoblot analysis confirmed strong CNS expression of hCx32 specifically in oligodendrocytes and correct localization forming GJs at cell bodies and along the myelin sheath. TG(+)Cx32/Cx47dKO mice generated by further crossing with Cx47KO mice showed that transgenic expression of hCx32 rescued the severe early phenotype of CNS demyelination in Cx32/Cx47dKO mice, resulting in marked improvement of behavioral abnormalities at 1 month of age, and preventing the early mortality. Furthermore, TG(+)Cx32/Cx47dKO mice showed significant improvement of myelination compared with Cx32/Cx47dKO CNS at 1 month of age, while the inflammatory and astrogliotic changes were fully reversed. Our study confirms that loss of oligodendrocyte GJs has cell autonomous effects and that re-establishment of GJ connectivity by replacement of least one GJ protein provides correction of the leukodystrophy phenotype.


Subject(s)
Connexins/metabolism , Demyelinating Diseases/metabolism , Gap Junctions/metabolism , Oligodendroglia/metabolism , Animals , Animals, Genetically Modified , Connexins/genetics , Demyelinating Diseases/genetics , Female , Gap Junctions/genetics , Humans , Male , Mice , Mice, Transgenic , Phenotype , Gap Junction beta-1 Protein
6.
J Gene Med ; 16(11-12): 364-73, 2014.
Article in English | MEDLINE | ID: mdl-25394283

ABSTRACT

BACKGROUND: Most leukodystrophies result from mutations in genes expressed in oligodendrocytes that may cause autonomous loss of function of cell structural proteins. Therefore, effective gene delivery to oligodendrocytes is necessary to develop future treatments. MATERIALS: To achieve this, we cloned a lentiviral vector in which the enhanced green fluorescent protein (EGFP) expression was driven by the oligodendrocyte specific 2,3-cyclic nucleotide 3-phosphodiesterase promoter. The vector was inserted into C57BL/6 neonatal mouse brain by combined intraventricular and parenchymal injections. RESULTS: Assessment of EGFP expression revealed a widespread distribution, specifically in cells of the oligodendrocyte linage, starting from postnatal day 6 (P6) in the subventricular zone and spreading through migrating oligodendrocyte precursors. By P30, it was detectable throughout the brain and persisted for at least 3 months, showing an increase both in the number of expressing cells and in intensity over time. EGFP expression was restricted to oligodendrocyte linage cells. On average, 20.3 ± 2.56% of all oligodendrocytes in different central nervous system areas were EGFP-positive, with regional variations. CONCLUSIONS: Lentiviral gene delivery using an oligodendrocyte-specific promoter may achieve widespread and long-lasting expression selectively in oligodendrocytes, offering a possibility for gene therapy in certain leukodystrophies, although the relatively low rates of oligodendrocyte transduction are a limitation that remains to be overcome.


Subject(s)
Genetic Therapy , Lentivirus/genetics , Oligodendroglia/metabolism , Animals , Brain/metabolism , Coculture Techniques , Female , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Male , Mice, Inbred C57BL , PC12 Cells , Rats , Sciatic Nerve/metabolism
7.
J Neuropathol Exp Neurol ; 73(9): 865-79, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25101702

ABSTRACT

Gap junctions are essential for glial cell function and have been increasingly implicated in multiple sclerosis (MS). Because increasing cortical abnormalities correlate with disease progression and cognitive dysfunction, we examined the expression of oligodendrocytic connexin32 (Cx32) and Cx47 and their astrocytic partners Cx30 and Cx43 in cortical lesions and normal-appearing gray matter (NAGM) in MS patients. Postmortem brain tissue samples from 9 MS cases were compared with 10 controls using real-time polymerase chain reaction, immunoblot, and immunohistochemical analyses. Connexin32 and Cx47 gap junction formation in oligodendrocytes was reduced within lesions, whereas Cx32 loss also extended to NAGM. In contrast, astrocytic Cx30 expression was increased within cortical lesions, whereas Cx43 was elevated in both lesions and NAGM. Diffuse microglial activation and marked astrogliotic changes accompanied these connexin abnormalities. Increased expression of Cx43 correlated with inflammatory load (r = 0.828, p = 0.042), whereas Cx32 expression correlated with longer disease duration and, therefore, milder course (r = 0.825, p = 0.043). Thus, there is a loss of intramyelin and intercellular oligodendrocyte gap junctions in MS gray matter lesions and NAGM, whereas interastrocytic gap junctions are increased, reflecting astrogliosis. These changes correlate with inflammation and disease duration and suggest that disconnection of oligodendrocytes from reactive astrocytes may play a role in failed remyelination and disease progression.


Subject(s)
Astrocytes/pathology , Gap Junctions/pathology , Gray Matter/pathology , Multiple Sclerosis/pathology , Oligodendroglia/pathology , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Connexins/genetics , Connexins/metabolism , Female , Gap Junctions/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Middle Aged , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Plaque, Amyloid/etiology , RNA, Messenger/metabolism
8.
Acta Neuropathol ; 123(6): 873-86, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22484441

ABSTRACT

Oligodendrocyte gap junctions (GJs) are vital for central nervous system myelination, but their involvement in multiple sclerosis (MS) pathology remains unknown. The aim of this study was to examine alterations of oligodendrocyte and related astrocyte GJs in MS lesions and normal-appearing white matter (NAWM). Post-mortem brain samples from 9 MS and 11 age-matched non-MS control patients were studied. Tissue sections that included both chronic active and inactive lesions were characterized neuropathologically with Luxol Fast Blue staining and immunostaining for myelin oligodendrocyte glycoprotein (MOG) and the microglial marker Iba1. We analyzed the expression of Cx32 and Cx47 in oligodendrocytes and of Cx43, the major astrocytic partner in oligodendrocyte-astrocyte (O/A) GJs by quantitative immunoblot and real-time PCR. Formation of GJ plaques was quantified by immunohistochemistry. Compared to control brains, both Cx32 and Cx47 GJ plaques and protein levels were reduced in and around MS lesions, while Cx43 was increased as part of astrogliosis. In the NAWM, Cx32 was significantly reduced along myelinated fibers whereas Cx47 showed increased expression mainly in oligodendrocyte precursor cells (OPCs). However, OPCs showed only limited connectivity to astrocytes. Cx43 showed modestly increased levels in MS NAWM compared to controls, while GJ plaque counts were unchanged. Our findings indicate that oligodendrocyte GJs are affected not only in chronic MS lesions but also in NAWM, where disruption of Cx32 GJs in myelinated fibers may impair myelin structure and function. Moreover, limited O/A GJ connectivity of recruited OPCs in the setting of persistent inflammation and astrogliosis may prevent differentiation and remyelination.


Subject(s)
Connexins/metabolism , Gap Junctions/pathology , Multiple Sclerosis/pathology , Nerve Fibers, Myelinated/pathology , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Connexins/genetics , Gap Junctions/metabolism , Humans , Microglia/metabolism , Microglia/pathology , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Oligodendroglia/metabolism
9.
Ann Neurol ; 72(2): 241-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22473710

ABSTRACT

OBJECTIVE: A study was undertaken to describe the clinical spectrum, voltage-gated potassium channel (VGKC) complex antibody specificities, and central nervous system localization of antibody binding in 29 patients diagnosed with Morvan syndrome (MoS). METHODS: Clinical data were collected using questionnaires. Radioimmunoassay, cell-based assays, and mouse brain immunohistochemistry were used to characterize the serum antibodies. RESULTS: Neuromyotonia (100%), neuropsychiatric features (insomnia 89.7%, confusion 65.5%, amnesia 55.6%, hallucinations 51.9%), dysautonomia (hyperhidrosis 86.2%, cardiovascular 48.3%), and neuropathic pain (62.1%) were the most common manifestations. A total of 93.1% of MoS patients were male. VGKC-complex antibodies were present in 23 of 29 (79%) MoS patients at referral; 24 of 27 available sera had CASPR2, LGI1, or both CASPR2 and LGI1 antibodies (3 also with contactin-2 antibodies). CASPR2 antibodies were generally higher titer than LGI1 antibodies. Tumors (41.4%), mainly thymomas, were associated with CASPR2 antibodies and a poor prognosis, whereas LGI1 antibodies were associated with serum hyponatremia. In brain tissue regions including the hypothalamus, raphe, and locus coeruleus, commercial antibodies to LGI1 bound to neuronal cell bodies including the antidiuretic hormone-secreting and orexin-secreting hypothalamic neurons, whereas CASPR2 commercial antibodies bound more often to the neuropil. MoS antibodies bound similarly, but there was evidence of additional antibodies in some sera that were not adsorbed by LGI1- or CASPR2-expressing cells and bound to mouse Caspr2(-/-) tissue. INTERPRETATION: MoS is clinically distinct from other VGKC-complex antibody-associated conditions, and usually is associated with high-titer CASPR2 antibodies, often accompanied by lower-titer LGI1 antibodies. CASPR2 and LGI1 antibodies bind to multiple brain regions, which helps to explain the multifocal clinical features of this disease, but other antibodies are likely to play a role in some patients and need to be characterized in future studies.


Subject(s)
Antibodies/blood , Potassium Channels, Voltage-Gated/immunology , Syringomyelia/blood , Syringomyelia/immunology , Syringomyelia/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Brain/metabolism , Brain/pathology , Contactin 2/immunology , Female , Humans , International Cooperation , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Membrane Proteins/immunology , Mice , Middle Aged , Nerve Tissue Proteins/immunology , Neurons/metabolism , Neuropeptides/pharmacology , Orexins , Pain/physiopathology , Protein Binding/drug effects , Proteins/immunology , Radioimmunoassay , Retrospective Studies , Serum/metabolism , Surveys and Questionnaires , Syringomyelia/therapy , Treatment Outcome , Young Adult
10.
J Neurol Sci ; 312(1-2): 52-6, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21906755

ABSTRACT

We describe a patient presenting with a combination of muscle fasciculations, paresthesias, hyperhidrosis, as well as insomnia, agitation and confusion. He went on to develop psychosis and respiratory failure requiring intensive care. Electromyography confirmed the presence of neuromyotonia and CSF showed mild pleocytosis. Routine testing for voltage-gated potassium channel complex (VGKC-complex) antibodies was highly positive, confirming the clinical diagnosis of Morvan's syndrome. The patient improved after treatment with intravenous immunoglobulin and methylprednisolone. Further investigation of the antigenic targets using immunohistochemistry and cell-based assays revealed that he had autoantibodies targeting Lgi1, Caspr2 and Contactin-2/Tag-1, all proteins known to be complexed with VGKC in peripheral nerves and CNS. This is the first case of Morvan's syndrome from Cyprus and illustrates the clinical features as well as the emerging complexity of antigenic targets involved in the pathogenesis.


Subject(s)
Autoantibodies/blood , Isaacs Syndrome/immunology , Myokymia/immunology , Potassium Channels, Voltage-Gated/immunology , Aged , Autoantibodies/biosynthesis , Contactin 2/immunology , Humans , Intracellular Signaling Peptides and Proteins , Isaacs Syndrome/drug therapy , Male , Membrane Proteins/immunology , Myokymia/drug therapy , Nerve Tissue Proteins/immunology , Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...