Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.563
Filter
Add more filters

Publication year range
1.
Cell ; 159(5): 1027-1041, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416943

ABSTRACT

Endocytic cargo and Rab GTPases are segregated to distinct domains of an endosome. These domains maintain their identity until they undergo fission to traffic cargo. It is not fully understood how segregation of cargo or Rab proteins is maintained along the continuous endosomal membrane or what machinery is required for fission. Endosomes form contact sites with the endoplasmic reticulum (ER) that are maintained during trafficking. Here, we show that stable contacts form between the ER and endosome at constricted sorting domains, and free diffusion of cargo is limited at these positions. We demonstrate that the site of constriction and fission for early and late endosomes is spatially and temporally linked to contact sites with the ER. Lastly, we show that altering ER structure and dynamics reduces the efficiency of endosome fission. Together, these data reveal a surprising role for ER contact in defining the timing and position of endosome fission.


Subject(s)
Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Microtubules/metabolism , Myelin Proteins/metabolism , Nogo Proteins , Time Factors
2.
Annu Rev Cell Dev Biol ; 30: 503-33, 2014.
Article in English | MEDLINE | ID: mdl-25288117

ABSTRACT

Myelination of axons in the nervous system of vertebrates enables fast, saltatory impulse propagation, one of the best-understood concepts in neurophysiology. However, it took a long while to recognize the mechanistic complexity both of myelination by oligodendrocytes and Schwann cells and of their cellular interactions. In this review, we highlight recent advances in our understanding of myelin biogenesis, its lifelong plasticity, and the reciprocal interactions of myelinating glia with the axons they ensheath. In the central nervous system, myelination is also stimulated by axonal activity and astrocytes, whereas myelin clearance involves microglia/macrophages. Once myelinated, the long-term integrity of axons depends on glial supply of metabolites and neurotrophic factors. The relevance of this axoglial symbiosis is illustrated in normal brain aging and human myelin diseases, which can be studied in corresponding mouse models. Thus, myelinating cells serve a key role in preserving the connectivity and functions of a healthy nervous system.


Subject(s)
Myelin Sheath/physiology , Adenosine Triphosphate/metabolism , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Axons/physiology , Central Nervous System/metabolism , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Cytoskeleton/ultrastructure , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Glucose/metabolism , Humans , Inflammation , Leukoencephalopathies/metabolism , Leukoencephalopathies/pathology , Mice , Microscopy, Electron , Myelin Proteins/physiology , Neuronal Plasticity , Oligodendroglia/physiology , Peripheral Nervous System/metabolism , Schwann Cells/physiology , Synaptic Transmission/physiology
3.
Am J Hum Genet ; 111(7): 1316-1329, 2024 07 11.
Article in English | MEDLINE | ID: mdl-38889728

ABSTRACT

Frontotemporal dementia (FTD) is the second most common cause of early-onset dementia after Alzheimer disease (AD). Efforts in the field mainly focus on familial forms of disease (fFTDs), while studies of the genetic etiology of sporadic FTD (sFTD) have been less common. In the current work, we analyzed 4,685 sFTD cases and 15,308 controls looking for common genetic determinants for sFTD. We found a cluster of variants at the MAPT (rs199443; p = 2.5 × 10-12, OR = 1.27) and APOE (rs6857; p = 1.31 × 10-12, OR = 1.27) loci and a candidate locus on chromosome 3 (rs1009966; p = 2.41 × 10-8, OR = 1.16) in the intergenic region between RPSA and MOBP, contributing to increased risk for sFTD through effects on expression and/or splicing in brain cortex of functionally relevant in-cis genes at the MAPT and RPSA-MOBP loci. The association with the MAPT (H1c clade) and RPSA-MOBP loci may suggest common genetic pleiotropy across FTD and progressive supranuclear palsy (PSP) (MAPT and RPSA-MOBP loci) and across FTD, AD, Parkinson disease (PD), and cortico-basal degeneration (CBD) (MAPT locus). Our data also suggest population specificity of the risk signals, with MAPT and APOE loci associations mainly driven by Central/Nordic and Mediterranean Europeans, respectively. This study lays the foundations for future work aimed at further characterizing population-specific features of potential FTD-discriminant APOE haplotype(s) and the functional involvement and contribution of the MAPT H1c haplotype and RPSA-MOBP loci to pathogenesis of sporadic forms of FTD in brain cortex.


Subject(s)
Apolipoproteins E , Frontotemporal Dementia , Genetic Predisposition to Disease , Genome-Wide Association Study , tau Proteins , Humans , Frontotemporal Dementia/genetics , tau Proteins/genetics , Apolipoproteins E/genetics , Male , Female , Aged , Polymorphism, Single Nucleotide , Genetic Loci , Middle Aged , Case-Control Studies , Myelin Proteins
4.
Mol Cell ; 74(6): 1215-1226.e4, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31053471

ABSTRACT

Programmed death ligand 1 (PD-L1, also called B7-H1) is an immune checkpoint protein that inhibits immune function through its binding of the programmed cell death protein 1 (PD-1) receptor. Clinically approved antibodies block extracellular PD-1 and PD-L1 binding, yet the role of intracellular PD-L1 in cancer remains poorly understood. Here, we discovered that intracellular PD-L1 acts as an RNA binding protein that regulates the mRNA stability of NBS1, BRCA1, and other DNA damage-related genes. Through competition with the RNA exosome, intracellular PD-L1 protects targeted RNAs from degradation, thereby increasing cellular resistance to DNA damage. RNA immunoprecipitation and RNA-seq experiments demonstrated that PD-L1 regulates RNA stability genome-wide. Furthermore, we developed a PD-L1 antibody, H1A, which abrogates the interaction of PD-L1 with CMTM6, thereby promoting PD-L1 degradation. Intracellular PD-L1 may be a potential therapeutic target to enhance the efficacy of radiotherapy and chemotherapy in cancer through the inhibition of DNA damage response and repair.


Subject(s)
B7-H1 Antigen/genetics , DNA Repair , DNA, Neoplasm/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Gene Expression Regulation, Neoplastic , Programmed Cell Death 1 Receptor/genetics , Animals , Antineoplastic Agents/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage , DNA, Neoplasm/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Gamma Rays/therapeutic use , HCT116 Cells , HeLa Cells , Humans , MARVEL Domain-Containing Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Myelin Proteins , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Proteolysis/drug effects , Proteolysis/radiation effects , RNA Stability/drug effects , RNA Stability/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 121(32): e2403652121, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39083419

ABSTRACT

Leucine-rich glioma-inactivated protein 1 (LGI1), a secretory protein in the brain, plays a critical role in myelination; dysfunction of this protein leads to hypomyelination and white matter abnormalities (WMAs). Here, we hypothesized that LGI1 may regulate myelination through binding to an unidentified receptor on the membrane of oligodendrocytes (OLs). To search for this hypothetic receptor, we analyzed LGI1 binding proteins through LGI1-3 × FLAG affinity chromatography with mouse brain lysates followed by mass spectrometry. An OL-specific membrane protein, the oligodendrocytic myelin paranodal and inner loop protein (OPALIN), was identified. Conditional knockout (cKO) of OPALIN in the OL lineage caused hypomyelination and WMAs, phenocopying LGI1 deficiency in mice. Biochemical analysis revealed the downregulation of Sox10 and Olig2, transcription factors critical for OL differentiation, further confirming the impaired OL maturation in Opalin cKO mice. Moreover, virus-mediated re-expression of OPALIN successfully restored myelination in Opalin cKO mice. In contrast, re-expression of LGI1-unbound OPALIN_K23A/D26A failed to reverse the hypomyelination phenotype. In conclusion, our study demonstrated that OPALIN on the OL membrane serves as an LGI1 receptor, highlighting the importance of the LGI1/OPALIN complex in orchestrating OL differentiation and myelination.


Subject(s)
Cell Differentiation , Intracellular Signaling Peptides and Proteins , Mice, Knockout , Oligodendroglia , Animals , Oligodendroglia/metabolism , Oligodendroglia/cytology , Mice , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Myelin Sheath/metabolism , Myelin Proteins/metabolism , Myelin Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 120(4): e2200057120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36649432

ABSTRACT

Antibody delivery to the CNS remains a huge hurdle for the clinical application of antibodies targeting a CNS antigen. The blood-brain barrier and blood-CSF barrier restrict access of therapeutic antibodies to their CNS targets in a major way. The very high amounts of therapeutic antibodies that are administered systemically in recent clinical trials to reach CNS targets are barely viable cost-wise for broad, routine applications. Though global CNS delivery of antibodies can be achieved by intrathecal application, these procedures are invasive. A non-invasive method to bring antibodies into the CNS reliably and reproducibly remains an important unmet need in neurology. In the present study, we show that intranasal application of a mouse monoclonal antibody against the neurite growth-inhibiting and plasticity-restricting membrane protein Nogo-A leads to a rapid transfer of significant amounts of antibody to the brain and spinal cord in intact adult rats. Daily intranasal application for 2 wk of anti-Nogo-A antibody enhanced growth and compensatory sprouting of corticofugal projections and functional recovery in rats after large unilateral cortical strokes. These findings are a starting point for clinical translation for a less invasive route of application of therapeutic antibodies to CNS targets for many neurological indications.


Subject(s)
Antibodies, Monoclonal , Myelin Proteins , Animals , Rats , Brain/metabolism , Myelin Proteins/metabolism , Nogo Proteins , Spinal Cord/metabolism , Antibodies, Monoclonal/administration & dosage , Administration, Intranasal
7.
Brain ; 147(9): 3113-3130, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38743588

ABSTRACT

Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common inherited peripheral neuropathy caused by a 1.5 Mb tandem duplication of chromosome 17 harbouring the PMP22 gene. This dose-dependent overexpression of PMP22 results in disrupted Schwann cell myelination of peripheral nerves. To obtain better insights into the underlying pathogenic mechanisms in CMT1A, we investigated the role of PMP22 duplication in cellular homeostasis in CMT1A mouse models and in patient-derived induced pluripotent stem cells differentiated into Schwann cell precursors (iPSC-SCPs). We performed lipidomic profiling and bulk RNA sequencing (RNA-seq) on sciatic nerves of two developing CMT1A mouse models and on CMT1A patient-derived iPSC-SCPs. For the sciatic nerves of the CMT1A mice, cholesterol and lipid metabolism was downregulated in a dose-dependent manner throughout development. For the CMT1A iPSC-SCPs, transcriptional analysis unveiled a strong suppression of genes related to autophagy and lipid metabolism. Gene ontology enrichment analysis identified disturbances in pathways related to plasma membrane components and cell receptor signalling. Lipidomic analysis confirmed the severe dysregulation in plasma membrane lipids, particularly sphingolipids, in CMT1A iPSC-SCPs. Furthermore, we identified reduced lipid raft dynamics, disturbed plasma membrane fluidity and impaired cholesterol incorporation and storage, all of which could result from altered lipid storage homeostasis in the patient-derived CMT1A iPSC-SCPs. Importantly, this phenotype could be rescued by stimulating autophagy and lipolysis. We conclude that PMP22 duplication disturbs intracellular lipid storage and leads to a more disordered plasma membrane owing to an alteration in the lipid composition, which might ultimately lead to impaired axo-glial interactions. Moreover, targeting lipid handling and metabolism could hold promise for the treatment of patients with CMT1A.


Subject(s)
Cell Membrane , Charcot-Marie-Tooth Disease , Homeostasis , Induced Pluripotent Stem Cells , Lipid Metabolism , Myelin Proteins , Schwann Cells , Schwann Cells/metabolism , Humans , Myelin Proteins/metabolism , Myelin Proteins/genetics , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Homeostasis/physiology , Animals , Mice , Lipid Metabolism/physiology , Cell Membrane/metabolism , Induced Pluripotent Stem Cells/metabolism , Sciatic Nerve/metabolism , Gene Duplication
8.
J Biol Chem ; 299(2): 102839, 2023 02.
Article in English | MEDLINE | ID: mdl-36581210

ABSTRACT

Data from gnomAD indicate that a missense mutation encoding the T118M variation in human peripheral myelin protein 22 (PMP22) is found in roughly one of every 75 genomes of western European lineage (1:120 in the overall human population). It is unusual among PMP22 variants that cause Charcot-Marie-Tooth (CMT) disease in that it is not 100% penetrant. Here, we conducted cellular and biophysical studies to determine why T118M PMP22 predisposes humans to CMT, but with only incomplete penetrance. We found that T118M PMP22 is prone to mistraffic but differs even from the WT protein in that increased expression levels do not result in a reduction in trafficking efficiency. Moreover, the T118M mutant exhibits a reduced tendency to form large intracellular aggregates relative to other disease mutants and even WT PMP22. NMR spectroscopy revealed that the structure and dynamics of T118M PMP22 resembled those of WT. These results show that the main consequence of T118M PMP22 in WT/T118M heterozygous individuals is a reduction in surface-trafficked PMP22, unaccompanied by formation of toxic intracellular aggregates. This explains the incomplete disease penetrance and the mild neuropathy observed for WT/T118M CMT cases. We also analyzed BioVU, a biobank linked to deidentified electronic medical records, and found a statistically robust association of the T118M mutation with the occurrence of long and/or repeated episodes of carpal tunnel syndrome. Collectively, our results illuminate the cellular effects of the T118M PMP22 variation leading to CMT disease and indicate a second disorder for which it is a risk factor.


Subject(s)
Charcot-Marie-Tooth Disease , Myelin Proteins , Humans , Charcot-Marie-Tooth Disease/genetics , Mutation, Missense , Myelin Proteins/genetics , Genetic Predisposition to Disease
9.
J Biol Chem ; 299(10): 105232, 2023 10.
Article in English | MEDLINE | ID: mdl-37690690

ABSTRACT

After adult mammalian central nervous system injury, axon regeneration is extremely limited or absent, resulting in persistent neurological deficits. Axon regeneration failure is due in part to the presence of inhibitory proteins, including NogoA (Rtn4A), from which two inhibitory domains have been defined. When these inhibitory domains are deleted, but an amino-terminal domain is still expressed in a gene trap line, mice show axon regeneration and enhanced recovery from injury. In contrast, when there is no amino-terminal Nogo-A fragment in the setting of inhibitory domain deletion, then axon regeneration and recovery are indistinguishable from WT. These data indicated that an amino-terminal Nogo-A fragment derived from the gene trap might promote axon regeneration, but this had not been tested directly and production of this fragment without gene targeting was unclear. Here, we describe posttranslation production of an amino-terminal fragment of Nogo-A from the intact gene product. This fragment is created by proteolysis near amino acid G214-N215 and levels are enhanced by axotomy. Furthermore, this fragment promotes axon regeneration in vitro and acts cell autonomously in neurons, in contrast to the inhibitory extracellular action of other Nogo-A domains.Proteins interacting with the amino-terminal Nogo-A fragment by immunoprecipitation include HSPA8 (HSC70, HSP7C). Suppression of HSPA8 expression by shRNA decreases axon regeneration from cerebral cortical neurons and overexpression increases axon regeneration. Moreover, the amino-terminal Nogo-A fragment increases HSPA8 chaperone activity. These data provide an explanation for varied results in different gene-targeted Nogo-A mice, as well as revealing an axon regeneration promoting domain of Nogo-A.


Subject(s)
Axons , Myelin Proteins , Animals , Mice , Axons/metabolism , Growth Inhibitors/metabolism , Mammals/metabolism , Myelin Proteins/genetics , Myelin Proteins/metabolism , Nerve Regeneration/physiology , Nogo Proteins/genetics , Nogo Proteins/metabolism , Proteolysis , Female , Mice, Inbred C57BL
10.
Neurobiol Dis ; 192: 106414, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38253209

ABSTRACT

Alteration in protein citrullination (PC), a common posttranslational modification (PTM), contributes to pathogenesis in various inflammatory disorders. We previously reported that PC and protein arginine deiminase 2 (PAD2), the predominant enzyme isoform that catalyzes this PTM in the central nervous system (CNS), are altered in mouse models of amyotrophic lateral sclerosis (ALS). We now demonstrate that PAD2 expression and PC are altered in human postmortem ALS spinal cord and motor cortex compared to controls, increasing in astrocytes while trending lower in neurons. Furthermore, PC is enriched in protein aggregates that contain the myelin proteins PLP and MBP in ALS. These results confirm our findings in ALS mouse models and suggest that altered PAD2 and PC contribute to neurodegeneration in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Citrullination , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/metabolism , Gliosis/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Myelin Proteins/metabolism , Myelin Sheath/pathology , Protein Aggregates , Protein-Arginine Deiminase Type 2/metabolism , Protein-Arginine Deiminases/metabolism , Proteins/metabolism , Spinal Cord/pathology
11.
Hippocampus ; 34(9): 491-502, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39091158

ABSTRACT

Hippocampal area CA2 has garnered attention in recent times owing to its significant involvement in social memory and distinctive plasticity characteristics. Research has revealed that the CA2 region demonstrates a remarkable resistance to plasticity, particularly in the Schaffer Collateral (SC)-CA2 pathway. In this study we investigated the role of Nogo-A, a well-known axon growth inhibitor and more recently discovered plasticity regulator, in modulating plasticity within the CA2 region. The findings demonstrate that blocking Nogo-A in male rat hippocampal slices facilitates the establishment of both short-term and long-term plasticity in the SC-CA2 pathway, while having no impact on the Entorhinal Cortical (EC)-CA2 pathway. Additionally, the study reveals that inhibiting Nogo-A enables association between the SC and EC pathways. Mechanistically, we confirm that Nogo-A operates through its well-known co-receptor, p75 neurotrophin receptor (p75NTR), and its downstream signaling factor such as Rho-associated protein kinase (ROCK), as their inhibition also allows plasticity induction in the SC-CA2 pathway. Additionally, the induction of long-term depression (LTD) in both the EC and SC-CA2 pathways led to persistent LTD, which was not affected by Nogo-A inhibition. Our study demonstrates the involvement of Nogo-A mediated signaling mechanisms in limiting synaptic plasticity within the CA2 region.


Subject(s)
CA2 Region, Hippocampal , Neuronal Plasticity , Nogo Proteins , Synapses , Animals , Nogo Proteins/metabolism , Male , Neuronal Plasticity/physiology , Synapses/physiology , Synapses/drug effects , Synapses/metabolism , CA2 Region, Hippocampal/physiology , CA2 Region, Hippocampal/metabolism , CA2 Region, Hippocampal/drug effects , Rats, Sprague-Dawley , Rats , rho-Associated Kinases/metabolism , rho-Associated Kinases/antagonists & inhibitors , Entorhinal Cortex/physiology , Entorhinal Cortex/metabolism , Receptors, Nerve Growth Factor/metabolism , Neural Pathways/physiology , Myelin Proteins/metabolism , Myelin Proteins/genetics , Nerve Tissue Proteins , Receptors, Growth Factor
12.
Eur J Neurosci ; 60(4): 4503-4517, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951719

ABSTRACT

Myelin sheath plays important roles in information conduction and nerve injury repair in the peripheral nerve system (PNS). Enhancing comprehension of the structure and components of the myelin sheath in the PNS during development would contribute to a more comprehensive understanding of the developmental and regenerative processes. In this research, the structure of sciatic nerve myelin sheath in C57BL/6 mice from embryonic day 14 (E14) to postnatal 12 months (12M) was observed with transmission electron microscopy. Myelin structure appeared in the sciatic nerve as early as E14, and the number and thickness of myelin lamellar gradually increased with the development until 12M. Transcriptome analysis was performed to show the expressions of myelin-associated genes and transcriptional factors involved in myelin formation. The genes encoding myelin proteins (Mag, Pmp22, Mpz, Mbp, Cnp and Prx) showed the same expression pattern, peaking at postnatal day 7 (P7) and P28 after birth, whereas the negative regulators of myelination (c-Jun, Tgfb1, Tnc, Cyr61, Ngf, Egr1, Hgf and Bcl11a) showed an opposite expression pattern. In addition, the expression of myelin-associated proteins and transcriptional factors was measured by Western blot and immunofluorescence staining. The protein expressions of MAG, PMP22, MPZ, CNPase and PRX increased from E20 to P14. The key transcriptional factor c-Jun co-localized with the Schwann cells Marker S100ß and decreased after birth, whereas Krox20/Egr2 increased during development. Our data characterized the structure and components of myelin sheath during the early developmental stages, providing insights for further understanding of PNS development.


Subject(s)
Mice, Inbred C57BL , Myelin Sheath , Sciatic Nerve , Animals , Myelin Sheath/metabolism , Sciatic Nerve/metabolism , Sciatic Nerve/growth & development , Sciatic Nerve/ultrastructure , Mice , Myelin Proteins/metabolism , Myelin Proteins/genetics
13.
Funct Integr Genomics ; 24(1): 10, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38221563

ABSTRACT

Thyroid cancer is the most common type of endocrine cancer. Chemokine-like factor (CKLF)-like MARVEL transmembrane domain containing 6 (CMTM6) is recognized as one of its potential immunotherapy targets. The purpose of this study was to investigate the role and molecular mechanism of CMTM6 in regulating the development of thyroid cancer cells. In this study, expression levels of CMTM6 and the sodium/iodide symporter (NIS) were detected by qRT-PCR. Additionally, colony formation assay and flow cytometry were used to detect cell proliferation and apoptosis, while expression levels of various proteins were assessed using Western blotting. Further, the apoptosis and invasion capacity of cells were investigated by scratch and transwell experiments. Finally, the effect of CMTM6 on the epithelial-mesenchymal transition (EMT) of thyroid cancer cells was determined by immunofluorescence assay, which measured the expression levels of epithelial and mesenchymal phenotypic markers. The results of qRT-PCR experiments showed that CMTM6 was highly expressed in thyroid cancer tissues and cells. In addition, knockdown of CMTM6 expression significantly increased NIS expression. Function experiments demonstrated that small interfering (si)-CMTM6 treatment inhibited the proliferation, migration, invasion, and EMT of thyroid cancer cells, while promoting apoptosis of FTC133 cells. Furthermore, mechanistic studies showed that mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase (ERK) phosphorylation were inhibited by si-CMTM6, as demonstrated by Western blot experiments. In conclusion, our findings demonstrated the role of CMTM6 in the metastasis of thyroid cancer. Briefly, CMTM6 exerts its tumor-promoting effect through the MAPK signaling pathway and could potentially be used as a valuable biomarker for thyroid cancer diagnosis and prognosis.


Subject(s)
MARVEL Domain-Containing Proteins , Myelin Proteins , Symporters , Thyroid Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Symporters/genetics , Symporters/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , MARVEL Domain-Containing Proteins/genetics , MARVEL Domain-Containing Proteins/metabolism , Myelin Proteins/genetics , Myelin Proteins/metabolism
14.
J Gene Med ; 26(5): e3685, 2024 May.
Article in English | MEDLINE | ID: mdl-38686653

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is identified as one of the most prevalent and malignant brain tumors, characterized by poor treatment outcomes and a limited prognosis. CMTM6, a membrane protein, has been found to upregulate the expression of programmed cell death 1 ligand 1 protein (PD-L1) and acts as an immune checkpoint inhibitor by inhibiting the programmed death 1 protein/PD-L1 signaling pathway. Recent research has demonstrated a high expression of CMTM6 in GBM, suggesting its potential role in influencing the pathogenesis and progression of GBM, as well as its association with immune cell infiltration in the tumor microenvironment. However, the underlying mechanism of CMTM6 in GBM requires further investigation. METHODS: Data from cancer patients in The Cancer Genome Atlas, Gene Expression Omnibus and Chinese Glioma Genome Atlas cohorts were consolidated for the current study. Through multi-omics analysis, the study systematically examined the expression profile of CMTM6, epigenetic modifications, prognostic significance, biological functions, potential mechanisms of action and alterations in the immune microenvironment. Additionally, the study investigated CMTM6 expression in GBM cell lines and normal cells using reverse transcription PCR and western blot analysis. The impact of CMTM6 on GBM cell proliferation, migration and invasion was evaluated using a combination of cell counting kit-8 assay, clone formation assay, 5-ethynyl-2'-deoxyuridine incorporation assay, wound healing assay and Transwell assay. In order to explore the mechanism of CMTM6, the Wnt/ß-catenin signaling pathway and autophagy-related genes were further verified through western blot analysis. RESULTS: CMTM6 is highly expressed in multiple tumors, particularly GBM. CMTM6 has been shown to be a valuable diagnostic and prognostic biomarker by various bioinformatics approaches. Additionally, CMTM6 plays a pivotal role in the pathogenesis of cancer, specifically GBM, by modulating various biological processes such as DNA methyltransferase expression, RNA modification, copy number variation, genomic heterogeneity, tumor stemness and DNA methylation. The findings of the experiment indicate a significant correlation between elevated CMTM6 expression and the proliferation, invasion, migration and autophagy of GBM cells, with potential key roles mediated through the Wnt/ß-catenin signaling pathway. Furthermore, CMTM6 is implicated in modulating tumor immune cell infiltration and is closely linked to the expression of various immune checkpoint inhibitors and immune modulators, particularly within the context of GBM. High levels of CMTM6 expression also enhance the responsiveness of GBM patients to radiotherapy and chemotherapy, thereby offering valuable insights for guiding treatment strategies for GBM. CONCLUSIONS: Autophagy-related CMTM6 is highly expressed in various types of cancer, especially GBM, and it can regulate GBM progression through the Wnt/ß-catenin signaling pathway and is capable of being used as an underlying target for the diagnosis, treatment selection and prognosis of patients with GBM.


Subject(s)
Autophagy , Biomarkers, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Glioblastoma , MARVEL Domain-Containing Proteins , Tumor Microenvironment , Wnt Signaling Pathway , Humans , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , MARVEL Domain-Containing Proteins/metabolism , MARVEL Domain-Containing Proteins/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Tumor Microenvironment/immunology , Cell Line, Tumor , Autophagy/genetics , Prognosis , Cell Proliferation , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Myelin Proteins/genetics , Myelin Proteins/metabolism , Cell Movement/genetics , beta Catenin/metabolism , beta Catenin/genetics
15.
Biochem Soc Trans ; 52(4): 1747-1756, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38979632

ABSTRACT

Underexpression, overexpression, and point mutations in peripheral myelin protein 22 (PMP22) cause most cases of Charcot-Marie-Tooth disease (CMTD). While its exact functions remain unclear, PMP22 is clearly essential for formation and maintenance of healthy myelin in the peripheral nervous system. This review explores emerging evidence for roles of PMP22 in cholesterol homeostasis. First, we highlight dysregulation of lipid metabolism in PMP22-based forms of CMTD and recently-discovered interactions between PMP22 and cholesterol biosynthesis machinery. We then examine data that demonstrates PMP22 and cholesterol co-traffic in cells and co-localize in lipid rafts, including how disease-causing PMP22 mutations result in aberrations in cholesterol localization. Finally, we examine roles for interactions between PMP22 and ABCA1 in cholesterol efflux. Together, this emerging body of evidence suggests that PMP22 plays a role in facilitating enhanced cholesterol synthesis and trafficking necessary for production and maintenance of healthy myelin.


Subject(s)
Charcot-Marie-Tooth Disease , Cholesterol , Homeostasis , Myelin Proteins , Schwann Cells , Cholesterol/metabolism , Humans , Schwann Cells/metabolism , Myelin Proteins/metabolism , Myelin Proteins/genetics , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/genetics , Animals , Myelin Sheath/metabolism , Lipid Metabolism , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Mutation
16.
J Peripher Nerv Syst ; 29(2): 202-212, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38581130

ABSTRACT

BACKGROUND: Caused by duplications of the gene encoding peripheral myelin protein 22 (PMP22), Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common hereditary neuropathy. Despite this shared genetic origin, there is considerable variability in clinical severity. It is hypothesized that genetic modifiers contribute to this heterogeneity, the identification of which may reveal novel therapeutic targets. In this study, we present a comprehensive analysis of clinical examination results from 1564 CMT1A patients sourced from a prospective natural history study conducted by the RDCRN-INC (Inherited Neuropathy Consortium). Our primary objective is to delineate extreme phenotype profiles (mild and severe) within this patient cohort, thereby enhancing our ability to detect genetic modifiers with large effects. METHODS: We have conducted large-scale statistical analyses of the RDCRN-INC database to characterize CMT1A severity across multiple metrics. RESULTS: We defined patients below the 10th (mild) and above the 90th (severe) percentiles of age-normalized disease severity based on the CMT Examination Score V2 and foot dorsiflexion strength (MRC scale). Based on extreme phenotype categories, we defined a statistically justified recruitment strategy, which we propose to use in future modifier studies. INTERPRETATION: Leveraging whole genome sequencing with base pair resolution, a future genetic modifier evaluation will include single nucleotide association, gene burden tests, and structural variant analysis. The present work not only provides insight into the severity and course of CMT1A, but also elucidates the statistical foundation and practical considerations for a cost-efficient and straightforward patient enrollment strategy that we intend to conduct on additional patients recruited globally.


Subject(s)
Charcot-Marie-Tooth Disease , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Humans , Adult , Male , Female , Middle Aged , Adolescent , Young Adult , Severity of Illness Index , Child , Myelin Proteins/genetics , Patient Selection , Phenotype , Aged , Genes, Modifier , Child, Preschool
17.
Brain ; 146(10): 4025-4032, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37337674

ABSTRACT

Copy number variation (CNV) may lead to pathological traits, and Charcot-Marie-Tooth disease type 1A (CMT1A), the commonest inherited peripheral neuropathy, is due to a genomic duplication encompassing the dosage-sensitive PMP22 gene. MicroRNAs act as repressors on post-transcriptional regulation of gene expression and in rodent models of CMT1A, overexpression of one such microRNA (miR-29a) has been shown to reduce the PMP22 transcript and protein level. Here we present genomic and functional evidence, for the first time in a human CNV-associated phenotype, of the 3' untranslated region (3'-UTR)-mediated role of microRNA repression on gene expression. The proband of the family presented with an early-onset, severe sensorimotor demyelinating neuropathy and harboured a novel de novo deletion in the PMP22 3'-UTR. The deletion is predicted to include the miR-29a seed binding site and transcript analysis of dermal myelinated nerve fibres using a novel platform, revealed a marked increase in PMP22 transcript levels. Functional evidence from Schwann cell lines harbouring the wild-type and mutant 3'-UTR showed significantly increased reporter assay activity in the latter, which was not ameliorated by overexpression of a miR-29a mimic. This shows the importance of miR-29a in regulating PMP22 expression and opens an avenue for therapeutic drug development.


Subject(s)
Charcot-Marie-Tooth Disease , MicroRNAs , Humans , Charcot-Marie-Tooth Disease/pathology , MicroRNAs/genetics , DNA Copy Number Variations , Myelin Proteins/genetics , Myelin Proteins/metabolism , Gene Expression
18.
Brain ; 146(7): 2885-2896, 2023 07 03.
Article in English | MEDLINE | ID: mdl-36511878

ABSTRACT

Charcot-Marie-Tooth disease is the most common inherited disorder of the PNS. CMT1A accounts for 40-50% of all cases and is caused by a duplication of the PMP22 gene on chromosome 17, leading to dysmyelination in the PNS. Patient-derived models to study such myelination defects are lacking as the in vitro generation of human myelinating Schwann cells has proved to be particularly challenging. Here, we present an induced pluripotent stem cell-derived organoid culture, containing various cell types of the PNS, including myelinating human Schwann cells, which mimics the human PNS. Single-cell analysis confirmed the PNS-like cellular composition and provides insight into the developmental trajectory. We used this organoid model to study disease signatures of CMT1A, revealing early ultrastructural myelin alterations, including increased myelin periodic line distance and hypermyelination of small axons. Furthermore, we observed the presence of onion-bulb-like formations in a later developmental stage. These hallmarks were not present in the CMT1A-corrected isogenic line or in a CMT2A iPSC line, supporting the notion that these alterations are specific to CMT1A. Downregulation of PMP22 expression using short-hairpin RNAs or a combinatorial drug consisting of baclofen, naltrexone hydrochloride and D-sorbitol was able to ameliorate the myelin defects in CMT1A-organoids. In summary, this self-organizing organoid model can capture biologically meaningful features of the disease and capture the physiological complexity, forms an excellent model for studying demyelinating diseases and supports the therapeutic approach of reducing PMP22 expression.


Subject(s)
Charcot-Marie-Tooth Disease , Induced Pluripotent Stem Cells , Humans , Myelin Sheath/metabolism , Induced Pluripotent Stem Cells/metabolism , Down-Regulation , Myelin Proteins/genetics , Myelin Proteins/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Organoids/metabolism , Schwann Cells
19.
Brain ; 146(9): 3608-3615, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37143322

ABSTRACT

The duplication of the peripheral myelin protein 22 (PMP22) gene causes a demyelinating type of neuropathy, commonly known as Charcot-Marie-Tooth disease type 1A (CMT1A). Development of effective drugs for CMT1A still remains as an unmet medical need. In the present study, we assessed the role of the transforming growth factor beta 4 (TGFß4)/Nodal axis in the pathogenesis of CMT1A. First, we identified PMP22 overexpression-induced Nodal expression in Schwann cells, which might be one of the downstream effectors in CMT1A. Administration of Nodal protein at the developmental stage of peripheral nerves induced the demyelinating phenotype in vivo. Second, we further isolated TGFß4 as an antagonist that could abolish Nodal-induced demyelination. Finally, we developed a recombinant TGFß4-fragment crystallizable (Fc) fusion protein, CX201, and demonstrated that its application had promyelinating efficacy in Schwann cells. CX201 administration improved the demyelinating phenotypes of CMT1A mouse models at both pre-symptomatic and post-symptomatic stages. These results suggest that the TGFß4/Nodal axis plays a crucial role in the pathogenesis of CMT1A and might be a potential therapeutic target for CMT1A.


Subject(s)
Charcot-Marie-Tooth Disease , Animals , Mice , Charcot-Marie-Tooth Disease/pathology , Myelin Proteins/metabolism , Schwann Cells , Phenotype , Transforming Growth Factor beta/metabolism
20.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Article in English | MEDLINE | ID: mdl-33893233

ABSTRACT

Peripheral myelin protein (PMP22) is an integral membrane protein that traffics inefficiently even in wild-type (WT) form, with only 20% of the WT protein reaching its final plasma membrane destination in myelinating Schwann cells. Misfolding of PMP22 has been identified as a key factor in multiple peripheral neuropathies, including Charcot-Marie-Tooth disease and Dejerine-Sottas syndrome. While biophysical analyses of disease-associated PMP22 mutants show altered protein stabilities, leading to reduced surface trafficking and loss of PMP22 function, it remains unclear how destabilization of PMP22 mutations causes mistrafficking. Here, native ion mobility-mass spectrometry (IM-MS) is used to compare the gas phase stabilities and abundances for an array of mutant PM22 complexes. We find key differences in the PMP22 mutant stabilities and propensities to form homodimeric complexes. Of particular note, we observe that severely destabilized forms of PMP22 exhibit a higher propensity to dimerize than WT PMP22. Furthermore, we employ lipid raft-mimicking SCOR bicelles to study PMP22 mutants, and find that the differences in dimer abundances are amplified in this medium when compared to micelle-based data, with disease mutants exhibiting up to 4 times more dimer than WT when liberated from SCOR bicelles. We combine our findings with previous cellular data to propose that the formation of PMP22 dimers from destabilized monomers is a key element of PMP22 mistrafficking.


Subject(s)
Myelin Proteins/metabolism , Peripheral Nervous System Diseases/physiopathology , Protein Transport/physiology , Cell Membrane/metabolism , Humans , Ion Mobility Spectrometry/methods , Mass Spectrometry/methods , Membrane Proteins/metabolism , Myelin Proteins/genetics , Myelin Proteins/physiology , Peripheral Nervous System Diseases/diagnostic imaging , Peripheral Nervous System Diseases/metabolism , Protein Folding , Protein Stability , Schwann Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL