Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
2.
Int J Mol Sci ; 24(12)2023 Jun 19.
Article in English | MEDLINE | ID: mdl-37373466

ABSTRACT

Oligodendrocytes generate myelin sheaths vital for the formation, health, and function of the central nervous system. Mounting evidence suggests that receptor tyrosine kinases (RTKs) are crucial for oligodendrocyte differentiation and myelination in the CNS. It was recently reported that discoidin domain receptor 1 (Ddr1), a collagen-activated RTK, is expressed in oligodendrocyte lineage. However, its specific expression stage and functional role in oligodendrocyte development in the CNS remain to be determined. In this study, we report that Ddr1 is selectively upregulated in newly differentiated oligodendrocytes in the early postnatal CNS and regulates oligodendrocyte differentiation and myelination. Ddr1 knock-out mice of both sexes displayed compromised axonal myelination and apparent motor dysfunction. Ddr1 deficiency alerted the ERK pathway, but not the AKT pathway in the CNS. In addition, Ddr1 function is important for myelin repair after lysolecithin-induced demyelination. Taken together, the current study described, for the first time, the role of Ddr1 in myelin development and repair in the CNS, providing a novel molecule target for the treatment of demyelinating diseases.


Subject(s)
Discoidin Domain Receptor 1 , Myelin Sheath , Oligodendroglia , Animals , Female , Male , Mice , Cell Differentiation , Central Nervous System , Discoidin Domain Receptor 1/genetics , Discoidin Domain Receptor 1/metabolism , Mice, Knockout , Myelin Sheath/metabolism , Neurogenesis , Oligodendroglia/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
3.
J Neurosci ; 43(24): 4405-4417, 2023 06 14.
Article in English | MEDLINE | ID: mdl-37188512

ABSTRACT

Although NG2 is known to be selectively expressed in oligodendrocyte precursor cells (OPCs) for many years, its expressional regulation and functional involvement in oligodendrocyte differentiation have remained elusive. Here, we report that the surface-bound NG2 proteoglycan can physically bind to PDGF-AA and enhances PDGF receptor alpha (PDGFRα) activation of downstream signaling. During differentiation stage, NG2 protein is cleaved by A disintegrin and metalloproteinase with thrombospondin motifs type 4 (Adamts4), which is highly upregulated in differentiating OPCs but gradually downregulated in mature myelinating oligodendrocytes. Genetic ablation of Adamts4 gene impedes NG2 proteolysis, leading to elevated PDGFRα signaling but impaired oligodendrocyte differentiation and axonal myelination in both sexes of mice. Moreover, Adamts4 deficiency also lessens myelin repair in adult brain tissue following Lysophosphatidylcholine-induced demyelination. Thus, Adamts4 could be a potential therapeutic target for enhancing oligodendrocyte differentiation and axonal remyelination in demyelinating diseases.SIGNIFICANCE STATEMENT NG2 is selectively expressed in OPCs and downregulated during differentiation stage. To date, the molecular mechanism underlying the progressive removal of NG2 surface proteoglycan in differentiating OPCs has been unknown. In this study, we demonstrate that ADAMTS4 released by differentiating OPCs cleaves surface NG2 proteoglycan, attenuates PDGFRα signaling, and accelerates oligodendrocyte differentiation. In addition, our study also suggests ADAMTS4 as a potential therapeutic target for promoting myelin recovery in demyelinating diseases.


Subject(s)
Demyelinating Diseases , Remyelination , Male , Female , Mice , Animals , Receptor, Platelet-Derived Growth Factor alpha , Myelin Sheath/metabolism , Proteoglycans/genetics , Oligodendroglia/metabolism , Cell Differentiation/physiology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism
4.
Cells ; 12(6)2023 03 08.
Article in English | MEDLINE | ID: mdl-36980172

ABSTRACT

Mounting evidence suggests that circular RNAs play important roles in the development and progression of cancers. However, their function in glioblastomas (GBM) is still unclear. By circRNA array analysis, we found that circXPO1 (hsa_circ_102737) was significantly upregulated in GBM, and qPCR analysis verified that the circXPO1 expression level was increased in both GBM tissues and cell lines. Functional studies demonstrated that the knockdown of circXPO1 in GBM cell lines repressed cell proliferation and migration; conversely, the overexpression of circXPO1 promoted the malignancy of GBM cells. In line with these findings, circXPO1 inhibition effectively suppressed gliomagenesis in the in situ transplantation model of nude mice. Through bioinformatic analyses and dual-luciferase reporter assays, we showed that circXPO1 directly bound to miR-7-5p, which acted as a tumor suppressor through the negative regulation of RAF1. In conclusion, our studies suggest that the circXPO1/miR-7-5p/RAF1 axis promotes brain tumor formation and may be a potential therapeutic target for GBM treatment.


Subject(s)
Brain Neoplasms , Glioblastoma , MicroRNAs , Animals , Mice , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Humans
5.
Mol Med ; 28(1): 50, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35508991

ABSTRACT

BACKGROUND: The conversion of astrocytes activated by nerve injuries to oligodendrocytes is not only beneficial to axonal remyelination, but also helpful for reversal of glial scar. Recent studies have shown that pathological niche promoted the Sox10-mediated astrocytic transdifferentiation to oligodendrocytes. The extracellular factors underlying the cell fate switching are not known. METHODS: Astrocytes were obtained from mouse spinal cord dissociation culture and purified by differential adherent properties. The lineage conversion of astrocytes into oligodendrocyte lineage cells was carried out by Sox10-expressing virus infection both in vitro and in vivo, meanwhile, epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR) inhibitor Gefitinib were adopted to investigate the function of EGF signaling in this fate transition process. Pharmacological inhibition analyses were performed to examine the pathway connecting the EGF with the expression of oligodendrogenic genes and cell fate transdifferentiation. RESULTS: EGF treatment facilitated the Sox10-induced transformation of astrocytes to O4+ induced oligodendrocyte precursor cells (iOPCs) in vitro. The transdifferentiation of astrocytes to iOPCs went through two distinct but interconnected processes: (1) dedifferentiation of astrocytes to astrocyte precursor cells (APCs); (2) transformation of APCs to iOPCs, EGF signaling was involved in both processes. And EGF triggered astrocytes to express oligodendrogenic genes Olig1 and Olig2 by activating extracellular signal-regulated kinase 1 and 2 (Erk1/2) pathway. In addition, we discovered that EGF can enhance astrocyte transdifferentiation in injured spinal cord tissues. CONCLUSIONS: These findings provide strong evidence that EGF facilitates the transdifferentiation of astrocytes to oligodendrocytes, and suggest that targeting the EGF-EGFR-Erk1/2 signaling axis may represent a novel therapeutic strategy for myelin repair in injured central nervous system (CNS) tissues.


Subject(s)
Astrocytes , Epidermal Growth Factor , Animals , Astrocytes/metabolism , Cell Differentiation , Cells, Cultured , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Mice , Oligodendroglia/metabolism
6.
J Genet Genomics ; 49(2): 132-144, 2022 02.
Article in English | MEDLINE | ID: mdl-34530169

ABSTRACT

Glioma is the most common type of tumor in the central nervous system, accounting for around 80% of all malignant brain tumors. Previous studies showed a significant association between nuclear morphology and the malignant progress of gliomas. By virtue of integrated proteomics and genomics analyses as well as experimental validations, we identify three nuclear lamin genes (LMNA, LMNB1, and LMNB2) that are significantly upregulated in glioma tissues compared with normal brain tissues. We show that elevated expressions of LMNB1, LMNB2, and LMNA in glioma cells are highly associated with the rapid progression of the disease and the knockdown of LMNB1, LMNB2, and LMNA dramatically suppresses glioma progression in both in vitro and in vivo mouse models. Moreover, the repression of glioma cell growth by lamin knockdown is mediated by the pRb-mediated G1-S inhibition. On the contrary, overexpression of lamins in normal human astrocytes dramatically induced nuclear morphological aberrations and accelerated cell growth. Together, our multi-omics-based analysis has revealed a previously unrecognized role of lamin genes in gliomagenesis, providing a strong support for the key link between aberrant tumor nuclear shape and the survival of glioma patients. Based on these findings, lamins are proposed to be potential oncogene targets for therapeutic treatments of brain tumors.


Subject(s)
Brain Neoplasms , Glioma , Animals , Brain Neoplasms/genetics , Genomics , Glioma/genetics , Humans , Mice , Nuclear Lamina/genetics , Nuclear Lamina/metabolism , Oncogenes
7.
Front Cell Neurosci ; 15: 751439, 2021.
Article in English | MEDLINE | ID: mdl-34630045

ABSTRACT

Myelination of neuronal axons in the central nervous system (CNS) by oligodendrocytes (OLs) enables rapid saltatory conductance and axonal integrity, which are crucial for normal brain functioning. Previous studies suggested that different subtypes of oligodendrocytes in the CNS form different types of myelin determined by the diameter of axons in the unit. However, the molecular mechanisms underlying the developmental association of different types of oligodendrocytes with different fiber sizes remain elusive. In the present study, we present the evidence that the intracellular Ca2+ release channel associated receptor (Itpr2) contributes to this developmental process. During early development, Itpr2 is selectively up-regulated in oligodendrocytes coinciding with the initiation of myelination. Functional analyses in both conventional and conditional Itpr2 mutant mice revealed that Itpr2 deficiency causes a developmental delay of OL differentiation, resulting in an increased percentage of CAII+ type I/II OLs which prefer to myelinate small-diameter axons in the CNS. The increased percentage of small caliber myelinated axons leads to an abnormal compound action potentials (CAP) in the optic nerves. Together, these findings revealed a previously unrecognized role for Itpr2-mediated calcium signaling in regulating the development of different types of oligodendrocytes.

8.
Mol Brain ; 14(1): 154, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615523

ABSTRACT

Studies on the development of central nervous system (CNS) primarily rely on the use of specific molecular markers for different types of neural cells. S100B is widely being used as a specific marker for astrocytes in the CNS. However, the specificity of its expression in astrocyte lineage has not been systematically investigated and thus has remained a lingering issue. In this study, we provide several lines of molecular and genetic evidences that S100B is expressed in both protoplasmic astrocytes and myelinating oligodendrocytes. In the developing spinal cord, S100B is first expressed in the ventral neuroepithelial cells, and later in ALDH1L1+/GS+ astrocytes in the gray matter. Meanwhile, nearly all the S100B+ cells in the white matter are SOX10+/MYRF+ oligodendrocytes. Consistent with this observation, S100B expression is selectively lost in the white matter in Olig2-null mutants in which oligodendrocyte progenitor cells (OPCs) are not produced, and dramatically reduced in Myrf-conditional knockout mutants in which OPCs fail to differentiate. Similar expression patterns of S100B are observed in the developing forebrain. Based on these molecular and genetic studies, we conclude that S100B is not a specific marker for astrocyte lineage; instead, it marks protoplasmic astrocytes in the gray matter and differentiating oligodendrocytes.


Subject(s)
Astrocytes/metabolism , Gray Matter/cytology , Oligodendroglia/metabolism , Prosencephalon/growth & development , S100 Calcium Binding Protein beta Subunit/biosynthesis , Spinal Cord/growth & development , Animals , Biomarkers , Brain/growth & development , Cell Lineage , Cytoplasm/metabolism , Glial Fibrillary Acidic Protein/analysis , Glutamate-Ammonia Ligase/analysis , Mice , Myelin Sheath/physiology , Neurons/metabolism , Organ Specificity , Oxidoreductases Acting on CH-NH Group Donors/analysis , Prosencephalon/cytology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , S100 Calcium Binding Protein beta Subunit/genetics , SOXE Transcription Factors/analysis , Spinal Cord/cytology
10.
Cell Div ; 15(1): 12, 2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33292330

ABSTRACT

BACKGROUND: Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) has opened new therapeutic possibilities. However, karyotypic abnormalities detected in iPSCs compromised their utility, especially chromosomal aberrations found at early passages raised serious safety concerns. The mechanism underlying the chromosomal abnormality in early-passage iPSCs is not known. METHODS: Human dermal fibroblasts (HDFs) were stimulated with KMOS (KLF4, cMYC, OCT4 and SOX2) proteins to enhance their proliferative capacity and many vigorous clones were obtained. Clonal reprogramming was carried out by KMOS mRNAs transfection to confirm the 'chromosomal mutagenicity' of reprogramming process. Subculturing was performed to examine karyotypic stability of iPSCs after the re-establishment of stemness. And antioxidant N-acetyl-cysteine (NAC) was added to the culture medium for further confirmming the mutagenicity in the first few days of reprogramming. RESULTS: Chromosomal aberrations were found in a small percentage of newly induced iPS clones by reprogramming transcription factors. Clonal reprogramming ruled out the aberrant chromosomes inherited from rare karyotypically abnormal parental cell subpopulation. More importantly, the antioxidant NAC effectively reduced the occurrence of chromosomal aberrations at the early stage of reprogramming. Once iPS cell lines were established, they restored karyotypic stability in subsequent subculturing. CONCLUSIONS: Our results provided the first line of evidence for the 'chromosomal mutagenicity' of reprogramming process.

11.
Neurosci Bull ; 36(12): 1474-1483, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33051817

ABSTRACT

In the developing spinal cord, the majority of oligodendrocyte progenitor cells (OPCs) are induced in the ventral neuroepithelium under the control of the Sonic Hedgehog (Shh) signaling pathway, whereas a small subset of OPCs are generated from the dorsal neuroepithelial cells independent of the Shh pathway. Although dorsally-derived OPCs (dOPCs) have been shown to participate in local axonal myelination in the dorsolateral regions during development, it is not known whether they are capable of migrating into the ventral region and myelinating ventral axons. In this study, we confirmed and extended the previous study on the developmental potential of dOPCs in the absence of ventrally-derived OPCs (vOPCs). In Nestin-Smo conditional knockout (cKO) mice, when ventral oligodendrogenesis was blocked, dOPCs were found to undergo rapid amplification, spread to ventral spinal tissue, and eventually differentiated into myelinating OLs in the ventral white matter with a temporal delay, providing genetic evidence that dOPCs are capable of myelinating ventral axons in the mouse spinal cord.


Subject(s)
Axons/physiology , Oligodendrocyte Precursor Cells , Spinal Cord/cytology , Animals , Cell Differentiation , Hedgehog Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodendrocyte Precursor Cells/physiology , White Matter/cytology
12.
Cell Mol Neurobiol ; 40(4): 653-662, 2020 May.
Article in English | MEDLINE | ID: mdl-31782037

ABSTRACT

In the developing central nervous system, the terminal differentiation of oligodendrocytes (OLs) is regulated by both extrinsic and intrinsic factors. Recent studies have suggested that the Notch-Hes signaling pathway influences the maturation of oligodendrocytes in culture and during development. However, the specific Notch receptors and their downstream effectors Hes genes that are involved in oligodendrocyte maturation have not been investigated systematically. In this study, we showed that Notch1 and Notch3 are expressed in oligodendrocyte precursor cells (OPCs) during gliogenesis, and Hes5 is the major Notch downstream transcription factor that is transiently expressed in OPCs. Overexpression of Notch intracellular domain (NICD) and Hes5 proteins in embryonic chicken spinal cord suppressed both the endogenous and Sox10-induced Mbp gene expression. Unexpectedly, overexpression of NICD/Hes5 did not inhibit Sox10 induction of Olig2 expression and Myrf induced Mbp expression, suggesting the differential inhibitory effects of NICD/Hes5 signaling on Sox10 activation of myelin-related genes and early progenitor genes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Receptors, Notch/metabolism , SOXE Transcription Factors/antagonists & inhibitors , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Chickens , Gene Expression Regulation, Developmental , Mice, Knockout , Myelin Basic Protein/metabolism , Oligodendroglia/metabolism , Receptors, Notch/genetics , SOXE Transcription Factors/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Stem Cells/metabolism
13.
Glia ; 68(2): 422-434, 2020 02.
Article in English | MEDLINE | ID: mdl-31605511

ABSTRACT

Elucidation of signaling pathways that control oligodendrocyte (OL) development is a prerequisite for developing novel strategies for myelin repair in neurological diseases. Despite the extensive work outlining the importance of Hedgehog (Hh) signaling in the commitment and generation of OL progenitor cells (OPCs), there are conflicting reports on the role of Hh signaling in regulating OL differentiation and maturation. In the present study, we systematically investigated OPC specification and differentiation in genetically modified mouse models of Smoothened (Smo), an essential component of the Hh signaling pathway in vertebrates. Through conditional gain-of-function strategy, we demonstrated that hyperactivation of Smo in neural progenitors induced transient ectopic OPC generation and precocious OL differentiation accompanied by the co-induction of Olig2 and Nkx2.2. After the commitment of OL lineage, Smo activity is not required for OL differentiation, and sustained expression of Smo in OPCs stimulated cell proliferation but inhibited terminal differentiation. These findings have uncovered the stage-specific regulation of OL development by Smo-mediated Hh signaling, providing novel insights into the molecular regulation of OL differentiation and myelin repair.


Subject(s)
Hedgehog Proteins/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Homeobox Protein Nkx-2.2 , Mice, Transgenic , Myelin Sheath/metabolism , Signal Transduction/physiology , Stem Cells/metabolism
14.
Glia ; 67(9): 1654-1666, 2019 09.
Article in English | MEDLINE | ID: mdl-31038233

ABSTRACT

Notch signaling has been implicated in the inhibition of oligodendrocyte differentiation and myelin gene expression during early development. However, inactivation of a particular Notch or Hes gene only produces a mild phenotype in oligodendrocyte development possibly due to the functional redundancies among closely related family members. To uncover the full role of Notch signaling in myelin development and regeneration, we generated the Sox10rtTA/+ ; TetO-dnMAML1 double transgenic mice in which expression of dominant negative Master-mind 1 (dnMAML1) gene can be selectively induced in oligodendrocyte precursor cells (OPCs) for complete blockade of Notch signaling. It is found that dnMAML1 expression leads to robust precocious OL differentiation and premature axonal myelination in the spinal cord, possibly by upregulating Nkx2.2 and downregulating Pdgfra expression. Unexpectedly, at late embryonic stages, dnMAML1 expression dramatically increased the number of OPCs, indicating a stage-dependent effect of Notch signaling on OPC proliferation. In addition, dnMAML1 also significantly enhances axonal remyelination following chemical-induced demyelination, providing a promising therapeutic target for lesion repair in demyelinating disease.


Subject(s)
Myelin Sheath/metabolism , Nerve Regeneration/physiology , Nuclear Proteins/metabolism , Oligodendrocyte Precursor Cells/metabolism , Spinal Cord/growth & development , Spinal Cord/metabolism , Transcription Factors/metabolism , Animals , Brain/growth & development , Brain/metabolism , Cell Proliferation/physiology , Demyelinating Diseases/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Mice, Transgenic , Neurogenesis/physiology , Nuclear Proteins/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Transcription Factors/genetics , Zebrafish Proteins/metabolism
15.
Neurosci Bull ; 34(3): 527-533, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29556912

ABSTRACT

Oligodendrocytes (OLs) are myelinating glial cells that form myelin sheaths around axons to ensure rapid and focal conduction of action potentials. Here, we found that an axonal outgrowth regulatory molecule, AATYK (apoptosis-associated tyrosine kinase), was up-regulated with OL differentiation and remyelination. We therefore studied its role in OL differentiation. The results showed that AATYK knockdown inhibited OL differentiation and the expression of myelin genes in vitro. Moreover, AATYK-deficiency maintained the proliferation status of OLs but did not affect their survival. Thus, AATYK is essential for the differentiation of OLs.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/genetics , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Embryo, Mammalian , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/metabolism , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Protein-Tyrosine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley
16.
Sci Rep ; 7(1): 337, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28336932

ABSTRACT

Myelin elaborated by oligodendrocytes (OLs) in the central nervous system (CNS) is required for saltatory conduction of action potentials along neuronal axons. We found that TMEFF2, a transmembrane protein with EGF-like and two follistatin-like domains, is selectively expressed in differentiating/myelinating OLs. Previous studies showed that TMEFF2 is capable of binding to PDGFA, which plays important roles in the proliferation, migration and differentiation of oligodendrocyte progenitor cells (OPCs). However, molecular and genetic analysis revealed that Tmeff2 is a weak binder of PDGFA, and not required for OL differentiation and myelin gene expression in vivo. Together, our data suggested that Tmeff2 is specifically upregulated in OLs, but dispensable for OL differentiation and maturation.


Subject(s)
Cell Differentiation , Membrane Proteins/biosynthesis , Oligodendroglia/physiology , Animals , Gene Expression Profiling , Membrane Proteins/genetics , Mice, Knockout
17.
Neurosci Bull ; 31(5): 517-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26242484

ABSTRACT

Oligodendrocytes (OLs) are glial cells that form myelin sheaths around axons in the central nervous system (CNS). Loss of the myelin sheath in demyelinating and neurodegenerative diseases can lead to severe impairment of movement. Understanding the extracellular signals and intracellular factors that regulate OL differentiation and myelination during development can help to develop novel strategies for enhancing myelin repair in neurological disorders. Here, we report that TAPP1 was selectively expressed in differentiating OL precursor cells (OPCs). TAPP1 knockdown promoted OL differentiation and myelin gene expression in culture. Conversely, over-expression of TAPP1 in immature OPCs suppressed their differentiation. Moreover, TAPP1 inhibition in OPCs altered the expression of Erk1/2 but not AKT. Taken together, our results identify TAPP1 as an important negative regulator of OPC differentiation through the Mek/Erk signaling pathway.


Subject(s)
Cell Differentiation , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology
18.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 28(2): 165-71, 2011 Apr.
Article in Chinese | MEDLINE | ID: mdl-21462128

ABSTRACT

Mutations in the mitochondrial DNA have been found to be one of the most important causes of sensorineural hearing loss. In particular, these mutations often occur in the mitochondrial 12S rRNA and tRNA genes. Of these, the homoplasmic A1555G and C1494T mutations in the 12S rRNA have been associated with both aminoglycoside induced and nonsyndromic hearing impairment in many families worldwide. Children carrying the A1555G or C1494T mutation are susceptible to the exposure of ototoxic drugs, thereby inducing or worsening hearing loss. Individuals harboring A1555G or C1494T mutation can also develop hearing loss even in the absence of aminoglycoside exposure. However, matrilineal relatives of intra-families or inter-families carrying the A1555G or C1494T mutation exhibit a wide range of severity, age-at-onset, and audiometric configuration of hearing impairment. These indicate that the A1555G or C1494T mutation is a primary factor underlying the development of deafness but insufficient to produce the clinical phenotype.Thus, other modifier factors, such as aminoglycoside(s), mitochondria l DNA haplotype(s) or nuclear modifier gene(s), play a role in the phenotypic expression of the deafness-associated mitochondrial 12S rRNA A1555G or C1494T mutation. In this review, we summarize the modifier factors for the phenotypic expression of deafness-associated 12S rRNA A1555G and C1494T mutations and propose the molecular pathogenetic mechanism of maternally inherited deafness.


Subject(s)
DNA, Mitochondrial/genetics , Deafness/genetics , Hearing Loss, Sensorineural/genetics , Mutation , Base Sequence , Humans , Molecular Sequence Data , Phenotype
19.
Mol Genet Metab ; 100(1): 57-64, 2010 May.
Article in English | MEDLINE | ID: mdl-20153673

ABSTRACT

Mutations in mitochondrial DNA (mtDNA) have been found to be one of the most important causes of sensorineural hearing loss. We report here a clinical, genetic, molecular and biochemical characterization of a Han Chinese pedigree with maternally transmitted nonsyndromic hearing impairment. Seven of nine matrilineal relatives exhibited a variable severity and age-at-onset (8 years old) of hearing loss. Mutational analysis of mtDNA identified the novel homoplasmic tRNA(Ser(UCN)) 7505T>C mutation and other 37 variants belonging to haplogroup F1. The 7505T>C mutation, which is absent in 449 Chinese controls, is located at a highly conserved base-pairing (10A-20U) of tRNA(Ser(UCN)). The abolishment of 10A-20U base-pairing likely alters the tRNA(Ser(UCN)) metabolism. Functional significant of this mutation was supported by approximately 65% reductions in the level of tRNA(Ser(UCN)) observed in the lymphoblastoid cell lines carrying the 7505T>C mutation, compared with the wild-type cell lines. This reduced tRNA level is below the proposed threshold to support a normal respiration in lymphoblastoid cells. Furthermore, the highly conserved tRNA(Ala) 5587T>C and Cytb C93Y variants may have a modifying role of deafness expression associated with the 7505T>C mutation. However, genotyping analysis of nuclear modifier gene TRMU and the prominent deafness-cause gene GJB2 failed to detect any mutations in the member of this family. These data strongly indicate that the novel tRNA(Ser(UCN)) 7505T>C mutation is involved in maternally transmitted hearing loss. However, other genetic, epigenetic or environmental factors may contribute to the phenotypic variability of this family. Our findings will be helpful for counseling families of maternally inherited hearing loss.


Subject(s)
DNA, Mitochondrial/genetics , Hearing Loss, Sensorineural/genetics , Adolescent , Age of Onset , Asian People/genetics , Connexin 26 , Connexins/genetics , Female , Humans , Male , Mitochondrial Proteins/genetics , Mutation , Pedigree , RNA, Transfer, Amino Acyl , tRNA Methyltransferases/genetics
20.
Mitochondrion ; 10(4): 380-90, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20100600

ABSTRACT

In this report, we investigated the frequency and spectrum of mitochondrial 12S rRNA variants in a large cohort of 1642 Han Chinese pediatric subjects with aminoglycoside-induced and nonsyndromic hearing loss. Mutational analysis of 12S rRNA gene in these subjects identified 68 (54 known and 14 novel) variants. The frequencies of known 1555A>G and 1494C>T mutations were 3.96% and 0.18%, respectively, in this cohort with nonsyndromic and aminoglycoside-induced hearing loss. Prevalence of other putative deafness-associated mutation at positions 1095 and 961 were 0.61% and 1.7% in this cohort, respectively. Furthermore, the 745A>G, 792C>T, 801A>G, 839A>G, 856A>G, 1027A>G, 1192C>T, 1192C>A, 1310C>T, 1331A>G, 1374A>G and 1452T>C variants conferred increased sensitivity to ototoxic drugs or nonsyndromic deafness as they were absent in 449 Chinese controls and localized at highly conserved nucleotides of this rRNA. However, other variants appeared to be polymorphisms. Moreover, 65 Chinese subjects carrying the 1555A>G mutation exhibited bilateral and sensorineural hearing loss. A wide range of severity, age-of-onset and audiometric configuration was observed among these subjects. In particular, the sloping and flat-shaped patterns were the common audiograms in individuals carrying the 1555A>G mutation. The phenotypic variability in subjects carrying these 12S rRNA mutations indicated the involvement of nuclear modifier genes, mitochondrial haplotypes, epigenetic and environmental factors in the phenotypic manifestation of these mutations. Therefore, our data demonstrated that mitochondrial 12S rRNA is the hot spot for mutations associated with aminoglycoside ototoxicity.


Subject(s)
Aminoglycosides/adverse effects , Hearing Loss/chemically induced , Hearing Loss/genetics , RNA, Ribosomal/genetics , RNA/genetics , Adolescent , Age Factors , Aminoglycosides/therapeutic use , Asian People , Child , Child, Preschool , Female , Gene Frequency , Genotype , Humans , Infant , Male , Point Mutation , RNA, Mitochondrial , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL