Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Neurochem ; 160(2): 234-255, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34816431

RESUMEN

The nervous system monitors the environment to maintain homeostasis, which can be affected by stressful conditions. Using mammalian models of chronic stress, we previously observed altered brain levels of GPM6A, a protein involved in neuronal morphology. However, GPM6A's role in systemic stress responses remains unresolved. The nematode Caenorhabditis elegans expresses a GPM6A ortholog, the neuronal membrane glycoprotein 1 (NMGP-1). Because of the shared features between nematode and mammalian nervous systems and the vast genetic tools available in C. elegans, we used the worm to elucidate the role of GPM6A in the stress response. We first identified nmgp-1 expression in different amphid and phasmid neurons. To understand the nmgp-1 role, we characterized the behavior of nmgp-1(RNAi) animals and two nmgp-1 mutant alleles. Compared to control animals, mutant and RNAi-treated worms exhibited increased recovery time from the stress-resistant dauer stage, altered SDS chemosensation and reduced egg-laying rate resulting in egg retention (bag-of-worms phenotype). Silencing of nmgp-1 expression induced morphological abnormalities in the ASJ sensory neurons, partly responsible for dauer exit. These results indicate that nmgp-1 is required for neuronal morphology and for behaviors associated with chemosensation. Finally, we propose nmgp-1 mutants as a tool to screen drugs for human nervous system pathologies.


Asunto(s)
Adaptación Fisiológica/fisiología , Conducta Animal/fisiología , Caenorhabditis elegans/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Femenino
2.
Malays J Med Sci ; 29(2): 31-42, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35528814

RESUMEN

Background: Schizophrenia is a multifactorial disease in which genetic factors play a greater role than other factors. The genes of importance in schizophrenia patients are the genes that encode for neurotransmitters associated with low minor allele frequency (MAF) scores. This study was aimed to determine the association of genetic variations in catechol-O-methyl transferase (COMT), Ras association domain family member 1 (RASSF1) and glycoprotein M6A (GPM6A) with the risk of paranoid schizophrenia (PS) in patients admitted to Prof HB Saanin Psychiatric Hospital, West Sumatra, Indonesia. Methods: Genotyping analysis through polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and PCR-amplification refractory mutation system (ARMS) was performed in 100 PS patients and 100 healthy controls. Chi-square and Fisher's exact tests were used to compare the frequencies of genotype and allotype between the PS and control groups. Odds ratio (OR) with 95% confidence interval (95% CI) were calculated to determine the relative risk of PS with respect to genetic variations. Results: Polymorphism rs13142920 in GPM6A was associated with significantly elevated risk of PS (P = 0.020; OR = 1.60 [95% CI: 1.08, 2.39]). However, COMT rs4680 and RASSF1 rs2073499 polymorphisms were not significantly associated with PS. Conclusion: The GPM6A rs13142920 polymorphism holds great potential as a genetic marker in PS patients.

3.
J Neurosci ; 37(15): 4046-4064, 2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28275160

RESUMEN

Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. To examine how signaling protein complexes are clustered in rafts, we focused on the functions of glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing mouse neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a GPM6a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a, such as Rufy3, Rap2, and Tiam2/STEF, accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation in neuronal development. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of neuronal polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.SIGNIFICANCE STATEMENT Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. We focused on glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.


Asunto(s)
Líquido Extracelular/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microdominios de Membrana/genética , Microdominios de Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Análisis por Conglomerados , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Embarazo
4.
J Cell Physiol ; 233(1): 673-687, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28332716

RESUMEN

The class IIa histone deacetylases (HDACs) play important roles in the central nervous system during diverse biological processes such as synaptic plasticity, axon regeneration, cell apoptosis, and neural differentiation. Although it is known that HDAC5 regulates neuronal differentiation, neither the physiological function nor the regulation of HDAC5 in neuronal differentiation is clear. Here, we identify HDAC5 as an inhibitor of neurite elongation and show that HDAC5 is regulated by the brain enriched microRNA miR-124 and miR-9. We discover that HDAC5 inhibits neurite extension both in differentiated P19 cells and primary neurons. We also show that the neuronal membrane glycoprotein GPM6A (M6a) is a direct target gene of HDAC5 regulated transcriptional factor MEF2C. HDAC5 inhibits neurite elongation, acting at least partially via a MEF2C/M6a signaling pathway. We also confirmed the miR-124/miR-9 regulated HDAC5-MEF2C-M6a pathway regulates neurite development in primary neurons. Thus, HDAC5 emerges as a cellular conductor of MEF2C and M6a activity and is regulated by miR-124 and miR-9 to control neurite development.


Asunto(s)
Células Madre Embrionarias/enzimología , Histona Desacetilasas/metabolismo , Glicoproteínas de Membrana/metabolismo , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/enzimología , Neuritas/enzimología , Neurogénesis , Animales , Regulación hacia Abajo , Células Madre Embrionarias/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Edad Gestacional , Células HEK293 , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Humanos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/efectos de los fármacos , Neuritas/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Transducción de Señal , Transfección
5.
J Hepatol ; 64(5): 1137-1146, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26806818

RESUMEN

BACKGROUND & AIMS: Contribution of hepatic stellate cells (HSCs), portal fibroblasts (PFs), and mesothelial cells (MCs) to myofibroblasts is not fully understood due to insufficient availability of markers and isolation methods. The present study aimed to isolate these cells, characterize their phenotypes, and examine their contribution to myofibroblasts in liver fibrosis. METHODS: Liver fibrosis was induced in Collagen1a1-green fluorescent protein (Col1a1(GFP)) mice by bile duct ligation (BDL), 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet, or CCl4 injections. Combining vitamin A (VitA) lipid autofluorescence and expression of GFP and glycoprotein M6a (GPM6A), we separated HSCs, PFs, and MCs from normal and fibrotic livers by fluorescence-activated cell sorting (FACS). RESULTS: Normal Col1a1(GFP) livers broadly expressed GFP in HSCs, PFs, and MCs. Isolated VitA+ HSCs expressed reelin, whereas VitA-GFP+GPM6A- PFs expressed ectonucleoside triphosphate diphosphohydrolase-2 and elastin. VitA-GFP+GPM6A+ MCs expressed keratin 19, mesothelin, and uroplakin 1b. Transforming growth factor (TGF)-ß1 treatment induced the transformation of HSCs, PFs, and MCs into myofibroblasts in culture. TGF-ß1 suppressed cyclin D1 mRNA expression in PFs but not in HSCs and MCs. In biliary fibrosis, PFs adjacent to the bile duct expressed α-smooth muscle actin. FACS analysis revealed that HSCs are the major source of GFP+ myofibroblasts in the injured Col1a1(GFP) mice after DDC or CCl4 treatment. Although PFs partly contributed to GFP+ myofibroblasts in the BDL model, HSCs were still dominant source of myofibroblasts. CONCLUSION: HSCs, PFs, and MCs have distinct phenotypes, and PFs partly contribute to myofibroblasts in the portal triad in biliary fibrosis.


Asunto(s)
Epitelio/patología , Células Estrelladas Hepáticas/patología , Cirrosis Hepática Experimental/patología , Hígado/patología , Vena Porta/patología , Animales , Células Cultivadas , Fibroblastos/patología , Citometría de Flujo , Mesotelina , Ratones , Proteína Reelina
6.
Cereb Cortex ; 25(11): 4111-25, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24917275

RESUMEN

The function of mature neurons critically relies on the developmental outgrowth and projection of their cellular processes. It has long been postulated that the neuronal glycoproteins M6a and M6b are involved in axon growth because these four-transmembrane domain-proteins of the proteolipid protein family are highly enriched on growth cones, but in vivo evidence has been lacking. Here, we report that the function of M6 proteins is required for normal axonal extension and guidance in vivo. In mice lacking both M6a and M6b, a severe hypoplasia of axon tracts was manifested. Most strikingly, the corpus callosum was reduced in thickness despite normal densities of cortical projection neurons. In single neuron tracing, many axons appeared shorter and disorganized in the double-mutant cortex, and some of them were even misdirected laterally toward the subcortex. Probst bundles were not observed. Upon culturing, double-mutant cortical and cerebellar neurons displayed impaired neurite outgrowth, indicating a cell-intrinsic function of M6 proteins. A rescue experiment showed that the intracellular loop of M6a is essential for the support of neurite extension. We propose that M6 proteins are required for proper extension and guidance of callosal axons that follow one of the most complex trajectories in the mammalian nervous system.


Asunto(s)
Corteza Cerebral/citología , Cuerpo Calloso/citología , Cuerpo Calloso/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/fisiología , Neuronas/citología , Factores de Edad , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/fisiología , Embrión de Mamíferos , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Técnicas In Vitro , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Proteolipídica de la Mielina/deficiencia , Proteína Proteolipídica de la Mielina/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo
7.
Hum Mutat ; 35(12): 1495-505, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25224183

RESUMEN

Glycoprotein M6A (GPM6A) is a neuronal transmembrane protein of the PLP/DM20 (proteolipid protein) family that associates with cholesterol-rich lipid rafts and promotes filopodia formation. We identified a de novo duplication of the GPM6A gene in a patient with learning disability and behavioral anomalies. Expression analysis in blood lymphocytes showed increased GPM6A levels. An increase of patient-derived lymphoblastoid cells carrying membrane protrusions supports a functional effect of this duplication. To study the consequences of GPM6A dosage alterations in an intact nervous system, we employed Drosophila melanogaster as a model organism. We found that knockdown of Drosophila M6, the sole member of the PLP family in flies, in the wing, and whole organism causes malformation and lethality, respectively. These phenotypes as well as the protrusions of patient-derived lymphoblastoid cells with increased GPM6A levels can be alleviated by cholesterol supplementation. Notably, overexpression as well as loss of M6 in neurons specifically compromises long-term memory in the courtship conditioning paradigm. Our findings thus indicate a critical role of correct GPM6A/M6 levels for cognitive function and support a role of the GPM6A duplication for the patient's phenotype. Together with other recent findings, this study highlights compromised cholesterol homeostasis as a recurrent feature in cognitive phenotypes.


Asunto(s)
Colesterol/fisiología , Trastornos del Conocimiento/genética , Dosificación de Gen , Glicoproteínas de Membrana/genética , Proteínas del Tejido Nervioso/genética , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Duplicación de Gen , Humanos , Hibridación Fluorescente in Situ , Locomoción , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Conducta Sexual Animal
8.
Cell Rep ; 43(9): 114762, 2024 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-39321020

RESUMEN

Adult mammary stem cells (aMaSCs) are vital to tissue expansion and remodeling during the process of postnatal mammary development. The protein C receptor (Procr) is one of the well-identified surface markers of multipotent aMaSCs. However, an understanding of the regulatory mechanisms governing Procr's protein stability remains incomplete. In this study, we identified Glycoprotein m6a (Gpm6a) as a critical protein for aMaSC activity modulation by using the Gpm6a knockout mouse model. Interestingly, we determined that Gpm6a depletion results in a reduction of Procr protein stability. Mechanistically, Gpm6a regulates Procr protein stability by mediating the formation of lipid rafts, a process requiring Zdhhc1 and Zdhhc2 to palmitate Gpm6a at Cys17,18 and Cys246 sites. Our findings highlight an important mechanism involving Zdhhc1- and Zdhhc2-mediated Gpm6a palmitoylation for the regulation of Procr stability, aMaSC activity, and postnatal mammary development.


Asunto(s)
Aciltransferasas , Lipoilación , Glándulas Mamarias Animales , Animales , Aciltransferasas/metabolismo , Aciltransferasas/genética , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/citología , Ratones , Femenino , Ratones Noqueados , Humanos , Microdominios de Membrana/metabolismo , Células Madre/metabolismo , Células Madre/citología , Estabilidad Proteica
9.
Biomolecules ; 13(4)2023 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-37189342

RESUMEN

The peripheral nervous system (PNS) has a unique ability for self-repair. Dorsal root ganglion (DRG) neurons regulate the expression of different molecules, such as neurotrophins and their receptors, to promote axon regeneration after injury. However, the molecular players driving axonal regrowth need to be better defined. The membrane glycoprotein GPM6a has been described to contribute to neuronal development and structural plasticity in central-nervous-system neurons. Recent evidence indicates that GPM6a interacts with molecules from the PNS, although its role in DRG neurons remains unknown. Here, we characterized the expression of GPM6a in embryonic and adult DRGs by combining analysis of public RNA-seq datasets with immunochemical approaches utilizing cultures of rat DRG explants and dissociated neuronal cells. M6a was detected on the cell surfaces of DRG neurons throughout development. Moreover, GPM6a was required for DRG neurite elongation in vitro. In summary, we provide evidence on GPM6a being present in DRG neurons for the first time. Data from our functional experiments support the idea that GPM6a could contribute to axon regeneration in the PNS.


Asunto(s)
Axones , Ganglios Espinales , Ratas , Animales , Axones/metabolismo , Ganglios Espinales/metabolismo , Células Cultivadas , Regeneración Nerviosa , Neuronas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proyección Neuronal
10.
Am J Cancer Res ; 13(3): 1118-1127, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37034204

RESUMEN

Different ethnic groups have different incidence rate of hepatocellular carcinoma (HCC). In addition to lifestyle and environmental factors, genetic susceptibility is also an important reason. In this study, we screened the immune related genes and stromal related genes in White and Asian liver cancer patients cohort of The Cancer Genome Atlas (TCGA) the using ESTIMATE algorithm. Hub genes that significantly associated with overall survival (OS) were selected from White and Asian liver cancer patients, respectively. In addition, we validated the functions of two hub genes, IL-18RAP and GPM6A, in vivo and in vitro. We confirmed different races have different tumor immune microenvironments. Immune microenvironment can influence and change the efficacy of immunotherapy for liver cancer patients.

11.
Cells ; 11(14)2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35883571

RESUMEN

Systematic recurrence of glioblastoma (GB) despite surgery and chemo-radiotherapy is due to GB stem cells (GBSC), which are particularly invasive and radioresistant. Therefore, there is a need to identify new factors that might be targeted to decrease GBSC invasive capabilities as well as radioresistance. Patient-derived GBSC were used in this study to demonstrate a higher expression of the glycoprotein M6a (GPM6A) in invasive GBSC compared to non-invasive cells. In 3D invasion assays performed on primary neurospheres of GBSC, we showed that blocking GPM6A expression by siRNA significantly reduced cell invasion. We also demonstrated a high correlation of GPM6A with the oncogenic protein tyrosine phosphatase, PTPRZ1, which regulates GPM6A expression and cell invasion. The results of our study also show that GPM6A and PTPRZ1 are crucial for GBSC sphere formation. Finally, we demonstrated that targeting GPM6A or PTPRZ1 in GBSC increases the radiosensitivity of GBSC. Our results suggest that blocking GPM6A or PTPRZ1 could represent an interesting approach in the treatment of glioblastoma since it would simultaneously target proliferation, invasion, and radioresistance.


Asunto(s)
Glioblastoma , Glicoproteínas de Membrana , Proteínas del Tejido Nervioso , Glioblastoma/genética , Glioblastoma/radioterapia , Humanos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/efectos de la radiación , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/efectos de la radiación , Tolerancia a Radiación , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores
12.
Prog Retin Eye Res ; 91: 101091, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35729001

RESUMEN

Albinism is a pigment disorder affecting eye, skin and/or hair. Patients usually have decreased melanin in affected tissues and suffer from severe visual abnormalities, including foveal hypoplasia and chiasmal misrouting. Combining our data with those of the literature, we propose a single functional genetic retinal signalling pathway that includes all 22 currently known human albinism disease genes. We hypothesise that defects affecting the genesis or function of different intra-cellular organelles, including melanosomes, cause syndromic forms of albinism (Hermansky-Pudlak (HPS) and Chediak-Higashi syndrome (CHS)). We put forward that specific melanosome impairments cause different forms of oculocutaneous albinism (OCA1-8). Further, we incorporate GPR143 that has been implicated in ocular albinism (OA1), characterised by a phenotype limited to the eye. Finally, we include the SLC38A8-associated disorder FHONDA that causes an even more restricted "albinism-related" ocular phenotype with foveal hypoplasia and chiasmal misrouting but without pigmentation defects. We propose the following retinal pigmentation pathway, with increasingly specific genetic and cellular defects causing an increasingly specific ocular phenotype: (HPS1-11/CHS: syndromic forms of albinism)-(OCA1-8: OCA)-(GPR143: OA1)-(SLC38A8: FHONDA). Beyond disease genes involvement, we also evaluate a range of (candidate) regulatory and signalling mechanisms affecting the activity of the pathway in retinal development, retinal pigmentation and albinism. We further suggest that the proposed pigmentation pathway is also involved in other retinal disorders, such as age-related macular degeneration. The hypotheses put forward in this report provide a framework for further systematic studies in albinism and melanin pigmentation disorders.


Asunto(s)
Albinismo , Melaninas , Humanos , Melaninas/genética , Melaninas/metabolismo , Mutación , Albinismo/genética , Retina/metabolismo , Pigmentación/genética
13.
Int J Biol Sci ; 18(2): 637-651, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35002514

RESUMEN

Background: Circular RNAs (circRNAs), which generally act as microRNA (miRNA) sponges to competitively regulate the downstream target genes of miRNA, play an essential role in cancer biology. However, few studies have been reported on the role of circRNA based competitive endogenous RNA (ceRNA) network in hepatocellular carcinoma (HCC). Herein, we aimed to screen and establish the circRNA/miRNA/mRNA networks related to the prognosis and progression of HCC and further explore the underlying mechanisms of tumorigenesis. Methods: GEO datasets GSE97332, GSE108724, and GSE101728 were utilized to screen the differentially expressed circRNAs (DE-circRNAs), DE-miRNAs, and DEmRNAs between HCC and matched para-carcinoma tissues. After six RNA-RNA predictions and five intersections between DE-RNAs and predicted RNAs, the survival-related RNAs were screened by the ENCORI analysis tool. The ceRNA networks were constructed using Cytoscape software, based on two models of up-regulated circRNA/down-regulated miRNA/up-regulated mRNA and down-regulated circRNA/up-regulated miRNA/down-regulated mRNA. The qRT-PCR assay was utilized for detecting the RNA expression levels in HCC cells and tissues. The apoptosis, Edu, wound healing, and transwell assays were performed to evaluate the effect of miR-106b-5p productions on the proliferation, invasion, and metastasis of HCC cells. In addition, the clone formation, cell cycle, and nude mice xenograft tumor assays were used to investigate the influence of hsa_circ_0001495 (circCCNB1) silencing and overexpression on the proliferation of HCC cells in vitro and in vivo. Furthermore, the mechanism of downstream gene DYNC1I1 and AKT/ERK signaling pathway via the circCCNB1/miR-106b-5p/GPM6A network in regulating the cell cycle was also explored. Results: Twenty DE-circRNAs with a genomic length less than 2000bp, 11 survival-related DE-miRNAs, and 61 survival-related DE-mRNAs were screened out and used to construct five HCC related ceRNA networks. Then, the circCCNB1/miR-106b-5p/GPM6A network was randomly selected for subsequent experimental verification and mechanism exploration at in vitro and in vivo levels. The expression of circCCNB1 and GPM6A were significantly down-regulated in HCC cells and cancer tissues, while miR-106b-5p expression was up-regulated. After transfections, miR-106b-5p mimics notably enhanced the proliferation, invasion, and metastasis of HCC cells, while the opposite was seen with miR-105b-5p inhibitor. In addition, circCCNB1 silencing promoted the clone formation ability, the cell cycle G1-S transition, and the growth of xenograft tumors of HCC cells via GPM6A downregulation. Subsequently, under-expression of GPM6A increased DYNC1I1 expression and activated the phosphorylation of the AKT/ERK pathway to regulate the HCC cell cycle. Conclusions: We demonstrated that circCCNB1 silencing promoted cell proliferation and metastasis of HCC cells by weakening sponging of oncogenic miR-106b-5p to induce GPM6A underexpression. DYNC1I1 gene expression was up-regulated and further led to activation of the AKT/ERK signaling pathway.


Asunto(s)
Carcinoma Hepatocelular/genética , Ciclina B1/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , ARN Circular/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Dineínas Citoplasmáticas/metabolismo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Biomol Struct Dyn ; 39(8): 2693-2701, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32248748

RESUMEN

Glycoprotein M6A, a stress related gene, plays an important role in synapse and filopodia formation. Filopodia formation is vital for development, immunity, angiogenesis, wound healing and metastasis. In this study, structural and functional analysis of high-risk SNPs associated with Glycoprotein M6-A were evaluated using six different bioinformatics tools. Results classified T210I, T134I, Y153H, I215T, F156L, T160I, I226T, R247W, R178C, W159R, N157S and P151L as deleterious mutants that are crucial for the structure and function of the protein causing malfunction of M6-a and ultimately leads to disease development. The three-dimensional structure of wild-type M6-a and mutant M6-a were also predicted. Furthermore, the effects of high risk substitutions were also analyzed with interaction with valproic acid. Based on structural models obtained, the binding pocket of ligand bound glycoprotein M6-A structure showed few core interacting residues which are different in the mutant models. Among all substitutions, F156L showed complete loss of binding pocket when interacting with valproic acid as compared to the wild type model. Up to the best of our knowledge this is the first comprehensive study where GPM6A mutations were analyzed. The mechanism of action of GPM6A is still not fully defined which limits the understanding of functional details encoding M6-A. Our results may help enlighten some molecular aspects underlying glycoprotein M6-A. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Proteínas del Tejido Nervioso , Polimorfismo de Nucleótido Simple , Humanos , Glicoproteínas de Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética
15.
Front Synaptic Neurosci ; 13: 661681, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34017241

RESUMEN

The cellular and molecular mechanisms underlying neuropsychiatric and neurodevelopmental disorders show that most of them can be categorized as synaptopathies-or damage of synaptic function and plasticity. Synaptic formation and maintenance are orchestrated by protein complexes that are in turn regulated in space and time during neuronal development allowing synaptic plasticity. However, the exact mechanisms by which these processes are managed remain unknown. Large-scale genomic and proteomic projects led to the discovery of new molecules and their associated variants as disease risk factors. Neuronal glycoprotein M6a, encoded by the GPM6A gene is emerging as one of these molecules. M6a has been involved in neuron development and synapse formation and plasticity, and was also recently proposed as a gene-target in various neuropsychiatric disorders where it could also be used as a biomarker. In this review, we provide an overview of the structure and molecular mechanisms by which glycoprotein M6a participates in synapse formation and maintenance. We also review evidence collected from patients carrying mutations in the GPM6A gene; animal models, and in vitro studies that together emphasize the relevance of M6a, particularly in synapses and in neurological conditions.

16.
Genes Dis ; 8(4): 521-530, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34179314

RESUMEN

Colorectal carcinoma (CRC) is one of the most common cancers, and is associated with a poor clinical outcome. The key genes and potential prognostic markers in colorectal carcinoma remain to be identified and explored for clinical application. DNA expression/methylation profiles were downloaded from the Gene Expression Omnibus (GEO) database to identify differentially expressed/methylated genes (DEGs and DEMs). A total of 255 genes and 372 genes were identified as being up-regulated and down-regulated, respectively, in GSE113513, GSE81558, and GSE89076. There were a total of 3350 hypermethylated genes and 443 hypomethylated genes identified in GSE48684. Twenty genes were found to be hypermethylated as well as down-regulated, and a functional enrichment analysis revealed that these genes were mainly involved in cancer-related pathways. Among these 20 genes, GPM6A, HAND2 and C2orf40 were related to poor outcomes in cancer patients based on a survival analysis. Concurrent decreases of GPM6A, HAND2 and C2orf40 protein expression were observed in highly-differentiated colorectal carcinoma tissues, and higher expression levels were found in undifferentiated or minimally-differentiated colorectal carcinoma tissues. In conclusion, 20 genes were found to be downregulated and hypermethylated in CRC, among which GPM6A, HAND2 and C2orf40 were explored for their potential prognostic value.

17.
Journal of Medical Research ; (12): 89-92,133, 2023.
Artículo en Zh | WPRIM | ID: wpr-1023575

RESUMEN

Objective To investigate the expression of glycoprotein M6A(GPM6A)in glioma specimens and its prognostic signifi-cance.Methods Bioinformatics methods were used to analyze the expression of GPM6A in 325 cases obtained from the Chinese Glioma Genome Atlas(CGGA)database.The STRING online database was used to screen and analyze the possible protein PPI interaction net-work and GO functional annotation that interact with GPM6A to explore its potential mechanism.The relationships between GPM6A and glioma cell purity and the level of CD8+T cell were assessed using the TIMER2.0database.Results The online analysis of the CGGA database showed that GPM6A was expressed in histopathological grades(WHO:G2,G3,G4)and histopathological types,and there were significant differences in GPM6A expression levels between all grades and all types of gliomas(Grades:F=50.25,P<0.001;Types:F=14.26,P<0.001);the expression level of GPM6A was closely related to molecular IDH mutation status,combined deletion of 1p19q,and patient age(all P<0.01),regardless of patient gender(t=1.45,P>0.05).The expression level of GPM6A in primary glioma was higher than that in secondary glioma,and the difference was not statistically significant(t=1.82,P>0.05).The survival time of patients with low GPM6A expression was significantly shortened(x2=69.79,P<0.001),but in glioblastoma(GBM)patients,regard-less of the level of GPM6A expression,there was no difference in survival time(x2=0.63,P>0.05).GO functional analysis showed that GPM6A was closely related to the functions of transmembrane protein transport,synapse formation and development,and information ex-change between cells in the nervous system.TIMER2.0database analysis showed that GPM6A was positively correlated with glioma tumor cell purity and the level of CD8+T cell.Conclusion The prognosis of glioma patients with low GPM6A expression is significantly worse than that of patients with high expression,and its expression level can be used as a predictor for evaluating the prognosis of glioma.

18.
Neurosci Res ; 128: 19-24, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29158160

RESUMEN

Neuronal development is composed of the complex steps which involve many signaling proteins. On the other hand, there are many proteins highly expressed in the differentiated neurons at developmental stages, but of which physiological functions are not precisely known so far. Glycoprotein 6a (GPM6a) currently belongs to such proteins. This protein has four-transmembrane domains and is a member of proteolipid protein family. Recently, we demonstrated that GPM6a is highly concentrated in lipid rafts of the developing neuron with its palmitoylation, and that this molecule is involved in rapid determination of the neuronal polarity, in response to laminin, an extracellular matrix protein (Honda et al., J Neurosci 37: 4046-64 [2017]). Since lipid rafts are membrane domains enriched in sphingolipids and cholesterol, have lower fluidity than other membrane areas and are believed to be signaling "hot-spots", and here, we discuss the functions of this protein in neuronal lipid raft signaling for neuronal development.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Microdominios de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Animales , Humanos , Lipoilación/fisiología
19.
J Mol Neurosci ; 65(3): 301-311, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29931501

RESUMEN

Prenatal stress (PS) induces molecular changes that alter neural connectivity, increasing the risk for neuropsychiatric disorders. Here we analyzed -in the hippocampus of adult rats exposed to PS- the epigenetic signature mediating the PS-induced neuroplasticity changes. Furthermore, using cultured hippocampal neurons, we investigated the effects on neuroplasticity of an epigenetic modulator. PS induced significant modifications in the mRNA levels of stress-related transcription factor MEF2A, SUV39H1 histone methyltransferase, and TET1 hydroxylase, indicating that PS modifies gene expression through chromatin remodeling. In in vitro analysis, histone acetylation inhibition with apicidin increased filopodium density, suggesting that the external regulation of acetylation levels might modulate neuronal morphology. These results offer a way to enhance neural connectivity that could be considered to revert PS effects.


Asunto(s)
Epigénesis Genética , Código de Histonas , Plasticidad Neuronal , Efectos Tardíos de la Exposición Prenatal/genética , Estrés Psicológico/genética , Animales , Células Cultivadas , Dioxigenasas/genética , Dioxigenasas/metabolismo , Femenino , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Masculino , Metiltransferasas/genética , Metiltransferasas/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Péptidos Cíclicos/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología
20.
Behav Brain Res ; 277: 254-63, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24768641

RESUMEN

The neuronal tetraspan proteins, M6A (Gpm6a) and M6B (Gpm6b), belong to the family of proteolipids that are widely expressed in the brain. We recently reported Gpm6a deficiency as a monogenetic cause of claustrophobia in mice. Its homolog proteolipid, Gpm6b, is ubiquitously expressed in neurons and oligodendrocytes. Gpm6b is involved in neuronal differentiation and myelination. It interacts with the N-terminal domain of the serotonin transporter (SERT) and decreases cell-surface expression of SERT. In the present study, we employed Gpm6b null mutant mice (Gpm6b(-/-)) to search for behavioral functions of Gpm6b. We studied male and female Gpm6b(-/-) mice and their wild-type (WT, Gpm6b(+/+)) littermates in an extensive behavioral test battery. Additionally, we investigated whether Gpm6b(-/-) mice exhibit changes in the behavioral response to a 5-HT2A/C receptor agonist. We found that Gpm6b(-/-) mice display completely normal sensory and motor functions, cognition, as well as social and emotionality-like (anxiety, depression) behaviors. On top of this inconspicuous behavioral profile, Gpm6b(-/-) mice of both genders exhibit a selective impairment in prepulse inhibition of the acoustic startle response. Furthermore, in contrast to WT mice that show the typical locomotion suppression and increase in grooming activity after intraperitoneal administration of DOI [(±)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane hydrochloride], Gpm6b(-/-) mice demonstrate a blunted behavioral response to this 5-HT2A/C receptor agonist. To conclude, Gpm6b deficiency impairs sensorimotor gating and modulates the behavioral response to a serotonergic challenge.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Filtrado Sensorial/fisiología , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Animales , Ansiedad/tratamiento farmacológico , Encéfalo/metabolismo , Femenino , Masculino , Glicoproteínas de Membrana/deficiencia , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Receptor de Serotonina 5-HT2A/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA