Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cancer Cell Int ; 20: 403, 2020.
Article in English | MEDLINE | ID: mdl-32855620

ABSTRACT

BACKGROUND: Long non-coding RNA (lncRNA) BM742401 is a tumor suppressor in gastric cancer and chronic lymphocytic leukemia. As the promoter and coding region of BM742401 are fully embedded in a CpG island, we hypothesized that BM742401 is a tumor suppressor lncRNA epigenetically silenced by promoter DNA methylation in multiple myeloma. METHODS: Methylation-specific PCR and quantitative bisulfite pyrosequencing were performed to detect the methylation of BM742401 in normal plasma cells, myeloma cell lines and primary myeloma samples. The expression of BM742401 was measured by qRT-PCR. The function of BM742401 in multiple myeloma cells was analyzed by lentivirus transduction followed by migration assay. RESULTS: BM742401 methylation was detected in 10 (66.7%) myeloma cell lines but not normal plasma cells, and inversely correlated with expression of BM742401. In primary samples, BM742401 methylation was detected in 3 (12.5%) monoclonal gammopathy of undetermined significance, 9 (15.8%) myeloma at diagnosis and 8 (17.0%) myeloma at relapse/progression. Moreover, BM742401 methylation at diagnosis was associated with inferior overall survival (median OS: 25 vs. 39 months; P = 0.0496). In myeloma cell line JJN-3, stable overexpression of BM742401 by lentivirus transduction resulted in reduced cell migration (P = 0.0001) but not impacting cell death or proliferation. CONCLUSIONS: This is the first report of tumor-specific methylation-mediated silencing of BM742401 in myeloma, which is likely an early event in myelomagenesis with adverse impact on overall survival. Moreover, BM742401 is a tumor suppressor lncRNA by inhibiting myeloma cell migration, hence implicated in myeloma plasma cell homing, metastasis and disease progression.

2.
Oncotarget ; 8(42): 71911-71923, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069756

ABSTRACT

Endoplasmic reticulum (ER) chaperone Prolyl 4-hydroxylase, beta polypeptide (P4HB) has previously been identified as a novel target for chemoresistance in glioblastoma multiforme (GBM). Yet its functional roles in glioma carcinogenesis remain elusive. In clinical analysis using human glioma specimens and Gene Expression Omnibus (GEO) profiles, we found that aberrant expression of P4HB was correlated with high-grade malignancy and an angiogenic phenotype in glioma. Furthermore, P4HB upregulation conferred malignant characteristics including proliferation, invasion, migration and angiogenesis in vitro, and increased tumor growth in vivo via the mitogen-activated protein kinase (MAPK) signaling pathway. Pathway analysis suggested genetic and pharmacologic inhibition of P4HB suppressed MAPK expression and its downstream targets were involved in angiogenesis and invasion. This is the first study that demonstrates the oncogenic roles of P4HB and its underlying mechanism in glioma. Since tumor invasion and Vascularisation are typical hallmarks in malignant glioma, our findings uncover a promising anti-glioma mechanism through P4HB-mediated retardation of MAPK signal transduction.

3.
Hum Mol Genet ; 26(16): 3202-3211, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28575308

ABSTRACT

The functional architecture of the human brain is greatly determined by the temporal and spatial regulation of the transcription process. However, the spatial and temporal transcriptional landscape of long non-coding RNAs (lncRNAs) during human brain development remains poorly understood. Here, we report the genome-wide lncRNA transcriptional analysis in an extensive series of 1340 post-mortem human brain specimens collected from 16 regions spanning the period from early embryo development to late adulthood. We discovered that lncRNA transcriptome dramatically changed during fetal development, while transited to a surprisingly relatively stable state after birth till the late adulthood. We also discovered that the transcription map of lncRNAs was spatially different, and that this spatial difference was developmentally regulated. Of the 16 brain regions explored (cerebellar cortex, thalamus, striatum, amygdala, hippocampus and 11 neocortex areas), cerebellar cortex showed the most distinct lncRNA expression features from all remaining brain regions throughout the whole developmental period, reflecting its unique developmental and functional features. Furthermore, by characterizing the functional modules and cellular processes of the spatial-temporal dynamic lncRNAs, we found that they were significantly associated with the RNA processing, neuron differentiation and synaptic signal transportation processes. Furthermore, we found that many lncRNAs associated with the neurodegenerative Alzheimer and Parkinson diseases were co-expressed in the fetal development of the human brain, and affected the convergent biological processes. In summary, our study provides a comprehensive map for lncRNA transcription dynamics in human brain development, which might shed light on the understanding of the molecular underpinnings of human brain function and disease.


Subject(s)
Brain/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Autopsy , Brain/metabolism , Embryonic Development/genetics , Gene Expression Regulation , Genome, Human , Humans , Regulatory Elements, Transcriptional , Spatio-Temporal Analysis , Transcriptome
4.
Target Oncol ; 12(3): 353-363, 2017 06.
Article in English | MEDLINE | ID: mdl-28493025

ABSTRACT

BACKGROUND: The long non-coding RNA CRNDE has emerged as an important regulator in carcinogenesis and cancer progression. While CRNDE has previously been found to be the most highly upregulated lncRNA in glioma, detailed information on its roles in regulating cancer cell growth remains limited. OBJECTIVE: In the present study, we aimed at exploring the functional roles and underlying mechanisms of CRNDE in glioma. METHODS: We applied microarray data analysis to determine the prognostic significance of CRNDE in glioma patients and its correlation with epidermal growth factor receptor (EGFR) activation. EGFR inhibition was used to confirm the role of EGFR in regulating CRNDE expression. Functional studies were performed upon CRNDE silencing to explore its role in gliomagenesis. RESULTS: We confirm that CRNDE acts as an oncogene that is highly up-regulated in glioma, and high CRNDE expression correlates with poor prognosis in glioma patients. We further demonstrate that the expression of CRNDE correlates with EGFR activation. EGF and EGFR tyrosine kinase inhibitor (TKI) enhance and block the up-regulation of CRNDE expression, respectively, suggesting that EGFR signaling may positively regulate CRNDE expression. Functional assays show that CRNDE depletion inhibits glioma cell growth both in vitro and in vivo, and is associated with induced cellular apoptosis with decreased Bcl2/Bax ratio. CONCLUSIONS: Our findings suggest that the aberrant expression of CRNDE may be mediated by activated EGFR signaling and play significant roles in gliomagenesis.


Subject(s)
ErbB Receptors/metabolism , Glioma/genetics , RNA, Long Noncoding/genetics , Animals , Carcinogenesis , Cell Growth Processes , Cell Line, Tumor , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Glioma/diagnosis , Glioma/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Staging , Prognosis , RNA, Small Interfering/genetics , Signal Transduction , Up-Regulation
5.
J Neurooncol ; 132(3): 393-400, 2017 05.
Article in English | MEDLINE | ID: mdl-28293765

ABSTRACT

The chemotherapeutic agent temozolomide (TMZ) is widely used in the treatment of glioblastoma multiforme (GBM). Rutin, a citrus flavonoid ecglycoside found in edible plants, has neuroprotective and anticancer activities. This study aimed to investigate the efficacy and the underlying mechanisms of rutin used in combination with TMZ in GBM. In vitro cell viability assay demonstrated that rutin alone had generally low cytotoxic effect, but it enhanced the efficacy of TMZ in a dose-dependent manner. Subcutaneous and orthotopic xenograft studies also showed that tumor volumes were significantly lower in mice receiving combined TMZ/Rutin treatment as compared to TMZ or rutin alone treatment. Moreover, immunoblotting analysis showed that TMZ activated JNK activity to induce protective response autophagy, which was blocked by rutin, resulting in decreased autophagy and increased apoptosis, suggesting that rutin enhances TMZ efficacy both in vitro and in vivo via inhibiting JNK-mediated autophagy in GBM. The combination rutin with TMZ may be a potentially useful therapeutic approach for GBM patient.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Brain Neoplasms/pathology , Glioblastoma/pathology , Rutin/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Synergism , Humans , Mice , Mice, Nude , Temozolomide , Xenograft Model Antitumor Assays
6.
Sci Rep ; 7: 41224, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28106169

ABSTRACT

Corneal wound healing involves a complex cascade of cytokine-controlled cellular events, including inflammatory and angiogenesis responses that are regulated by transcriptional chromatin remodeling. Nuclear Ubiquitous Casein and cyclin-dependent Kinase Substrate (NUCKS) is a key chromatin modifier and transcriptional regulator of metabolic signaling. In this study, we investigated the role of NUCKS in corneal wound healing by comparing its effects on corneal alkali burn in NUCKS knockout (NKO) and NUCKS wild-type (NWT) mice. Our data showed that following alkali-injury, inhibition of NUCKS (NKO) accelerated ocular resurfacing and suppressed neovascularization; the cytokine profile of alkali burned corneas in NKO mice showed suppressed expression of inflammation cytokines (IL1A &IL1B); upregulated expression of antiangiogenic factor (Pigment Epithelium-derived Factor; PEDF); and downregulated expression of angiogenic factor (Vascular Endothelial Growth Factor, VEGF); in vitro, following LPS-induced NFκB activation, NKO corneal cells showed reduced expression of IL6, IP10 and TNFα. In vitro, corneal epithelial cells showed reduced NF-κb activation on silencing of NUCKS and corresponding NFκB-mediated cytokine expression was reduced. Here, we illustrate that inhibition of NUCKS played a role in cytokine modulation and facilitated corneal recovery. This reveals a potential new effective strategy for ocular burn treatment.


Subject(s)
Burns, Chemical , Corneal Injuries/chemically induced , Eye Burns/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Animals , Cells, Cultured , Corneal Injuries/genetics , Corneal Injuries/metabolism , Cytokines/metabolism , Disease Models, Animal , Epithelium, Corneal/cytology , Epithelium, Corneal/metabolism , Eye Burns/chemically induced , Eye Burns/metabolism , Eye Proteins/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Mice , NF-kappa B/metabolism , Nerve Growth Factors/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Serpins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Wound Healing
7.
Anticancer Res ; 35(12): 6487-95, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26637861

ABSTRACT

BACKGROUND: Glioma stem cells are associated for temozolomide-resistance in glioblastoma. Adducin 3 (ADD3) is a cytoskeletal protein associated with chemoresistance but its role in glioblastoma has not been investigated. MATERIALS AND METHODS: Using an in vitro model of glioblastoma cells with acquired temozolomide resistance (D54-MG-R), the expressions of ADD3 and cancer stem cell markers were compared to those in temozolomide-sensitive glioblastoma cells (D54-MG-S). Immunofluorescence staining was used to investigate the expression patterns of ADD3 and cancer stem cell markers in temozolomide resistance and neurospheres of glioblastoma. RESULTS: Chemoresistant cells were found to have up-regulation of ADD3 and CD133 expression. A sub-population of D54-MG-R cells and glioma neurospheres exhibited coexpression of ADD3 with CD133. CONCLUSION: To our knowledge, this is the first report of a possible link between cytoskeletal protein expression, cancer stem cell phenotype and temozolomide resistance in human glioblastoma. This report lays the foundation for further investigation for ADD3 as a potential biomarker and therapeutic target in temozolomide-resistant glioma cells.


Subject(s)
Antigens, CD/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Calmodulin-Binding Proteins/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , AC133 Antigen , Antineoplastic Agents, Alkylating/administration & dosage , Apoptosis , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/therapeutic use , Humans , Temozolomide
8.
PLoS One ; 10(7): e0131288, 2015.
Article in English | MEDLINE | ID: mdl-26131692

ABSTRACT

A variety of pluripotency reprogramming frequencies from different somatic cells has been observed, indicating cell origin is a critical contributor for efficiency of pluripotency reprogramming. Identifying the cell sources for efficient induced pluripotent stem cells (iPSCs) generation, and defining its advantages or disadvantages on reprogramming, is therefore important. Human ocular tissue-derived conjunctival epithelial cells (OECs) exhibited endogenous expression of reprogramming factors OCT4A (the specific OCT 4 isoform on pluripotency reprogramming) and SOX2. We therefore determined whether OECs could be used for high efficiency of iPSCs generation. We compared the endogenous expression levels of four pluripotency factors and the pluripotency reprograming efficiency of human OECs with that of ocular stromal cells (OSCs). Real-time PCR, microarray analysis, Western blotting and immunostaining assays were employed to compare OECiPSCs with OSCiPSCs on molecular bases of reprogramming efficiency and preferred lineage-differentiation potential. Using the traditional KMOS (KLF4, C-MYC, OCT4 and SOX2) reprogramming protocol, we confirmed that OECs, endogenously expressing reprogramming factors OCT4A and SOX2, yield very high efficiency of iPSCs generation (~1.5%). Furthermore, higher efficiency of retinal pigmented epithelial differentiation (RPE cells) was observed in OECiPSCs compared to OSCiPSCs or skin fibroblast iMR90iPSCs. The findings in this study suggest that conjunctival-derived epithelial (OECs) cells can be easier converted to iPSCs than conjunctival-derived stromal cells (OSCs). This cell type may also have advantages in retinal pigmented epithelial differentiation.


Subject(s)
Cellular Reprogramming , Conjunctiva/cytology , Epithelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/genetics
9.
Invest Ophthalmol Vis Sci ; 56(2): 711-21, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25574050

ABSTRACT

PURPOSE: Recently, RAP1 (Telomeric Repeat Binding Factor 2, Interacting Protein [TERF2IP]) was discovered as a modulator that selectively regulates nuclear factor light chain kappa enhancer of activated B cells (NFκB) signaling. The roles of RAP1 in regulation of inflammation and angiogenesis for corneal recovery following corneal injury remain poorly understood. The effects of RAP1 deletion on corneal resurfacing and neovascularization in a corneal alkali burn mouse model were examined. METHODS: Corneal defects and neovascularization were induced in vivo by infliction of an alkali burn to the cornea with 1 N sodium hydroxide solution in RAP1 knock-out (RKO) and wild-type (RWT) mice. Corneal resurfacing was evaluated using slit-lamp biomicroscopy. Neovascularization following injury was evaluated by bright view biomicroscopy and immunofluorescence staining with the endothelial marker platelet endothelial cell adhesion molecule (PECAM). The cytokine profiles of corneal tissue involved in inflammation and neovascularization following injury was compared between RKO and RWT mice. Corneal epithelial cells were isolated for classic scratch wound healing assay and further testing with lipopolysaccharide stimulation. RESULTS: Resurfacing of the burned cornea was accelerated and angiogenesis was suppressed, faster recovery of corneal epithelial cells from classic scratch wound healing and superior tolerance of lipopolysaccharides challenge was observed in the RKO compared to RWT. Molecular investigation revealed that deletion of RAP1 reduced upregulation of inflammatory cytokine (IL1A), finely regulated the expression of angiogenic factor (VEGF), and antiangiogenic factor (PEDF), following injury for better corneal recovery. CONCLUSIONS: Deficiency of RAP1 facilitates corneal recovery after injury. Specificity of RAP1 inhibition may lead to design of specific inhibitors of NFκB in the treatment of ocular injuries.


Subject(s)
Burns, Chemical/metabolism , Corneal Injuries/metabolism , Corneal Neovascularization/genetics , Eye Burns/metabolism , Telomere-Binding Proteins/genetics , Up-Regulation , Wound Healing , Alkalies/toxicity , Animals , Burns, Chemical/complications , Burns, Chemical/pathology , Corneal Injuries/complications , Corneal Injuries/pathology , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Disease Models, Animal , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Eye Burns/complications , Eye Burns/pathology , Mice , Mice, Knockout , RNA, Messenger/genetics , Shelterin Complex , Signal Transduction , Telomere-Binding Proteins/biosynthesis
10.
Cell Transplant ; 21(10): 2225-39, 2012.
Article in English | MEDLINE | ID: mdl-22776744

ABSTRACT

Although transplantation of adult bone marrow mesenchymal stem cells (BM-MSCs) holds promise in the treatment for pulmonary arterial hypertension (PAH), the poor survival and differentiation potential of adult BM-MSCs have limited their therapeutic efficiency. Here, we compared the therapeutic efficacy of human embryonic stem cell-derived MSCs (hESC-MSCs) with adult BM-MSCs for the treatment of PAH in an animal model. One week following monocrotaline (MCT)-induced PAH, mice were randomly assigned to receive phosphate-buffered saline (MCT group); 3.0×10(6) human BM-derived MSCs (BM-MSCs group) or 3.0×10(6) hESC-derived MSCs (hESC-MSCs group) via tail vein injection. At 3 weeks post-transplantation, the right ventricular systolic pressure (RVSP), degree of RV hypertrophy, and medial wall thickening of pulmonary arteries were lower=, and pulmonary capillary density was higher in the hESC-MSC group as compared with BM-MSC and MCT groups (all p < 0.05). At 1 week post-transplantation, the number of engrafted MSCs in the lungs was found significantly higher in the hESC-MSC group than in the BM-MSC group (all p < 0.01). At 3 weeks post-transplantation, implanted BM-MSCs were undetectable whereas hESC-MSCs were not only engrafted in injured pulmonary arteries but had also undergone endothelial differentiation. In addition, protein profiling of hESC-MSC- and BM-MSC-conditioned medium revealed a differential paracrine capacity. Classification of these factors into bioprocesses revealed that secreted factors from hESC-MSCs were preferentially involved in early embryonic development and tissue differentiation, especially blood vessel morphogenesis. We concluded that improved cell survival and paracrine capacity of hESC-MSCs provide better therapeutic efficacy than BM-MSCs in the treatment for PAH.


Subject(s)
Embryonic Stem Cells/cytology , Hypertension, Pulmonary/surgery , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cell Growth Processes , Cell Line , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Embryonic Stem Cells/metabolism , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred ICR , Mice, SCID
11.
Graefes Arch Clin Exp Ophthalmol ; 249(10): 1439-48, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21866334

ABSTRACT

With the development of stem cell technology, stem cell-based therapy for retinal degeneration has been proposed to restore the visual function. Many animal studies and some clinical trials have shown encouraging results of stem cell-based therapy in retinal degenerative diseases. While stem cell-based therapy is a promising strategy to replace damaged retinal cells and ultimately cure retinal degeneration, there are several important challenges which need to be overcome before stem cell technology can be applied widely in clinical settings. In this review, different types of donor cell origins used in retinal treatments, potential target cell types for therapy, methods of stem cell delivery to the eye, assessments of potential risks in stem cell therapy, as well as future developments of retinal stem cells therapy, will be discussed.


Subject(s)
Retinal Degeneration/surgery , Stem Cell Transplantation/trends , Humans , Treatment Outcome
12.
Histol Histopathol ; 26(8): 953-63, 2011 08.
Article in English | MEDLINE | ID: mdl-21692029

ABSTRACT

Kiaa0319L is a novel protein encoded by a recently discovered gene KIAA0319-like(L) that may be associated with reading disability. Little is known about the characteristics of this protein and its distribution in the brain. We investigated here expression of this protein in adult mice, using an antibody specific for human and rodent Kiaa0319L. In the brain, Kiaa0319L was localized strongly in the olfactory bulb, and strong expression was found in other regions, including hippocampus, cerebellum, diencephalon and the cerebral cortex. Immunohistochemistry confirmed expression in these brain regions, and showed further that the protein was expressed preferentially in neurons in layer IV and VI of the neocortex, CA1 and CA2 subfields of the hippocampus and a subpopulation of neurons in CA3 and dentate gyrus. Furthermore, the protein was confined to dendrites of CA1 neurons in the stratum radiatum, but not those in the stratum oriens, and in astrocytes within the hippocampus. In the cerebellum, the protein was observed in the molecular layer and a fraction of Purkinje neurons. These findings confirmed expression of Kiaa0319L in brain regions that are involved in reading performance, supporting its possible involvement in reading disability. The specific patterns of localization in the neocortex, hippocampus and cerebellum suggest further that this protein may be related to other biological processes in a subpopulation of neurons within these regions, eg. formation and maintenance of polarity in the neuron.


Subject(s)
Brain/metabolism , Dyslexia/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Animals , Blotting, Western , Brain/cytology , Cerebellum/cytology , Cerebellum/metabolism , Computational Biology/methods , Dendrites/metabolism , Dyslexia/genetics , Female , Hippocampus/cytology , Hippocampus/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Protein Structure, Tertiary , Receptors, Cell Surface
13.
Cell Mol Neurobiol ; 31(1): 27-35, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20697954

ABSTRACT

To identify the putative interacting partners for Kiaa0319-like protein. KIAA0319-like, located near the dyslexia susceptibility locus, DYX8 in chromosome 1p34.3, has been suggested as a positional candidate for developmental dyslexia due to its homology with another gene, KIAA0319 which has been strongly established as a candidate gene for developmental dyslexia. Previous research has shown that a single marker, rs7523017 (P = 0.042) has been associated with developmental dyslexia by a Canadian group. There is little functional information about this gene and protein. In this article, we put forward further evidence that support Kiaa0319-like is a candidate for this disorder. A yeast-2-hybrid screen and co-immunopreciptiation assays were performed to find protein interacting partners of KIAA0319L. A human cortex immunohistochemistry assay was performed to show the colocalization of Kiaa0319-like and its specific interacting partner in cells. Nogo Receptor 1 (NgR1), an axon guidance receptor, was identified to have physical interactions with Kiaa0319-like protein. These two proteins interact predominantly in the cytoplasmic granules of cortical neurons in the human brain cortex. Based on this data, it can be concluded that Kiaa0319-like protein interacts with Nogo Receptor 1, supporting the idea that Kiaa0319-like protein participates in axon guidance.


Subject(s)
Axons/metabolism , Axons/physiology , Dyslexia , Myelin Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Cell Surface/metabolism , Biopsy , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/pathology , Dyslexia/genetics , Dyslexia/metabolism , Dyslexia/pathology , GPI-Linked Proteins/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nogo Receptor 1 , Nuclear Proteins/genetics , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...