Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 284
Filter
1.
Exp Neurol ; 347: 113896, 2022 01.
Article in English | MEDLINE | ID: mdl-34662541

ABSTRACT

Presenilins (PS) form the active subunit of the gamma-secretase complex, which mediates the proteolytic clearance of a broad variety of type-I plasma membrane proteins. Loss-of-function mutations in PSEN1/2 genes are the leading cause of familial Alzheimer's disease (fAD). However, the PS/gamma-secretase substrates relevant for the neuronal deficits associated with a loss of PS function are not completely known. The members of the neurexin (Nrxn) family of presynaptic plasma membrane proteins are candidates to mediate aspects of the synaptic and memory deficits associated with a loss of PS function. Previous work has shown that fAD-linked PS mutants or inactivation of PS by genetic and pharmacological approaches failed to clear Nrxn C-terminal fragments (NrxnCTF), leading to its abnormal accumulation at presynaptic terminals. Here, we generated transgenic mice that selectively recreate the presynaptic accumulation of NrxnCTF in adult forebrain neurons, leaving unaltered the function of PS/gamma-secretase complex towards other substrates. Behavioral characterization identified selective impairments in NrxnCTF mice, including decreased fear-conditioning memory. Electrophysiological recordings in medial prefrontal cortex-basolateral amygdala (mPFC-BLA) of behaving mice showed normal synaptic transmission and uncovered specific defects in synaptic facilitation. These data functionally link the accumulation of NrxnCTF with defects in associative memory and short-term synaptic plasticity, pointing at impaired clearance of NrxnCTF as a new mediator in AD.


Subject(s)
Association Learning/physiology , Calcium-Binding Proteins/biosynthesis , Memory Disorders/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Neuronal Plasticity/physiology , Presenilins/biosynthesis , Prosencephalon/metabolism , Age Factors , Animals , Calcium-Binding Proteins/genetics , Fear/physiology , Fear/psychology , Gene Expression Regulation , Humans , Male , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Cell Adhesion Molecules/genetics , Presenilin-1/biosynthesis , Presenilin-1/genetics , Presenilin-2/biosynthesis , Presenilin-2/genetics , Presenilins/genetics , Presynaptic Terminals/metabolism
2.
Mol Cell Neurosci ; 112: 103614, 2021 04.
Article in English | MEDLINE | ID: mdl-33845123

ABSTRACT

Homozygous Dab1 yotari mutant mice, Dab1yot (yot/yot) mice, have an autosomal recessive mutation of Dab1 and show reeler-like phenotype including histological abnormality of the cerebellum, hippocampus, and cerebral cortex. We here show abnormal hippocampal development of yot/yot mice where granule cells and pyramidal cells fail to form orderly rows but are dispersed diffusely in vague multiplicative layers. Possibly due to the positioning failure of granule cells and pyramidal cells and insufficient synaptogenesis, axons of the granule cells did not extend purposefully to connect with neighboring regions in yot/yot mice. We found that both hippocampal granule cells and pyramidal cells of yot/yot mice expressed proteins reactive with the anti-Dab1 antibody. We found that Y198- phosphorylated Dab1 of yot/yot mice was greatly decreased. Accordingly the downstream molecule, Akt was hardly phosphorylated. Especially, synapse formation was defective and the distribution of neurons was scattered in hippocampus of yot/yot mice. Some of neural cell adhesion molecules and hippocampus associated transcription factors of the neurons were expressed aberrantly, suggesting that the Reelin-Dab1 signaling pathway seemed to be importantly involved in not only neural migration as having been shown previously but also neural maturation and/or synaptogenesis of the mice. It is interesting to clarify whether the defective neural maturation is a direct consequence of the dysfunctional Dab1, or alternatively secondarily due to the Reelin-Dab1 intracellular signaling pathways.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Hippocampus/abnormalities , Mice, Mutant Strains/abnormalities , Nerve Tissue Proteins/physiology , Serine Endopeptidases/physiology , Signal Transduction/physiology , Animals , Cell Adhesion Molecules, Neuronal/deficiency , Cell Movement , Enzyme Activation , Extracellular Matrix Proteins/deficiency , Genes, Recessive , Hippocampus/embryology , Hippocampus/metabolism , Hippocampus/pathology , Homozygote , Mice , Mice, Mutant Strains/genetics , Mice, Mutant Strains/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Reelin Protein , Serine Endopeptidases/deficiency , Synapses/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
3.
Fertil Steril ; 113(1): 176-186, 2020 01.
Article in English | MEDLINE | ID: mdl-32033718

ABSTRACT

OBJECTIVE: To characterize the role of steroid hormone and antihormone exposure on neurotrimin (NTM) expression in human leiomyoma and myometrial tissue and cells. DESIGN: Laboratory study of placebo and ulipristal acetate (UPA)-treated patient tissue. In vitro assessment of immortalized myometrial and leiomyoma cell lines after hormone and antihormone exposure. SETTING: Academic research center. PATIENT(S): Not applicable. INTERVENTIONS(S): Exposure of leiomyoma cell lines to 17ß-E2, medroxyprogesterone acetate (MPA), UPA, and fulvestrant. MAIN OUTCOME MEASURE(S): Messenger RNA expression quantified with the use of RNASeq analysis and quantitative real-time polymerase chain reaction (qRT-PCR). Protein levels quantified by means of Western blot analysis. Immunohistochemistry (IHC) on placebo- and UPA-treated patient uterine tissue specimens. RESULT(S): Expression of NTM in human uterine leiomyoma specimens according to RNASeq was increased compared with myometrium (5.22 ± 0.57-fold), which was confirmed with the use of qRT-PCR (1.95 ± 0.05). Furthermore, NTM protein was elevated in leiomyoma tissue compared with matched myometrium (2.799 ± 0.575). IHC revealed increased staining intensity in leiomyoma surgical specimens compared with matched myometrium of placebo patients. Western blot analysis in immortalized leiomyoma cell lines demonstrated an up-regulation of NTM protein expression (2.4 ± 0.04). Treatment of leiomyoma cell lines with 17ß-E2 yielded a 1.98 ± 0.11-fold increase in NTM protein expression; however, treatment with fulvestrant showed no significant change compared with control. Leiomyoma cell lines demonstrated a 1.91 ± 0.97-fold increase in NTM protein expression after progesterone treatment. RNASeq analysis demonstrated a reduced expression in patient leiomyoma after UPA treatment (0.75 ± 0.14). Treatment of leiomyoma cells with UPA demonstrated a reduced total NTM protein amount (0.54 ± 0.31) in patients, which was confirmed with the use of IHC (UPA10 147.2 ± 9.40, UPA20 182.8 ± 8.98). In vitro studies with UPA treatment revealed a concentration-dependent effect that supported these findings. CONCLUSION(S): NTM, a neural cell adhesion molecule, is increased in leiomyoma compared with myometrium in patient tissue and in vitro models after estrogen and progesterone treatment. Down-regulation of expression occurs after UPA treatment, but not after fulvestrant exposure. CLINICAL TRIAL REGISTRATION NUMBER: NCT00290251.


Subject(s)
Contraceptive Agents, Female/pharmacology , Gonadal Steroid Hormones/pharmacology , Hormone Antagonists/pharmacology , Leiomyoma/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Biomarkers/metabolism , Cell Line, Tumor , Contraceptive Agents, Female/therapeutic use , Double-Blind Method , Estradiol/pharmacology , Estradiol/therapeutic use , Female , GPI-Linked Proteins/agonists , GPI-Linked Proteins/biosynthesis , Gonadal Steroid Hormones/therapeutic use , Hormone Antagonists/therapeutic use , Humans , Leiomyoma/drug therapy , Leiomyoma/pathology , Neural Cell Adhesion Molecules/agonists , Norpregnadienes/pharmacology , Norpregnadienes/therapeutic use
4.
Prostate ; 80(4): 352-364, 2020 03.
Article in English | MEDLINE | ID: mdl-31905248

ABSTRACT

BACKGROUND: Signal regulatory protein ß1 (SIRPB1) is a signal regulatory protein member of the immunoglobulin superfamily and is capable of modulating receptor tyrosine kinase-coupled signaling. Copy number variations at the SIRPB1 locus were previously reported to associate with prostate cancer aggressiveness in patients, however, the role of SIRPB1 in prostate carcinogenesis is unknown. METHODS: Fluorescence in situ hybridization and laser-capture microdissection coupled with quantitative polymerase chain reaction was utilized to determine SIRPB1 gene amplification and messenger RNA expression in prostate cancer specimens. The effect of knockdown of SIRPB1 by RNA interference in PC3 prostate cancer cells on cell growth in colony formation assays and cell mobility in wound-healing, transwell assays, and cell cycle analysis was determined. Overexpression of SIPRB1 in C4-2 prostate cancer cells on cell migration, invasion, colony formation and cell cycle progression and tumor take rate in xenografts was also determined. Western blot assay of potential downstream SIRPB1 pathways was also performed. RESULTS: SIRPB1 gene amplification was detected in up to 37.5% of prostate cancer specimens based on in silico analysis of several publicly available datasets. SIRPB1 gene amplification and overexpression were detected in prostate cancer specimens. The knockdown of SIRPB1 significantly suppressed cell growth in colony formation assays and cell mobility. SIRPB1 knockdown also induced cell cycle arrest during the G0 /G1 phase and enhancement of apoptosis. Conversely, overexpression of SIPRB1 in C4-2 prostate cancer cells significantly enhanced cell migration, invasion, colony formation, and cell cycle progression and increased C4-2 xenograft tumor take rate in nude mice. Finally, this study presented evidence for SIRPB1 regulation of Akt phosphorylation and showed that Akt inhibition could abolish SIRPB1 stimulation of prostate cancer cell proliferation. CONCLUSIONS: These results suggest that SIRPB1 is a potential oncogene capable of activating Akt signaling to stimulate prostate cancer proliferation and could be a biomarker for patients at risk of developing aggressive prostate cancer.


Subject(s)
Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Enzyme Activation , Gene Amplification , Heterografts , Humans , Male , Mice , Mice, Nude , Neural Cell Adhesion Molecules/biosynthesis , PC-3 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
5.
Cell Rep ; 27(13): 3752-3759.e4, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31242409

ABSTRACT

Neurexins are key synaptic organizers that are expressed in thousands of alternatively spliced isoforms. Because transsynaptic neurexin interactions with different postsynaptic molecules are largely isoform dependent, a cell type-level census of different neurexin isoforms could predict molecular interactions relating to synapse identity and function. Using single-cell transcriptomics to study the origin of neurexin diversity in multiple murine mature and embryonic cell types, we have discovered shared neurexin expression patterns in developmentally related cells. By comparing neurexin profiles in immature embryonic neurons, we show that neurexin profiles are specified during early development and remain unchanged throughout neuronal maturation. Thus, our findings reveal ontogenetic stability and provide a cell type-level census of neurexin isoform expression in the cortex.


Subject(s)
Alternative Splicing , Cerebral Cortex/metabolism , Neural Cell Adhesion Molecules , RNA-Seq , Single-Cell Analysis , Animals , Cerebral Cortex/cytology , Female , Male , Mice , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Optogenetics , Protein Stability
6.
J Neurosci ; 39(5): 814-832, 2019 01 30.
Article in English | MEDLINE | ID: mdl-30530861

ABSTRACT

Neurons in the murine olfactory epithelium (OE) differ by the olfactory receptor they express as well as other molecular phenotypes that are regionally restricted. These patterns can be precisely regenerated following epithelial injury, suggesting that spatial cues within the tissue can direct neuronal diversification. Nonetheless, the permanency and mechanism of this spatial patterning remain subject to debate. Via transplantation of stem and progenitor cells from dorsal OE into ventral OE, we demonstrate that, in mice of both sexes, nonautonomous spatial cues can direct the spatially circumscribed differentiation of olfactory sensory neurons. The vast majority of dorsal transplant-derived neurons express the ventral marker OCAM (NCAM2) and lose expression of NQO1 to match their new location. Single-cell analysis also demonstrates that OSNs adopt a fate defined by their new position following progenitor cell transplant, such that a ventral olfactory receptor is expressed after stem and progenitor cell engraftment. Thus, spatially constrained differentiation of olfactory sensory neurons is plastic, and any bias toward an epigenetic memory of place can be overcome.SIGNIFICANCE STATEMENT Spatially restricted differentiation of olfactory sensory neurons is both key to normal olfactory function and a challenging example of biological specificity. That the stem cells of the olfactory epithelium reproduce the organization of the olfactory periphery to a very close approximation during lesion-induced regeneration begs the question of whether stem cell-autonomous genomic architecture or environmental cues are responsible. The plasticity demonstrated after transfer to a novel location suggests that cues external to the transplanted stem and progenitor cells confer neuronal identity. Thus, a necessary prerequisite is satisfied for using engraftment of olfactory stem and progenitor cells as a cellular therapeutic intervention to reinvigorate neurogenesis whose exhaustion contributes to the waning of olfaction with age.


Subject(s)
Olfactory Mucosa/cytology , Olfactory Receptor Neurons/physiology , Animals , Cell Differentiation/physiology , Cues , Epigenesis, Genetic , Female , Male , Mice , Mice, Inbred C57BL , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Neural Stem Cells , Neurogenesis/physiology , Neuronal Plasticity , Stem Cell Transplantation
7.
Int J Cardiol ; 241: 322-325, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28343767

ABSTRACT

Chronic intramyocardial inflammation (inflammatory cardiomyopathy/DCMi) is linked to the pathogenesis of dilated cardiomyopathy (DCM). Neural cell adhesion molecule (NCAM) is involved in orchestrating cardiac muscle morphogenesis, but is down-regulated after embryogenesis. We investigated NCAM expression in adult DCM hearts, its possible association with DCMi-parameters, and with cardiomyocyte hypertrophy (CMH). Endomyocardial biopsies (EMBs) from DCM patients (n=85; n=37 females; age: 48±19years; LVEF <40%) and controls from non-cardiac deaths were immunostained for DCMi markers and for NCAM expression, and quantified by digital image analysis (DIA). NCAM expression on the intercalated discs and the sarcolemma was confirmed in n=46 (54%) of the DCM-EMBs. In the 17 controls, NCAM expression was confined to scattered intramyocardial nerves, but was absent on cardiomyocytes. DIA-quantified area fraction (AF) of NCAM was significantly (p=0.0001) higher in the DCM hearts (0.0044±0.017) compared with the controls (0.0006±0.0004). Multivariate analysis of DIA-quantified NCAM-AF revealed significant associations with infiltrates (CD18+, CD11a/LFA-1+, CD11b/Mac-1+, TNFα+, CD3+) and with endothelial cell adhesion molecules (CAM; CD54/ICAM-1 and CD29; p<0.05). The mean cardiomyocyte diameter (MCD) correlated highly significantly (p<0.01) with NCAM-AF, ICAM-1-AF, CD29-AF, CD18+ and TNFa+ infiltrates, and was associated less significantly (p<0.05) with CD3+, CD11a/LFA-1+, and CD11b/Mac-1+ infiltrates. In conclusion, NCAM-expression in ca. 50% of adult DCM hearts is associated with CMH, and may be induced by inflammatory pathways.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Adult , Aged , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Female , Gene Expression , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Middle Aged , Myocardium/pathology , Myocytes, Cardiac/pathology , Neural Cell Adhesion Molecules/genetics
8.
Epilepsy Behav ; 69: 18-23, 2017 04.
Article in English | MEDLINE | ID: mdl-28222338

ABSTRACT

Axonal sprouting and synaptic reorganization are the primary pathophysiological characteristics of epilepsy. Recent studies demonstrated that synaptic adhesion-like molecule 3 (SALM3) is highly expressed in the central nervous system and plays important roles in neurite outgrowth, branching, and axon guidance, mechanisms that are also observed in epilepsy. However, the expression of SALM3 in the epileptic brain and the effect of SALM3 in the pathogenesis of epilepsy remain unclear. The aims of this study were to investigate SALM3 expression in rat models of epilepsy and to explore the functional significance of SALM3 in epilepsy. We demonstrated that SALM3 was expressed at significantly higher levels in epileptic rats compared with controls. Inhibition of SALM3 by SALM3 shRNA inhibited status epilepticus in the acute stage of disease and decreased spontaneous recurrent seizures in the Lithium-pilocarpine model of chronic stages of epilepsy. Consistent with these findings, SALM3 shRNA significantly prolonged the latent period in the PTZ kindling model. Our study suggests that the overexpression of SALM3 might be associated with epileptogenesis and that selectively inhibiting SALM3 may have therapeutic potential in treating epilepsy.


Subject(s)
Brain/metabolism , Disease Models, Animal , Epilepsy/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Seizures/metabolism , Animals , Brain/physiopathology , Epilepsy/chemically induced , Epilepsy/physiopathology , Kindling, Neurologic/metabolism , Male , Membrane Glycoproteins , Nerve Tissue Proteins , Pilocarpine/toxicity , Random Allocation , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/physiopathology
9.
Neurosci Lett ; 644: 76-82, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28237805

ABSTRACT

The adult rodent piriform cortex has been reported to harbor immature neurons that express markers associated with neurodevelopment and plasticity, namely polysialylated neural cell adhesion molecule (PSA-NCAM) and doublecortin (DCX). We characterized the expression of PSA-NCAM and DCX across the rostrocaudal axis of the rat piriform cortex and observed higher numbers of PSA-NCAM and DCX positive cells in the posterior subdivision. As observed in the rat piriform cortex, Nestin-GFP reporter mice also revealed a similar gradient of GFP-positive cells with an increasing rostro-caudal gradient of expression. Given the extensive noradrenergic innervation of the piriform cortex and its role in regulating piriform cortex function and synaptic plasticity, we addressed the influence of norepinephrine (NE) on piriform cortex plasticity marker expression. Depletion of NE by treatment with the noradrenergic neurotoxin DSP-4 significantly increased the number of DCX and PSA-NCAM immunopositive cells in the piriform cortex of adult rats. Similarly, DSP-4 treated Nestin-GFP reporter mice revealed a robust induction of GFP-positive cells within the piriform cortex following NE depletion. Genetic loss of NE in dopamine ß-hydroxylase knockout (Dbh -/-) mice phenocopied the effects of DSP-4, with an increase noted in PSA-NCAM and DCX positive cells in the piriform cortex. Further, chronic α2-adrenergic receptor stimulation with the agonist guanabenz increased PSA-NCAM and DCX positive cells in the piriform cortex of adult rats and GFP-positive cells in the piriform cortex of Nestin-GFP mice. By contrast, chronic α2-adrenergic receptor blockade with the antagonist yohimbine reduced PSA-NCAM and DCX positive cells in the piriform cortex of adult rats. Our results provide novel evidence for a role of NE in regulating the expression of plasticity markers, including PSA-NCAM, DCX, and nestin, within the adult mouse and rat piriform cortex.


Subject(s)
Neuronal Plasticity/physiology , Norepinephrine/metabolism , Piriform Cortex/metabolism , Animals , Biomarkers/analysis , Doublecortin Domain Proteins , Doublecortin Protein , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/biosynthesis , Nestin/biosynthesis , Neural Cell Adhesion Molecules/biosynthesis , Neuropeptides/biosynthesis , Rats , Rats, Wistar
10.
Int J Gynecol Cancer ; 27(2): 239-245, 2017 02.
Article in English | MEDLINE | ID: mdl-27984374

ABSTRACT

OBJECTIVE: Neural cell adhesion molecule (CD56) has been proposed as a potential marker for neuroendocrine differentiation in carcinomas, together with synaptophysin and chromogranin A. However, CD56 immunoreactivity by itself can be found in a broad variety of tumors, including ovarian neoplasms. CD56 has recently been suggested as a potential target for antibody-based therapy. However, for ovarian carcinoma, there is only limited data available regarding the pattern of CD56 immunoreactivity, coexpression of neuroendocrine markers, and correlation with histological types and clinical parameters. METHODS: In our study, we therefore evaluated CD56 staining by immunohistochemistry on a tissue micrroarray with 206 ovarian carcinomas, including 151 high-grade serous, 7 low-grade serous, 32 endometrioid, 11 clear cell, 5 mucinous, as well as 33 atypically proliferating serous tumors/serous borderline tumors. RESULTS: At least focal CD56 immunoreactivity was observed in 65% of carcinomas of all histological types. Moderate staining with at least 10% positive cells was found in 44 (28%) high-grade serous carcinomas (HGSOCs), 2 (29%) low-grade serous and 3(9%) endometrioid carcinomas. Strong immunoreactivity was limited to 10 (7%) HGSOCs. There was no correlation with the expression of chromogranin or synaptophysin. Serous borderline tumors showed only weak and focal staining in 11 (33%). Expression of CD56 overall was significantly associated with high-grade and advanced stage. In the subgroup of HGSOCs, CD56 expression was associated with reduced overall survival (median 30 vs. 47 months, P = 0.039, log rank, univariate analysis). CONCLUSIONS: CD56 (neural cell adhesion molecule) is frequently expressed in ovarian carcinomas and is significantly associated with HGSOC and advanced tumor stage. Due to its lack of correlation with neuroendocrine differentiation, CD56 expression is of limited diagnostic value, but may rather serve as a marker for tumor progression or as a potential therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Neural Cell Adhesion Molecules/biosynthesis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Aged , Cell Differentiation/physiology , Cohort Studies , Female , Humans , Immunohistochemistry , Neoplasm Grading , Neoplasm Staging , Tissue Array Analysis
11.
Elife ; 52016 12 13.
Article in English | MEDLINE | ID: mdl-27960072

ABSTRACT

The unique anatomical and functional features of principal and interneuron populations are critical for the appropriate function of neuronal circuits. Cell type-specific properties are encoded by selective gene expression programs that shape molecular repertoires and synaptic protein complexes. However, the nature of such programs, particularly for post-transcriptional regulation at the level of alternative splicing is only beginning to emerge. We here demonstrate that transcripts encoding the synaptic adhesion molecules neurexin-1,2,3 are commonly expressed in principal cells and interneurons of the mouse hippocampus but undergo highly differential, cell type-specific alternative splicing. Principal cell-specific neurexin splice isoforms depend on the RNA-binding protein Slm2. By contrast, most parvalbumin-positive (PV+) interneurons lack Slm2, express a different neurexin splice isoform and co-express the corresponding splice isoform-specific neurexin ligand Cbln4. Conditional ablation of Nrxn alternative splice insertions selectively in PV+ cells results in elevated hippocampal network activity and impairment in a learning task. Thus, PV-cell-specific alternative splicing of neurexins is critical for neuronal circuit function.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Hippocampus/physiology , Nerve Tissue Proteins/biosynthesis , Neural Cell Adhesion Molecules/biosynthesis , Neurons/physiology , Animals , Calcium-Binding Proteins , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/genetics , Protein Precursors/metabolism , RNA-Binding Proteins/metabolism
12.
Neuron ; 91(5): 1052-1068, 2016 Sep 07.
Article in English | MEDLINE | ID: mdl-27608760

ABSTRACT

Mutations in a synaptic organizing pathway contribute to autism. Autism-associated mutations in MDGA2 (MAM domain containing glycosylphosphatidylinositol anchor 2) are thought to reduce excitatory/inhibitory transmission. However, we show that mutation of Mdga2 elevates excitatory transmission, and that MDGA2 blocks neuroligin-1 interaction with neurexins and suppresses excitatory synapse development. Mdga2(+/-) mice, modeling autism mutations, demonstrated increased asymmetric synapse density, mEPSC frequency and amplitude, and altered LTP, with no change in measures of inhibitory synapses. Behavioral assays revealed an autism-like phenotype including stereotypy, aberrant social interactions, and impaired memory. In vivo voltage-sensitive dye imaging, facilitating comparison with fMRI studies in autism, revealed widespread increases in cortical spontaneous activity and intracortical functional connectivity. These results suggest that mutations in MDGA2 contribute to altered cortical processing through the dual disadvantages of elevated excitation and hyperconnectivity, and indicate that perturbations of the NRXN-NLGN pathway in either direction from the norm increase risk for autism.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Cerebral Cortex/physiology , Cognition/physiology , GPI-Linked Proteins/physiology , Haploinsufficiency/physiology , Neural Cell Adhesion Molecules/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Disks Large Homolog 4 Protein , Excitatory Postsynaptic Potentials/physiology , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Guanylate Kinases/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/physiology , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Receptors, AMPA/metabolism , Receptors, AMPA/physiology , Synapses/metabolism
13.
Oncotarget ; 7(30): 48027-48037, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27384995

ABSTRACT

Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and a major cause of cancer mortality. Chemotherapy resistance remains a major challenge for treating advanced CRC. Therefore, the identification of targets that induce drug resistance is a priority for the development of novel agents to overcome resistance. Dragon (also known as RGMb) is a member of the repulsive guidance molecule (RGM) family. We previously showed that Dragon expression increases with CRC progression in human patients. In the present study, we found that Dragon inhibited apoptosis and increased viability of CMT93 and HCT116 cells in the presence of oxaliplatin. Dragon induced resistance of xenograft tumor to oxaliplatinin treatment in mice. Mechanistically, Dragon inhibited oxaliplatin-induced JNK and p38 MAPK activation, and caspase-3 and PARP cleavages. Our results indicate that Dragon may be a novel target that induces drug resistance in CRC.


Subject(s)
Cell Adhesion Molecules, Neuronal/biosynthesis , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Nerve Tissue Proteins/biosynthesis , Neural Cell Adhesion Molecules/biosynthesis , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , HCT116 Cells , Humans , Mice , Mice, Inbred C57BL , Oxaliplatin , Xenograft Model Antitumor Assays
14.
Neuron ; 89(3): 480-93, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26844831

ABSTRACT

Cell recognition molecules are key regulators of neural circuit assembly. The Dscam family of recognition molecules in Drosophila has been shown to regulate interactions between neurons through homophilic repulsion. This is exemplified by Dscam1 and Dscam2, which together repel dendrites of lamina neurons, L1 and L2, in the visual system. By contrast, here we show that Dscam2 directs dendritic targeting of another lamina neuron, L4, through homophilic adhesion. Through live imaging and genetic mosaics to dissect interactions between specific cells, we show that Dscam2 is required in L4 and its target cells for correct dendritic targeting. In a genetic screen, we identified Dscam4 as another regulator of L4 targeting which acts with Dscam2 in the same pathway to regulate this process. This ensures tiling of the lamina neuropil through heterotypic interactions. Thus, different combinations of Dscam proteins act through distinct mechanisms in closely related neurons to pattern neural circuits.


Subject(s)
Dendrites/physiology , Drosophila Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Neural Cell Adhesion Molecules/physiology , Alleles , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , Mosaicism , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics
15.
Pharmazie ; 71(10): 583-587, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-29441926

ABSTRACT

TAK-242 (resatorvid), a novel small-molecule cyclohexene derivative, inhibits TLR4 signaling selectively. TAK-242 blocked the Toll-like receptor (TLR) 4-triggered inflammatory signaling by binding directly to a specific amino acid Cys747 in the intracellular domain of TLR4. The present study was designed to examine the effects of TAK-242 on vascular inflammatory responses in human coronary artery endothelial cells (HCAECs) challenged by lipopolysaccharide (LPS, a TLR4 ligand). The results show that TAK-242 attenuated the LPS-induced expression of interleukin (IL)-6, IL-8 and monocyte chemoattractant protein 1 both at the transcription and translation levels in HCAECs. LPS-induced endothelial cell adhesion molecules, intercellular adhesion molecular-1 and vascular cell adhesion molecule-1 expressions were also reduced by treatment with TAK-242. In addition, coincubation with TAK-242 did not effect the expression of TLR4 in LPS-activated HCAECs. Furthermore, TAK-242 efficiently suppressed LPS-induced phosphorylation of nuclear factor κB (NF-κB) and IL-1 associated kinase-1 (IRAK-1) in HCAECs. These findings show that TAK-242 can suppress endothelial cell inflammation, suggesting that TAK-242 might be suitable for development as a therapeutic agent for inflammatory cardiovascular disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Coronary Vessels/drug effects , Endothelial Cells/drug effects , Lipopolysaccharides/antagonists & inhibitors , Sulfonamides/pharmacology , Coronary Vessels/cytology , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/prevention & control , Interleukin-1 Receptor-Associated Kinases/biosynthesis , Interleukin-1 Receptor-Associated Kinases/genetics , NF-kappa B/drug effects , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , RNA, Messenger/biosynthesis , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors
16.
Indian J Med Res ; 142(4): 391-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26609030

ABSTRACT

BACKGROUND & OBJECTIVES: Cancer stem cells (CSCs) may be responsible for tumour recurrence and resistance to chemotherapy in hepatocellular carcinoma (HCC). This study was carried out to evaluate the association between histological parameters and liver CSCs (LCSC) in HCC, and to compare distribution of liver CSCs in HCC associated with and without hepatitis B virus (HBV) infection. METHODS: Seventy nine tumours (49 surgical resections from 46 patients, and 30 from autopsy) were reviewed. Immunohistochemical staining for the LCSC marker EpCAM (epithelial cell adhesion molecule), liver progenitor cell (LPC) markers CK19 (cytokeratin 19) and neural cell adhesion molecule (NCAM) were performed and were associated with histological features of tumour behaviour. RESULTS: Thirty three tumours (41.8%) showed positive staining for EpCAM. CK19 and NCAM expression were seen in 26 (32.9%) and four (5.1%) tumours, respectively. The expression of EpCAM and CK19 was significantly associated with each other ( P<0.001). EpCAM expression was significantly associated with clinical and histological features indicating aggressive tumour behaviour, including younger age of onset, higher serum alpha foetoprotein (AFP) levels, tumour cell dedifferentiation, increased mitotic activity, and vascular invasiveness. There was no significant difference in expression of EpCAM, CK19 and NCAM between HBV positive and negative HCC. INTERPRETATION & CONCLUSIONS: The LCSC marker EpCAM was expressed in less than half of HCC, was independent of HBV aetiology, and was strongly associated with clinical and histological features of aggressive tumour behaviour. Positive staining for CK19 suggests a possible LPC origin of the EpCAM positive HCCs.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Hepatocellular/genetics , Keratin-19/biosynthesis , Liver Neoplasms/genetics , Neoplastic Stem Cells , Adult , Aged , Autopsy , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Female , Gene Expression Regulation, Neoplastic , Hepatitis B/complications , Hepatitis B/genetics , Hepatitis B/pathology , Hepatitis B virus/genetics , Hepatitis B virus/pathogenicity , Humans , Keratin-19/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Male , Middle Aged , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , alpha-Fetoproteins/biosynthesis
17.
Biogerontology ; 16(6): 811-23, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26385499

ABSTRACT

Sarcopenia is a major contributor to the loss of independence and deteriorating quality of life in elderly individuals, it manifests as a decline in skeletal muscle mass and strength beyond the age of 65. Muscle fibre atrophy is a major contributor to sarcopenia and the most severely atrophic fibres are commonly found in elderly muscles to have permanently lost their motor nerve input. By contrast with elderly fibres, when fibres in young animals lose their motor input they normally mount a response to induce restoration of nerve contact, and this is mediated in part by upregulated expression of the nerve cell adhesion molecule (NCAM). Therefore, skeletal muscles appear to progressively lose their ability to become reinnervated, and here we have investigated whether this decline occurs via loss of the muscle's ability to upregulate NCAM in response to denervation. We performed partial denervation (by peripheral nerve crush) of the extensor digitorum longus muscle of the lower limb in both young and elderly mice. We used immunohistochemistry to compare relative NCAM levels at denervated and control innervated muscle fibres, focused on measurements at neuromuscular junctional, extra-junctional and cytoplasmic locations. Muscle fibres in young animals responded to denervation with significant (32.9%) increases in unpolysialylated NCAM at extra-junctional locations, but with no change in polysialylated NCAM. The same partial denervation protocol applied to elderly animals resulted in no significant change in either polysialylated or unpolysialylated NCAM at junctional, extra-junctional or cytoplasmic locations, therefore muscle fibres in young mice upregulated NCAM in response to denervation but fibres in elderly mice failed to do so. Elevation of NCAM levels is likely to be an important component of the muscle fibre's ability to attract or reattract a neural input, so we conclude that the presence of increasing numbers of long-term denervated fibres in elderly muscles is due, at least in part, to the fibre's declining ability to mount a normal response to loss of motor input.


Subject(s)
Aging/metabolism , Muscle Denervation , Muscle Fibers, Skeletal/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Aging/pathology , Animals , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Sarcopenia/etiology , Sarcopenia/metabolism , Sarcopenia/pathology , Up-Regulation
18.
Carcinogenesis ; 36(9): 1071-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26153023

ABSTRACT

Rhabdomyosarcoma represents about 50% of soft-tissue sarcomas and 10% of malignant solid tumors in childhood. Embryonal rhabdomyosarcoma (ERMS) is the most frequent subtype, suggested to have an origin in muscle precursor cells that fail to exit the cell cycle and terminally differentiate mainly because of overexpression of the transcription factor, PAX7, which sustains proliferation, migration and invasiveness in ERMS cells. Artesunate (ARS) is a semi-synthetic derivative of artemisinin (ART), a natural compound well known as an antimalarial drug. However, ART and its derivatives have been found efficacious even as anticancer drugs that induce cell cycle arrest and/or apoptosis in several kinds of cancer. Here, we show that ARS dose-dependently induces DNA damage and apoptosis in ERMS cell lines. Production of reactive oxygen species (ROS) and activation of p38 MAPK have a central role in triggering ARS-mediated apoptosis in ERMS cells; indeed either the antioxidant, N-acetylcysteine or the p38 MAPK inhibitor, SB203580, protects ERMS cells from ARS-induced apoptosis. Moreover, ARS treatment in ERMS cells ROS-dependently induces the expression of the myo-miRs, miR-133a and miR-206, which are down-regulated in RMS, and reduces PAX7 protein levels. Finally, ARS upregulates the expression of the adhesion molecules, NCAM and integrin ß1, and reduces migration and invasiveness of ERMS cells in vitro, and ARS treatment reduces of about 50% the growth of ERMS xenografts in vivo. Our results are the first evidence of efficacy of ART derivatives in restraining ERMS growth in vivo, and suggest ARS as a potential candidate for therapeutic treatment of ERMS.


Subject(s)
Apoptosis/drug effects , Artemisinins/pharmacology , Reactive Oxygen Species/metabolism , Rhabdomyosarcoma, Embryonal/drug therapy , Rhabdomyosarcoma, Embryonal/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Artemisinins/therapeutic use , Artesunate , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Damage/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Integrin beta1/biosynthesis , Mice , MicroRNAs/biosynthesis , Neoplasm Invasiveness , Neoplasm Transplantation , Neural Cell Adhesion Molecules/biosynthesis , PAX7 Transcription Factor/biosynthesis , Pyridines/pharmacology , Transplantation, Heterologous , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
19.
J Neurochem ; 134(4): 618-28, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25951919

ABSTRACT

Leucine-rich repeat transmembrane proteins (LRRTMs) are single-spanning transmembrane proteins that belong to the family of synaptically localized adhesion molecules that play various roles in the formation, maturation, and function of synapses. LRRTMs are highly localized in the post-synaptic density; however, the mechanisms and significance of LRRTM synaptic clustering remain unclear. Here, we focus on the intracellular domain of LRRTMs and investigate its role in cell surface expression and synaptic clustering. The deletion of 55-56 residues in the cytoplasmic tail caused significantly reduced synaptic clustering of LRRTM1-4 in rat hippocampal neurons, whereas it simultaneously resulted in augmented LRRTM1-2 cell surface expression. A series of deletions and further single amino acid substitutions in the intracellular domain of LRRTM2 demonstrated that a previously uncharacterized sequence at the region of -16 to -13 from the C-terminus was responsible for efficient synaptic clustering and proper cell surface trafficking of LRRTMs. Furthermore, the clustering-deficient LRRTM2 mutant lost the ability to promote the accumulation of post-synaptic density protein-95 (PSD-95). These results suggest that trafficking to the cell surface and synaptic clustering of LRRTMs are regulated by a specific mechanism through this novel sequence in the intracellular domain that underlies post-synaptic molecular assembly and maturation. Leucine-rich repeat transmembrane proteins (LRRTMs) are synaptic cell adhesion molecules promoting synapse formation. LRRTMs are highly localized in the postsynaptic density. We report amino acid sequence YxxC in the intracellular domain of LRRTMs is responsible for the postsynaptic localization of LRRTMs. This novel amino acid sequence of LRRTMs facilitates synapse maturation. We propose this regulated synaptic clustering of LRRTMs by the intracellular domain presents a novel molecular mechanism of synapse maturation.


Subject(s)
Hippocampus/metabolism , Intracellular Membranes/metabolism , Neurons/metabolism , Proteins/metabolism , Synapses/metabolism , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Cells, Cultured , Chickens , HEK293 Cells , Humans , Leucine-Rich Repeat Proteins , Membrane Proteins , Mice , Nerve Tissue Proteins , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Proteins/genetics , Rats , Rats, Sprague-Dawley , Synapses/genetics
20.
PLoS One ; 10(4): e0122337, 2015.
Article in English | MEDLINE | ID: mdl-25875008

ABSTRACT

The proliferation and differentiation of neural stem cells are tightly controlled by intrinsic and extrinsic cues. Cell adhesion molecules are increasingly recognized as regulators of these processes. Here we report the expression of the olfactory cell adhesion molecule (OCAM/NCAM2/RNCAM) during mouse spinal cord development and in neural stem cells cultured as neurospheres. OCAM is also weakly expressed in the dormant adult stem cell niche around the central canal and is overexpressed after spinal cord injury. Both transmembrane (TM) and glycosylphosphatidylinositol (GPI)-linked isoforms are present in neurospheres. Electron microscopy and internalisation experiments revealed a dynamic trafficking of OCAM between the membrane and intracellular compartments. After differentiation, OCAM remains in neurons and oligodendrocytes whereas no expression is detected in astrocytes. Using OCAM knockout (KO) mice, we found that mutant spinal cord stem cells showed an increased proliferation and self-renewal rates although no effect on differentiation was observed. This effect was reversed by lentivirus-mediated re-introduction of OCAM. Mechanistically, we identified the ErbB2/Neu/HER2 protein as being implicated in the enhanced proliferation of mutant cells. ErbB2 protein expression and phosphorylation level were significantly increased in KO cells whereas no difference was observed at the mRNA level. Overexpression of ErbB2 in wild-type and mutant cells also increased their growth while reintroduction of OCAM in mutant cells reduced the level of phosphorylated ErbB2. These results indicate that OCAM exerts a posttranscriptional control on the ErbB2 signalling in spinal cord stem cells. This study adds further support for considering cell adhesion molecules as regulators of the ErbB signalling.


Subject(s)
Embryonic Stem Cells/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Receptor, ErbB-2/biosynthesis , Spinal Cord/metabolism , Animals , Cell Adhesion/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Neural Cell Adhesion Molecules/genetics , RNA, Messenger/biosynthesis , Receptor, ErbB-2/genetics , Signal Transduction/genetics , Spinal Cord/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...