Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cereb Cortex ; 33(8): 4779-4793, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-36169576

RESUMO

A microdeletion of approximately 2.4 Mb at the 8p23 terminal region has been identified in a Taiwanese autistic boy. Among the products transcribed/translated from genes mapped in this region, the reduction of DLGAP2, a postsynaptic scaffold protein, might be involved in the pathogenesis of autism spectrum disorder (ASD). DLGAP2 protein was detected in the hippocampus yet abolished in homozygous Dlgap2 knockout (Dlgap2 KO) mice. In this study, we characterized the hippocampal phenotypes in Dlgap2 mutant mice. Dlgap2 KO mice exhibited impaired spatial memory, indicating poor hippocampal function in the absence of DLGAP2. Aberrant expressions of postsynaptic proteins, including PSD95, SHANK3, HOMER1, GluN2A, GluR2, mGluR1, mGluR5, ßCAMKII, ERK1/2, ARC, BDNF, were noticed in Dlgap2 mutant mice. Further, the spine density was increased in Dlgap2 KO mice, while the ratio of mushroom-type spines was decreased. We also observed a thinner postsynaptic density thickness in Dlgap2 KO mice at the ultrastructural level. These structural changes found in the hippocampus of Dlgap2 KO mice might be linked to impaired hippocampus-related cognitive functions such as spatial memory. Mice with Dlgap2 deficiency, showing signs of intellectual disability, a common co-occurring condition in patients with ASD, could be a promising animal model which may advance our understanding of ASD.


Assuntos
Transtorno do Espectro Autista , Animais , Camundongos , Espinhas Dendríticas/metabolismo , Hipocampo/metabolismo , Transtornos da Memória , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Modelos Genéticos , Proteínas do Tecido Nervoso/metabolismo , Memória Espacial , Coluna Vertebral/metabolismo , Sinapses/metabolismo
2.
Int J Mol Sci ; 21(14)2020 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-32664674

RESUMO

Connective tissue growth factor (CTGF) plays important roles in the development and regeneration of the connective tissue, yet its function in the nervous system is still not clear. CTGF is expressed in some distinct regions of the brain, including the dorsal endopiriform nucleus (DEPN) which has been recognized as an epileptogenic zone. We generated a forebrain-specific Ctgf knockout (FbCtgf KO) mouse line in which the expression of Ctgf in the DEPN is eliminated. In this study, we adopted a pentylenetetrazole (PTZ)-induced seizure model and found similar severity and latencies to death between FbCtgf KO and WT mice. Interestingly, there was a delay in the seizure reactions in the mutant mice. We further observed reduced c-fos expression subsequent to PTZ treatment in the KO mice, especially in the hippocampus. While the densities of astrocytes and microglia in the hippocampus were kept constant after acute PTZ treatment, microglial morphology was different between genotypes. Our present study demonstrated that in the FbCtgf KO mice, PTZ failed to increase neuronal activity and microglial response in the hippocampus. Our results suggested that inhibition of Ctgf function may have a therapeutic potential in preventing the pathophysiology of epilepsy.


Assuntos
Astrócitos/fisiologia , Fator de Crescimento do Tecido Conjuntivo/deficiência , Genes fos , Microglia/fisiologia , Prosencéfalo/metabolismo , Convulsões/fisiopatologia , Animais , Astrócitos/efeitos dos fármacos , Contagem de Células , Claustrum/efeitos dos fármacos , Claustrum/metabolismo , Fator de Crescimento do Tecido Conjuntivo/fisiologia , Convulsivantes/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Pentilenotetrazol/toxicidade , Prosencéfalo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , Convulsões/induzido quimicamente , Convulsões/genética , Convulsões/patologia
3.
J Vasc Res ; 49(1): 24-32, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21986482

RESUMO

BACKGROUND/AIMS: Restenosis after a percutaneous coronary intervention (PCI) during treatment for coronary artery disease is closely related to smooth muscle cell (SMC) proliferation and migration. In this study, we investigated the effects of caffeic acid phenethyl ester (CAPE) and its underlying mechanism on human coronary SMCs (HCSMCs) after platelet-derived growth factor-BB (PDGF-BB) stimulation in vitro. METHODS AND RESULTS: The results showed that CAPE inhibited proliferation and migration, and induced apoptosis. Concomitantly, CAPE inhibited activation of AKT1, MEK1 and ERK1/2 signaling molecules at 10-60 min after CAPE treatment. As revealed by flow cytometry, DNA fragmentation and TUNEL assay, the cells accumulated at the sub-G(1) phase, and cell apoptosis was observed after 30 and 90 µM CAPE treatment for 72 h. CAPE triggered the release of cytochrome c from mitochondria to cytosol, upregulated the proapoptotic gene Bax and downregulated the antiapoptotic gene Bcl-2. Upregulation of caspase-9 and caspase-3 indicated that CAPE precipitated the mitochondrion-dependent apoptotic signaling pathway. CONCLUSIONS: These results provide a molecular explanation for the antiproliferation, antimigration and proapoptotic effects of CAPE on HCSMCs after PDGF-BB stimulation.


Assuntos
Apoptose/efeitos dos fármacos , Ácidos Cafeicos/farmacologia , Vasos Coronários/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Álcool Feniletílico/análogos & derivados , Proteínas Proto-Oncogênicas c-sis/farmacologia , Becaplermina , Caspases/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Vasos Coronários/fisiologia , Citocromos c/metabolismo , Humanos , Músculo Liso Vascular/citologia , Álcool Feniletílico/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/análise
4.
Transl Psychiatry ; 12(1): 411, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-36163151

RESUMO

Mood disorders are an important public health issue and recent advances in genomic studies have indicated that molecules involved in neurodevelopment are causally related to mood disorders. BLM-s (BCL-2-like molecule, small transcript isoform), a BH3-only proapoptotic BCL-2 family member, mediates apoptosis of postmitotic immature neurons during embryonic cortical development, but its role in the adult brain is unknown. To better understand the physiological role of Blm-s gene in vivo, we generated a Blm-s-knockout (Blm-s-/-) mouse. The Blm-s-/- mice breed normally and exhibit grossly normal development. However, global depletion of Blm-s is highly associated with depression- and anxiety-related behaviors in adult mutant mice with intact learning and memory capacity. Functional magnetic resonance imaging of adult Blm-s-/- mice reveals reduced connectivity mainly in the ventral dentate gyrus (vDG) of the hippocampus with no alteration in the dorsal DG connectivity and in total hippocampal volume. At the cellular level, BLM-s is expressed in DG granule cells (GCs), and Blm-s-/- mice show reduced dendritic complexity and decreased spine density in mature GCs. Electrophysiology study uncovers that mature vGCs in adult Blm-s-/- DG are intrinsically more excitable. Interestingly, certain genetic variants of the human Blm homologue gene (VPS50) are significantly associated with depression traits from publicly resourced UK Biobank data. Taken together, BLM-s is required for the hippocampal mood control function. Loss of BLM-s causes abnormality in the electrophysiology and morphology of GCs and a disrupted vDG neural network, which could underlie Blm-s-null-associated anxiety and depression.


Assuntos
Hipocampo , Neurogênese , Adulto , Animais , Apoptose , Giro Denteado , Hipocampo/diagnóstico por imagem , Humanos , Camundongos , Neurogênese/genética , Neurônios , Proteínas Proto-Oncogênicas c-bcl-2 , RecQ Helicases
5.
Sci Rep ; 10(1): 10857, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616804

RESUMO

PHRF1 is involved in transforming growth factor ß (TGF-ß) signaling to constrain the formation of acute promyelocytic leukemia (APL) in mouse APL models. PHRF1 also participates in modulating non-homologous end-joining. However, the role of PHRF1 in mammalian dendrite architecture and synaptic plasticity is unclear. Here, we investigated the role of PHRF1 in dendritic formation in the murine hippocampus using Camk2a promoter driven-iCre recombinase to conduct a PHRF1 conditional knockout, namely PHRF1Δ/Δ, in the forebrain region. PHRF1Δ/Δ mice developed normally, but exhibited anxiety-like behaviors and displayed defective spatial memory. Alterations of dendritic complexity in apical and basal dendrites of pyramidal neurons were noticed in PHRF1Δ/Δ mutants. Furthermore, electrical stimulation in the hippocampal CA1 region after the TGF-ß1 treatment showed a reduced synaptic plasticity in PHRF1Δ/Δ mice. Immunoblotting analysis indicated that PHRF1 ablation affected the TGF-ß signaling. Collectively, our results demonstrate that PHRF1 is important for the dendritic architecture and required for spatial memory formation in the hippocampus.


Assuntos
Dendritos/química , Hipocampo/metabolismo , Proteínas de Membrana/fisiologia , Células Piramidais/metabolismo , Domínios RING Finger/fisiologia , Memória Espacial/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Dendritos/fisiologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Plasticidade Neuronal , Células Piramidais/citologia , Transdução de Sinais , Fator de Crescimento Transformador beta/genética
6.
Front Neuroanat ; 13: 75, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31417371

RESUMO

Myotonic dystrophy (Dystrophia Myotonica; DM) is the most common adult-onset muscular dystrophy and its brain symptoms seriously affect patients' quality of life. It is caused by extended (CTG)n expansions at 3'-UTR of DMPK gene (DM type 1, DM1) or (CCTG)n repeats in the intron 1 of CNBP gene (DM type 2, DM2) and the sequestration of Muscleblind-like (MBNL) family proteins by transcribed (CUG)n RNA hairpin is the main pathogenic mechanism for DM. The MBNL proteins are splicing factors regulating posttranscriptional RNA during development. Previously, Mbnl knockout (KO) mouse lines showed molecular and phenotypic evidence that recapitulate DM brains, however, detailed morphological study has not yet been accomplished. In our studies, control (Mbnl1 +/+; Mbnl2 cond/cond; Nestin-Cre -/-), Mbnl2 conditional KO (2KO, Mbnl1 +/+; Mbnl2 cond/cond; Nestin-Cre +/-) and Mbnl1/2 double KO (DKO, Mbnl1 ΔE3/ΔE3; Mbnl2 cond/cond; Nestin-Cre +/-) mice were generated by crossing three individual lines. Immunohistochemistry for evaluating density and distribution of cortical neurons; Golgi staining for depicting the dendrites/dendritic spines; and electron microscopy for analyzing postsynaptic ultrastructure were performed. We found distributional defects in cortical neurons, reduction in dendritic complexity, immature dendritic spines and alterations of postsynaptic densities (PSDs) in the mutants. In conclusion, loss of function of Mbnl1/2 caused fundamental defects affecting neuronal distribution, dendritic morphology and postsynaptic architectures that are reminiscent of predominantly immature and fetal phenotypes in DM patients.

7.
Front Neuroanat ; 13: 16, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30842729

RESUMO

Connective tissue growth factor (CTGF) is a secreted extracellular matrix-associated protein, which play a role in regulating various cellular functions. Although the expression of CTGF has been reported in the cortical subplate, its function is still not clear. Thus, to explore the significance of CTGF in the brain, we created a forebrain-specific Ctgf knockout (FbCtgf KO) mouse model. By crossing Ctgf fl/fl mice with Emx1-Cre transgenic mice, in which the expression of Cre is prenatally initiated, the full length Ctgf is removed in the forebrain structures. In young adult (2-3 months old) FbCtgf KO mice, subplate markers such as Nurr1 and Cplx3 are still expressed in the cortical layer VIb; however, the density of the subplate neurons is increased. Interestingly, in these mutants, we found a reduced structural complexity in the subplate neurons. The distribution patterns of neurons and glial cells, examined by immunohistochemistry, are comparable between genotypes in the somatosensory cortex. However, increased densities of mature oligodendrocytes, but not immature ones, were noticed in the external capsule underneath the cortical layer VIb in young adult FbCtgf KO mice. The features of myelinated axons in the external capsule were then examined using electron microscopy. Unexpectedly, the thickness of the myelin sheath was reduced in middle-aged (>12 months old), but not young adult FbCtgf KO mice. Our results suggest a secretory function of the subplate neurons, through the release of CTGF, which regulates the density and dendritic branching of subplate neurons as well as the maturation and function of nearby oligodendrocytes in the white matter.

8.
Sci Rep ; 7: 45646, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28368047

RESUMO

Vilse/Arhgap39 is a Rho GTPase activating protein (RhoGAP) and utilizes its WW domain to regulate Rac/Cdc42-dependent morphogenesis in Drosophila and murine hippocampal neurons. However, the function of Vilse in mammalian dendrite architecture and synaptic plasticity remained unclear. In the present study, we aimed to explore the possible role of Vilse in dendritic structure and synaptic function in the brain. Homozygous knockout of Vilse resulted in premature embryonic lethality in mice. Changes in dendritic complexity and spine density were noticed in hippocampal neurons of Camk2a-Cre mediated forebrain-specific Vilse knockout (VilseΔ/Δ) mice. VilseΔ/Δ mice displayed impaired spatial memory in water maze and Y-maze tests. Electrical stimulation in hippocampal CA1 region revealed that the synaptic transmission and plasticity were defected in VilseΔ/Δ mice. Collectively, our results demonstrate that Vilse is essential for embryonic development and required for spatial memory.


Assuntos
Região CA1 Hipocampal/fisiologia , Dendritos/fisiologia , Proteínas Ativadoras de GTPase/metabolismo , Plasticidade Neuronal/fisiologia , Células Piramidais/fisiologia , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Dendritos/metabolismo , Feminino , Proteínas Ativadoras de GTPase/genética , Genes Letais/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Plasticidade Neuronal/genética , Células Piramidais/metabolismo , Memória Espacial/fisiologia , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia
9.
PLoS One ; 7(11): e49102, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23145084

RESUMO

BACKGROUND: PPARγ and RBP4 are known to regulate lipid and glucose metabolism and insulin resistance. The influences of PPARγ (C1431T and Pro12Ala) and RBP4 (-803GA) polymorphisms on metabolic syndrome in HIV-infected patients receiving anti-retroviral therapy were examined in this study. MATERIALS AND METHODS: A cross-sectional study of HIV-1 infected adults with antiretroviral therapy for more than one year in the National Cheng Kung University Hospital was conducted. The gene polymorphisms were determined by quantitative PCR. RESULTS: Ninety-one patients were included in the study. Eighty-two (90.1%) patients were males with a mean age of 44.4 years. For the C1431T polymorphism in PPARγ, while patients with the T allele (48.4%) had trends toward lower rate of hypertriglyceridemia, the borderline significance together with insignificant power did not support the protective effect of the T allele against development of hypertriglyceridemia. For the Pro12Ala polymorphism in PPARγ, although patients with the Pro/Ala genotype (8.8%) had a higher level of serum LDL (138.0 vs. 111.5 mg/dl, P = 0.04) and trends toward higher rates of hypercholesterolemia and serum LDL>110 mg/dl, these variables were found to be independent of the Pro/Ala genotype in the multivariate analysis. For the -803GA polymorphism in RBP4, patients with the A allele (23.1%) more often had insulin resistance (HOMA>3.8; 33.3 vs. 8.7%, P = 0.01) and more often received anti-hypoglycemic drugs (14.3 vs. 1.4%, P = 0.04). The detrimental effect of the A allele in RBP4 -803GA polymorphism on development of insulin resistance was supported by the multivariate analysis adjusting for covariates. CONCLUSION: The impacts of PPARγ C1431T and Pro12Ala polymorphisms on metabolism in HIV-infected patients are not significant. RBP4 -803GA polymorphism has increased risk of insulin resistance in HIV-infected patients with anti-retroviral therapy.


Assuntos
Infecções por HIV , Síndrome Metabólica , PPAR gama/genética , Proteínas Plasmáticas de Ligação ao Retinol/genética , Adulto , Alelos , Terapia Antirretroviral de Alta Atividade , Estudos Transversais , Feminino , Predisposição Genética para Doença , Genótipo , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Infecções por HIV/metabolismo , HIV-1/genética , Humanos , Resistência à Insulina/genética , Masculino , Síndrome Metabólica/complicações , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/metabolismo , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA