Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 617(Pt 1): 8-15, 2022 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-35660877

RESUMEN

Mouse embryonic stem cells (mESCs) can be maintained in a pluripotent state under R2i culture conditions that inhibit the TGF-ß and ERK signaling pathways. BMP4 is another member of the TGF-ß family that plays a crucial role in maintaining the pluripotency state of mESCs. It has been reported that inhibition of BMP4 caused the death of R2i-grown cells. In this study, we used the loss-of-function approach to investigate the role of BMP4 signaling in mESC self-renewal. Inhibition of this pathway with Noggin and dorsomorphin, two bone morphogenetic protein (BMP) antagonists, elicited a quick death of the R2i-grown cells. We showed that the canonical pathway of BMP4 (BMP/SMAD) was dispensable for self-renewal and maintaining pluripotency of these cells. Transcriptome analysis of the BMPi-treated cells revealed that the p53 signaling and two adhesion (AD) and apoptotic mitochondrial change (MT) pathways could be involved in the cell death of the BMPi-treated cells. According to our results, inhibition of BMP4 signaling caused a decrease in cell adhesion and ECM detachment, which triggered anoikis in the R2i-grown cells. Altogether, these findings demonstrate that endogenous BMP signaling is required for the survival of mESCs under the R2i condition.


Asunto(s)
Células Madre Embrionarias de Ratones , Transducción de Señal , Animales , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Sistema de Señalización de MAP Quinasas , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
Int J Mol Sci ; 23(24)2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-36555270

RESUMEN

Acute myocardial infarction (AMI) is one of the major leading causes of death in humans globally. Recently, increased levels of recruited macrophages and AGE-albumin were observed in the hearts of humans and animals with acute myocardial infarction. Thus, the purposes of this study were to investigate whether the elevated levels of AGE-albumin from activated macrophage cells are implicated in ischemia-induced cardiomyocyte death and to develop therapeutic strategies for AMI based on its underlying molecular mechanisms with respect to AGEs. The present study demonstrated that activated macrophages and AGE-albumin were observed in heart tissues obtained from humans and rats with AMI incidences. In the cellular model of AMI, it was found that increased expression of AGE-albumin was shown to be co-localized with macrophages, and the presence of AGE-albumin led to increased expression of RAGE through the mitogen-activated protein kinase pathway. After revealing cardiomyocyte apoptosis induced by toxicity of the AGE-RAGE system, sRAGE-secreting MSCs were generated using the CRISPR/Cas9 platform to investigate the therapeutic effects of sRAGE-MSCs in an AMI rat model. Gene-edited sRAGE-MSCs showed greater therapeutic effects against AMI pathogenesis in rat models compared to mock MSCs, and promising results of the functional improvement of stem cells could result in significant improvements in the clinical management of cardiovascular diseases.


Asunto(s)
Infarto del Miocardio , Roedores , Humanos , Ratas , Animales , Infarto del Miocardio/metabolismo , Transducción de Señal , Reperfusión , Albúminas
3.
Mol Cell Proteomics ; 17(9): 1670-1684, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29848781

RESUMEN

Despite the progress in safety and efficacy of cell replacement therapy with pluripotent stem cells (PSCs), the presence of residual undifferentiated stem cells or proliferating neural progenitor cells with rostral identity remains a major challenge. Here we report the generation of a LIM homeobox transcription factor 1 alpha (LMX1A) knock-in GFP reporter human embryonic stem cell (hESC) line that marks the early dopaminergic progenitors during neural differentiation to find reliable membrane protein markers for isolation of midbrain dopaminergic neurons. Purified GFP positive cells in vitro exhibited expression of mRNA and proteins that characterized and matched the midbrain dopaminergic identity. Further quantitative proteomics analysis of enriched LMX1A+ cells identified several membrane-associated proteins including a polysialylated embryonic form of neural cell adhesion molecule (PSA-NCAM) and contactin 2 (CNTN2), enabling prospective isolation of LMX1A+ progenitor cells. Transplantation of human-PSC-derived purified CNTN2+ progenitors enhanced dopamine release from transplanted cells in the host brain and alleviated Parkinson's disease-related phenotypes in animal models. This study establishes an efficient approach for purification of large numbers of human-PSC-derived dopaminergic progenitors for therapeutic applications.


Asunto(s)
Biomarcadores/metabolismo , Membrana Celular/metabolismo , Separación Celular/métodos , Neuronas Dopaminérgicas/trasplante , Células Madre Embrionarias/citología , Enfermedad de Parkinson/terapia , Animales , Diferenciación Celular , Contactina 2/metabolismo , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas con Homeodominio LIM/metabolismo , Enfermedad de Parkinson/patología , Proteómica , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Factores de Transcripción/metabolismo
4.
J Proteome Res ; 17(12): 4320-4324, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30113170

RESUMEN

We performed proteomic analyses of human olfactory epithelial tissue to identify missing proteins using liquid chromatography-tandem mass spectrometry. Using a next-generation proteomic pipeline with a < 1.0% false discovery rate at the peptide and protein levels, we identified 3731 proteins, among which five were missing proteins (P0C7M7, P46721, P59826, Q658L1, and Q8N434). We validated the identified missing proteins using the corresponding synthetic peptides. No olfactory receptor (OR) proteins were detected in olfactory tissue, suggesting that detection of ORs would be very difficult. We also identified 49 and 50 alternative splicing variants mapped at the neXtProt and GENCODE databases, respectively, and 2000 additional single amino acid variants. This data set is available at the ProteomeXchange consortium via PRIDE repository (PXD010025).


Asunto(s)
Mucosa Olfatoria/química , Proteómica/métodos , Empalme Alternativo , Secuencia de Aminoácidos , Variación Genética , Humanos , Péptidos/análisis
5.
Expert Rev Proteomics ; 15(11): 911-922, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30358457

RESUMEN

INTRODUCTION: Human embryonic stem cells (hESCs) have unique biological features and attributes that make them attractive in various areas of biomedical research. With heightened applications, there is an ever increasing need for advancement of proteome analysis. Membrane proteins are one of the most important subset of hESC proteins as they can be used as surface markers. Areas covered: This review discusses commonly used surface markers of hESCs, and provides in-depth analysis of available hESC membrane proteome reports and the existence of these markers in many other cell types, especially cancer cells. Appreciating, existing ambiguity in the definition of a membrane protein, we have attempted a meta analysis of the published membrane protein reports of hESCs by using a combination of protein databases and prediction tools to find the most confident plasma membrane proteins in hESCs. Furthermore, responsiveness of plasma membrane proteins to differentiation has been discussed based on available transcriptome profiling data bank. Expert commentary: Combined transcriptome and membrane proteome analysis highlighted additional proteins that may eventually find utility as new cell surface markers.


Asunto(s)
Biomarcadores/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Proteínas de la Membrana/metabolismo , Biomarcadores/análisis , Biotina/metabolismo , Adhesión Celular , Membrana Celular/metabolismo , Enzimas/metabolismo , Perfilación de la Expresión Génica , Humanos , Canales Iónicos/metabolismo , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Proteómica/métodos , Fracciones Subcelulares
6.
Brain Behav Immun ; 66: 347-358, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28760504

RESUMEN

Alzheimer's disease (AD), which is the most commonly encountered neurodegenerative disease, causes synaptic dysfunction and neuronal loss due to various pathological processes that include tau abnormality and amyloid beta (Aß) accumulation. Aß stimulates the secretion and the synthesis of Receptor for Advanced Glycation End products (RAGE) ligand by activating microglial cells, and has been reported to cause neuronal cell death in Aß1-42 treated rats and in mice with neurotoxin-induced Parkinson's disease. The soluble form of RAGE (sRAGE) is known to reduce inflammation, and to decrease microglial cell activation and Aß deposition, and thus, it protects from neuronal cell death in AD. However, sRAGE protein has too a short half-life for therapeutic purposes. We developed sRAGE-secreting umbilical cord derived mesenchymal stem cells (sRAGE-MSCs) to enhance the inhibitory effects of sRAGE on Aß deposition and to reduce the secretion and synthesis of RAGE ligands in 5xFAD mice. In addition, these cells improved the viability of injected MSCs, and enhanced the protective effects of sRAGE by inhibiting the binding of RAGE and RAGE ligands in 5xFAD mice. These findings suggest sRAGE protein from sRAGE-MSCs has better protection against neuronal cell death than sRAGE protein or single MSC treatment by inhibiting the RAGE cell death cascade and RAGE-induce inflammation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apoptosis , Encéfalo/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Trasplante de Células Madre de Sangre del Cordón Umbilical , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Ratones Transgénicos , Microglía/metabolismo , Receptor para Productos Finales de Glicación Avanzada/genética
7.
Biochim Biophys Acta ; 1854(7): 788-97, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25770686

RESUMEN

The hippocampus is one of the most essential components of the human brain and plays an important role in learning and memory. The hippocampus has drawn great attention from scientists and clinicians due to its clinical importance in diseases such as Alzheimer's disease (AD), non-AD dementia, and epilepsy. Understanding the function of the hippocampus and related disease mechanisms requires comprehensive knowledge of the orchestration of the genome, epigenome, transcriptome, proteome, and post-translational modifications (PTMs) of proteins. The past decade has seen remarkable advances in the high-throughput sequencing techniques that are collectively called next generation sequencing (NGS). NGS enables the precise analysis of gene expression profiles in cells and tissues, allowing powerful and more feasible integration of expression data from the gene level to the protein level, even allowing "-omic" level assessment of PTMs. In addition, improved bioinformatics algorithms coupled with NGS technology are finally opening a new era for scientists to discover previously unidentified and elusive proteins. In the present review, we will focus mainly on the proteomics of the human hippocampus with an emphasis on the integrated analysis of genomics, epigenomics, transcriptomics, and proteomics. Finally, we will discuss our perspectives on the potential and future of proteomics in the field of hippocampal biology. This article is part of a Special Issue entitled: Neuroproteomics: Applications in Neuroscience and Neurology.


Asunto(s)
Enfermedad de Alzheimer , Epigenómica/métodos , Epilepsia , Hipocampo/metabolismo , Proteínas del Tejido Nervioso , Proteómica/métodos , Transcriptoma , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/patología , Hipocampo/patología , Humanos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Procesamiento Proteico-Postraduccional/genética
8.
Genome Res ; 23(8): 1283-94, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23674306

RESUMEN

Several studies have sought systematically to identify protein subcellular locations, but an even larger task is to map which of these proteins conditionally relocates in disease (the mislocalizome). Here, we report an integrative computational framework for mapping conditional location and mislocation of proteins on a proteome-wide scale, called a conditional location predictor (CoLP). Using CoLP, we mapped the locations of over 10,000 proteins in normal human brain and in glioma. The prediction showed 0.9 accuracy using 100 location tests of 20 randomly selected proteins. Of the 10,000 proteins, over 150 have a strong likelihood of mislocation under glioma, which is striking considering that few mislocation events have been identified in this disease previously. Using immunofluorescence and Western blotting in both primary cells and tissues, we successfully experimentally confirmed 15 mislocations. The most common type of mislocation occurs between the endoplasmic reticulum and the nucleus; for example, for RNF138, TLX3, and NFRKB. In particular, we found that the gene for the mislocating protein GFRA4 had a nonsynonymous point mutation in exon 2. Moreover, redirection of GFRA4 to its normal location, the plasma membrane, led to marked reductions in phospho-STAT3 and proliferation of glioma cells. This framework has the potential to track changes in protein location in many human diseases.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Proteoma/metabolismo , Encéfalo/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular , Progresión de la Enfermedad , Ontología de Genes , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Glioma/patología , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Humanos , Cinesinas/metabolismo , Anotación de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-ret/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas , Proteínas de Pez Cebra
9.
EMBO Rep ; 15(7): 801-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24825475

RESUMEN

Hypothalamic leptin signaling plays a central role in maintaining body weight homeostasis. Here, we show that clusterin/ApoJ, recently identified as an anorexigenic neuropeptide, is an important regulator in the hypothalamic leptin signaling pathway. Coadministration of clusterin potentiates the anorexigenic effect of leptin and boosts leptin-induced hypothalamic Stat3 activation. In cultured neurons, clusterin enhances receptor binding and subsequent endocytosis of leptin. These effects are mainly mediated through the LDL receptor-related protein-2 (Lrp2). Notably, inhibition of hypothalamic clusterin, Lrp2 or endocytosis abrogates anorexia and hypothalamic Stat3 activation caused by leptin. These findings propose a novel regulatory mechanism in central leptin signaling pathways.


Asunto(s)
Clusterina/metabolismo , Endocitosis/fisiología , Leptina/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Transducción de Señal , Animales , Clusterina/deficiencia , Clusterina/genética , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Unión Proteica , Receptores de Leptina/metabolismo
10.
Brain ; 138(Pt 12): 3610-22, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26490328

RESUMEN

Currently there is no neuroprotective or neurorestorative therapy for Parkinson's disease. Here we report that transient receptor potential vanilloid 1 (TRPV1) on astrocytes mediates endogenous production of ciliary neurotrophic factor (CNTF), which prevents the active degeneration of dopamine neurons and leads to behavioural recovery through CNTF receptor alpha (CNTFRα) on nigral dopamine neurons in both the MPP(+)-lesioned or adeno-associated virus α-synuclein rat models of Parkinson's disease. Western blot and immunohistochemical analysis of human post-mortem substantia nigra from Parkinson's disease suggests that this endogenous neuroprotective system (TRPV1 and CNTF on astrocytes, and CNTFRα on dopamine neurons) might have relevance to human Parkinson's disease. Our results suggest that activation of astrocytic TRPV1 activates endogenous neuroprotective machinery in vivo and that it is a novel therapeutic target for the treatment of Parkinson's disease.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuroprotección , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Sustancia Negra/metabolismo , Animales , Subunidad alfa del Receptor del Factor Neurotrófico Ciliar/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Femenino , Humanos , Regeneración Nerviosa , Enfermedad de Parkinson/fisiopatología , Ratas , Sustancia Negra/citología , Sustancia Negra/patología , Canales Catiónicos TRPV/metabolismo
11.
J Proteome Res ; 14(1): 214-23, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25384129

RESUMEN

Microglial activation in the central nervous system is a key event in the neuroinflammation that accompanies neurodegenerative diseases such as Alzheimer's disease (AD). Among cytokines involved in microglial activation, amyloid ß (Aß) peptide is known to be a key molecule in the induction of diverse inflammatory products, which may lead to chronic inflammation in AD. However, proteomic studies of microglia in AD are limited due to lack of proper cell or animal model systems. In this study, we performed a proteomic analysis of Aß-stimulated human microglial cells using SILAC (stable isotope labeling with amino acids in cell culture) combined with LC-MS/MS. Results showed that 60 proteins increased or decreased their abundance by 1.5 fold or greater. Among these, ER-resident proteins such as SERPINH1, PDIA6, PDIA3, and PPIB were revealed to be key molecular biomarkers of human microglial activation by validation of the proteomic results by immunostaining, PCR, ELISA, and Western blot. Taken together, our data suggest that ER proteins play an essential role in human microglial activation by Aß and may be important molecular therapeutic targets for treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Microglía/fisiología , Proteoma/metabolismo , Enfermedad de Alzheimer/metabolismo , Secuencia de Aminoácidos , Animales , Biomarcadores/metabolismo , Línea Celular , Expresión Génica , Ontología de Genes , Humanos , Ratones , Datos de Secuencia Molecular , Mapeo de Interacción de Proteínas , Proteoma/genética , Proteómica , Espectrometría de Masas en Tándem
13.
Stem Cell Res ; 77: 103426, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38678980

RESUMEN

GATA6 is expressed during early embryogenesis and localizes to endoderm- and mesoderm-derived tissues during later embryogenesis. Here, we established a human induced pluripotent stem cell (hiPSC) line expressing EGFP under GATA6 gene. EGFP coding sequence was introduced into the C-terminus of GATA6 in KSCBi017-A hiPSCs through homologous recombination using CRISPR/Cas9 system. The successfully edited line, KSCBi017-A-1, was selected and confirmed by sequencing. The line had a normal karyotype and exhibited potential to differentiate into three germ layers while it expressed EGFP upon endoderm induction. KSCBi017-A-1 cells can be used to monitor the expression of GATA6 during differentiation. This cell line is available from Korea National Stem Cell Bank.


Asunto(s)
Sistemas CRISPR-Cas , Factor de Transcripción GATA6 , Proteínas Fluorescentes Verdes , Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Humanos , Factor de Transcripción GATA6/metabolismo , Factor de Transcripción GATA6/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Fluorescentes Verdes/genética , Línea Celular , Diferenciación Celular
14.
Stem Cell Res ; 76: 103358, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38447455

RESUMEN

Parkinson's disease is a degenerative brain disorder characterized by dopamine neuronal degeneration and dopamine transporter loss. In this study, we generated an induced pluripotent stem cell (iPSC) line, KNIHi001-A, from the peripheral blood mononuclear cells (PBMCs) of a 76-year-old man with Parkinson's disease. The non-integrating Sendai virus was used to reprogram iPSCs. iPSCs exhibit pluripotent markers, a normal karyotype, viral clearance, and the ability to differentiate into the three germ layers.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Masculino , Humanos , Anciano , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Parkinson/metabolismo , Leucocitos Mononucleares/metabolismo , Estratos Germinativos/metabolismo , Virus Sendai/genética , Reprogramación Celular , Diferenciación Celular/fisiología
15.
J Proteome Res ; 12(1): 97-105, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23286719

RESUMEN

Human chromosome 11 is the third gene-rich chromosome having 1304 protein-coding genes. According to the GeneCards, this chromosome contains 240 genes related to diseases, as it is well known as a disease-rich chromosome. Although there are many protein-coding genes, the proteomic identification ratio is rather low. As a model study, human hippocampal tissues from patients suffering from Alzheimer's disease and epilepsy were prepared to evaluate the gene-centric statistics related to the gene expression and disorders of chromosome 11. A total of 8828 protein coding genes from brain tissues were extensively off-gel fractionated and profiled by a high resolution mass spectrometer with collision induced dissociation and electron transfer dissociation. Five-hundred twenty-three of the proteins from brain tissues were determined to belong to chromosome 11, representing 37% of the proteins reported in the Global Proteome Machine Database. We extracted gene clusters from a specific biological process or molecular function in gene ontology, among which the olfactory receptor genes showed the largest cluster on chromosome 11. Analysis of the proteome data set from the hippocampus provides a significant network associated with genes and proteins and leads to new insights into the biological and genetic mechanisms of chromosome 11-specific diseases such as Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Cromosomas Humanos Par 11 , Epilepsia , Hipocampo/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 11/metabolismo , Bases de Datos de Proteínas , Epilepsia/genética , Epilepsia/metabolismo , Expresión Génica , Humanos , Persona de Mediana Edad , Proteínas/clasificación , Proteínas/genética , Proteínas/metabolismo , Proteoma
16.
Cell Mol Neurobiol ; 33(5): 615-24, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23605681

RESUMEN

Aging is an inevitable process that occurs in the whole body system accompanying with many functional and morphological changes. Inflammation is known as one of age-related factors, and inflammatory changes could enhance mortality risk. In this study, we compared immunoreactivities of inflammatory cytokines, such as interleukin (IL)-2 (a pro-inflammatory cytokine), its receptor (IL-2R), IL-4 (an anti-inflammatory cytokine), and its receptor (IL-4R) in the cervical and lumbar spinal cord of young adult (2-3 years old) and aged (10-12 years old) beagle dogs using immunohistochemistry and western blotting. IL-2 and IL-2R-immunoreactive nerve cells were found throughout the gray matter of the cervical and lumbar spinal cord of young adult and aged dogs. In the spinal cord neurons of the aged dog, immunoreactivity and protein levels were apparently increased compared with those in the young adult dog. Change patterns of IL-4- and IL-4R-immunoreactive cells and their protein levels were also similar to those in IL-2 and IL-2R; however, IL-4 and IL-4R immunoreactivity in the periphery of the neuronal cytoplasm in the aged dog was much stronger than that in the young adult dog. These results indicate that the increase of inflammatory cytokines and their receptors in the aged spinal cord might be related to maintaining a balance of inflammatory reaction in the spinal cord during normal aging.


Asunto(s)
Envejecimiento/patología , Inflamación/patología , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Médula Espinal/patología , Animales , Western Blotting , Perros , Inmunohistoquímica , Receptores de Interleucina-2/metabolismo , Receptores de Interleucina-4/metabolismo , Médula Espinal/metabolismo
17.
Neurochem Res ; 38(5): 1045-54, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23504294

RESUMEN

Beta-catenin, a transcription factor, plays a critical role in cell survival and degradation after stroke. In this study, we examined changes of expression in beta-catenin in the hippocampal CA1 region of the gerbil following 5 min of transient cerebral ischemia. We observed neuronal damage using cresyl violet staining, neuronal nuclei immunohistochemistry and Fluro-Jade B immunofluorescence. Four days after ischemia-reperfusion (I-R), most of pyramidal cells in the CA1 region were damaged. In addition, early damage in dendrites was detected 1 day after I-R by immunohistochemical staining for microtubule-associated protein 2 (MAP-2), and MAP-2 immunoreactivity was hardly detected in the CA1 region 4 days after I-R. We found that beta-catenin (a synapse-enriched cell adhesion molecule) was well expressed in dendrites before I-R. Its immunoreactivity was well colocalized with MAP-2. Chronological change of beta-catenin immunoreactivity was novelty in the present study. Twelve hours after I-R, its immunoreactivity was decreased in the stratum radiatum of the CA1 region, however, its immunoreactivity was increased 1 and 2 days after I-R, and decreased sharply 4 days after I-R. However, we did not find any change in beta-catenin immunoreactivity in the CA2 and CA3 region. In brief, we suggest that early change of beta-catenin expression in the stratum pyramidale of ischemic hippocampal CA1 region is associated with early dendrite damage following transient cerebral ischemia.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Ataque Isquémico Transitorio/metabolismo , beta Catenina/metabolismo , Animales , Gerbillinae , Masculino
18.
Neurochem Res ; 38(9): 1980-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23836293

RESUMEN

Apripiprazole (APZ) is well known as an atypical antipsychotic and antidepressant. In the present study, we investigated effects of APZ on cell proliferation and neuronal differentiation in the dentate gyrus (DG) of the adolescent mouse using BruU, Ki-67 and doublecortin (DCX) immunohistochemistry. BruU, Ki-67 and DCX-positive (+) cells were easily detected in the subgranular zone of the DG in the vehicle- and APZ-treated group. We found that in the 8 mg/kg APZ-treated group numbers of Ki-67(+), DCX(+) and BrdU(+)/DCX(+) cells were significantly increased compared with those in the vehicle-treated group. We also found that maturation and complexity of DCX(+) dendrites in the 8 mg/kg APZ-treated group was well improved compared with those in the vehicle-treated group. In addition, markedly decreased lipid peroxidation and increased superoxide dismutase 2 (SOD2) level were observed in the DG of the 8 mg/kg APZ-treated group. Our present findings indicate that APZ can enhance cell proliferation and neuroblast differentiation, particularly maturation and complexity of neuroblast dendrites, in the DG via decreasing lipid peroxidation and increasing SOD2 level.


Asunto(s)
Antipsicóticos/farmacología , Giro Dentado/efectos de los fármacos , Neuronas/efectos de los fármacos , Piperazinas/farmacología , Quinolonas/farmacología , Superóxido Dismutasa/metabolismo , Animales , Aripiprazol , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Giro Dentado/citología , Giro Dentado/enzimología , Proteína Doblecortina , Masculino , Ratones , Ratones Endogámicos ICR
19.
Genome Res ; 19(9): 1622-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19470904

RESUMEN

We present the first Korean individual genome sequence (SJK) and analysis results. The diploid genome of a Korean male was sequenced to 28.95-fold redundancy using the Illumina paired-end sequencing method. SJK covered 99.9% of the NCBI human reference genome. We identified 420,083 novel single nucleotide polymorphisms (SNPs) that are not in the dbSNP database. Despite a close similarity, significant differences were observed between the Chinese genome (YH), the only other Asian genome available, and SJK: (1) 39.87% (1,371,239 out of 3,439,107) SNPs were SJK-specific (49.51% against Venter's, 46.94% against Watson's, and 44.17% against the Yoruba genomes); (2) 99.5% (22,495 out of 22,605) of short indels (< 4 bp) discovered on the same loci had the same size and type as YH; and (3) 11.3% (331 out of 2920) deletion structural variants were SJK-specific. Even after attempting to map unmapped reads of SJK to unanchored NCBI scaffolds, HGSV, and available personal genomes, there were still 5.77% SJK reads that could not be mapped. All these findings indicate that the overall genetic differences among individuals from closely related ethnic groups may be significant. Hence, constructing reference genomes for minor socio-ethnic groups will be useful for massive individual genome sequencing.


Asunto(s)
Pueblo Asiatico/genética , Genoma Humano/genética , Análisis de Secuencia de ADN/métodos , Biología Computacional/métodos , Bases de Datos Genéticas , Femenino , Genómica/métodos , Humanos , Mutación INDEL , Corea (Geográfico) , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple/genética , Estándares de Referencia
20.
Electrophoresis ; 33(24): 3756-63, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23161002

RESUMEN

Microglial cells act as the first and main form of active immune defense in the central nervous system related to inflammation and neurodegenerative disease. Lipopolysaccharide (LPS) induces many genes encoding inflammatory mediators, including cytokines such as tumor necrosis factor-α, interleukin-1ß, (IL-1ß), and IL-6, chemokines, and prostaglandins in microglial cells. Quantitative proteomics methods with isobaric chemical labeling using tandem mass tags and 2D-nano LC-ESI-MS/MS were used to systematically analyze proteomic changes in microglia responding to LPS stimulation. As a result, we found that the expression level of 21 proteins in human microglial cells changed after activation. Among those, one of the strong mitogen-activated protein kinase (MAPK) regulator proteins, CMPK1 was highly upregulated after LPS stimulation in human microglial cells. We detected and validated upregulation of MAPK including ERK1/2, p38, and SAPK/JNK by immunohistochemistry and Western blotting. NFκB, strong transcription factor of CMPK1, was translocated to the nucleus from the cytosol by high contents screening after LPS stimulation. Taken together, we conclude that MAPK signaling plays an important role in LPS-induced human microglial activation related to inflammatory response.


Asunto(s)
Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Microglía/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteoma/efectos de los fármacos , Secuencia de Aminoácidos , Línea Celular , Electroforesis en Gel Bidimensional , Activación Enzimática/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Microglía/citología , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Proteómica/métodos , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA