Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(32): e2204779119, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914128

RESUMEN

Earlier work has shown that siRNA-mediated reduction of the SUPT4H or SUPT5H proteins, which interact to form the DSIF complex and facilitate transcript elongation by RNA polymerase II (RNAPII), can decrease expression of mutant gene alleles containing nucleotide repeat expansions differentially. Using luminescence and fluorescence assays, we identified chemical compounds that interfere with the SUPT4H-SUPT5H interaction and then investigated their effects on synthesis of mRNA and protein encoded by mutant alleles containing repeat expansions in the huntingtin gene (HTT), which causes the inherited neurodegenerative disorder, Huntington's Disease (HD). Here we report that such chemical interference can differentially affect expression of HTT mutant alleles, and that a prototypical chemical, 6-azauridine (6-AZA), that targets the SUPT4H-SUPT5H interaction can modify the biological response to mutant HTT gene expression. Selective and dose-dependent effects of 6-AZA on expression of HTT alleles containing nucleotide repeat expansions were seen in multiple types of cells cultured in vitro, and in a Drosophila melanogaster animal model for HD. Lowering of mutant HD protein and mitigation of the Drosophila "rough eye" phenotype associated with degeneration of photoreceptor neurons in vivo were observed. Our findings indicate that chemical interference with DSIF complex formation can decrease biochemical and phenotypic effects of nucleotide repeat expansions.


Asunto(s)
Azauridina , Proteína Huntingtina , Enfermedad de Huntington , Proteínas Mutantes , Mutación , Proteínas Nucleares , Fenotipo , Proteínas Represoras , Factores de Elongación Transcripcional , Alelos , Animales , Azauridina/farmacología , Células Cultivadas , Expansión de las Repeticiones de ADN , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Mediciones Luminiscentes , Proteínas Mutantes/biosíntesis , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Células Fotorreceptoras de Invertebrados/efectos de los fármacos , Proteínas Represoras/metabolismo , Factores de Elongación Transcripcional/metabolismo
2.
J Vet Pharmacol Ther ; 47(1): 36-47, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37593974

RESUMEN

Toltrazuril (TZR) is currently the only registered chemotherapeutic drug in the European Union for the treatment of Cystoisospora suis. This study investigated the comparative pharmacokinetics and tissue concentration-time profiles of TZR and its active metabolite, toltrazuril sulfone (TZR-SO2 ), after oral (per os, p.o.) and intramuscular (i.m.) administration to suckling piglets. Following a single administration of TZR orally at 50 mg/piglet or intramuscularly at 45 mg/piglet, higher concentrations of TZR and TZR-SO2 were observed in all three investigated tissues after p.o. administration. The mean TZR concentration in serum peaked at 14 µg/mL (34.03 h) and 5.36 µg/mL (120 h), while TZR-SO2 peaked at 14.12 µg/mL (246 h) and 9.92 µg/mL (330 h) after p.o. and i.m. administration, respectively. TZR was undetectable in the liver after p.o. administration (18 days) and in the jejunum (24 days) after i.m. injection, while TZR-SO2 was still detectable in all three tissues after 36 days regardless of administration routes. This study showed that p.o. formulation exhibited faster absorption and higher serum/tissue TZR/TZR-SO2 concentrations than i.m. formulation. Both formulations generated sufficient therapeutic concentrations in the serum and jejunum, and sustained enough time to protect against Cystoisospora suis infection in the piglets.


Asunto(s)
Coccidiostáticos , Animales , Porcinos , Administración Oral , Triazinas , Sulfonas , Inyecciones Intramusculares/veterinaria
3.
Emerg Infect Dis ; 29(8): 1634-1637, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37486207

RESUMEN

A CTX-M-65‒producing Salmonella enterica serovar Infantis clone, probably originating in Latin America and initially reported in the United States, has emerged in Taiwan. Chicken meat is the most likely primary carrier. Four of the 9 drug resistance genes have integrated into the chromosome: blaCTX-M-65, tet(A), sul1, and aadA1.


Asunto(s)
Salmonella enterica , beta-Lactamasas , Estados Unidos , Animales , Serogrupo , Taiwán/epidemiología , beta-Lactamasas/genética , Salmonella enterica/genética , Cromosomas , Antibacterianos/farmacología , Pollos , Plásmidos , Farmacorresistencia Bacteriana Múltiple/genética
4.
IUBMB Life ; 74(8): 812-825, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35102668

RESUMEN

Recent advances in induced pluripotent stem cell (iPSC) technology have allowed researchers to generate neurodegenerative disease-specific iPSCs and use the cells to derive a variety of relevant cell populations for laboratory modeling and drug testing. Nevertheless, these efforts have faced challenges related to immaturity and lack of complex developmental niches in the derived cell populations, limiting the utility of these in vitro models of neurodegenerative disease. Such limitations may be overcome by using human iPSC technology to generate three-dimensional (3D) brain organoids, which better recapitulate in vivo tissue architecture than traditional neuronal cultures to provide more complex and representative disease models and drug testing systems. In this review, we focus on the application of pluripotent stem cell-derived central nervous system (CNS) organoids to model neurodegenerative diseases. We first summarize recent progress in generating and characterizing various CNS organoids from pluripotent stem cells. We then review the application of CNS organoids for modeling several different human neurodegenerative diseases. We also describe several novel pathological mechanisms and drugs that were studied using patient iPSC-derived CNS organoids. Finally, we discuss remaining challenges and emerging opportunities for the use of 3D brain organoids for in vitro modeling of CNS development and neurodegeneration.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades Neurodegenerativas , Sistema Nervioso Central , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Organoides/patología , Organoides/fisiología
5.
Mov Disord ; 37(10): 2008-2020, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35997316

RESUMEN

BACKGROUND: Huntington's disease (HD) is a neurodegenerative disease caused by CAG-repeat expansions (>36) in exon 1 of HTT, which dysregulates multiple cellular machineries. Translin-associated protein X (TRAX) is a scaffold protein with diverse functions, including suppressing the microRNA (miRNA)-mediated silencing by degrading pre-miRNA. To date, the role of TRAX in neurodegenerative diseases remains unknown. OBJECTIVES: We delineated the role of TRAX upregulation during HD progression. METHODS: Expression of TRAX in the brains of humans and three mouse models with HD were analyzed by immunohistochemistry staining, western blot, and quantitative reverse transcription-polymerase chain reaction. Adeno-associated viruses harboring TRAX short hairpin RNA were intrastriatally injected into HD mice to downregulate TRAX. HD-like symptoms were analyzed by behavioral and biochemical assessments. The miRNA-sequencing and RNA-sequencing analyses were used to identify the TRAX- regulated miRNA-messenger RNA (mRNA) axis during HD progression. The identified gene targets were validated biochemically in mouse and human striatal cells. RESULTS: We discovered that TRAX was upregulated in the brains of HD patients and three HD mouse models. Downregulation of TRAX enhanced 83 miRNAs (including miR-330-3p, miR-496a-3p) and subsequently changed the corresponding mRNA networks critical for HD pathogenesis (eg, DARPP-32 and brain-derived neurotrophic factor). Disruption of the TRAX-mediated miRNA-mRNA axis accelerated the progression of HD-like symptoms, including the degeneration of motor function, accumulation of mHTT aggregates, and shortened neurite outgrowth. CONCLUSIONS: We demonstrated that TRAX upregulation is authentic and protective in HD. Our study provides a novel layer of regulation for HD pathogenesis and may lead to the development of new therapeutic strategies for HD. © 2022 International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Huntington , MicroARNs , Enfermedades Neurodegenerativas , Animales , Humanos , Ratones , Factor Neurotrófico Derivado del Encéfalo , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Enfermedad de Huntington/metabolismo , MicroARNs/genética , Neuroprotección , ARN Mensajero , ARN Interferente Pequeño
6.
Neurobiol Dis ; 160: 105531, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34634461

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a motor neuron disease characterized by progressive degeneration of motor neurons. Mislocalization of TAR DNA-binding protein 43 (TDP-43) is an early event in the formation of cytoplasmic TDP-43-positive inclusions in motor neurons and a hallmark of ALS. However, the underlying mechanism and the pathogenic impact of this mislocalization are relatively unexplored. We previously reported that abnormal AMPK activation mediates TDP-43 mislocalization in motor neurons of humans and mice with ALS. In the present study, we hypothesized that other nuclear proteins are mislocalized in the cytoplasm of motor neurons due to the AMPK-mediated phosphorylation of importin-α1 and subsequently contribute to neuronal degeneration in ALS. To test this hypothesis, we analyzed motor neurons of sporadic ALS patients and found that when AMPK is activated, importin-α1 is abnormally located in the nucleus. Multiple integrative molecular and cellular approaches (including proteomics, immunoprecipitation/western blot analysis, immunohistological evaluations and gradient analysis of preribosomal complexes) were employed to demonstrate that numerous RNA binding proteins are mislocalized in a rodent motor neuron cell line (NSC34) and human motor neurons derived from iPSCs during AMPK activation. We used comparative proteomic analysis of importin-α1 complexes that were immunoprecipitated with a phosphorylation-deficient mutant of importin-α1 (importin-α1-S105A) and a phosphomimetic mutant of importin-α1 (importin-α1-S105D) to identify 194 proteins that have stronger affinity for the unphosphorylated form than the phosphorylated form of importin-α1. Furthermore, GO and STRING analyses suggested that RNA processing and protein translation is the major machinery affected by abnormalities in the AMPK-importin-α1 axis. Consistently, the expression of importin-α1-S105D alters the assembly of preribosomal complexes and increases cell apoptosis. Collectively, we propose that by impairing importin-α1-mediated nuclear import, abnormal AMPK activation in motor neurons alters the cellular distribution of many RNA-binding proteins, which pathogenically affect multiple cellular machineries in motor neurons and contribute to ALS pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Neuronas Motoras/metabolismo , Proteínas de Unión al ARN/metabolismo , Médula Espinal/metabolismo , Adulto , Anciano , Esclerosis Amiotrófica Lateral/genética , Animales , Apoptosis/fisiología , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteómica , Proteínas de Unión al ARN/genética
7.
Hum Mol Genet ; 28(23): 3880-3894, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31518394

RESUMEN

Pompe disease (OMIM # 232300) is a glycogen storage disease caused by autosomal recessive mutations of the gene encoding alpha-1,4-glucosidase (GAA; EC 3.2.1.20). Despite the relatively effective employment of enzyme replacement therapy, some critical medical issues still exist in patients with this disease, including the persistence of abnormalities in the central nervous system (CNS), probably because of the inability of the recombinant GAA to pass through the blood-brain barrier. To address this issue, identification of more therapeutic agents that target the CNS of patients with Pompe disease may be required. In this study, we derived neuronal cells from Pompe disease-induced pluripotent stem cells (Pom-iPSCs) and proved that they are able to recapitulate the hallmark cellular and biochemical phenotypes of Pompe disease. Using the Pom-iPSC-derived neurons as an in vitro drug-testing model, we then identified three compounds, ebselen, wortmannin and PX-866, with therapeutic potential to alleviate Pompe disease-associated pathological phenotypes in the neurons derived from Pom-iPSCs. We confirmed that all three compounds were able to enhance the GAA activity in the Pom-iPSC-derived neurons. Moreover, they were able to enhance the GAA activity in several important internal organs of GAA-deficient mice when co-injected with recombinant human GAA, and we found that intraperitoneal injection of ebselen was able to promote the GAA activity of the GAA-heterozygous mouse brain. Our results prove the usefulness of Pom-iPSC-derived neuronal populations for identifying new compounds with therapeutic potential.


Asunto(s)
Azoles/administración & dosificación , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Compuestos de Organoselenio/administración & dosificación , alfa-Glucosidasas/metabolismo , Animales , Azoles/farmacología , Barrera Hematoencefálica , Encéfalo/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Gonanos/farmacología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Inyecciones Intraperitoneales , Isoindoles , Masculino , Ratones , Células-Madre Neurales/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Wortmanina/farmacología , alfa-Glucosidasas/genética
8.
PLoS Pathog ; 14(7): e1007166, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30016363

RESUMEN

CCCH-type zinc-finger antiviral protein (ZAP) is a host factor that restricts the infection of many viruses mainly through RNA degradation, translation inhibition and innate immune responses. So far, only one flavivirus, yellow fever virus, has been reported to be ZAP-resistant. Here, we investigated the antiviral potential of human ZAP (isoform ZAP-L and ZAP-S) against three flaviviruses, Japanese encephalitis virus (JEV), dengue virus (DENV) and Zika virus (ZIKV). Infection of JEV but not DENV or ZIKV was blocked by ZAP overexpression, and depletion of endogenous ZAP enhanced JEV replication. ZAP hampered JEV translation and targeted viral RNA for 3'-5' RNA exosome-mediated degradation. The zinc-finger motifs of ZAP were essential for RNA targeting and anti-JEV activity. JEV 3'-UTR, especially in the region with dumbbell structures and high content of CG dinucleotide, was mapped to bind ZAP and confer sensitivity to ZAP. In summary, we identified JEV as the first ZAP-sensitive flavivirus. ZAP may act as an intrinsic antiviral factor through specific RNA binding to fight against JEV infection.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Proteínas de Unión al ARN/inmunología , Humanos
9.
J Biomed Sci ; 27(1): 49, 2020 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-32264890

RESUMEN

Recent transcriptome analyses have revealed that noncoding RNAs (ncRNAs) are broadly expressed in mammalian cells and abundant in the CNS, with tissue and cell type-specific expression patterns. Moreover, ncRNAs have been found to intricately and dynamically regulate various signaling pathways in neurodegeneration. As such, some antisense transcripts and microRNAs are known to directly affect neurodegeneration in disease contexts. The functions of ncRNAs in pathogenesis are unique for each disorder, as are the pertinent networks of ncRNA/miRNA/mRNA that mediate these functions. Thus, further understanding of ncRNA biogenesis and effects might aid the discovery of diagnostic biomarkers or development of effective therapeutics for neurodegenerative disorders. Here, we review the ncRNAs that have so far been identified in major neurodegenerative disease etiology and the mechanisms that link ncRNAs with disease-specific phenotypes, such as HTT aggregation in HD, α-synuclein in PD, and Aß plaques and hyperphosphorylated Tau in AD. We also summarize the known lncRNA/miRNA/mRNA networks that participate in neurodegenerative diseases, and we discuss ncRNA-related treatments shown to delay disease onset and prolong lifespan in rodent models.


Asunto(s)
Perfilación de la Expresión Génica , Enfermedades Neurodegenerativas/genética , ARN no Traducido/genética , Animales , Biomarcadores/análisis , Humanos , Ratones , ARN no Traducido/metabolismo , ARN no Traducido/uso terapéutico , Ratas
10.
Nucleic Acids Res ; 46(7): 3671-3691, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29385530

RESUMEN

Transcriptionally non-co-linear (NCL) transcripts can originate from trans-splicing (trans-spliced RNA; 'tsRNA') or cis-backsplicing (circular RNA; 'circRNA'). While numerous circRNAs have been detected in various species, tsRNAs remain largely uninvestigated. Here, we utilize integrative transcriptome sequencing of poly(A)- and non-poly(A)-selected RNA-seq data from diverse human cell lines to distinguish between tsRNAs and circRNAs. We identified 24,498 NCL events and found that a considerable proportion (20-35%) of them arise from both tsRNAs and circRNAs, representing extensive alternative trans-splicing and cis-backsplicing in human cells. We show that sequence generalities of exon circularization are also observed in tsRNAs. Recapitulation of NCL RNAs further shows that inverted Alu repeats can simultaneously promote the formation of tsRNAs and circRNAs. However, tsRNAs and circRNAs exhibit quite different, or even opposite, expression patterns, in terms of correlation with the expression of their co-linear counterparts, expression breadth/abundance, transcript stability, and subcellular localization preference. These results indicate that tsRNAs and circRNAs may play different regulatory roles and analysis of NCL events should take the joint effects of different NCL-splicing types and joint effects of multiple NCL events into consideration. This study describes the first transcriptome-wide analysis of trans-splicing and cis-backsplicing, expanding our understanding of the complexity of the human transcriptome.


Asunto(s)
Empalme Alternativo/genética , ARN/genética , Trans-Empalme/genética , Transcriptoma/genética , Exones/genética , Perfilación de la Expresión Génica , Humanos , Empalme del ARN/genética , ARN Circular
11.
J Neuroinflammation ; 16(1): 187, 2019 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-31606043

RESUMEN

BACKGROUND: The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor activated by environmental agonists and dietary tryptophan metabolites for the immune response and cell cycle regulation. Emerging evidence suggests that AHR activation after acute stroke may play a role in brain ischemic injury. However, whether AHR activation alters poststroke astrogliosis and neurogenesis remains unknown. METHODS: We adopted conditional knockout of AHR from nestin-expressing neural stem/progenitor cells (AHRcKO) and wild-type (WT) mice in the permanent middle cerebral artery occlusion (MCAO) model. WT mice were treated with either vehicle or the AHR antagonist 6,2',4'-trimethoxyflavone (TMF, 5 mg/kg/day) intraperitoneally. The animals were examined at 2 and 7 days after MCAO. RESULTS: The AHR signaling pathway was significantly upregulated after stroke. Both TMF-treated WT and AHRcKO mice showed significantly decreased infarct volume, improved sensorimotor, and nonspatial working memory functions compared with their respective controls. AHR immunoreactivities were increased predominantly in activated microglia and astrocytes after MCAO compared with the normal WT controls. The TMF-treated WT and AHRcKO mice demonstrated significant amelioration of astrogliosis and microgliosis. Interestingly, these mice also showed augmentation of neural progenitor cell proliferation at the ipsilesional neurogenic subventricular zone (SVZ) and the hippocampal subgranular zone. At the peri-infarct cortex, the ipsilesional SVZ/striatum, and the hippocampus, both the TMF-treated and AHRcKO mice demonstrated downregulated IL-1ß, IL-6, IFN-γ, CXCL1, and S100ß, and concomitantly upregulated Neurogenin 2 and Neurogenin 1. CONCLUSION: Neural cell-specific AHR activation following acute ischemic stroke increased astrogliosis and suppressed neurogenesis in adult mice. AHR inhibition in acute stroke may potentially benefit functional outcomes likely through reducing proinflammatory gliosis and preserving neurogenesis.


Asunto(s)
Encéfalo/metabolismo , Gliosis/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Accidente Cerebrovascular/metabolismo , Factores de Edad , Animales , Encéfalo/patología , Gliosis/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Accidente Cerebrovascular/patología
12.
J Biomed Sci ; 26(1): 29, 2019 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-31027496

RESUMEN

Circular RNAs (circRNAs) are closed long non-coding RNAs, in which the 5' and 3' termini are covalently linked by back-splicing of exons from a single pre-mRNA. Emerging evidence indicates that circRNAs are broadly expressed in mammalian cells and show cell type- or tissue-specific expression patterns. Importantly, circRNAs have been shown to participate in regulating various biological processes. Functionally, circRNAs can influence cellular physiology through various molecular mechanisms, such as serving as a decoy for microRNAs or RNA-binding proteins to modulate gene expression or translation of regulatory proteins. The biogenesis of circRNAs is known to be tightly regulated by cis- (intronic complementary sequences) and/or trans-factors (splicing factors) that constitute a cell- and context-dependent regulatory layer in the control of gene expression. However, our understanding of the regulation and function of circRNAs is still limited. In this review, we summarize the current progress in elucidating the functional roles, mechanisms and biogenesis of circRNAs. We also discuss the relationship between regulation and formation of circRNAs.


Asunto(s)
Regulación de la Expresión Génica , ARN Largo no Codificante/fisiología , ARN/fisiología , Factores de Transcripción/metabolismo , Transcriptoma , Animales , Humanos , ARN/genética , ARN Circular , ARN Largo no Codificante/genética , Factores de Transcripción/genética
13.
Mol Psychiatry ; 23(12): 2375-2390, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29298990

RESUMEN

Translin-associated protein X (TRAX) is a scaffold protein with various functions and has been associated with mental illnesses, including schizophrenia. We have previously demonstrated that TRAX interacts with a Gsα protein-coupled receptor, the A2A adenosine receptor (A2AR), and mediates the function of this receptor in neuritogenesis. In addition, stimulation of the A2AR markedly ameliorates DNA damage evoked by elevated oxidative stress in neurons derived from induced pluripotent stem cells (iPSCs). Here, we report that glycogen synthase kinase 3 beta (GSK3ß) and disrupted-in-schizophrenia 1 (DISC1) are two novel interacting proteins of TRAX. We present evidence to suggest that the stimulation of A2AR markedly facilitated DNA repair through the TRAX/DISC1/GSK3ß complex in a rat neuronal cell line (PC12), primary mouse neurons, and human medium spiny neurons derived from iPSCs. A2AR stimulation led to the inhibition of GSK3ß, thus dissociating the TRAX/DISC1/GSK3ß complex and facilitating the non-homologous end-joining pathway (NHEJ) by enhancing the activation of a DNA-dependent protein kinase via phosphorylation at Thr2609. Similarly, pharmacological inhibition of GSK3ß by SB216763 also facilitated the TRAX-mediated repair of oxidative DNA damage. Collectively, GSK3ß binds with TRAX and negatively affects its ability to facilitate NHEJ repair. The suppression of GSK3ß by A2AR activation or a GSK3ß inhibitor releases TRAX for the repair of oxidative DNA damage. Our findings shed new light on the molecular mechanisms underlying diseases associated with DNA damage and provides a novel target (i.e., the TRAX/DISC1/GSK3ß complex) for future therapeutic development for mental disorders.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Receptor de Adenosina A2A/metabolismo , Animales , Proteínas Portadoras/genética , Reparación del ADN , Proteínas de Unión al ADN/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/fisiología , Hipocampo/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuritas , Neuronas/metabolismo , Células PC12 , Fosforilación , Ratas , Receptor de Adenosina A2A/genética , Transducción de Señal
14.
Cell Mol Life Sci ; 75(18): 3339-3351, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29961157

RESUMEN

With dual capacities for unlimited self-renewal and pluripotent differentiation, pluripotent stem cells (PSCs) give rise to many cell types in our body and PSC culture systems provide an unparalleled opportunity to study early human development and disease. Accumulating evidence indicates that the molecular mechanisms underlying pluripotency maintenance in PSCs involve many factors. Among these regulators, recent studies have shown that long non-coding RNAs (lncRNAs) can affect the pluripotency circuitry by cooperating with master pluripotency-associated factors. Additionally, trans-spliced RNAs, which are generated by combining two or more pre-mRNA transcripts to produce a chimeric RNA, have been identified as regulators of various biological processes, including human pluripotency. In this review, we summarize and discuss current knowledge about the roles of lncRNAs, including trans-spliced lncRNAs, in controlling pluripotency.


Asunto(s)
ARN Largo no Codificante/metabolismo , Trans-Empalme/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal , Factores de Transcripción/metabolismo
15.
J Vet Pharmacol Ther ; 47(1): 62-63, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38047430
16.
J Proteome Res ; 17(12): 4138-4151, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30203655

RESUMEN

Human embryonic stem cells (hESCs) have the capacity for self-renewal and multilineage differentiation, which are of clinical importance for regeneration medicine. Despite the significant progress of hESC study, the complete hESC proteome atlas, especially the surface protein composition, awaits delineation. According to the latest release of neXtProt database (January 17, 2018; 19 658 PE1, 2, 3, and 4 human proteins), membrane proteins present the major category (1047; 48%) among all 2186 missing proteins (MPs). We conducted a deep subcellular proteomics analysis of hESCs to identify the nuclear, cytoplasmic, and membrane proteins in hESCs and to mine missing membrane proteins in the very early cell status. To our knowledge, our study achieved the largest data set with confident identification of 11 970 unique proteins (1% false discovery rate at peptide, protein, and PSM levels), including the most-comprehensive description of 6 138 annotated membrane proteins in hESCs. Following the HPP guideline, we identified 26 gold (neXtProt PE2, 3, and 4 MPs) and 87 silver (potential MP candidates with a single unique peptide detected) MPs, of which 69 were membrane proteins, and the expression of 21 gold MPs was further verified either by multiple reaction monitoring mass spectrometry or by matching synthetic peptides in the Peptide Atlas database. Functional analysis of the MPs revealed their potential roles in the pluripotency-related pathways and the lineage- and tissue-specific differentiation processes. Our proteome map of hESCs may provide a rich resource not only for the identification of MPs in the human proteome but also for the investigation on self-renewal and differentiation of hESC. All mass spectrometry data were deposited in ProteomeXchange via jPOST with identifier PXD009840.


Asunto(s)
Células Madre Embrionarias Humanas/química , Proteínas de la Membrana/análisis , Proteoma/análisis , Diferenciación Celular , Linaje de la Célula , Humanos , Membranas Intracelulares/química , Proteómica/métodos
17.
Artículo en Inglés | MEDLINE | ID: mdl-29581119

RESUMEN

We identified 20 to 22 resistance genes, carried in four incompatibility groups of plasmids, in each of five genetically closely related Salmonella enterica serovar Typhimurium strains recovered from humans, pigs, and chickens. The genes conferred resistance to aminoglycosides, chloramphenicol, sulfonamides, trimethoprim, tetracycline, fluoroquinolones, extended-spectrum cephalosporins and cefoxitin, and azithromycin. This study demonstrates the transmission of multidrug-resistant Salmonella strains among humans and food animals and may be the first identification of mphA in azithromycin-resistant Salmonella strains in Taiwan.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Plásmidos/genética , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética , Animales , Azitromicina/farmacología , Pollos , Humanos , Pruebas de Sensibilidad Microbiana , Familia de Multigenes/genética , Salmonelosis Animal , Salmonella typhimurium/aislamiento & purificación , Porcinos , Taiwán , Secuenciación Completa del Genoma
18.
Proc Natl Acad Sci U S A ; 112(39): 12199-204, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26371318

RESUMEN

The timing of cortical neurogenesis has a major effect on the size and organization of the mature cortex. The deletion of the LIM-homeodomain transcription factor Lhx2 in cortical progenitors by Nestin-cre leads to a dramatically smaller cortex. Here we report that Lhx2 regulates the cortex size by maintaining the cortical progenitor proliferation and delaying the initiation of neurogenesis. The loss of Lhx2 in cortical progenitors results in precocious radial glia differentiation and a temporal shift of cortical neurogenesis. We further investigated the underlying mechanisms at play and demonstrated that in the absence of Lhx2, the Wnt/ß-catenin pathway failed to maintain progenitor proliferation. We developed and applied a mathematical model that reveals how precocious neurogenesis affected cortical surface and thickness. Thus, we concluded that Lhx2 is required for ß-catenin function in maintaining cortical progenitor proliferation and controls the timing of cortical neurogenesis.


Asunto(s)
Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas con Homeodominio LIM/metabolismo , Modelos Neurológicos , Neurogénesis/genética , Neurogénesis/fisiología , Factores de Transcripción/metabolismo , Animales , Simulación por Computador , Cartilla de ADN/genética , Galactósidos , Regulación del Desarrollo de la Expresión Génica/fisiología , Células HEK293 , Humanos , Hibridación in Situ , Indoles , Proteínas con Homeodominio LIM/genética , Luciferasas , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/genética , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
19.
Hum Mol Genet ; 24(21): 6066-79, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26264576

RESUMEN

Huntington's disease (HD) is an autosomal-dominant degenerative disease caused by a cytosine-adenine-guanine trinucleotide expansion in the Huntingtin (htt) gene. The most vulnerable brain areas to mutant HTT-evoked toxicity are the striatum and cortex. In spite of the extensive efforts that have been devoted to the characterization of HD pathogenesis, no disease-modifying therapy for HD is currently available. The A2A adenosine receptor (A2AR) is widely distributed in the brain, with the highest level observed in the striatum. We previously reported that stimulation of the A2AR triggers an anti-apoptotic effect in a rat neuron-like cell line (PC12). Using a transgenic mouse model (R6/2) of HD, we demonstrated that A2AR-selective agonists effectively ameliorate several major symptoms of HD. In the present study, we show that human iPSCs can be successfully induced to differentiate into DARPP32-positive, GABAergic neurons which express the A2AR in a similar manner to striatal medium spiny neurons. When compared with those derived from control subjects (CON-iPSCs), these HD-iPSC-derived neurons exhibited a higher DNA damage response, based on the observed expression of γH2AX and elevated oxidative stress. This is a critical observation, because oxidative damage and abnormal DNA damage/repair have been reported in HD patients. Most importantly, stimulation of the A2AR using selective agonists reduced DNA damage and oxidative stress-induced apoptosis in HD-iPSC-derived neurons through a cAMP/PKA-dependent pathway. These findings support our hypothesis that human neurons derived from diseased iPSCs might serve as an important platform to investigate the beneficial effects and underlying mechanisms of A2AR drugs.


Asunto(s)
Neuronas GABAérgicas/patología , Enfermedad de Huntington/patología , Degeneración Nerviosa , Células Madre Pluripotentes/patología , Receptor de Adenosina A2A/metabolismo , Adulto , Apoptosis , Caspasa 3/metabolismo , Línea Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Daño del ADN , Fosfoproteína 32 Regulada por Dopamina y AMPc/biosíntesis , Femenino , Neuronas GABAérgicas/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Peróxido de Hidrógeno , Recién Nacido , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Células Madre Pluripotentes/metabolismo , Adulto Joven
20.
Artículo en Inglés | MEDLINE | ID: mdl-28416545

RESUMEN

We detected the colistin resistance gene mcr-1 in four Salmonella serovars isolated from humans and animals with diarrhea. The resistance gene was carried on different plasmids. One mcr-1-carrying conjugative plasmid, a variant of pHNSHP45, was disseminated among Salmonella isolates recovered from humans, pigs, and chickens.


Asunto(s)
Antibacterianos/farmacología , Plásmidos/genética , Salmonella/efectos de los fármacos , Animales , Pollos , Diarrea/microbiología , Humanos , Pruebas de Sensibilidad Microbiana , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA