Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci ; 347: 122651, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38642844

RESUMEN

Calcium ion (Ca2+) dysregulation is one of the main causes of neuronal cell death and brain damage after cerebral ischemia. During ischemic stroke, the ability of neurons to maintain Ca2+ homeostasis is compromised. Ca2+ regulates various functions of the nervous system, including neuronal activity and adenosine triphosphate (ATP) production. Disruptions in Ca2+ homeostasis can trigger a cascade of events, including activation of the unfolded protein response (UPR) pathway, which is associated with endoplasmic reticulum (ER) stress and mitochondrial dysfunction. This response occurs when the cell is unable to manage protein folding within the ER due to various stressors, such as a high influx of Ca2+. Consequently, the UPR is initiated to restore ER function and alleviate stress, but prolonged activation can lead to mitochondrial dysfunction and, ultimately, cell death. Hence, precise regulation of Ca2+ within the cell is mandatory. The ER and mitochondria are two such organelles that maintain intracellular Ca2+ homeostasis through various calcium-operating channels, including ryanodine receptors (RyRs), inositol trisphosphate receptors (IP3Rs), sarco/endoplasmic reticulum calcium ATPases (SERCAs), the mitochondrial Na+/Ca2+ exchanger (NCLX), the mitochondrial calcium uniporter (MCU) and voltage-dependent anion channels (VDACs). These channels utilize Ca2+ sequestering and release mechanisms to maintain intracellular Ca2+ homeostasis and ensure proper cellular function and survival. The present review critically evaluates the significance of Ca2+ and its physiological role in cerebral ischemia. We have compiled recent findings on calcium's role and emerging treatment strategies, particularly targeting mitochondria and the endoplasmic reticulum, to address Ca2+ overload in cerebral ischemia.


Asunto(s)
Calcio , Muerte Celular , Accidente Cerebrovascular Isquémico , Neuronas , Humanos , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Animales , Calcio/metabolismo , Neuronas/metabolismo , Neuronas/patología , Mitocondrias/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Respuesta de Proteína Desplegada , Señalización del Calcio/fisiología , Homeostasis
2.
Inflammopharmacology ; 32(1): 287-305, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37991660

RESUMEN

Pulmonary fibrosis is a devastating disorder distinguished by redundant inflammation and matrix accumulation in the lung interstitium. The early inflammatory cascade coupled with recurring tissue injury orchestrates a set of events marked by perturbed matrix hemostasis, deposition of matrix proteins, and remodeling in lung tissue. Numerous investigations have corroborated a direct correlation between the NLR family pyrin domain-containing 3 (NLRP3) activation and the development of pulmonary fibrosis. Dysregulated activation of NLRP3 within the pulmonary microenvironment exacerbates inflammation and may incite fibrogenic responses. Nevertheless, the precise mechanisms through which the NLRP3 inflammasome elicits pro-fibrogenic responses remain inadequately defined. Contemporary findings suggest that the pro-fibrotic consequences stemming from NLRP3 signaling primarily hinge on the action of interleukin-1ß (IL-1ß). IL-1ß instigates IL-1 receptor signaling, potentiating the activity of transforming growth factor-beta (TGF-ß). This signaling cascade, in turn, exerts influence over various transcription factors, including SNAIL, TWIST, and zinc finger E-box-binding homeobox 1 (ZEB 1/2), which collectively foster myofibroblast activation and consequent lung fibrosis. Here, we have connected the dots to illustrate how the NLRP3 inflammasome orchestrates a multitude of signaling events, including the activation of transcription factors that facilitate myofibroblast activation and subsequent lung remodeling. In addition, we have highlighted the prominent role played by various cells in the formation of myofibroblasts, the primary culprit in lung fibrosis. We also provided a concise overview of various compounds that hold the potential to impede NLRP3 inflammasome signaling, thus offering a promising avenue for the treatment of pulmonary fibrosis.


Asunto(s)
Inflamasomas , Fibrosis Pulmonar , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Inflamación , Factores de Transcripción
3.
ACS Chem Neurosci ; 15(1): 31-55, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38118278

RESUMEN

Neuroinflammation is a key pathophysiological feature of stroke-associated brain injury. A local innate immune response triggers neuroinflammation following a stroke via activating inflammasomes. The nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome has been heavily implicated in stroke pathobiology. Following a stroke, several stimuli have been suggested to trigger the assembly of the NLRP3 inflammasome. Recent studies have advanced the understanding and revealed several new players regulating NLRP3 inflammasome-mediated neuroinflammation. This article discussed recent advancements in NLRP3 assembly and highlighted stroke-induced mitochondrial dysfunction as a major checkpoint to regulating NLRP3 activation. The NLRP3 inflammasome activation leads to caspase-1-dependent maturation and release of IL-1ß, IL-18, and gasdermin D. In addition, genetic or pharmacological inhibition of the NLRP3 inflammasome activation and downstream signaling has been shown to attenuate brain infarction and improve the neurological outcome in experimental models of stroke. Several drug-like small molecules targeting the NLRP3 inflammasome are in different phases of development as novel therapeutics for various inflammatory conditions, including stroke. Understanding how these molecules interfere with NLRP3 inflammasome assembly is paramount for their better optimization and/or development of newer NLRP3 inhibitors. In this review, we summarized the assembly of the NLRP3 inflammasome and discussed the recent advances in understanding the upstream regulators of NLRP3 inflammasome-mediated neuroinflammation following stroke. Additionally, we critically examined the role of the NLRP3 inflammasome-mediated signaling in stroke pathophysiology and the development of therapeutic modalities to target the NLRP3 inflammasome-related signaling for stroke treatment.


Asunto(s)
Lesiones Encefálicas , Accidente Cerebrovascular , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedades Neuroinflamatorias , Accidente Cerebrovascular/tratamiento farmacológico , Interleucina-1beta/metabolismo
4.
Drug Discov Today ; 28(11): 103792, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37783431

RESUMEN

Stroke, a debilitating condition with limited treatment options, presents a significant therapeutic challenge. A comprehensive grasp of stroke pathophysiology is imperative for designing newer and more effective therapeutic approaches. Notably, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway has emerged as a central orchestrator of the poststroke immune response. It regulates pivotal processes, including immune cell activation, cytokine production, neuroinflammation, apoptosis, and tissue regeneration. Modulating this pathway shows immense potential in improving stroke outcomes, necessitating the development of selective inhibitors and activators. This review provides an overview of the cGAS-STING pathway's role in ischemic stroke and explores emerging therapies, including cGAS and STING inhibitors and STING agonist preconditioning. It also addresses challenges like specificity, timing, and off-target effects.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Accidente Cerebrovascular/tratamiento farmacológico , Nucleotidiltransferasas , Transducción de Señal , Inmunidad
5.
Expert Opin Drug Discov ; 18(10): 1133-1149, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37537969

RESUMEN

INTRODUCTION: Ischemic stroke is a significant global health challenge with limited treatment options. Neuroinflammation, driven by microglial activation, plays a critical role in stroke pathophysiology. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway has emerged as a key player in microglial activation, sterile neuroinflammation, and cell death following stroke. Understanding the interplay between this pathway and stroke pathophysiology is crucial for exploring newer therapeutics for stroke patients. AREAS COVERED: This review discusses the pivotal role of the cGAS-STING pathway in ischemic stroke. It explores the interplay between cGAS-STING activation, neuroinflammation, microglia activation, M2 polarization, neutrophil infiltration, and cytokine release. Additionally, the authors examine its contributions to various cell death programs (pyroptosis, apoptosis, necroptosis, lysosomal cell death, autophagy, and ferroptosis). The review summarizes recent studies on targeting cGAS-STING signaling in stroke, highlighting the therapeutic potential of small molecule inhibitors and RNA-based approaches in mitigating neuroinflammation, preventing cell death, and improving patient outcomes. EXPERT OPINION: Understanding cGAS-STING signaling in ischemic stroke offers an exciting avenue for drug discovery. Targeting this pathway holds promise for developing novel therapeutics that effectively mitigate neuroinflammation, prevent cell death, and enhance patient outcomes. Further research and development of therapeutic strategies are warranted to fully exploit the potential of this pathway as a therapeutic target for stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Enfermedades Neuroinflamatorias , Accidente Cerebrovascular/tratamiento farmacológico , Descubrimiento de Drogas , Nucleotidiltransferasas
6.
Drug Discov Today ; 28(6): 103583, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37028501

RESUMEN

Sirtuin 3 (SIRT3), a mitochondrial deacetylase expressed preferentially in high-metabolic-demand tissues including the brain, requires NAD+ as a cofactor for catalytic activity. It regulates various processes such as energy homeostasis, redox balance, mitochondrial quality control, mitochondrial unfolded protein response, biogenesis, dynamics and mitophagy by altering protein acetylation status. Reduced SIRT3 expression or activity causes hyperacetylation of hundreds of mitochondrial proteins, which has been linked with neurological abnormalities, neuro-excitotoxicity and neuronal cell death. A body of evidence has suggested, SIRT3 activation as a potential therapeutic modality for age-related brain abnormalities and neurodegenerative disorders.


Asunto(s)
Enfermedades Neurodegenerativas , Sirtuina 3 , Humanos , Sirtuina 3/metabolismo , Proteínas Mitocondriales/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Biología
7.
Drug Discov Today ; 28(5): 103555, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36931386

RESUMEN

Tailoring drug products to personalized medicines poses challenges for conventional dosage forms. The prominent reason is the restricted availability of flexible dosage strengths in the market. Inappropriate dosage strengths lead to adverse drug reactions or compromised therapeutic effects. The situation worsens when the drug has a narrow therapeutic window. To overcome these challenges, data-enriched edible pharmaceuticals (DEEP) are novel concepts for designing solid oral products. DEEP have individualized doses and information embedded in quick response (QR) code form. When data are presented in a QR code, the information is printed with edible ink that contains the drug in tailored doses required for the patients.


Asunto(s)
Sistemas de Liberación de Medicamentos , Medicina de Precisión , Humanos , Preparaciones Farmacéuticas , Tecnología Farmacéutica , Formas de Dosificación
8.
Cell ; 185(16): 3008-3024.e16, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35870449

RESUMEN

Here, we report inducible mosaic animal for perturbation (iMAP), a transgenic platform enabling in situ CRISPR targeting of at least 100 genes in parallel throughout the mouse body. iMAP combines Cre-loxP and CRISPR-Cas9 technologies and utilizes a germline-transmitted transgene carrying a large array of individually floxed, tandemly linked gRNA-coding units. Cre-mediated recombination triggers expression of all the gRNAs in the array but only one of them per cell, converting the mice to mosaic organisms suitable for phenotypic characterization and also for high-throughput derivation of conventional single-gene perturbation lines via breeding. Using gRNA representation as a readout, we mapped a miniature Perturb-Atlas cataloging the perturbations of 90 genes across 39 tissues, which yields rich insights into context-dependent gene functions and provides a glimpse of the potential of iMAP in genome decoding.


Asunto(s)
Sistemas CRISPR-Cas , ARN Guía de Kinetoplastida , Animales , Sistemas CRISPR-Cas/genética , Edición Génica , Genoma , Ratones , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Transgenes
9.
Pharmacol Res ; 175: 106018, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34863823

RESUMEN

The nuclear factor erythroid-derived 2-related factor 2 (NFE2L2/Nrf2) is a pivotal facilitator of cytoprotective responses against the oxidative/electrophilic insults. Upon activation, Nrf2 induces transcription of a wide range of cytoprotective genes having antioxidant response element (ARE) in their promoter region. Dysfunction in Nrf2 signaling has been linked to the pathogenesis of AD and several studies have suggested that boosting Nrf2 expression/activity by genetic or pharmacological approaches is beneficial in AD. Among the diverse mechanisms that regulate the Nrf2 signaling, miRNAs-mediated regulation of Nrf2 has gained much attention in recent years. Several miRNAs have been reported to directly repress the post-transcriptional expression of Nrf2 and thereby negatively regulate the Nrf2-dependent cellular cytoprotective response in AD. Moreover, several Nrf2 targeting miRNAs are misregulated in AD brains. This review is focused on the role of misregulated miRNAs that directly target Nrf2, in AD pathophysiology. Here, alongside a general description of functional interactions between miRNAs and Nrf2, we have reviewed the evidence indicating the possible role of these miRNAs in AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , MicroARNs , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Humanos , Factor 2 Relacionado con NF-E2/química , Transcripción Genética
10.
Mol Neurobiol ; 58(9): 4575-4587, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34110602

RESUMEN

Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 or COVID-19 has been declared as a pandemic disease by the World Health Organization (WHO). Globally, this disease affected 159 million of the population and reported ~ 3.3 million deaths to the current date (May 2021). There is no definitive treatment strategy that has been identified, although this disease has prevailed in its current form for the past 18 months. The main challenges in the (SARS-CoV)-2 infections are in identifying the heterogeneity in viral strains and the plausible mechanisms of viral infection to human tissues. In parallel to the investigations into the patho-mechanism of SARS-CoV-2 infection, understanding the fundamental processes underlying the clinical manifestations of COVID-19 is very crucial for designing effective therapies. Since neurological symptoms are very apparent in COVID-19 infected patients, here, we tried to emphasize the involvement of redox imbalance and subsequent mitochondrial dysfunction in the progression of the COVID-19 infection. It has been articulated that mitochondrial dysfunction is very apparent and also interlinked to neurological symptoms in COVID-19 infection. Overall, this article provides an in-depth overview of redox imbalance and mitochondrial dysfunction involvement in aggravating COVID-19 infection and its probable contribution to the neurological manifestation of the disease.


Asunto(s)
COVID-19/complicaciones , Mitocondrias/fisiología , SARS-CoV-2/patogenicidad , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/metabolismo , Sistema Nervioso Central/virología , Reposicionamiento de Medicamentos , Endotelio Vascular/fisiopatología , Endotelio Vascular/virología , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Modelos Biológicos , Nervio Olfatorio/virología , Especificidad de Órganos , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Pandemias , SARS-CoV-2/fisiología , Proteínas Virales/fisiología , Tropismo Viral , Viremia/complicaciones , Virulencia , Internalización del Virus
12.
Curr Neuropharmacol ; 19(11): 1984-2011, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33588734

RESUMEN

According to the World Health Organization, Traumatic brain injury (TBI) is the major cause of death and disability and will surpass the other diseases by the year 2020. Patients who suffer TBI face many difficulties which negatively affect their social and personal life. TBI patients suffer from changes in mood, impulsivity, poor social judgment and memory deficits. Both open and closed head injuries have their own consequences. Open head injury associated problems are specific in nature e.g. loss of motor functions whereas closed head injuries are diffused in nature like poor memory, problems in concentration etc. Brain injury may have a detrimental effect on the biochemical processes responsible for the homeostatic and physiological disturbances in the brain. Although significant research has been done in order to decrease the overall TBI-related mortality, many individuals suffer from a life-long disability. In this article, we have discussed the causes of TBI, its consequence and the pathobiology of secondary injury. We have also tried to discuss the evidence-based strategies which are shown to decline the devastating consequences of TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Encéfalo , Lesiones Encefálicas/etiología , Lesiones Encefálicas/terapia , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/terapia , Humanos , Trastornos de la Memoria
13.
Pharmacol Res ; 167: 105414, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33524540

RESUMEN

Cardiac fibrosis is characterized by excessive deposition of extracellular matrix proteins and myofibroblast differentiation. Our previous findings have implicated resistin in cardiac fibrosis; however, the molecular mechanisms underlying this process are still unclear. Here we investigated the role of resistin in fibroblast-to-myofibroblast differentiation and elucidated the pathways involved in this process. Fibroblast-to-myofibroblast transdifferentiation was induced with resistin or TGFß1 in NIH-3T3 and adult cardiac fibroblasts. mRNA and protein expression of fibrotic markers were analyzed by qPCR and immunoblotting. Resistin-knockout mice, challenged with a high-fat diet (HFD) for 20 weeks to stimulate cardiac impairment, were analyzed for cardiac function and fibrosis using histologic and molecular methods. Cardiac fibroblasts stimulated with resistin displayed increased fibroblast-to-myofibroblast conversion, with increased levels of αSma, col1a1, Fn, Ccn2 and Mmp9, with remarkable differences in the actin network appearance. Mechanistically, resistin promotes fibroblast-to-myofibroblast transdifferentiation and fibrogenesis via JAK2/STAT3 and JNK/c-Jun signaling pathways, independent of TGFß1. Resistin-null mice challenged with HFD showed an improvement in cardiac function and a decrease in tissue fibrosis and reduced mRNA levels of fibrogenic markers. These findings are the first to delineate the role of resistin in the process of cardiac fibroblast-to-myofibroblast differentiation via JAK/STAT3 and JNK/c-Jun pathways, potentially leading to stimulation of cardiac fibrosis.


Asunto(s)
Transdiferenciación Celular/fisiología , Fibroblastos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Miocitos Cardíacos/metabolismo , Resistina/farmacología , Factor de Transcripción STAT3/metabolismo , Animales , Transdiferenciación Celular/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Janus/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Células 3T3 NIH , Resistina/deficiencia
14.
Nat Commun ; 11(1): 2781, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493900

RESUMEN

Mutations disrupting regulatory T (Treg) cell function can cause IPEX and IPEX-related disorders, but whether established disease can be reversed by correcting these mutations is unclear. Treg-specific deletion of the chromatin remodeling factor Brg1 impairs Treg cell activation and causes fatal autoimmunity in mice. Here, we show with a reversible knockout model that re-expression of Brg1, in conjunction with the severe endogenous proinflammatory environment, can convert defective Treg cells into powerful, super-activated Treg cells (SuperTreg cells) that can resolve advanced autoimmunity,  with  Brg1 re-expression in a minor fraction of Treg cells sufficient for the resolution in some cases. SuperTreg cells have enhanced trafficking and regulatory capabilities, but become deactivated as the inflammation subsides, thus avoiding excessive immune suppression. We propose a simple, robust yet safe gene-editing-based therapy for IPEX and IPEX-related disorders that exploits the defective Treg cells and the inflammatory environment pre-existing in the patients.


Asunto(s)
Diabetes Mellitus Tipo 1/congénito , Diarrea/inmunología , Enfermedades Genéticas Ligadas al Cromosoma X/inmunología , Enfermedades del Sistema Inmune/congénito , Linfocitos T Reguladores/inmunología , Alelos , Animales , Citocinas/metabolismo , ADN Helicasas/deficiencia , Diabetes Mellitus Tipo 1/inmunología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Enfermedades del Sistema Inmune/inmunología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/deficiencia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CXCR3/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Linfocitos T Reguladores/efectos de los fármacos , Tamoxifeno/farmacología , Factores de Transcripción/deficiencia
16.
J Immunol ; 190(2): 737-47, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23293358

RESUMEN

Synthetic regulatory proteins such as tetracycline (tet)-controlled transcription factors are potentially useful for repression as well as ectopic activation of endogenous genes and also for probing their regulatory mechanisms, which would offer a versatile genetic tool advantageous over conventional gene targeting methods. In this study, we provide evidence supporting this concept using Cd4 as a model. CD4 is expressed in double-positive and CD4 cells but irreversibly silenced in CD8 cells. The silencing is mediated by heterochromatin established during CD8 lineage development via transient action of the Cd4 silencer; once established, the heterochromatin becomes self-perpetuating independently of the Cd4 silencer. Using a tet-sensitive Cd4 allele harboring a removable Cd4 silencer, we found that a tet-controlled repressor recapitulated the phenotype of Cd4-deficient mice, inhibited Cd4 expression in a reversible and dose-dependent manner, and could surprisingly replace the Cd4 silencer to induce irreversible Cd4 silencing in CD8 cells, thus suggesting the Cd4 silencer is not the (only) determinant of heterochromatin formation. In contrast, a tet-controlled activator reversibly disrupted Cd4 silencing in CD8 cells. The Cd4 silencer impeded this disruption but was not essential for its reversal, which revealed a continuous role of the silencer in mature CD8 cells while exposing a remarkable intrinsic self-regenerative ability of heterochromatin after forced disruption. These data demonstrate an effective approach for gene manipulation and provide insights into the epigenetic Cd4 regulatory mechanisms that are otherwise difficult to obtain.


Asunto(s)
Antígenos CD4/genética , Epigénesis Genética , Regulación de la Expresión Génica , Transcripción Genética , Alelos , Animales , Linfocitos T CD8-positivos/metabolismo , Orden Génico , Silenciador del Gen , Marcación de Gen , Ratones , Ratones Noqueados , Fenotipo , Elementos Silenciadores Transcripcionales , Linfocitos T/metabolismo
17.
Development ; 140(4): 843-52, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23325759

RESUMEN

Environmental factors can stably perturb the epigenome of exposed individuals and even that of their offspring, but the pleiotropic effects of these factors have posed a challenge for understanding the determinants of mitotic or transgenerational inheritance of the epigenetic perturbation. To tackle this problem, we manipulated the epigenetic states of various target genes using a tetracycline-dependent transcription factor. Remarkably, transient manipulation at appropriate times during embryogenesis led to aberrant epigenetic modifications in the ensuing adults regardless of the modification patterns, target gene sequences or locations, and despite lineage-specific epigenetic programming that could reverse the epigenetic perturbation, thus revealing extraordinary malleability of the fetal epigenome, which has implications for 'metastable epialleles'. However, strong transgenerational inheritance of these perturbations was observed only at transgenes integrated at the Col1a1 locus, where both activating and repressive chromatin modifications were heritable for multiple generations; such a locus is unprecedented. Thus, in our inducible animal models, mitotic inheritance of epigenetic perturbation seems critically dependent on the timing of the perturbation, whereas transgenerational inheritance additionally depends on the location of the perturbation. In contrast, other parameters examined, particularly the chromatin modification pattern and DNA sequence, appear irrelevant.


Asunto(s)
Cromatina/metabolismo , Colágeno Tipo I/genética , Epigénesis Genética/fisiología , Patrón de Herencia/fisiología , Modelos Biológicos , Fenotipo , Animales , Antígenos CD4/genética , Cromatina/genética , Inmunoprecipitación de Cromatina , Cadena alfa 1 del Colágeno Tipo I , Epigénesis Genética/genética , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Patrón de Herencia/genética , Ratones , Ratones Transgénicos , Transgenes/genética
18.
BMC Biol ; 10: 96, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23198860

RESUMEN

BACKGROUND: Conditional gene knockout (cKO) mediated by the Cre/LoxP system is indispensable for exploring gene functions in mice. However, a major limitation of this method is that gene KO is not reversible. A number of methods have been developed to overcome this, but each method has its own limitations. RESULTS: We describe a simple method we have named LOFT [LoxP-flippase (FLP) recognition target (FRT) Trap], which is capable of reversible cKO and free of the limitations associated with existing techniques. This method involves two alleles of a target gene: a standard floxed allele, and a multi-functional allele bearing an FRT-flanked gene-trap cassette, which inactivates the target gene while reporting its expression with green fluorescent protein (GFP); the trapped allele is thus a null and GFP reporter by default, but is convertible into a wild-type allele. The floxed and trapped alleles can typically be generated using a single construct bearing a gene-trap cassette doubly flanked by LoxP and FRT sites, and can be used independently to achieve conditional and constitutive gene KO, respectively. More importantly, in mice bearing both alleles and also expressing the Cre and FLP recombinases, sequential function of the two enzymes should lead to deletion of the target gene, followed by restoration of its expression, thus achieving reversible cKO. LOFT should be generally applicable to mouse genes, including the growing numbers of genes already floxed; in the latter case, only the trapped alleles need to be generated to confer reversibility to the pre-existing cKO models. LOFT has other applications, including the creation and reversal of hypomorphic mutations. In this study we proved the principle of LOFT in the context of T-cell development, at a hypomorphic allele of Baf57/Smarce1 encoding a subunit of the chromatin-remodeling Brg/Brahma-associated factor (BAF) complex. Interestingly, the FLP used in the current work caused efficient reversal in peripheral T cells but not thymocytes, which is advantageous for studying developmental epigenetic programming of T-cell functions, a fundamental issue in immunology. CONCLUSIONS: LOFT combines well-established basic genetic methods into a simple and reliable method for reversible gene targeting, with the flexibility of achieving traditional constitutive and conditional KO.


Asunto(s)
Técnicas de Inactivación de Genes , Ingeniería Genética/métodos , Alelos , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Regulación de la Expresión Génica/fisiología , Marcación de Gen , Vectores Genéticos , Integrasas , Ratones , Ratones Noqueados , Subunidades de Proteína
19.
Drug Discov Today ; 17(13-14): 793-804, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22406696

RESUMEN

Increased expression of endothelin (ET) peptide and its receptors following ischemic stroke is found to regulate many critical aspects of stroke pathophysiology. Many attempts have been made to target ET receptors in various animal models of stroke, but it is very difficult to draw a definite line of conclusion, because these studies differ in many aspects, such as animal model, treatment schedule, parameters and techniques used for assessing these parameters. A meta-analysis of all studies showed a significant reduction in the lesion volume and improvement in functional outcome in focal cerebral ischemia. ET(A) receptor antagonists appear to offer an essential advantage of multiple neuroprotective mechanisms, including prevention of blood-brain barrier disruption and leukocyte infiltration.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Fármacos Neuroprotectores/uso terapéutico , Receptores de Endotelina/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Isquemia Encefálica/complicaciones , Modelos Animales de Enfermedad , Descubrimiento de Drogas/tendencias , Antagonistas de los Receptores de Endotelina , Humanos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/efectos adversos , Receptores de Endotelina/agonistas , Receptores de Endotelina/genética , Transducción de Señal , Accidente Cerebrovascular/etiología
20.
Curr Neurovasc Res ; 8(3): 236-45, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21722092

RESUMEN

PPARγ agonist; 2-(Benzoylphenyl)-O-[2-(methyl-2-pyridinylamino) ethyl]-L-tyrosine (GW1929) in focal cerebral ischemic-reperfusion (IR) injury in rats. Focal cerebral IR injury resulted significant brain infarction and neurological deficits in rats. A significant increase in various inflammatory mediators like COX-2, iNOS, MMP-9, TNFα and IL-6 and massive apoptotic DNA fragmentation was also observed in the IR challenged brains. GW1929 treatment significantly attenuated the neurological damage in focal cerebral IR injury. Neuroprotective effects of GW1929 were found to be associated with significant reduction in the COX-2, iNOS, MMP-9, TNFα and IL-6 levels. Together, we have also evaluated the effects of Pioglitazone, a clinically available thiazolidinedione PPARγ agonist, against focal cerebral IR injury. Like GW1929, Pioglitazone also showed beneficial effects in cerebral IR injury associated neurological damage but at the higher dose as compared to GW1929. Neuroprotective effects of PPARγ agonists were found to be associated with significant reduction in TUNEL positive cells in IR challenged brain. In summary, these results suggested the neuroprotective potential of PPARγ agonists in cerebral IR injury and these effects may be attributed to their anti-inflammatory and anti-apoptotic potential.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzofenonas/farmacología , Isquemia Encefálica/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/tratamiento farmacológico , Tirosina/análogos & derivados , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Inflamación/metabolismo , Inflamación/patología , Masculino , PPAR gamma/agonistas , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Tirosina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...