Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 149(19): 1501-1515, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38223978

RESUMEN

BACKGROUND: During the neonatal stage, the cardiomyocyte undergoes a constellation of molecular, cytoarchitectural, and functional changes known collectively as cardiomyocyte maturation to increase myocardial contractility and cardiac output. Despite the importance of cardiomyocyte maturation, the molecular mechanisms governing this critical process remain largely unexplored. METHODS: We leveraged an in vivo mosaic knockout system to characterize the role of Carm1, the founding member of protein arginine methyltransferase, in cardiomyocyte maturation. Using a battery of assays, including immunohistochemistry, immuno-electron microscopy imaging, and action potential recording, we assessed the effect of loss of Carm1 function on cardiomyocyte cell growth, myofibril expansion, T-tubule formation, and electrophysiological maturation. Genome-wide transcriptome profiling, H3R17me2a chromatin immunoprecipitation followed by sequencing, and assay for transposase-accessible chromatin with high-throughput sequencing were used to investigate the mechanisms by which CARM1 (coactivator-associated arginine methyltransferase 1) regulates cardiomyocyte maturation. Finally, we interrogated the human syntenic region to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks for single-nucleotide polymorphisms associated with human heart diseases. RESULTS: We report that mosaic ablation of Carm1 disrupts multiple aspects of cardiomyocyte maturation cell autonomously, leading to reduced cardiomyocyte size and sarcomere thickness, severe loss and disorganization of T tubules, and compromised electrophysiological maturation. Genomics study demonstrates that CARM1 directly activates genes that underlie cardiomyocyte cytoarchitectural and electrophysiological maturation. Moreover, our study reveals significant enrichment of human heart disease-associated single-nucleotide polymorphisms in the human genomic region syntenic to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks. CONCLUSIONS: This study establishes a critical and multifaceted role for CARM1 in regulating cardiomyocyte maturation and demonstrates that deregulation of CARM1-dependent cardiomyocyte maturation gene expression may contribute to human heart diseases.


Asunto(s)
Epigénesis Genética , Miocitos Cardíacos , Proteína-Arginina N-Metiltransferasas , Animales , Humanos , Ratones , Diferenciación Celular/genética , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo
2.
Semin Cell Dev Biol ; 118: 144-149, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33994094

RESUMEN

Heart malformation is the leading cause of human birth defects, and many of the congenital heart diseases (CHDs) originate from genetic defects that impact cardiac development and maturation. During development, the vertebrate heart undergoes a series of complex morphogenetic processes that increase its ability to pump blood. One of these processes leads to the formation of the sheet-like muscular projections called trabeculae. Trabeculae increase cardiac output and permit nutrition and oxygen uptake in the embryonic myocardium prior to coronary vascularization without increasing heart size. Cardiac trabeculation is also crucial for the development of the intraventricular fast conduction system. Alterations in cardiac trabecular development can manifest as a variety of congenital defects such as left ventricular noncompaction. In this review, we discuss the latest advances in understanding the molecular and cellular mechanisms underlying cardiac trabecular development.


Asunto(s)
Miocardio/patología , Miocitos Cardíacos/metabolismo , Humanos , Miocitos Cardíacos/citología
3.
EMBO Rep ; 22(11): e52901, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34523214

RESUMEN

Cardiac regeneration occurs primarily through proliferation of existing cardiomyocytes, but also involves complex interactions between distinct cardiac cell types including non-cardiomyocytes (non-CMs). However, the subpopulations, distinguishing molecular features, cellular functions, and intercellular interactions of non-CMs in heart regeneration remain largely unexplored. Using the LIGER algorithm, we assemble an atlas of cell states from 61,977 individual non-CM scRNA-seq profiles isolated at multiple time points during regeneration. This analysis reveals extensive non-CM cell diversity, including multiple macrophage (MC), fibroblast (FB), and endothelial cell (EC) subpopulations with unique spatiotemporal distributions, and suggests an important role for MC in inducing the activated FB and EC subpopulations. Indeed, pharmacological perturbation of MC function compromises the induction of the unique FB and EC subpopulations. Furthermore, we developed computational algorithm Topologizer to map the topological relationships and dynamic transitions between functional states. We uncover dynamic transitions between MC functional states and identify factors involved in mRNA processing and transcriptional regulation associated with the transition. Together, our single-cell transcriptomic analysis of non-CMs during cardiac regeneration provides a blueprint for interrogating the molecular and cellular basis of this process.


Asunto(s)
Miocitos Cardíacos , Pez Cebra , Animales , Proliferación Celular/genética , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Corazón/fisiología , Miocitos Cardíacos/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
5.
Phytopathology ; 111(7): 1152-1157, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33289404

RESUMEN

Tetrandrine (TET) is a potent calcium channel blocker used to treat hypertension and inflammation. Currently, TET is predominantly used to treat a variety of human diseases, and there is little information regarding the use of TET against plant pathogens. In this study, we explored the antifungal activity of TET on a plant pathogen, Botrytis cinerea. We show that administration of low concentrations of TET effectively inhibited hyphal growth of fungus grown on potato dextrose agarose and decreased the virulence of B. cinerea in tomato plants. Real-time PCR revealed that the expression of drug efflux pump-related genes (alcohol dehydrogenase 1, multidrug/pheromone exporter, pleiotropic drug resistance protein 1, and synaptic vesicle transporter) were downregulated in the presence of TET. Finally, we show that TET acts synergistically with iprodione, resulting in increased inhibition of B. cinerea both in vitro and in vivo. These results indicate that TET might act as an effective antifungal agent in reducing gray mold disease.


Asunto(s)
Antifúngicos , Botrytis , Antifúngicos/farmacología , Bencilisoquinolinas , Enfermedades de las Plantas , Virulencia
6.
Plant Dis ; 105(5): 1356-1364, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33258433

RESUMEN

Photinia (Photinia × fraseri Dress) is a well-known green plant that has high ornamental value and is widely distributed around the world. An outbreak of typical bud blight disease was observed between May and August in photinia in 2017 in Qingdao, China. The causal agent for this blight was subsequently isolated from symptomatic samples and identified as Nothophoma quercina based on morphological characterization and molecular analyses (ITS, LSU, RPB2, and TUB2). Results of pathogenicity tests on isolated fungi also supported the conclusion that N. quercina is the pathogen responsible for this condition. To our knowledge, this is the first report of bud blight on P. fraseri caused by N. quercina in China.


Asunto(s)
Ascomicetos , Photinia , Ascomicetos/genética , China
7.
Int J Mol Sci ; 22(16)2021 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-34445647

RESUMEN

Unveiling the molecular features in the heart is essential for the study of heart diseases. Non-cardiomyocytes (nonCMs) play critical roles in providing structural and mechanical support to the working myocardium. There is an increasing amount of single-cell RNA-sequencing (scRNA-seq) data characterizing the transcriptomic profiles of nonCM cells. However, no tool allows researchers to easily access the information. Thus, in this study, we develop an open-access web portal, ExpressHeart, to visualize scRNA-seq data of nonCMs from five laboratories encompassing three species. ExpressHeart enables comprehensive visualization of major cell types and subtypes in each study; visualizes gene expression in each cell type/subtype in various ways; and facilitates identifying cell-type-specific and species-specific marker genes. ExpressHeart also provides an interface to directly combine information across datasets, for example, generating lists of high confidence DEGs by taking the intersection across different datasets. Moreover, ExpressHeart performs comparisons across datasets. We show that some homolog genes (e.g., Mmp14 in mice and mmp14b in zebrafish) are expressed in different cell types between mice and zebrafish, suggesting different functions across species. We expect ExpressHeart to serve as a valuable portal for investigators, shedding light on the roles of genes on heart development in nonCM cells.


Asunto(s)
Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Ventrículos Cardíacos/metabolismo , Internet , Macrófagos/metabolismo , Pericitos/metabolismo , Transcriptoma , Algoritmos , Animales , Perfilación de la Expresión Génica , Humanos , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Programas Informáticos , Pez Cebra
8.
Circulation ; 139(23): 2668-2684, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30832495

RESUMEN

BACKGROUND: The adult mammalian cardiomyocytes lose their proliferative capacity, which is responsible for cardiac dysfunction and heart failure following injury. The molecular mechanisms underlying the attenuation of adult cardiomyocyte proliferation remain largely unknown. Because long noncoding RNAs (lncRNAs) have a critical role in the development of cardiovascular problems, we investigated whether lncRNAs have any role in the regulation of cardiomyocyte proliferation and cardiac repair. METHODS: Using bioinformatics and initial analysis, we identified an lncRNA, named CPR (cardiomyocyte proliferation regulator), that has a potential regulatory role in cardiomyocyte proliferation. For in vivo experiments, we generated CPR knockout and cardiac-specific CPR-overexpressing mice. In isolated cardiomyocytes, we used adenovirus for silencing (CPR-small interfering RNA) or overexpressing CPR. To investigate the mechanisms of CPR function in cardiomyocyte proliferation, we performed various analyses including quantitative reverse transcription-polymerase chain reaction, Western blot, histology, cardiac function (by echocardiography), transcriptome analyses (microarray assay), RNA pull-down assay, and chromatin immunoprecipitation assay. RESULTS: CPR level is comparatively higher in the adult heart than in the fetal stage. The silencing of CPR significantly increased cardiomyocyte proliferation in postnatal and adult hearts. Moreover, CPR deletion restored the heart function after myocardial injury, which was evident from increased cardiomyocyte proliferation, improvement of myocardial function, and reduced scar formation. In contrast, the neonatal cardiomyocyte proliferation and cardiac regeneration were remarkably suppressed in CPR-overexpressing mice or adeno-associated virus serotype 9-CPR-overexpressing heart. These results indicate that CPR acts as a negative regulator of cardiomyocyte proliferation and regeneration. Next, we found that CPR targets minichromosome maintenance 3, an initiator of DNA replication and cell cycle progression, to suppress cardiomyocyte proliferation. CPR silenced minichromosome maintenance 3 expression through directly interacting and recruiting DNMT3A to its promoter cysteine-phosphate-guanine sites, as evident from decreased minichromosome maintenance 3 promoter methylation and increased minichromosome maintenance 3 expression in CPR knocked-down cardiomyocytes and CPR knockout mouse heart. These results were confirmed in CPR-overexpressing cardiomyocytes and CPR-overexpressing mouse heart. CONCLUSIONS: Together, our findings identified that CPR is a suppressor of cardiomyocyte proliferation and indicated that lncRNAs take part in the regulation of cardiomyocyte proliferation and cardiac repair. Our study provides an lncRNA-based therapeutic strategy for effective cardiac repair and regeneration.


Asunto(s)
Proliferación Celular , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/metabolismo , Regeneración , Animales , Animales Recién Nacidos , Sitios de Unión , Ciclo Celular , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Componente 3 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 3 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Regiones Promotoras Genéticas , ARN Largo no Codificante/genética , Transducción de Señal
9.
Mol Cancer ; 18(1): 123, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31399034

RESUMEN

Piwi interacting RNAs (piRNAs) constitute novel small non-coding RNA molecules of approximately 24-31 nucleotides in length that often bind to members of the piwi protein family to play regulatory roles. Recently, emerging evidence suggests that in addition to the mammalian germline, piRNAs are also expressed in a tissue-specific manner in a variety of human tissues and modulate key signaling pathways at the transcriptional or post-transcriptional level. In addition, a growing number of studies have shown that piRNA and PIWI proteins, which are abnormally expressed in various cancers, may serve as novel biomarkers and therapeutic targets for tumor diagnostics and treatment. However, the functions of piRNAs in cancer and their underlying mechanisms remain incompletely understood. In this review, we discuss current findings regarding piRNA biogenetic processes, functions, and emerging roles in cancer, providing new insights regarding the potential applications of piRNAs and piwi proteins in cancer diagnosis and clinical treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Animales , Biomarcadores de Tumor , Epigénesis Genética , Perfilación de la Expresión Génica , Histonas , Humanos , Modelos Biológicos , Neoplasias/metabolismo , ARN Interferente Pequeño/metabolismo
10.
Cell Mol Life Sci ; 75(2): 291-300, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28913665

RESUMEN

Loss of functional cardiomyocytes is a major underlying mechanism for myocardial remodeling and heart diseases, due to the limited regenerative capacity of adult myocardium. Apoptosis, programmed necrosis, and autophagy contribute to loss of cardiac myocytes that control the balance of cardiac cell death and cell survival through multiple intricate signaling pathways. In recent years, non-coding RNAs (ncRNAs) have received much attention to uncover their roles in cell death of cardiovascular diseases, such as myocardial infarction, cardiac hypertrophy, and heart failure. In addition, based on the view that mitochondrial morphology is linked to three types of cell death, ncRNAs are able to regulate mitochondrial fission/fusion of cardiomyocytes by targeting genes involved in cell death pathways. This review focuses on recent progress regarding the complex relationship between apoptosis/necrosis/autophagy and ncRNAs in the context of myocardial cell death in response to stress. This review also provides insight into the treatment for heart diseases that will guide novel therapies in the future.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Enfermedades Cardiovasculares/genética , Miocitos Cardíacos/metabolismo , ARN no Traducido/genética , Animales , Enfermedades Cardiovasculares/metabolismo , Regulación de la Expresión Génica , Humanos , Modelos Genéticos , Necrosis , ARN no Traducido/metabolismo
11.
BMC Microbiol ; 18(1): 123, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30285616

RESUMEN

Following the publication of this article [1], the authors noticed that they mistakenly introduced duplicate images in Figure 6A during the preparation of figures. They apologize for any confusion that brought to the readers and have corrected the figure. This correction does not change any statement or conclusion drawn from the data.

12.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2871-2881, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28782654

RESUMEN

The pathogenesis of cardiac hypertrophy is tightly associated with mitochondrial dysfunction. Disequilibrium of mitochondrial dynamic is one of the main drivers in the pathological processes during development of various cardiac diseases. However, the effect of mitochondrial dynamics on cardiac hypertrophy remains largely unclear. MicroRNAs (miRNAs) are small noncoding RNAs that can switch off expression of many genes. Mitochondrial anchored protein ligase (MAPL) is a small ubiquitin-like modifier (SUMO) E3 ligase, which is an important contributor in mitochondrial fission process. In this study, we found that hypertrophic agonist phenylephrine (PE) enhanced the expression of MAPL and promoted mitochondrial fission, while it decreased the expression of mitochondrial fusion protein2 (Mfn2) in hypertrophic cardiomyocytes. Silencing expression of MAPL by siRNA attenuated PE-induced depletion of Mfn2 and increase of mitochondrial fission as well as hypertrophic response in cultured primary cardiomyocytes. MiR-485-5p is screened as a candidate inhibitor of MAPL. Overexpression of miR-485-5p blocked mitochondrial fission and hypertrophy induced by PE through inhibiting MAPL expression and increasing the level of Mfn2 in cultured primary cardiomyocytes. In mice model of cardiac hypertrophy induced by PE, the administration of miR-485-5p agomir significantly decreased the PE induced increase in the expression of MAPL and hypertrophic markers (ANP and ß-MHC) along with protection of cardiac structure and function. Together, this study exhibits a novel signaling axis composed of miR-485-5p/MAPL/Mfn2, which regulates mitochondrial machinery and cardiac hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , MicroARNs/metabolismo , Mitocondrias Cardíacas/metabolismo , Dinámicas Mitocondriales , Proteínas Mitocondriales/biosíntesis , Miocitos Cardíacos/metabolismo , Ubiquitina-Proteína Ligasas/biosíntesis , Animales , Cardiomegalia/patología , GTP Fosfohidrolasas , Regulación Enzimológica de la Expresión Génica , Proteínas de la Membrana/metabolismo , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley
13.
PLoS Pathog ; 11(4): e1004801, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25837042

RESUMEN

Genome dynamics of pathogenic organisms are driven by pathogen and host co-evolution, in which pathogen genomes are shaped to overcome stresses imposed by hosts with various genetic backgrounds through generation of a variety of isolates. This same principle applies to the rice blast pathogen Magnaporthe oryzae and the rice host; however, genetic variations among different isolates of M. oryzae remain largely unknown, particularly at genome and transcriptome levels. Here, we applied genomic and transcriptomic analytical tools to investigate M. oryzae isolate 98-06 that is the most aggressive in infection of susceptible rice cultivars. A unique 1.4 Mb of genomic sequences was found in isolate 98-06 in comparison to reference strain 70-15. Genome-wide expression profiling revealed the presence of two critical expression patterns of M. oryzae based on 64 known pathogenicity-related (PaR) genes. In addition, 134 candidate effectors with various segregation patterns were identified. Five tested proteins could suppress BAX-mediated programmed cell death in Nicotiana benthamiana leaves. Characterization of isolate-specific effector candidates Iug6 and Iug9 and PaR candidate Iug18 revealed that they have a role in fungal propagation and pathogenicity. Moreover, Iug6 and Iug9 are located exclusively in the biotrophic interfacial complex (BIC) and their overexpression leads to suppression of defense-related gene expression in rice, suggesting that they might participate in biotrophy by inhibiting the SA and ET pathways within the host. Thus, our studies identify novel effector and PaR proteins involved in pathogenicity of the highly aggressive M. oryzae field isolate 98-06, and reveal molecular and genomic dynamics in the evolution of M. oryzae and rice host interactions.


Asunto(s)
Evolución Biológica , Genoma Fúngico , Magnaporthe/genética , Magnaporthe/patogenicidad , Oryza/microbiología , Enfermedades de las Plantas/microbiología , Virulencia/genética , Secuencia de Bases , Proteínas Fúngicas/genética , Perfilación de la Expresión Génica , Genes Fúngicos/genética , Estudio de Asociación del Genoma Completo , Datos de Secuencia Molecular , Enfermedades de las Plantas/genética , Reacción en Cadena de la Polimerasa
14.
Clin Exp Pharmacol Physiol ; 44(4): 431-440, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28008641

RESUMEN

Myocardial apoptosis play a vital role in pathogenesis of cardiovascular diseases. The intrinsic pathway of apoptosis (mitochondrial apoptosis pathway) and abnormal mitochondrial fission and fusion have a detrimental effect on cells under a variety of intracellular stresses including hypoxia, oxidative stress, drug toxicity or DNA damage and contributes to the development of heart failure (HF), myocardial infarction (MI), diabetic cardiomyopathy and ischaemia/reperfusion injury (I/R). MicroRNAs (miRNAs) are endogenous short non-coding RNAs, which target 3'-untranslated region of mRNA to switch off gene expression. They play crucial roles in regulating complicated cardiac signalling and transcriptional events during cardiac development as well as in diseased condition. In this review, we summarize the molecular mechanism of mitochondrial apoptosis in cardiac cells and influence of miRNAs on them. MiRNAs regulate cardiac mitochondrial apoptosis by exert their effects on mitochondrial fission and fusion, reactive oxygen species (ROS) generation and Ca2+ homeostasis, Bcl-2 family members, and other mitochondrial function proteins. This advancement in understanding mechanism of cardiac cells death provides us new therapy targets for cardiovascular diseases associated with mitochondrial dysfunctions.


Asunto(s)
Apoptosis/genética , MicroARNs/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocardio/citología , Animales , Humanos , Mitocondrias Cardíacas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
15.
Eur Heart J ; 37(33): 2602-11, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26802132

RESUMEN

AIMS: Sustained cardiac hypertrophy accompanied by maladaptive cardiac remodelling represents an early event in the clinical course leading to heart failure. Maladaptive hypertrophy is considered to be a therapeutic target for heart failure. However, the molecular mechanisms that regulate cardiac hypertrophy are largely unknown. METHODS AND RESULTS: Here we show that a circular RNA (circRNA), which we term heart-related circRNA (HRCR), acts as an endogenous miR-223 sponge to inhibit cardiac hypertrophy and heart failure. miR-223 transgenic mice developed cardiac hypertrophy and heart failure, whereas miR-223-deficient mice were protected from hypertrophic stimuli, indicating that miR-223 acts as a positive regulator of cardiac hypertrophy. We identified ARC as a miR-223 downstream target to mediate the function of miR-223 in cardiac hypertrophy. Apoptosis repressor with CARD domain transgenic mice showed reduced hypertrophic responses. Further, we found that a circRNA HRCR functions as an endogenous miR-223 sponge to sequester and inhibit miR-223 activity, which resulted in the increase of ARC expression. Heart-related circRNA directly bound to miR-223 in cytoplasm and enforced expression of HRCR in cardiomyocytes and in mice both exhibited attenuated hypertrophic responses. CONCLUSIONS: These findings disclose a novel regulatory pathway that is composed of HRCR, miR-223, and ARC. Modulation of their levels provides an attractive therapeutic target for the treatment of cardiac hypertrophy and heart failure.


Asunto(s)
Insuficiencia Cardíaca , Animales , Cardiomegalia , Ratones , Ratones Transgénicos , MicroARNs , Miocitos Cardíacos
16.
Int J Mol Sci ; 18(4)2017 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-28362341

RESUMEN

MicroRNAs (miRNAs) are a class of small single-stranded and highly conserved non-coding RNAs, which are closely linked to cardiac disorders such as myocardial infarction (MI), cardiomyocyte hypertrophy, and heart failure. A growing number of studies have demonstrated that miRNAs determine the fate of the heart by regulating cardiac cell death and regeneration after MI. A deep understanding of the pathophysiology of miRNA dependent regulatory pathways in these processes is required. The role of miRNAs as diagnostic, prognostic, and therapeutic targets also needs to be explored in order to utilize them in clinical settings. This review summarizes the role of miRNAs in myocardial infarction and focuses mainly on their influence on cardiomyocyte regeneration and cell death including apoptosis, necrosis, and autophagy. In addition, the targets of pro- and anti-MI miRNAs are comparatively described. In particular, the possibilities of miRNA-based diagnostic and therapeutic strategies for myocardial infarction are discussed in this review.


Asunto(s)
Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , MicroARNs/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/metabolismo , Animales , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/terapia , Humanos , Modelos Cardiovasculares , Modelos Genéticos , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/terapia , Miocitos Cardíacos/patología , Investigación Biomédica Traslacional/métodos , Investigación Biomédica Traslacional/tendencias
17.
Curr Genet ; 62(1): 47-51, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26228744

RESUMEN

Plant diseases cause extensive yield loss of crops worldwide, and secretory 'warfare' occurs between plants and pathogenic organisms all the time. Filamentous plant pathogens have evolved the ability to manipulate host processes and facilitate colonization through secreting effectors inside plant cells. The stresses from hosts and environment can drive the genome dynamics of plant pathogens. Remarkable advances in plant pathology have been made owing to these adaptable genome regions of several lineages of filamentous phytopathogens. Characterization new effectors and interaction analyses between pathogens and plants have provided molecular insights into the plant pathways perturbed during the infection process. In this mini-review, we highlight promising approaches of identifying novel effectors based on the genome plasticity. We also discuss the interaction mechanisms between plants and their filamentous pathogens and outline the possibilities of effector gene expression under epigenetic control that will be future directions for research.


Asunto(s)
Hongos/genética , Genómica , Interacciones Huésped-Patógeno/genética , Enfermedades de las Plantas/genética , Enfermedades de las Plantas/microbiología , Plantas/genética , Plantas/microbiología , Evolución Biológica , Resistencia a la Enfermedad , Epigénesis Genética , Hongos/inmunología , Regulación Fúngica de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Genoma Fúngico , Genoma de Planta , Genómica/métodos , Interacciones Huésped-Patógeno/inmunología , Enfermedades de las Plantas/inmunología , Plantas/inmunología
18.
New Phytol ; 209(4): 1655-67, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26522477

RESUMEN

Soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) mediate cellular membrane fusion and intracellular vesicle trafficking in eukaryotic cells, and are critical in the growth and development of pathogenic fungi such as Magnaporthe oryzae which causes rice blast. Rice blast is thought to involve distinct SNARE-mediated transport and secretion of fungal effector proteins into the host to modulate rice immunity. We have previously characterized two SNARE proteins, secretory protein (MoSec22) and vesicle-associated membrane protein (MoVam7), as being important in cellular transport and pathogenicity. Here, we show that syntaxin 8 (MoSyn8), a Qc-SNARE protein homolog, also plays important roles in growth, conidiation, and pathogenicity. The MoSYN8 deletion mutant (∆Mosyn8) mutant exhibits defects in endocytosis and F-actin organization, appressorium turgor pressure generation, and host penetration. In addition, the ∆Mosyn8 mutant cannot elaborate biotrophic invasion of the susceptible rice host, or secrete avirulence factors Avr-Pia (corresponding to the rice resistance gene Pia) and Avrpiz-t (the cognate Avr gene for the resistance gene Piz-t) proteins. Our study of MoSyn8 advances our understanding of SNARE proteins in effector secretion which underlies the normal physiology and pathogenicity of M. oryzae, and it sheds new light on the mechanism of the blight disease caused by M. oryzae.


Asunto(s)
Espacio Intracelular/metabolismo , Magnaporthe/metabolismo , Magnaporthe/patogenicidad , Oryza/microbiología , Enfermedades de las Plantas/microbiología , Proteínas Qa-SNARE/metabolismo , Esporas Fúngicas/metabolismo , Actinas/metabolismo , Transporte Biológico , Endocitosis , Endosomas/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Genes Fúngicos , Magnaporthe/genética , Magnaporthe/crecimiento & desarrollo , Mutación/genética , Presión , Reproducción , Virulencia/genética
19.
Appl Microbiol Biotechnol ; 100(8): 3655-66, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26810198

RESUMEN

Orotate phosphoribosyl transferase (OPRTase) plays an important role in de novo and salvage pathways of nucleotide synthesis and is widely used as a screening marker in genetic transformation. However, the function of OPRTase in plant pathogens remains unclear. In this study, we characterized an ortholog of Saccharomyces cerevisiae Ura5, the OPRTase MoPyr5, from the rice blast fungus Magnaporthe oryzae. Targeted gene disruption revealed that MoPyr5 is required for mycelial growth, appressorial turgor pressure and penetration into plant tissues, invasive hyphal growth, and pathogenicity. Interestingly, the ∆Mopyr5 mutant is also involved in mycelial surface hydrophobicity. Exogenous uridine 5'-phosphate (UMP) restored vegetative growth and rescued the defect in pathogenicity on detached barley and rice leaf sheath. Collectively, our results show that MoPyr5 is an OPRTase for UMP biosynthesis in M. oryzae and indicate that UTP biosynthesis is closely linked with vegetative growth, cell wall integrity, and pathogenicity of fungus. Our results also suggest that UMP biosynthesis would be a good target for the development of novel fungicides against M. oryzae.


Asunto(s)
Proteínas Fúngicas/metabolismo , Magnaporthe/enzimología , Magnaporthe/patogenicidad , Orotato Fosforribosiltransferasa/metabolismo , Oryza/microbiología , Enfermedades de las Plantas/microbiología , Uridina Monofosfato/biosíntesis , Proteínas Fúngicas/genética , Magnaporthe/genética , Magnaporthe/crecimiento & desarrollo , Orotato Fosforribosiltransferasa/genética , Esporas Fúngicas/enzimología , Esporas Fúngicas/genética , Esporas Fúngicas/crecimiento & desarrollo , Esporas Fúngicas/patogenicidad , Virulencia
20.
BMC Microbiol ; 15: 37, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25885817

RESUMEN

BACKGROUND: The Myb super-family of proteins contain a group of functionally diverse transcriptional activators found in plant, animal and fungus. Myb proteins are involved in cell proliferation, differentiation and apoptosis, and have crucial roles in telomeres. The purpose of this study was to characterize the biological function of Myb1 protein in the rice blast fungus Magnaporthe oryzae. RESULTS: We identified the Saccharomyces cerevisiae BAS1 homolog MYB1 in M. oryzae, named MoMyb1. MoMyb1 encodes a protein of 322 amino acids and has two SANT domains and is well conserved in various organisms. Targeted gene deletion of MoMYB1 resulted in a significant reduction in vegetative growth and showed defects in conidiation and conidiophore development. Quantitative RT-PCR analysis revealed that the transcription levels of several conidiophore-related genes were apparently decreased in the ΔMomyb1 mutant. Inoculation with mycelia mats displayed that the virulence of the ΔMomyb1 mutant was not changed on rice leaves but was non-pathogenic on rice roots in comparison to the wild type Guy11. In addition, ∆Momyb1 mutants showed increased resistance to osmotic stresses but more sensitive to cell wall stressor calcofluor white (CFW). Further analysis revealed that MoMyb1 has an important role in the cell wall biosynthesis pathway. CONCLUSION: This study provides the evidence that MoMyb1 is a key regulator involved in conidiogenesis, stress response, cell wall integrity and pathogenesis on rice roots in the filamentous phytopathogen M. oryzae.


Asunto(s)
Magnaporthe/crecimiento & desarrollo , Oryza/microbiología , Enfermedades de las Plantas/microbiología , Proteínas de Unión a Telómeros/metabolismo , Factores de Virulencia/metabolismo , Eliminación de Gen , Perfilación de la Expresión Génica , Magnaporthe/genética , Magnaporthe/fisiología , Hojas de la Planta/microbiología , Raíces de Plantas/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Esporas Fúngicas/crecimiento & desarrollo , Proteínas de Unión a Telómeros/genética , Virulencia , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA