Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Circ Res ; 133(3): 237-251, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37345556

ABSTRACT

BACKGROUND: Progressive cardiac fibrosis leads to ventricular wall stiffness, cardiac dysfunction, and eventually heart failure, but the underlying mechanism remains unexplored. PDCD5 (programmed cell death 5) ubiquitously expresses in tissues, including the heart; however, the role of PDCD5 in cardiac fibrosis is largely unknown. Therefore, this study aims at exploring the possible role and underlying mechanisms of PDCD5 in the pathogenesis of cardiac fibrosis. METHODS AND RESULTS: PDCD5 levels were found to be elevated in the serum obtained from patients with cardiac fibrosis, in fibrotic mice heart tissues after myocardial infarction, and in cardiac fibroblasts stimulated by Ang II (angiotensin II)- or TGF-ß1 (transforming growth factor-ß1). Overexpression of PDCD5 in cardiac fibroblasts or treatment with PDCD5 protein reduced the expression of profibrogenic proteins in response to TGF-ß1 stimulation, while knockdown of PDCD5 increased fibrotic responses. It has been demonstrated that SMAD3, a protein that is also known as mothers against decapentaplegic homolog 3, directly upregulated PDCD5 during cardiac fibrosis. Subsequently, the increased PDCD5 promoted HDAC3 (histone deacetylase 3) ubiquitination, thus, inhibiting HDAC3 to reduce fibrotic responses. Fibroblast-specific knock-in of PDCD5 in mice ameliorated cardiac fibrosis after myocardial infarction and enhanced cardiac function, and these protective effects were eliminated by AAV9-mediated HDAC3 overexpression. CONCLUSIONS: The findings of this study demonstrated that PDCD5 is upregulated by SMAD3 during cardiac fibrosis, which subsequently ameliorated progressive fibrosis and cardiac dysfunction through HDAC3 inhibition. Thus, this study suggests that PDCD5 functions as a negative feedback factor on fibrotic signaling pathways and might serve as a potential therapeutic target to suppress the progression of fibrotic responses.


Subject(s)
Myocardial Infarction , Transforming Growth Factor beta1 , Mice , Animals , Transforming Growth Factor beta1/metabolism , Myocardial Infarction/metabolism , Heart , Fibroblasts/metabolism , Apoptosis , Fibrosis , Smad3 Protein/metabolism , Myocardium/metabolism
2.
Biochem Biophys Res Commun ; 667: 162-169, 2023 07 30.
Article in English | MEDLINE | ID: mdl-37229825

ABSTRACT

OBJECTIVES: Cardiac hypertrophy is the heart's compensatory response stimulated by various pathophysiological factors. However, prolonged cardiac hypertrophy poses a significant risk of progression to heart failure, lethal arrhythmias, and even sudden cardiac death. For this reason, it is crucial to effectively prevent the occurrence and development of cardiac hypertrophy. CMTM is a superfamily of human chemotaxis, which is involved in immune response and tumorigenesis. CMTM3 expressed widely in tissues, including the heart, but its cardiac function remains unclear. This research aims to explore the effect and mechanism of CMTM3 in the development of cardiac hypertrophy. METHODS AND RESULTS: We generated a Cmtm3 knockout mouse model (Cmtm3-/-) as the loss-of-function approach. CMTM3 deficiency induced cardiac hypertrophy and further exacerbated hypertrophy and cardiac dysfunction stimulated by Angiotensin Ⅱ infusion. In Ang Ⅱ-infusion stimulated hypertrophic hearts and phenylephrine-induced hypertrophic neonatal cardiomyocytes, CMTM3 expression significantly increased. However, adenovirus-mediated overexpression of CMTM3 inhibited the hypertrophy of rat neonatal cardiomyocytes induced by PE stimulation. In terms of mechanism, RNA-seq data revealed that Cmtm3 knockout-induced cardiac hypertrophy was related to MAPK/ERK activation. In vitro, CMTM3 overexpression significantly inhibited the increased phosphorylation of p38 and ERK induced by PE stimulation. CONCLUSIONS: CMTM3 deficiency induces cardiac hypertrophy and aggravates hypertrophy and impaired cardiac function stimulated by angiotensin Ⅱ infusion. The expression of CMTM3 increases during cardiac hypertrophy, and the increased CMTM3 can inhibit further hypertrophy of cardiomyocytes by inhibiting MAPK signaling. Thus, CMTM3 plays a negative regulatory effect in the occurrence and development of cardiac hypertrophy.


Subject(s)
Cardiomegaly , Chemokines , MARVEL Domain-Containing Proteins , Animals , Mice , Cardiomegaly/metabolism , MARVEL Domain-Containing Proteins/genetics , MARVEL Domain-Containing Proteins/metabolism , Chemokines/genetics , Chemokines/metabolism , Gene Knockout Techniques , Angiotensin II/metabolism , Myocytes, Cardiac/metabolism , Up-Regulation , Phenylephrine , Rats , p38 Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Heart
3.
Front Physiol ; 13: 1047909, 2022.
Article in English | MEDLINE | ID: mdl-36467681

ABSTRACT

Background: Hemorrhagic shock (HS) is a type of hypovolemic shock characterized by hemodynamic instability, tissue hypoperfusion and cellular hypoxia. In pathophysiology, the gradual accumulation of reactive oxygen species (ROS) damages the mitochondria, leading to irreversible cell damage and the release of endogenous damage-associated molecular patterns (DAMPs) including mitochondrial DAMPs (MTDs), eventually triggering the inflammatory response. The novel mitochondria-targeted antioxidant SkQ1 (Visomitin) effectively eliminate excessive intracellular ROS and exhibits anti-inflammatory effects; however, the specific role of SkQ1 in HS has not yet been explicated. Methods and results: A 40% fixed-blood-loss HS rat model was established in this study. Transmission electron microscopy showed that after HS, the myocardial mitochondrial ultrastructure was damaged and the mtDNA release in circulation was increased and the differentially expressed genes were significantly enriched in mitochondrial and ROS-related pathways. Mitochondria-targeted antioxidant SkQ1 attenuated the increased ROS induced by HS in myocardial tissues and by oxygen-glucose deprivation (OGD) in cardiomyocytes. Ultrastructurally, SkQ1 protected the myocardial mitochondrial structure and reduced the release of the peripheral blood mtDNA after HS. RNA-seq transcriptome analysis showed that 56.5% of the inflammation-related genes, which altered after HS, could be significantly reversed after SkQ1 treatment. Moreover, ELISA indicated that SkQ1 significantly reversed the HS-induced increases in the TNF-α, IL-6, and MCP-1 protein levels in rat peripheral blood. Conclusion: HS causes damage to the rat myocardial mitochondrial structure, increases mtDNA release and ROS contents, activates the mitochondrial and ROS-related pathways, and induces systemic inflammatory response. The mitochondrial antioxidant SkQ1 can improve rat myocardial mitochondria ultrastructure, reduce mtDNA and ROS contents, and decrease inflammation by protecting myocardial mitochondria, thereby playing a novel protective role in HS.

4.
Front Cell Dev Biol ; 10: 792774, 2022.
Article in English | MEDLINE | ID: mdl-35252172

ABSTRACT

Cardiac diseases compose a fatal disease category worldwide. Over the past decade, high-throughput transcriptome sequencing of bulk heart tissues has widened our understanding of the onset and progression of cardiac diseases. The recent rise of single-cell RNA sequencing (scRNA-seq) technology further enables deep explorations of their molecular mechanisms in a cell-type-specific manner. However, due to technical difficulties in performing scRNA-seq on heart tissues, there are still few scRNA-seq studies on cardiac diseases. In this study, we demonstrate that an effective alternative could be cell-type-specific computational reconstruction of bulk transcriptomes. An integrative bulk transcriptome dataset covering 110 samples from 12 studies was first constructed by re-analysis of raw sequencing data derived from the heart tissues of four common cardiac disease mouse models (myocardial infarction, dilated cardiomyopathy, hypertrophic cardiomyopathy, and arrhythmogenic right ventricular cardiomyopathy). Based on the single-cell reference covering four major cardiac component cell types and 22 immune cell subtypes, for each sample, the bulk transcriptome was reconstructed into cellular compositions and cell-type-specific expression profiles by CIBERSORTx. Variations in the estimated cell composition revealed elevated abundances of fibroblast and monocyte during myocardial infarction, which were further verified by our flow cytometry experiment. Moreover, through cell-type-specific differential gene expression and pathway enrichment analysis, we observed a series of signaling pathways that mapped to specific cell type in diseases, like MAPK and EGFR1 signaling pathways in fibroblasts in myocardial infarction. We also found an increased expression of several secretory proteins in monocytes which may serve as regulatory factors in cardiac fibrosis. Finally, a ligand-receptor analysis identified key cell types which may serve as hubs in cellular communication in cardiac diseases. Our results provide novel clues for the cell-type-specific signatures of cardiac diseases that would promote better understanding of their pathophysiological mechanisms.

5.
J Exp Bot ; 72(8): 2947-2964, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33476364

ABSTRACT

High temperature often leads to failure of grain filling in rice (Oryza sativa) causing yield loss, but the underlying mechanisms are still not elucidated. Here, we report that two genes encoding seed-specific NAM/ATAF/CUC (NAC) domain transcription factors, ONAC127 and ONAC129, are responsive to heat stress and involved in the grain filling process of rice. ONAC127 and ONAC129 are dominantly expressed in the pericarp and can form a heterodimer during rice grain filling. CRISPR/Cas9 induced mutants and overexpression lines were then generated to investigate the function of these two transcription factors. Interestingly, both knock-out and overexpression plants showed incomplete grain filling and shrunken grains, which became more severe under heat stress. Transcriptome analysis revealed that ONAC127 and ONAC129 mainly regulate stimulus response and nutrient transport. ChIP-seq analysis identified that the direct target genes of ONAC127 and ONAC129 in developing rice seeds include monosaccharide transporter gene OsMST6, sugar transporter gene OsSWEET4, calmodulin-like protein gene OsMSR2 and AP2/ERF factor gene OsEATB. These results suggest that ONAC127 and ONAC129 regulate grain filling by affecting sugar transportation and abiotic stress responses. Overall, this study demonstrates a transcriptional regulatory network with ONAC127 and ONAC129 coordinating multiple pathways to modulate seed development and heat stress responses at rice reproductive stages.


Subject(s)
Heat Shock Transcription Factors , Oryza , Plant Proteins , Seeds/growth & development , Edible Grain/genetics , Edible Grain/metabolism , Gene Expression Regulation, Plant , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/genetics , Oryza/genetics , Oryza/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism
6.
J Exp Bot ; 70(15): 3765-3780, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31211389

ABSTRACT

Starch and storage proteins, the primary storage substances of cereal endosperm, are a major source of food for humans. However, the transcriptional regulatory networks of the synthesis and accumulation of storage substances remain largely unknown. Here, we identified a rice endosperm-specific gene, NF-YC12, that encodes a putative nuclear factor-Y transcription factor subunit C. NF-YC12 is expressed in the aleurone layer and starchy endosperm during grain development. Knockout of NF-YC12 significantly decreased grain weight as well as altering starch and protein accumulation and starch granule formation. RNA-sequencing analysis revealed that in the nf-yc12 mutant genes related to starch biosynthesis and the metabolism of energy reserves were enriched in the down-regulated category. In addition, starch and protein contents in seeds differed between NF-YC12-overexpression lines and the wild-type. NF-YC12 was found to interact with NF-YB1. ChIP-qPCR and yeast one-hybrid assays showed that NF-YC12 regulated the rice sucrose transporter OsSUT1 in coordination with NF-YB1 in the aleurone layer. In addition, NF-YC12 was directly bound to the promoters of FLO6 (FLOURY ENDOSPERM6) and OsGS1;3 (glutamine synthetase1) in developing endosperm. This study demonstrates a transcriptional regulatory network involving NF-YC12, which coordinates multiple pathways to regulate endosperm development and the accumulation of storage substances in rice seeds.


Subject(s)
Oryza/metabolism , Plant Proteins/metabolism , Seeds/metabolism , Transcription Factors/metabolism , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Gene Expression Regulation, Plant , Oryza/genetics , Plant Proteins/genetics , Seeds/genetics , Transcription Factors/genetics
7.
Plant Sci ; 280: 219-227, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30824000

ABSTRACT

Grain size and shape are important factors in determining the grain yield. In this study, OsNF-YC10, a member of the NF-Y transcription factor family encoding a putative histone transcription factor, was cloned and characterized. qRT-PCR and mRNA in situ hybridization analysis revealed that OsNF-YC10 was highly expressed in endosperm and spikelet hull at late developmental stages. The results showed that OsNF-YC10 was a nuclear protein showing transcription activation activity. The osnf-yc10 lines, produced using CRISPR/Cas9 technology, showed narrow, thin and light grains. Cytological experiments revealed significantly reduced cell number of spikelet hull in osnf-yc10 lines compared with that in WT. Narrow, thin, and light grains were found consistently in OsNF-YC10 RNAi transgenic lines. Moreover, the number of cells decreased in the grain-width direction than WT. These results indicated that OsNF-YC10 plays an important role in determining grain size and shape. OsNF-YC10 was further revealed to influence the expression of GW8 (a positive regulator of grain width), GW7 (a negative regulator of grain width) and cell cycle-regulated genes CYCD4, CYCA2.1, CYCB2.1, CYCB2.2, E2F2. Taken together, it is suggested that OsNF-YC10 regulates the grains size and shape by influencing the cell proliferation of spikelet hulls.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Plant , Oryza/genetics , Transcription Factors/metabolism , Cell Nucleus/metabolism , Endosperm/genetics , Endosperm/growth & development , Endosperm/physiology , Organ Specificity , Oryza/growth & development , Oryza/physiology , Phenotype , Plant Proteins/genetics , Plant Proteins/metabolism , Plants, Genetically Modified , RNA Interference , Seeds/genetics , Seeds/growth & development , Seeds/physiology , Transcription Factors/genetics , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...