Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Theranostics ; 10(23): 10680-10696, 2020.
Article in English | MEDLINE | ID: mdl-32929374

ABSTRACT

Background: Oxidative stress has emerged as an essential factor in the pathogenesis of intestinal ischemia/reperfusion (I/R) injury. The adaptor protein p66Shc is a key regulator of reactive oxygen species (ROS) generation and a mediator of I/R damage in the intestine, but the upstream mechanisms that directly regulate p66Shc expression during intestinal I/R remain largely unknown. Recent studies have suggested that noncoding RNAs, such as circular RNAs (circRNAs), are important players in physiological and pathological processes based on their versatile regulatory roles in gene expression. The aim of this study was to elucidate the contribution of p66Shc to oxidative damage in intestinal I/R and to investigate the regulation of p66Shc by circRNA sponges. Methods: Intestinal I/R was induced in mice via superior mesenteric artery (SMA) occlusion. A miR-339-5p agomir or circ-protein kinase C beta (PRKCB) siRNA was injected intravenously before I/R challenge. In addition, Caco-2 cells were subjected to hypoxia/reoxygenation (H/R) in vitro to simulate an in vivo I/R model. Results:In vitro, p66Shc deficiency significantly reduced H/R-induced ROS overproduction by attenuating mitochondrial superoxide anion (O2-) levels, suppressing NADPH oxidase activity and enhancing antioxidant enzyme expression. Moreover, miR-339-5p was identified to directly regulate p66Shc expression in the intestine. Furthermore, we found that a circRNA transcribed from the PRKCB gene, named circ-PRKCB, acted as an endogenous miR-339-5p sponge to regulate p66Shc expression. circ-PRKCB silencing or miR-339-5p overexpression significantly downregulated p66Shc expression and attenuated oxidative stress levels and I/R injury in vivo and in vitro. Notably, the increased circ-PRKCB levels and decreased miR-339-5p levels associated with murine intestinal I/R were consistent with those in patients with intestinal infarction. Conclusions: Our findings reveal a crucial role for the circ-PRKCB/miR-339-5p/p66Shc signaling pathway in regulating oxidative stress in the I/R intestine. This pathway may be a potential therapeutic target for intestinal I/R injury.


Subject(s)
Intestinal Mucosa/blood supply , MicroRNAs/metabolism , RNA, Circular/metabolism , Reperfusion Injury/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Animals , Disease Models, Animal , Gene Knockdown Techniques , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Male , Mice , MicroRNAs/agonists , MicroRNAs/genetics , Mitochondria/drug effects , Mitochondria/pathology , Oxidative Stress/drug effects , Oxidative Stress/genetics , RNA, Circular/genetics , RNA, Small Interfering/administration & dosage , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
2.
Pharmacol Res ; 160: 105197, 2020 10.
Article in English | MEDLINE | ID: mdl-32942015

ABSTRACT

Parkin is a crucial protein that promotes the clearance of damaged mitochondria via mitophagy in neuron, and parkin mutations result in autosomal-recessive Parkinson's disease (AR-PD). However, the exact mechanisms underlying the regulation of Parkin-mediated mitophagy in PD remain unclear. In this study, PD models were generated through incubation of SH-SY5Y cells with 1-methyl-4-phenylpyridinium ion (MPP+, 1.5 mM for 24 h) and intraperitoneal injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP, 30 mg/kg for five consecutive days) in mice. A Bioinformatics database was used to identify Parkin-targeting microRNAs (miRNAs). Then, miR-103a-3p agomir, miR-103a-3p antagomir and Parkin siRNA were used to assess the effects of miR-103a-3p/Parkin/Ambra1 signaling-mediated mitophagy in PD in vitro and in vivo. The protein and mRNA levels of Parkin and Ambra1 were significantly decreased, while miR-103a-3p, which is a highly expressed miRNA in the human brain, was obviously increased in PD mouse and SH-SY5Y cell models. Moreover, miR-103a-3p suppressed Parkin expression by targeting a conserved binding site in the 3'-untranslated region (UTR) of Parkin mRNA. Importantly, miR-103a-3p inhibition resulted in neuroprotective effects and improved mitophagy in vitro and in vivo, whereas Parkin siRNA strongly abolished these effects. These findings suggested that miR-103a-3p inhibition has neuroprotective effects in PD, which may be involved in regulating mitophagy through the Parkin/Ambra1 pathway. Modulating miR-103a-3p levels may be an applicable therapeutic strategy for PD.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , MicroRNAs/genetics , Mitophagy/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions/genetics , Animals , Cell Line , Computational Biology , Dopamine/metabolism , Humans , MPTP Poisoning/drug therapy , MPTP Poisoning/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Point Mutation , RNA, Small Interfering/pharmacology
3.
Mol Ther Nucleic Acids ; 21: 751-763, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32781430

ABSTRACT

We previously found that inhibition of p66Shc confers protection against hepatic stellate cell (HSC) activation during liver fibrosis. However, the effect of p66Shc on HSC proliferation, as well as the mechanism by which p66Shc is modulated, remains unknown. Here, we elucidated the effect of p66Shc on HSC proliferation and evaluated microRNA (miRNA)-p66Shc-mediated reactive oxidative species (ROS) generation in liver fibrosis. An in vivo model of carbon tetrachloride (CCl4)-induced liver fibrosis in rats and an LX-2 cell model were developed. p66Shc expression was significantly upregulated in rats with CCl4-induced liver fibrosis and in human fibrotic livers. Additionally, p66Shc knockdown in vitro attenuated mitochondrial ROS generation and HSC proliferation. Interestingly, p66Shc promoted HSC proliferation via ß-catenin dephosphorylation in vitro. MicroRNA (miR)-203a-3p, which was identified by microarray and bioinformatics analyses, directly inhibited p66Shc translation and attenuated HSC proliferation in vitro. Importantly, p66Shc was found to play an indispensable role in the protective effect of miR-203a-3p. Furthermore, carnosic acid (CA), the major antioxidant compound extracted from rosemary leaves, protected against CCl4-induced liver fibrosis through the miR-203a-3p/p66Shc axis. Collectively, these results suggest that p66Shc, which is directly suppressed by miR-203a-3p, is a key regulator of liver fibrosis. This finding may lead to the development of therapeutic targets for liver fibrosis.

4.
Toxicol Appl Pharmacol ; 403: 115125, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32659284

ABSTRACT

Epithelial-mesenchymal transition (EMT) is regulated by reactive oxygen species (ROS) in liver fibrosis. p66Shc is a redox enzyme, but its role of EMT is unclear in liver fibrosis. Long noncoding RNAs (lncRNAs) have been implicated as important regulators in numerous physiological and pathological processes and generally acting as a microRNA (miRNA) sponge to regulate gene expression. The aim of the current study was to evaluate the contribution of p66Shc to EMT in liver fibrosis and the regulation of p66Shc by lncRNA sponge. In vivo, p66Shc silencing prevented carbon tetrachloride (CCl4)-induced EMT as evidenced by the upregulation of E-cadherin, downregulation of Vimentin and N-cadherin, and inhibition of oxidative stress and extracellular matrix (ECM) components. Moreover, in vitro, TGF-ß1 significantly enhanced ECM components, as well as the development of the EMT phenotype. These effects were abrogated by p66Shc downregulation and aggravated by p66Shc overexpression. Mechanistically, p66Shc contributed to EMT via mediating ROS, as evidenced by p66Shc downregulation inhibiting EMT under exogenous hydrogen peroxide (H2O2) stimulation. Furthermore, we found that molecule interacting with CasL2 (Mical2) lncRNA functioned as an endogenous miR-203a-3p sponge to regulate p66Shc expression. Both Mical2 silencing and miR-203a-3p agomiR treatment downregulated p66Shc expression, thus suppressing EMT in vivo and in vitro. Notably, the increased p66Shc and Mical2 levels and decreased miR-203a-3p levels in murine fibrosis were consistent with those in patients with liver fibrosis. In sum, our study reveals that p66Shc is critical for liver fibrosis and that Mical2, miR-203a-3p and p66Shc compose a novel regulatory pathway in liver fibrosis.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Animals , Carbon Tetrachloride Poisoning , Cell Line , Down-Regulation , Gene Expression Regulation , Gene Silencing , Hepatocytes , Humans , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mice , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Random Allocation , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
5.
World J Gastroenterol ; 26(15): 1758-1774, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32351292

ABSTRACT

BACKGROUND: Intestinal ischemia reperfusion (I/R) occurs in various diseases, such as trauma and intestinal transplantation. Excessive reactive oxygen species (ROS) accumulation and subsequent apoptotic cell death in intestinal epithelia are important causes of I/R injury. PTEN-induced putative kinase 1 (PINK1) and phosphorylation of dynamin-related protein 1 (DRP1) are critical regulators of ROS and apoptosis. However, the correlation of PINK1 and DRP1 and their function in intestinal I/R injury have not been investigated. Thus, examining the PINK1/DRP1 pathway may help to identify a protective strategy and improve the patient prognosis. AIM: To clarify the mechanism of the PINK1/DRP1 pathway in intestinal I/R injury. METHODS: Male C57BL/6 mice were used to generate an intestinal I/R model via superior mesenteric artery occlusion followed by reperfusion. Chiu's score was used to evaluate intestinal mucosa damage. The mitochondrial fission inhibitor mdivi-1 was administered by intraperitoneal injection. Caco-2 cells were incubated in vitro in hypoxia/reoxygenation conditions. Small interfering RNAs and overexpression plasmids were transfected to regulate PINK1 expression. The protein expression levels of PINK1, DRP1, p-DRP1 and cleaved caspase 3 were measured by Western blotting. Cell viability was evaluated using a Cell Counting Kit-8 assay and cell apoptosis was analyzed by TUNEL staining. Mitochondrial fission and ROS were tested by MitoTracker and MitoSOX respectively. RESULTS: Intestinal I/R and Caco-2 cell hypoxia/reoxygenation decreased the expression of PINK1 and p-DRP1 Ser637. Pretreatment with mdivi-1 inhibited mitochondrial fission, ROS generation, and apoptosis and ameliorated cell injury in intestinal I/R. Upon PINK1 knockdown or overexpression in vitro, we found that p-DRP1 Ser637 expression and DRP1 recruitment to the mitochondria were associated with PINK1. Furthermore, we verified the physical combination of PINK1 and p-DRP1 Ser637. CONCLUSION: PINK1 is correlated with mitochondrial fission and apoptosis by regulating DRP1 phosphorylation in intestinal I/R. These results suggest that the PINK1/DRP1 pathway is involved in intestinal I/R injury, and provide a new approach for prevention and treatment.


Subject(s)
Dynamins/metabolism , Mesenteric Ischemia/pathology , Protein Kinases/metabolism , Reperfusion Injury/pathology , Animals , Apoptosis/genetics , Caco-2 Cells , Cell Hypoxia , Disease Models, Animal , Gene Knockdown Techniques , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Intestine, Small/blood supply , Intestine, Small/pathology , Male , Mesenteric Artery, Superior/surgery , Mesenteric Ischemia/etiology , Mice , Mitochondria/pathology , Mitochondrial Dynamics/genetics , Phosphorylation/genetics , Protein Kinases/genetics , RNA, Small Interfering/metabolism , Reperfusion Injury/etiology , Serine/metabolism
6.
Oxid Med Cell Longev ; 2019: 3527809, 2019.
Article in English | MEDLINE | ID: mdl-31428222

ABSTRACT

Alcohol abuse has become common worldwide and has been recognized as a major cause of chronic alcoholic liver disease (ALD). ALD encompasses a complex process that includes a broad scope of hepatic lesions, ranging from steatosis to cirrhosis. In particular, reactive oxygen species (ROS) are mainly involved. Numerous studies have shown that p66shc plays a significant role in ALD. Protocatechuic acid (PCA), a dihydroxybenzoic acid that is naturally found in green tea, vegetables, and fruits, has efficient free radical scavenging effects. In this study, we aimed to assess the protective effect of PCA on ALD and to evaluate the microRNA- (miRNA-) p66shc-mediated reduction of ROS formation in ALD. Our results demonstrated that PCA treatment significantly decreased p66shc expression and downstream ROS formation in ALD. miR-219a-5p, which was identified by bioinformatics and experimental analysis, was enhanced by PCA and subsequently suppressed p66shc expression. Importantly, p66shc played an essential role in the protection of PCA-stimulated miR-219a-5p overexpression. Overall, these findings show that PCA-stimulated miR-219a-5p expression mitigates ALD by reducing p66shc-mediated ROS formation. This study may contribute to the development of therapeutic interventions for ALD.


Subject(s)
Hydroxybenzoates/pharmacology , Liver Diseases, Alcoholic/drug therapy , MicroRNAs/metabolism , Protective Agents/therapeutic use , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , 3' Untranslated Regions , Animals , Cell Survival/drug effects , Down-Regulation/drug effects , Ethanol/toxicity , Glutathione/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hydroxybenzoates/therapeutic use , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Male , Mice , MicroRNAs/chemistry , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 1/chemistry , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Superoxide Dismutase/metabolism
7.
Theranostics ; 9(5): 1510-1522, 2019.
Article in English | MEDLINE | ID: mdl-30867846

ABSTRACT

Background: p66Shc is a redox enzyme that mediates mitochondrial reactive oxygen species (ROS) generation. p66Shc inhibition confers protection against liver injury, however, its functional contribution to liver fibrosis remains unclear. The aim of this study is to explore the involvement of p66Shc in liver fibrosis and underlying mechanism of p66Shc by focusing on mitochondrial ROS. Methods: p66Shc-silenced mice were injected with carbon tetrachloride (CCl4). Primary hepatic stellate cells (HSCs) were performed with p66Shc silencing or overexpression prior to TGF-ß1 stimulation. Results: p66Shc expression was progressively elevated in mice with CCl4-induced liver fibrosis, and p66Shc silencing in vivo significantly attenuated fibrosis development, reducing liver damage, oxidative stress and HSC activation, indicated by the decreased α-SMA, CTGF and TIMP1 levels. Furthermore, in primary HSCs, p66Shc-mediated mitochondrial ROS production played a vital role in mitochondrial morphology and cellular metabolism. Knockdown of p66Shc significantly inhibited mitochondrial ROS production and NOD-like receptor protein 3 (NLRP3) inflammasome activation, which were closely associated with HSC activation, indicated by the decreased α-SMA, CTGF and TIMP1 levels. However, p66Shc overexpression exerted the opposite effects, which were suppressed by a specific mitochondrial ROS scavenger (mito-TEMPO). More importantly, p66Shc expression was significantly increased in human with liver fibrosis, accompanied by NLRP3 inflammasome activation. Conclusions: p66Shc is a key regulator of liver fibrosis by mediating mitochondrial ROS production, which triggers NLRP3 inflammasome activation.


Subject(s)
Hepatic Stellate Cells/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Animals , Carbon Tetrachloride/administration & dosage , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Inflammasomes/metabolism , Liver Cirrhosis/chemically induced , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
8.
World J Gastroenterol ; 25(7): 824-836, 2019 Feb 21.
Article in English | MEDLINE | ID: mdl-30809082

ABSTRACT

BACKGROUND: Intestinal ischemia reperfusion (I/R) injury is a serious but common pathophysiological process of many diseases, resulting in a high mortality rate in clinical practice. Ubiquitin-specific protease 22 (USP22) acts as regulator of cell cycle progression, proliferation, and tumor invasion. Depleted USP22 expression has been reported to contribute to arrested cell cycle and disrupted generation of differentiated cell types in crypts and villi. However, the role of USP22 in intestinal damage recovery has not been investigated. Therefore, elucidation of the underlying mechanism of USP22 in intestinal I/R injury may help to improve the tissue repair and patient prognosis in clinical practice. AIM: To investigate the role of USP22 in intestinal cell proliferation and regeneration after intestinal I/R injury. METHODS: An animal model of intestinal I/R injury was generated in male Sprague-Dawley rats by occlusion of the superior mesenteric artery followed by reperfusion. Chiu's scoring system was used to grade the damage to the intestinal mucosa. An in vitro model was developed by incubating rat intestinal epithelial IEC-6 cells in hypoxia/reoxygenation conditions in order to simulate I/R in vivo. siRNA and overexpression plasmid were used to regulate the expression of USP22. USP22, Cyclin D1, and proliferating cell nuclear antigen (PCNA) expression levels were measured by Western blot analysis and immunohistochemistry staining. Cell survival (viability) and cell cycle were evaluated using the Cell Counting Kit-8 and flow cytometry, respectively. RESULTS: USP22 expression was positively correlated with the expression levels of PCNA and Cyclin D1 both in vivo and in vitro, which confirmed that USP22 was involved in cell proliferation and intestinal regeneration after intestinal I/R injury. Decreased levels of Cyclin D1 and cell cycle arrest were observed in the USP22 knockdown group (P < 0.05), while opposite results were observed in the USP22 overexpression group (P < 0.05). In addition, increased expression of USP22 was related to improved intestinal pathology or IEC-6 cell viability after I/R or hypoxia/reoxygenation. These results suggested that USP22 may exert a protective effect on intestinal I/R injury by regulating cell proliferation and facilitating tissue regeneration. CONCLUSION: USP22 is correlated with promoting intestinal cell proliferation and accelerating intestinal tissue regeneration after intestinal I/R injury and may serve as a potential target for therapeutic development for tissue repair during intestinal I/R injury.


Subject(s)
Cell Proliferation , Deubiquitinating Enzymes/metabolism , Intestinal Mucosa/pathology , Regeneration , Reperfusion Injury/pathology , Ubiquitin-Specific Proteases/metabolism , Animals , Cell Line , Deubiquitinating Enzymes/genetics , Disease Models, Animal , Humans , Intestinal Mucosa/cytology , Male , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/etiology
9.
Cell Death Differ ; 26(11): 2284-2299, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30737476

ABSTRACT

Ferroptosis is a recently identified form of regulated cell death defined by the iron-dependent accumulation of lipid reactive oxygen species. Ferroptosis has been studied in various diseases such as cancer, Parkinson's disease, and stroke. However, the exact function and mechanism of ferroptosis in ischemia/reperfusion (I/R) injury, especially in the intestine, remains unknown. Considering the unique conditions required for ferroptosis, we hypothesize that ischemia promotes ferroptosis immediately after intestinal reperfusion. In contrast to conventional strategies employed in I/R studies, we focused on the ischemic phase. Here we verified ferroptosis by assessing proferroptotic changes after ischemia along with protein and lipid peroxidation levels during reperfusion. The inhibition of ferroptosis by liproxstatin-1 ameliorated I/R-induced intestinal injury. Acyl-CoA synthetase long-chain family member 4 (ACSL4), which is a key enzyme that regulates lipid composition, has been shown to contribute to the execution of ferroptosis, but its role in I/R needs clarification. In the present study, we used rosiglitazone (ROSI) and siRNA to inhibit ischemia/hypoxia-induced ACSL4 in vivo and in vitro. The results demonstrated that ACSL4 inhibition before reperfusion protected against ferroptosis and cell death. Further investigation revealed that special protein 1 (Sp1) was a crucial transcription factor that increased ACSL4 transcription by binding to the ACSL4 promoter region. Collectively, this study demonstrates that ferroptosis is closely associated with intestinal I/R injury, and that ACSL4 has a critical role in this lethal process. Sp1 is an important factor in promoting ACSL4 expression. These results suggest a unique and effective mechanistic approach for intestinal I/R injury prevention and treatment.


Subject(s)
Coenzyme A Ligases/metabolism , Ferroptosis/physiology , Intestines/injuries , Reperfusion Injury/pathology , Sp1 Transcription Factor/metabolism , Animals , Caco-2 Cells , Cell Line, Tumor , Coenzyme A Ligases/antagonists & inhibitors , Coenzyme A Ligases/genetics , DNA-Binding Proteins/metabolism , Ferroptosis/drug effects , Humans , Intestines/pathology , Lipid Peroxidation/physiology , Mice , Mice, Inbred C57BL , Models, Animal , Promoter Regions, Genetic/genetics , Quinoxalines/pharmacology , Reactive Oxygen Species/metabolism , Rosiglitazone/pharmacology , Spiro Compounds/pharmacology
10.
Shock ; 52(3): 387-395, 2019 09.
Article in English | MEDLINE | ID: mdl-30216227

ABSTRACT

Intestinal ischemia/reperfusion (I/R)-induced systemic inflammation leads to multiple organ dysfunction syndrome. Previous studies have indicated that the NOD-like receptor protein (NLRP)3 inflammasome modulates intestinal inflammation; however, the pathophysiological mechanisms remain unclear. Autophagy is a critical metabolic mechanism that promotes cellular survival following ischemic injury. Recently, basal autophagy has been implicated in the alleviation of extensive inflammation. However, the role of autophagy in NLRP3 inflammasome activation in intestinal I/R-induced inflammatory injury remains undefined. In the present study, we examined whether NLRP3 inflammasome activation is induced in mice subjected to intestinal I/R injury, which is measured as increased apoptosis-associated speck-like protein containing a CARD levels, caspase-1 activity, and interleukin-1ß (IL-1ß) secretion. Importantly, the in-vitro results showed that NLRP3 knockdown decreases proinflammatory cytokine production and increases resistance to hypoxia/reoxygenation (H/R)-triggered inflammation. Subsequently, we demonstrated a critical role for autophagy in suppressing intestinal I/R-induced NLRP3 inflammasome activation in vivo. Furthermore, we showed that the loss of autophagy activates inflammasome-mediated IL-1ß secretion, which aggravates H/R injury, and NLRP3 knockdown reverses these effects. Collectively, these results directly implicated the homeostatic process of autophagy and NLRP3 inflammasome in ischemic bowel disease and identified a novel pathway for therapeutic intervention in intestinal I/R.


Subject(s)
Autophagic Cell Death , Inflammasomes/metabolism , Intestinal Diseases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/metabolism , Animals , Gene Knockdown Techniques , Inflammasomes/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology
11.
Apoptosis ; 23(11-12): 667-678, 2018 12.
Article in English | MEDLINE | ID: mdl-30284149

ABSTRACT

Epithelial apoptosis is an important factor in intestinal ischemia-reperfusion (I/R) injury. Heat shock factor 1 (HSF1) is a classical stress response factor that directly regulates the transcription of heat shock proteins (HSPs) under stress conditions. Although HSPs are involved in protecting the intestine against I/R, the mechanism whereby HSF1 is regulated in I/R is poorly understood. Here, we show that the ubiquitin ligase FBXW7 targets HSF1 for ubiquitination and degradation in intestinal I/R. In this study, we found that FBXW7 expression was upregulated at the transcriptional level in intestinal mucosae subjected to I/R. In Caco-2 and IEC-6 cells subjected to hypoxia/reoxygenation (H/R), a high FBXW7 level led to excessive HSF1 ubiquitination and degradation. FBXW7 knockdown attenuated HSF1 ubiquitination and downregulation and accelerated HSPB1 and HSP70 expression. In addition, FBXW7 deletion alleviated the apoptosis of intestinal epithelial cells, as evidenced by decreased activation of caspase-3 and caspase-9. The results suggest that FBXW7 suppression protects against intestinal I/R, at least partly through the HSF1/HSP pathway. These findings indicate that FBXW7 may be a potential therapeutic target for inhibiting intestinal mucosa apoptosis during intestinal I/R.


Subject(s)
F-Box-WD Repeat-Containing Protein 7/metabolism , Heat Shock Transcription Factors/metabolism , Intestines/pathology , Reperfusion Injury/prevention & control , Ubiquitination , Animals , Apoptosis , Caco-2 Cells , Cell Line , Cell Nucleus/metabolism , Disease Models, Animal , F-Box-WD Repeat-Containing Protein 7/genetics , Gene Deletion , Gene Knockdown Techniques , Heat-Shock Proteins/metabolism , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice, Inbred C57BL , Phosphorylation , Rats , Reperfusion Injury/genetics , Signal Transduction , Transcriptional Activation
12.
Cell Physiol Biochem ; 48(4): 1579-1594, 2018.
Article in English | MEDLINE | ID: mdl-30071511

ABSTRACT

BACKGROUND/AIMS: Ischemic postconditioning (iPoC) represents a promising strategy to mitigate ischemia/reperfusion (I/R) injury of the intestine, yet the mechanisms of this treatment remain to be elucidated. Circular RNAs (circRNAs), a novel class of endogenous non-coding RNAs, have recently been recognized as important regulators of gene expression and pathological processes. Here, we aimed to investigate the expression patterns of circRNAs after intestinal I/R with and without iPoC and, furthermore, to explore the potential mechanisms of iPoC in relation to the differentially expressed circRNAs. METHODS: The global circRNA and mRNA expression profiles in mouse intestinal mucosa were initially screened by microarray (n = 3 per group) and quantitative real-time PCR was used to validate the expression pattern of circRNAs and mRNAs. Bioinformatics analysis including Gene ontology, KEGG pathway analysis, microRNA binding sites identification and circRNA-miRNA-mRNA network construction were utilized for in-depth mechanism exploration. RESULTS: There were 4 up- and 58 downregulated circRNAs as well as 322 up- and 199 downregulated mRNAs in the intestinal I/R group compared with the sham group, whereas compared with I/R, iPoC treatment significantly upregulated 12 circRNAs and 129 mRNAs and downregulated 21 circRNAs and 174 mRNAs. The expression levels of a randomly selected set of 6 circRNAs and 5 mRNAs were successfully validated by qRT-PCR. Through a systematic comparison of the direction of circRNA expression changes in all groups, we identified two circRNAs, circRNA_012412 and circRNA_016863, that may be closely associated with the protective mechanisms of iPoC. Finally, four possible circRNA_012412/circRNA_016863-miRNA-mRNA pathways were predicted, which may play important roles in endogenous protective signaling in iPoC. CONCLUSIONS: This study was the first to comprehensively delineate the expression profiles of circRNAs in a mouse model of intestinal I/R and iPoC and provides novel clues for understanding the mechanisms of iPoC against intestinal I/R injury.


Subject(s)
Intestines/pathology , RNA/metabolism , Reperfusion Injury/pathology , Animals , Computational Biology , Disease Models, Animal , Down-Regulation , Ischemic Postconditioning , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Circular , RNA, Messenger/metabolism , Reperfusion Injury/genetics , Tumor Necrosis Factor-alpha/blood , Up-Regulation
13.
Cell Death Dis ; 9(5): 483, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29706629

ABSTRACT

Autophagy is an essential cytoprotective response against pathologic stresses that selectively degrades damaged cellular components. Impaired autophagy contributes to organ injury in multiple diseases, including ischemia/reperfusion (I/R), but the exact mechanism by which impaired autophagy is regulated remains unclear. Several researchers have demonstrated that microRNAs (miRNAs) negatively regulate autophagy by targeting autophagy-related genes (ATGs). Therefore, the effect of ATG-related miRNAs on I/R remains a promising research avenue. In our study, we found that autophagy flux is impaired during intestinal I/R. A miRNA microarray analysis showed that miR-665-3p was highly expressed in the I/R group, which was confirmed by qRT-PCR. Then, we predicted and proved that miR-665-3p negatively regulates ATG4B expression in Caco-2 and IEC-6 cells. In ileum biopsy samples from patients with intestinal infarction, there was an inverse correlation between miR-665-3p and ATG4B expression, which supports the in vitro findings. Moreover, based on miR-665-3p regulation of autophagy in response to hypoxia/reoxygenation in vitro, gain-of-function and loss-of-function approaches were used to investigate the therapeutic potential of miR-665-3p. Additionally, we provide evidence that ATG4B is indispensable for protection upon inhibition of miR-665-3p. Finally, we observed that locked nucleic acid-modified inhibition of miR-665-3p in vivo alleviates I/R-induced systemic inflammation and apoptosis via recovery of autophagic flux. Our study highlights miR-665-3p as a novel small molecule that regulates autophagy by targeting ATG4B, suggesting that miR-665-3p inhibition may be a potential therapeutic approach against inflammation and apoptosis for the clinical treatment of intestinal I/R.


Subject(s)
Apoptosis , Autophagy-Related Proteins/metabolism , Autophagy , Cysteine Endopeptidases/metabolism , Ileum/blood supply , Ileum/metabolism , Inflammation/prevention & control , MicroRNAs/metabolism , Reperfusion Injury/prevention & control , Animals , Autophagy-Related Proteins/genetics , Caco-2 Cells , Cysteine Endopeptidases/genetics , Disease Models, Animal , Enterocytes/metabolism , Enterocytes/pathology , Gene Expression Regulation , Humans , Ileum/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
14.
Toxicol Appl Pharmacol ; 350: 21-31, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29729281

ABSTRACT

In recent years, alcoholic liver disease (ALD) has emerged as a growing public health problem worldwide. ß-catenin plays an important role in the growth, development, regeneration and metabolic activity of the liver. Salvianolic acid A (SalA) is a water-soluble component from the root extract of Salvia miltiorrhiza Bunge, and its effect on ALD has not yet been investigated. This study aimed to investigate the effect of SalA on chronic alcohol-induced liver injury and to explore the role of SIRT1-mediated ß-catenin deacetylation in such an effect. In this study, SalA treatment significantly alleviated the accumulation of lipid droplets and reduced the plasma alanine aminotransferase (ALT), aspartate aminotransferase (AST), total cholesterol (TC), triglyceride (TG), alcohol and ammonia levels in rats. SalA enhanced ethanol and ammonia metabolism and maintained mitochondrial homeostasis. Moreover, SalA restored the activity of the major ethanol-metabolizing enzymes and oxidative stress functions in the liver. Importantly, we found that SalA treatment effectively inhibited the ethanol-mediated decrease in nuclear ß-catenin by upregulating SIRT1 in the liver. SIRT1 then deacetylated ß-catenin to promote its accumulation in the nucleus, thereby preventing alcohol-induced liver injury. The results demonstrate that the SIRT1/ß-catenin pathway is a key therapeutic target in liver injury caused by chronic alcohol exposure and that SalA protects against alcohol-induced liver injury via the SIRT1-mediated deacetylation of ß-catenin.


Subject(s)
Caffeic Acids/therapeutic use , Cell Nucleolus/metabolism , Lactates/therapeutic use , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/metabolism , Sirtuin 1/metabolism , beta Catenin/metabolism , Animals , Caffeic Acids/pharmacology , Cell Nucleolus/drug effects , Chronic Disease , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Lactates/pharmacology , Liver Diseases, Alcoholic/pathology , Male , Mice , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley
15.
Cell Death Dis ; 9(3): 411, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540663

ABSTRACT

Impairment in gut barrier function induced by intestinal ischemia/reperfusion (I/R) injury is associated with high morbidity and mortality. Intestinal barrier function requires the tight coordination of epithelial migration, proliferation and differentiation. We previously observed that nuclear receptor-related protein 1 (nurr1)-mediated proliferative pathway was impaired in intestinal I/R injury. Here, we aimed to assess the effect of nurr1 on intestinal barrier function and to evaluate microRNA (miRNA)-nurr1-mediated restoration of intestinal barrier function in intestinal I/R injury. We induced an in vivo intestinal I/R injury mouse model by clamping and then releasing the superior mesenteric artery. We also performed an in vitro study in which we exposed Caco-2 and IEC-6 cells to hypoxia/reoxygenation (H/R) conditions to stimulate intestinal I/R injury. Our results demonstrated that nurr1 regulated intestinal epithelial development and barrier function after intestinal I/R injury. miR-381-3p, which directly suppressed nurr1 translation, was identified by microarray and bioinformatics analysis. miR-381-3p inhibition enhanced intestinal epithelial proliferation and barrier function in vitro and in vivo and also attenuated remote organ injury and improved survival. Importantly, nurr1 played an indispensable role in the protective effect of miR-381-3p inhibition. Collectively, these findings show that miR-381-3p inhibition mitigates intestinal I/R injury by enhancing nurr1-mediated intestinal epithelial proliferation and barrier function. This discovery may lead to the development of therapeutic interventions for intestinal I/R injury.


Subject(s)
Intestinal Mucosa/metabolism , MicroRNAs/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Reperfusion Injury/genetics , Animals , Caco-2 Cells , Cell Proliferation , Gene Knockdown Techniques , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/surgery , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology
16.
Toxicol Appl Pharmacol ; 339: 172-180, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29253500

ABSTRACT

Carnosic acid (CA), a major bioactive component in rosemary extract, has many biological and pharmaceutical activities. Smad3 acetylation can regulate the transcription of type I α2 collagen (COL1A2), which is the major component of the extracellular matrix (ECM). The aim of the current study was to evaluate whether CA inhibits COL1A2 transcription via the reduction of Smad3 acetylation against liver fibrosis. The results showed that CA treatment significantly suppressed COL1A2 transcription and markedly decreased the deposition of ECM induced by dimethylamine (DMN) in rats. Importantly, the suppression of COL1A2 transcription following CA treatment depended on the reduction of Smad3 acetylation via the activation of Sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide+ (NAD+)-dependent deacetylase. SIRT1 siRNA increased the acetylation of Smad3 and blocked CA-down-regulated Smad3 deacetylation. Notably, CA-mediated AMP-activated protein kinase-α1 (AMPKα1) activation not only increased AMPKα1 phosphorylation but also increased SIRT1 expression, thus leading to a significant reduction in Smad3 acetylation. Furthermore, CA-mediated SIRT1 activation was inhibited by AMPKα1 siRNA. Collectively, CA can inhibit the transcription of COL1A2 through SIRT1-mediated Smad3 deacetylation, and the activation of SIRT1 by CA involves the AMPKα1/SIRT1 pathway in liver fibrosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Abietanes/pharmacology , Collagen Type I/metabolism , Sirtuin 1/metabolism , Smad3 Protein/metabolism , Transcription, Genetic/physiology , Acetylation/drug effects , Animals , Antioxidants/pharmacology , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Liver Cirrhosis/metabolism , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects
17.
Pharmacol Res ; 128: 327-337, 2018 02.
Article in English | MEDLINE | ID: mdl-29106960

ABSTRACT

Quiescent hepatic stellate cell (HSC) activation and subsequent conversion into myofibroblasts is the central event in hepatic fibrosis pathogenesis. Epithelial-mesenchymal transition (EMT), another vital participant in liver fibrosis, has the potential to initiate HSC activation, which promotes abundant myofibroblast production. Previous studies suggest that Enhancer of Zeste Homolog 2 (EZH2) plays a significant role in myofibroblast transdifferentiation; however, the underlying mechanisms remain largely unaddressed. Carnosol (CS), a compound extracted from rosemary, displays multiple pharmacological activities. This study aimed to investigate the signaling mechanisms underlying EZH2 inhibition and the anti-fibrotic effect of CS in liver fibrosis. We found that CS significantly inhibited CCl4- and TGFß1-induced liver fibrosis and reduced both HSC activation and EMT. EZH2 knockdown also prevented these processes induced by TGFß1 in HSCs and AML-12 cells. Interestingly, the protective effect of CS was positively associated with Sirtuin 1 (SIRT1) activation and accompanied by EZH2 inhibition. SIRT1 knockdown attenuated the EZH2 inhibition induced by CS and increased EZH2 acetylation, which enhanced its stability. Conversely, upon TGFß1 exposure, SIRT1 activation significantly reduced the level of EZH2 acetylation; however, EZH2 overexpression prevented the SIRT1 activation that primed myofibroblast inhibition, indicating that EZH2 is a target of SIRT1. Thus, SIRT1/EZH2 regulation could be used as a new therapeutic strategy for fibrogenesis. Together, this study provides evidence of activation of the SIRT1/EZH2 pathway by CS that inhibits myofibroblast generation, and thus, CS may represent an attractive candidate for anti-fibrotic clinical therapy.


Subject(s)
Abietanes/pharmacology , Abietanes/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Enhancer of Zeste Homolog 2 Protein/metabolism , Liver Cirrhosis/drug therapy , Sirtuin 1/metabolism , Animals , Carbon Tetrachloride , Cell Line , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Enhancer of Zeste Homolog 2 Protein/genetics , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Rats, Sprague-Dawley , Sirtuin 1/genetics , Transforming Growth Factor beta1
18.
Cell Death Dis ; 8(7): e2935, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28726782

ABSTRACT

Intestinal ischemia-reperfusion (I/R) is a common clinical problem that occurs during various clinical pathological processes. Excessive apoptosis has an indispensable role in intestinal I/R injury. Tumor necrosis factor receptor-associated factor 2 (TRAF2) and PKCζ have an essential role in apoptosis. Here, we aimed to investigate the effects of PKCζ and TRAF2 and to explore the correlation between PKCζ and TRAF2 in intestinal I/R injury. Mice were subjected to intestinal I/R injury in vivo. In vitro experiments were conducted by treating Caco-2 cells with hypoxia/reoxygenation (H/R) stimulation to simulate intestinal I/R. Intestinal tissue samples and Caco-2 cells were examined using various approaches. Intestinal I/R induced the membrane translocation and phosphorylation of PKCζ. Pretreatment with the PKCζ activator phosphatidylcholine remarkably attenuated gut injury by suppressing apoptosis. H/R induced PKCζ to combine with TRAF2, which was phosphorylated by PKCζ at Ser55, but not at Ser11, under intestinal I/R or H/R conditions. In addition, TRAF2 Ser55 phosphorylation increased cell survival by inhibiting cell apoptosis in the H/R model. Mechanistically, TRAF2 Ser55 phosphorylation promoted NF-κB activation but suppressed c-Jun activation in Caco-2 cells under H/R conditions. The results of this study demonstrate that the PKCζ/TRAF2 pathway represents a novel protective mechanism against intestinal I/R injury. Therefore, the PKCζ/TRAF2 pathway is a novel target for potential treatments of intestinal I/R injury-related diseases.


Subject(s)
Intestinal Diseases/metabolism , Protein Kinase C/metabolism , Reperfusion Injury/metabolism , TNF Receptor-Associated Factor 2/metabolism , Animals , Caco-2 Cells , Humans , Intestinal Diseases/pathology , Intestinal Diseases/prevention & control , Male , Mice , Phosphorylation , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control
19.
Clin Sci (Lond) ; 131(8): 759-773, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28232511

ABSTRACT

Intestinal epithelial oxidative stress and apoptosis constitute key pathogenic mechanisms underlying intestinal ischemia/reperfusion (I/R) injury. We previously reported that the adaptor 66 kDa isoform of the adaptor molecule ShcA (p66Shc)-mediated pro-apoptotic pathway was activated after intestinal I/R. However, the upstream regulators of the p66Shc pathway involved in intestinal I/R remain to be fully identified. Here, we focused on the role of a prolyl-isomerase, peptidyl-prolyl cis-trans isomerase (Pin1), in the regulation of p66Shc activity during intestinal I/R. Intestinal I/R was induced in rats by superior mesenteric artery (SMA) occlusion. Juglone (Pin1 inhibitor) or vehicle was injected intraperitoneally before I/R challenge. Caco-2 cells were exposed to hypoxia/reoxygenation (H/R) in vitro to simulate an in vivo I/R model. We found that p66Shc was significantly up-regulated in the I/R intestine and that this up-regulation resulted in the accumulation of intestinal mitochondrial reactive oxygen species (ROS) and massive epithelial apoptosis. Moreover, intestinal I/R resulted in elevated protein expression and enzyme activity of Pin1 as well as increased interaction between Pin1 and p66Shc. This Pin1 activation was responsible for the translocation of p66Shc to the mitochondria during intestinal I/R, as Pin1 suppression by juglone or siRNA markedly blunted p66Shc mitochondrial translocation and the subsequent ROS generation and cellular apoptosis. Additionally, Pin1 inhibition alleviated gut damage and secondary lung injury, leading to improvement of survival after I/R. Collectively, our findings demonstrate for the first time that Pin1 inhibition protects against intestinal I/R injury, which could be partially attributed to the p66Shc-mediated mitochondrial apoptosis pathway. This may represent a novel prophylactic target for intestinal I/R injury.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Intestines/blood supply , Naphthoquinones/therapeutic use , Reperfusion Injury/prevention & control , Src Homology 2 Domain-Containing, Transforming Protein 1/antagonists & inhibitors , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Intestinal Mucosa/metabolism , Intestines/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy/methods , Naphthoquinones/pharmacology , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/physiology , Translocation, Genetic
20.
J Mol Med (Berl) ; 95(1): 83-95, 2017 01.
Article in English | MEDLINE | ID: mdl-27553040

ABSTRACT

Intestinal ischemia/reperfusion (I/R) injury is a potentially life-threatening condition that can cause injuries to remote organs at the end stage. The damage caused by intestinal I/R insult induces changes in the barrier functions of the intestine, and the intrinsic mechanism of regeneration is often insufficient to restore barrier functions, as indicated by the high mortality rate of patients experiencing intestinal I/R injury. However, little is known about the mechanisms of intestinal regeneration after I/R injury. Here, we reported that nuclear receptor-related protein 1 (Nurr1), a nuclear orphan receptor, was induced during intestinal regeneration after I/R. Our findings showed that Nurr1 expression was consistent with the expression of Ki-67 and phosphorylated histone H3 (pH 3) in the intestine after I/R injury. Nurr1 knockdown led to G1-phase arrest mediated by p21 (Waf1/Cip1) activation, but Nurr1 overexpression reduced the proportion of IEC-6 cells in G1 phase as a result of p21 inhibition in a p53-independent manner. Using chromatin immunoprecipitation assays, luciferase assays, and mutational analysis, we demonstrated that Nurr1 directly inhibited the transcription of p21. These results define a novel Nurr1/p21 pathway that is involved in intestinal regeneration after I/R injury. These findings provide novel molecular insights into the pathogenesis of intestinal regeneration after I/R and possibly support the development of new potential therapies for intestinal I/R injury. KEY MESSAGE: Nurr1 was induced during intestinal regeneration after I/R injury. Nurr1 promoted proliferation of intestinal epithelial cells after H/R injury. Nurr1 inhibited p21 expression in a p53-independent manner. Nurr1 inhibited p21 gene transcription by binding to p21 promoter directly.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation , Intestines/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Regeneration , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Animals , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Humans , Male , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Regeneration/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...