Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167140, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38548092

ABSTRACT

Heart failure (HF) is one of the major causes of death among diabetic patients. Although studies have shown that curcumin analog C66 can remarkably relieve diabetes-associated cardiovascular and kidney complications, the role of SJ-12, SJ-12, a novel curcumin analog, in diabetic cardiomyopathy and its molecular targets are unknown. 7-week-old male C57BL/6 mice were intraperitoneally injected with single streptozotocin (STZ) (160 mg/kg) to develop diabetic cardiomyopathy (DCM). The diabetic mice were then treated with SJ-12 via gavage for two months. Body weight, fast blood glucose, cardiac utrasonography, myocardial injury markers, pathological morphology of the heart, hypertrophic and fibrotic markers were assessed. The potential target of SJ-12 was evaluated via RNA-sequencing analysis. The O-GlcNAcylation levels of SP1 were detected via immunoprecipitation. SJ-12 effectively suppressed myocardial hypertrophy and fibrosis, thereby preventing heart dysfunction in mice with STZ-induced heart failure. RNA-sequencing analysis revealed that SJ-12 exerted its therapeutic effects through the modulation of the calcium signaling pathway. Furthermore, SJ-12 reduced the O-GlcNAcylation levels of SP1 by inhibiting O-linked N-acetylglucosamine transferase (OGT). Also, SJ-12 stabilized Sarcoplasmic/Endoplasmic Reticulum Calcium ATPase 2a (SERCA2a), a crucial regulator of calcium homeostasis, thus reducing hypertrophy and fibrosis in mouse hearts and cultured cardiomyocytes. However, the anti-fibrotic effects of SJ-12 were not detected in SERCA2a or OGT-silenced cardiomyocytes, indicating that SJ-12 can prevent DCM by targeting OGT-dependent O-GlcNAcylation of SP1.These findings indicate that SJ-12 can exert cardioprotective effects in STZ-induced mice by reducing the O-GlcNAcylation levels of SP1, thus stabilizing SERCA2a and reducing myocardial fibrosis and hypertrophy. Therefore, SJ-12 can be used for the treatment of diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Mice, Inbred C57BL , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animals , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/drug therapy , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Male , Mice , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/drug therapy , Streptozocin , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Fibrosis , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Calcium Signaling/drug effects
2.
Cell Mol Life Sci ; 81(1): 18, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38195959

ABSTRACT

Prolonged stimulation of ß-adrenergic receptor (ß-AR) can lead to sympathetic overactivity that causes pathologic cardiac hypertrophy and fibrosis, ultimately resulting in heart failure. Recent studies suggest that abnormal protein ubiquitylation may contribute to the pathogenesis of cardiac hypertrophy and remodeling. In this study, we demonstrated that deficiency of a deubiquitinase, Josephin domain-containing protein 2 (JOSD2), ameliorated isoprenaline (ISO)- and myocardial infarction (MI)-induced cardiac hypertrophy, fibrosis, and dysfunction both in vitro and in vivo. Conversely, JOSD2 overexpression aggravated ISO-induced cardiac pathology. Through comprehensive mass spectrometry analysis, we identified that JOSD2 interacts with Calcium-calmodulin-dependent protein kinase II (CaMKIIδ). JOSD2 directly hydrolyzes the K63-linked polyubiquitin chains on CaMKIIδ, thereby increasing the phosphorylation of CaMKIIδ and resulting in calcium mishandling, hypertrophy, and fibrosis in cardiomyocytes. In vivo experiments showed that the cardiac remodeling induced by JOSD2 overexpression could be reversed by the CaMKIIδ inhibitor KN-93. In conclusion, our study highlights the role of JOSD2 in mediating ISO-induced cardiac remodeling through the regulation of CaMKIIδ ubiquitination, and suggests its potential as a therapeutic target for combating the disease. Please check and confirm that the authors and their respective affiliations have been correctly identified and amend if necessary. All have been checked.


Subject(s)
Heart Failure , Myocytes, Cardiac , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cardiomegaly/chemically induced , Fibrosis , Heart Failure/chemically induced , Isoproterenol/pharmacology , Ventricular Remodeling
3.
J Minim Access Surg ; 20(2): 180-186, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37706409

ABSTRACT

INTRODUCTION: Laparoendoscopic single-site inguinal lymphadenectomy (LESS-IL), a minimally invasive technique, has been reported in patients with vulvar or vaginal cancer regarding its safety and feasibility. However, the long-term outcomes, especially oncologic outcomes, are still lacking. We aimed to evaluate the long-term outcomes of LESS-IL to confirm its safety further. PATIENTS AND METHODS: Data were prospectively collected from patients with vulvar or vaginal cancer who underwent LESS-IL at our institution between July 2018 and June 2021. The patients were followed up for at least 12 months. All procedures were performed according to treatment standards. Short- and long-term complications and oncologic outcomes were analysed. RESULTS: A total of 16 patients undergoing 28 LESS-IL procedures were identified, amongst whom 4 underwent unilateral LESS-IL. The median numbers of excised groin lymph nodes were 9.0 (6.5-11.8) and 10.5 (8.3-12.0) in each left and right groin, respectively. Short-term complications occurred in 4 (25%) patients, including 18.7% lymphocele and 6.3% wound infection. Long-term complications regarding lower-limb lymphoedema appeared in 6 (37.5%) patients. Most short- and long-term complications were Clavien-Dindo 1 or 2, accounting for 90% of all post-operative issues. After a median follow-up of 27 (21.3-35.8) months, only 1 (6.3%) patient had isolated inguinal recurrence at 13 months postoperatively. No local or distant recurrence occurred. CONCLUSION: Our results suggest that LESS-IL is associated with little incidence of complications and promising oncologic outcomes, further demonstrating the safety and feasibility of the LESS-IL technique in patients requiring IL.

4.
Acta Pharmacol Sin ; 45(4): 765-776, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38110583

ABSTRACT

Hypertensive renal disease (HRD) contributes to the progression of kidney dysfunction and ultimately leads to end-stage renal disease. Understanding the mechanisms underlying HRD is critical for the development of therapeutic strategies. Deubiquitinating enzymes (DUBs) have been recently highlighted in renal pathophysiology. In this study, we investigated the role of a DUB, OTU Domain-Containing Protein 1 (OTUD1), in HRD models. HRD was induced in wild-type or Otud1 knockout mice by chronic infusion of angiotensin II (Ang II, 1 µg/kg per min) through a micro-osmotic pump for 4 weeks. We found that OTUD1 expression levels were significantly elevated in the kidney tissues of Ang II-treated mice. Otud1 knockout significantly ameliorated Ang II-induced HRD, whereas OTUD1 overexpression exacerbated Ang II-induced kidney damage and fibrosis. Similar results were observed in TCMK-1 cells but not in SV40 MES-13 cells following Ang II (1 µM) treatment. In Ang II-challenged TCMK-1 cells, we demonstrated that OTUD1 bound to CDK9 and induced CDK9 deubiquitination: OTUD1 catalyzed K63 deubiquitination on CDK9 with its Cys320 playing a critical role, promoting CDK9 phosphorylation and activation to induce inflammatory responses and fibrosis in kidney epithelial cells. Administration of a CDK9 inhibitor NVP-2 significantly ameliorated Ang II-induced HRD in mice. This study demonstrates that OTUD1 mediates HRD by targeting CDK9 in kidney epithelial cells, suggesting OTUD1 is a potential target in treating this disease.


Subject(s)
Hypertension, Renal , Kidney , Nephritis , Ubiquitin-Specific Proteases , Animals , Mice , Angiotensin II/metabolism , Epithelial Cells/metabolism , Fibrosis , Hypertension, Renal/enzymology , Hypertension, Renal/pathology , Kidney/pathology , Mice, Inbred C57BL , Mice, Knockout , Nephritis/enzymology , Nephritis/pathology , Ubiquitin-Specific Proteases/metabolism , Disease Models, Animal
5.
Biomed Pharmacother ; 169: 115880, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37956481

ABSTRACT

BACKGROUND: Enhanced levels of angiotensin-2 (Ang-II) causes hypertensive heart failure (HHF) through non-hemodynamical and hemodynamical alterations. 20(S)-ginsenoside Rh2 (20(S)-Rh2) is a natural ginseng compound with numerous cardiovascular benefits. This investigation elucidates the influence of 20(S)-Rh2 on Ang-II-induced heart failure and cardiac alterations. METHODS: Ang-II was administered in C57BL/6 mice for 4 weeks to induce HHF. In the last 2 weeks of treatment, 20(S)-Rh2 was orally administered in mice to assess the potential 20(S)-Rh2 mechanism. Subsequently, RNA sequencing was carried out. RESULTS: It was indicated that 20(S)-Rh2 suppresses myocardial fibrosis, hypertrophy, and inflammation, thereby inhibiting cardiac disruption in Ang-II-challenged mice without affecting blood pressure. According to the RNA sequencing data, this cardio-protective effect was linked with the (JNK)/AP 1 pathway. 20(S)-Rh2 alleviated heart tissue and cardiomyocytes inflammation by inhibiting the Ang-II-mediated JNK/AP-1 pathway. Within cardiomyocytes, JNK or AP-1 absence abolished the anti-inflammatory effects of 20(S)-Rh2. CONCLUSION: This study investigation indicated that 20(S)-Rh2 prevents cardiovascular dysfunction induced by Ang-II induced by decreasing JNK-regulated inflammatory responses, providing evidence for its use as an efficient regimen for HHF.


Subject(s)
Heart Failure , Hypertension , Mice , Animals , Transcription Factor AP-1/metabolism , Angiotensin II/pharmacology , Ventricular Remodeling , Mice, Inbred C57BL , Heart Failure/drug therapy , Heart Failure/prevention & control , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Hypertension/chemically induced , Hypertension/drug therapy
6.
Int Immunopharmacol ; 120: 110408, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37276830

ABSTRACT

Increased level of Angiotensin II (Ang II) contributes to hypertensive heart failure via -hemodynamic and non-hemodynamic actions. Ginsenoside Rg5 (Rg5) occurs naturally in ginseng, which has shown various benefits for cardiovascular diseases. This study evaluated Rg5's effects on Ang II-caused cardiac remodeling and heart failure. C57BL/6 mice developed hypertensive cardiac failure after four weeks of Ang II infusion. The mice were administered Rg5 via oral gavage for the last two weeks to investigate the potential mechanism of Rg5. RNA sequencing of heart tissues was performed for mechanistic studies. It was discovered that Rg5 inhibited cardiac inflammation, myocardial fibrosis, and hypertrophy, and prevented cardiac malfunction in mice challenged with Ang II, without altering blood pressure. RNA sequencing showed that Rg5's cardioprotective effect involves the JNK/AP-1 signaling pathway. Rg5 diminished inflammation in mice hearts and cultured cardiomyocytes by blocking Ang II-activated JNK/AP-1 pathway. In the absence of JNK or AP-1 in cardiomyocytes, the anti-inflammatory effects of Rg5 were nullified. The study found that Rg5 preserved the hearts of Ang II-induced mice by reducing JNK-mediated inflammatory responses, suggesting that Rg5 is an effective therapy for hypertensive heart failure.


Subject(s)
Heart Failure , Hypertension , Mice , Animals , Transcription Factor AP-1/metabolism , Angiotensin II , Mice, Inbred C57BL , Heart Failure/drug therapy , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Hypertension/chemically induced , Hypertension/drug therapy , Arrhythmias, Cardiac
7.
Theranostics ; 13(7): 2263-2280, 2023.
Article in English | MEDLINE | ID: mdl-37153745

ABSTRACT

Rationale: Understanding the molecular mechanisms of deleterious cardiac remodeling is important for the development of treatments for heart failure. Recent studies have highlighted a role of deubiquitinating enzymes in cardiac pathophysiology. In the present study, we screened for alteration of deubiquitinating enzymes in experimental models of cardiac remodeling, which indicated a potential role of OTU Domain-Containing Protein 1 (OTUD1). Methods: Wide-type or OTUD1 knockout mice with chronic angiotensin II infusion and transverse aortic constriction (TAC) were utilized to develop cardiac remodeling and heart failure. We also overexpressed OTUD1 in mouse heart with AAV9 vector to validate the function of OTUD1. LC-MS/MS analysis combined with Co-IP was used to identify the interacting proteins and substrates of OTUD1. Results: We found that OTUD1 is elevated in mouse heart tissues following chronic angiotensin II administration. OTUD1 knockout mice were significantly protected against angiotensin II-induced cardiac dysfunction, hypertrophy, fibrosis and inflammatory response. Similar results were obtained in the TAC model. Mechanistically, OTUD1 bounds to the SH2 domain of STAT3 and causes deubiquitination of STAT3. Cysteine at position 320 of OTUD1 exerts K63 deubiquitination to promote STAT3 phosphorylation and nuclear translocation, thereby increasing STAT3 activity to induce inflammatory responses, fibrosis, and hypertrophy in cardiomyocytes. Finally, OTUD1 overexpression by AAV9 vector increases Ang II-induced cardiac remodeling in mice and OTUD1-regulated responses can be inhibited by blocking STAT3. Conclusion: Cardiomyocyte OTUD1 promotes pathological cardiac remodeling and dysfunction by deubiquitinating STAT3. These studies have highlighted a novel role of OTUD1 in hypertensive heart failure and identified STAT3 as a target of OTUD1 in mediating these actions.


Subject(s)
Heart Failure , Myocytes, Cardiac , Animals , Mice , Angiotensin II/pharmacology , Chromatography, Liquid , Deubiquitinating Enzymes/metabolism , Fibrosis , Heart Failure/metabolism , Hypertrophy/metabolism , Hypertrophy/pathology , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Tandem Mass Spectrometry , Ventricular Remodeling/physiology , STAT3 Transcription Factor
8.
Int J Comput Assist Radiol Surg ; 18(2): 303-312, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36319921

ABSTRACT

PURPOSE: To address the difficulties of M-mode ultrasound images classification in pneumothorax diagnosis and the shortcomings of existing neural network algorithms in this field, we proposed an M-mode ultrasound images classification model based on Disturbed Meta-Pseudo-Labels (D-MPL). METHODS: An M-mode ultrasound image augmentation system was designed to make the model more robust and generalizable. In D-MPL, teacher-generated pseudo-labeling was first taught to students through a soft mask, and additional disturbance data were added to the teacher network. As the loss of the teacher network continues to decline, disturbance data were injected to improve the generalization of the model to cope with image differences across patients in clinical settings. RESULTS: We compared the proposed model with four commonly used models, including MPL, EfficientnetB2, Inception V3, and Resnet101, in order to confirm its efficacy. Our model has an average specificity of 98.28%, sensitivity of 98.22%, F1-score of 98.23%, and AUC of 98.10%, according to the experiment findings, and its comprehensive performance is better than the above four models. CONCLUSION: The results demonstrated our model's superiority over the competition and its greater. The model proposed in this study is expected to assist doctors in the diagnosis of pneumothorax as an auxiliary mean.


Subject(s)
Pneumothorax , Humans , Algorithms , Diagnosis, Computer-Assisted/methods , Echocardiography , Neural Networks, Computer , Pneumothorax/diagnostic imaging
9.
Biomed Pharmacother ; 149: 112917, 2022 May.
Article in English | MEDLINE | ID: mdl-36068777

ABSTRACT

BACKGROUND AND PURPOSE: An overdose of acetaminophen (APAP) causes acute liver damage and lead to liver failure. Therefore, it is of great clinical significance to find drugs for the treatment of APAP-induced liver injury. Diacerein is clinically used drug for the treatment of osteoarthritis. Here, we evaluate the pharmacological effects and potential mechanisms of diacerein in APAP-induced liver injury. METHODS AND RESULTS: C57BL/6 mice were treated with diacerein by gavage, followed by intraperitoneal injection of APAP (400 mg/kg) to induce acute liver injury in mice. RNA-sequencing analysis and in vitro kinase assay were performed to explore the underlying mechanisms of diacerein. The experimental results showed that pretreatment with diacerein could inhibit APAP-induced elevation of serum AST and ALT levels, hepatic histopathological damage, oxidative stress, hepatocyte death, and mitochondrial damage in mice. The RNA-sequencing analysis and in vitro kinase assay indicated that indicating that JNK (c-Jun N-terminal kinase) is involved in that liver-protective effects of Diacerein. Diacerein could directly and selectively inhibit JNK kinase phosphorylation in cell-free system. We further confirmed that diacerein inhibits APAP-activated JNK pathway to reduce injury response in mouse livers and cultured AML12 cells. Deficiency of JNK in AML12 cells abolished the anti-injury effects of diacerein. CONCLUSION: Our experimental results suggest that diacerein protects APAP-induced liver injury by the inhibition of JNK kinase phosphorylation, rendering diacerein may serve as a potential therapeutic drug for the prevention of acute liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Chemical and Drug Induced Liver Injury , Acetaminophen/pharmacology , Animals , Anthraquinones , Apoptosis , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes , JNK Mitogen-Activated Protein Kinases/metabolism , Liver , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Oxidative Stress , RNA/metabolism
10.
Phytomedicine ; 106: 154387, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36027716

ABSTRACT

BACKGROUND: Heart failure is a common event in the course of hypertension. Recent studies have highlighted the key role of the non-hemodynamic activity of angiotensin II (Ang II) in hypertension-related cardiac inflammation and remodeling. A naturally occurring compound, diacerein, exhibits anti-inflammatory activities in various systems. HYPOTHESIS/PURPOSE: In this study, we have examined the potential effects of diacerein on Ang II-induced heart failure. METHODS: C57BL/6 mice were administered Ang II by micro-osmotic pump infusion for 4 weeks to develop hypertensive heart failure. Mice were treated with diacerein by gavage for final 2 weeks. RNA-sequencing analysis was performed to explore the potential mechanism of diacerein. RESULTS: We found that diacerein could inhibit inflammation, myocardial fibrosis, and hypertrophy to prevent heart dysfunction, without the alteration of blood pressure. To explore the potential mechanism of diacerein, RNA-sequencing analysis was performed, indicating that MAPKs/c-Myc pathway is involved in that cardioprotective effects of Diacerein. We further confirmed that diacerein inhibits Ang II-activated MAPKs/c-Myc pathway to reduce inflammatory response in mouse hearts and cultured cardiomyocytes. Deficiency of MAPKs or c-Myc in cardiomyocytes abolished the anti-inflammatory effects of diacerein. CONCLUSION: Our results indicate that diacerein protects hearts in Ang II-induced mice through inhibiting MAPKs/c-Myc-mediated inflammatory responses, rendering diacerein a potential therapeutic candidate agent for hypertensive heart failure.


Subject(s)
Cardiomyopathies , Heart Failure , Hypertension , Angiotensin II/pharmacology , Animals , Anthraquinones , Cardiomegaly/chemically induced , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cardiomyopathies/metabolism , Fibrosis , Heart Failure/metabolism , Hypertension/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocytes, Cardiac , RNA , Ventricular Remodeling
11.
Biomed Pharmacother ; 150: 113075, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35658238

ABSTRACT

Heart failure is a fairly common outcome of hypertension. Recent studies have highlighted the key role of the non-hemodynamic activity of angiotensin II (Ang II) in hypertensive heart failure via inducing cardiac inflammation. Drugs that disrupt Ang II-induced cardiac inflammation may have clinical utility in the treatment of hypertensive heart failure. A naturally occurring compound, corynoline, exhibit anti-inflammatory activities in other systems. C57BL/6 mice were injected with Ang II via a micro-osmotic pump for four weeks to develop hypertensive heart failure. The mice were treated with corynoline by gavage for two weeks. RNA-sequencing analysis was performed to explore the potential mechanism of corynoline. We found that corynoline could inhibit inflammation, myocardial fibrosis, and hypertrophy to prevent heart dysfunction, without the alteration of blood pressure. RNA-sequencing analysis indicates that the PPARα pathway is involved Ang II-induced cardiac fibrosis and cardiac remodeling. Corynoline reversed Ang II-induced PPARα inhibition both in vitro and in vivo. We further found that corynoline increases the interaction between PPARα and P65 to inhibit the NF-κB pro-inflammatory pathway in H9c2 cells. Our studies show that corynoline relieves Ang II-induced hypertensive heart failure by increasing the interaction between PPARα and P65 to inhibit the NF-κB pathway.


Subject(s)
Heart Failure , Hypertension , Angiotensin II/pharmacology , Animals , Berberine Alkaloids , Fibrosis , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/prevention & control , Hypertension/chemically induced , Hypertension/complications , Hypertension/drug therapy , Inflammation , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , PPAR alpha , RNA
12.
Nanomaterials (Basel) ; 10(7)2020 Jul 04.
Article in English | MEDLINE | ID: mdl-32635490

ABSTRACT

In this study, hexagonal-packed Si nanorods (SiNRs) arrays were fabricated and conjugated with Au nanoparticles (AuNPs) in different spatial distributions for surface-enhanced Raman spectroscopy (SERS). The AuNPs were functionalized on the bottom of SiNRs (B-SiNRs@AuNPs), top of SiNRs (T-SiNRs@AuNPs) and sides of SiNRs (S-SiNRs@AuNPs), respectively. Our results demonstrated that the SiNRs conjugated with AuNPs on the sides achieved high reproducibility in detection of R6G molecules, while the AuNPs on the top of the SiNRs obtained the strongest Raman enhancement. In addition, the substrate with S-SiNRs@AuNPs obtained the highest spatial uniformity of enhancement. The finite-difference time-domain simulation gave further evidence that the incident light could be confined in the space of SiNRs arrays and yield a zero-gap enhancement coupled with the AuNPs. Our study provided a spatially tunable SiNRs@AuNPs substrate with high sensitivity and reproducibility in molecular detection.

SELECTION OF CITATIONS
SEARCH DETAIL
...