1
|
Hu SJ, Chen GC, Wang FY, Fang YQ, Wang SQ, Song ZL, Zhao ZH, Zhang QL, Meng XY, Zhang QY, Qin LP. Network pharmacology analysis uncovers the mechanism of Shudihuang-Shanzhuyu herb pair in prevention and treatment of diabetic osteoporosis via PI3K/AKT pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119581. [PMID: 40054640 DOI: 10.1016/j.jep.2025.119581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetic osteoporosis (DOP) is a complication of diabetes characterized by reduced bone mass and increased fracture risk. Shudihuang (Rehmanniae Radix Praeparata, RR) and Shanzhuyu (Corni Fructus, CF) form a classical herb pair known as RR-CF in traditional Chinese medicine (TCM) for nourishing Yin and tonifying the kidney, and have long been used for the treatment of diabetes and OP in TCM clinical practise. However, the potential mechanism underlying the preventive and therapeutic effects of RR-CF on DOP has not been clarified. AIM OF THE STUDY This study aimed to explore the protective effects of RR-CF on bone loss caused by diabetes and elucidate the underlying action mechanism. METHODS The chemical constituents in RR-CF were detected using UPLC-Q-Exactive-MS. Type 1 diabetes mellitus (T1DM) was induced in rats by injecting streptozotocin, followed by administration of RR-CF extracts for 10 weeks. Bone mineral density, morphometric bone parameters, and serum and urine biochemical markers were analyzed using Micro-CT and ELISA kits. An in vitro osteoblastic injury model was constructed by subjecting MC3T3-E1 cells to high glucose and used to evaluate the effects of the RR-CF on osteoblastic bone formation. The anti-DOP mechanism of RR-CF was explored by network pharmacologic analysis and then verified in osteoblasts damaged by high glucose. RESULTS A total of 56 compounds were identified in RR-CF. Treatment with RR-CF extracts improved the bone microstructure and mineral density in the T1DM rats, and decreased the level of urine deoxypyridinoline and serum carboxyl terminal peptide of type I procollagen. The network pharmacology analysis identified cornuside, hydroxygenkwanin, acteoside, catalpol and echinacoside as the potential active components of RR-CF against DOP by interacting with the key node genes such as AKT1, EGFR, TNF, MMP9 and HSP90α. Further GO and KEGG enrichment analysis suggested that the therapeutic effects of RR, CF and RR-CF seemed to be related to the regulation of hormones, inflammation and metabolism, as well as signaling transductions of PI3K-AKT, IL-17, TNF, MAPK and estrogen signaling pathways. RR-CF promoted osteoblast differentiation and bone formation in the MC3T3-E1 cells by regulating PI3K-AKT signaling pathway. CONCLUSION RR-CF herb pair inhibits bone loss caused by high glucose by regulating the PI3K-AKT signaling pathways.
Collapse
Affiliation(s)
- Si-Jing Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Gao-Ce Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Fang-Yuan Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ying-Qi Fang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Si-Qi Wang
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210000, China
| | - Zi-le Song
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zi-Hui Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Quan-Long Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiong-Yu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Qiao-Yan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Lu-Ping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
2
|
Rajabloo Y, Latifi H, Akhlaghipour I, Taghehchian N, Moghbeli M. MicroRNA-409: Molecular functions and clinical applications in cancer. Biochem Biophys Rep 2024; 38:101728. [PMID: 38737729 PMCID: PMC11087923 DOI: 10.1016/j.bbrep.2024.101728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/30/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024] Open
Abstract
Late diagnosis is one of the main reasons for high mortality rates in cancer patients. Therefore, investigating the molecular mechanisms involved in tumor progression can improve the cancer diagnosis in the early stages of the tumor progression. MicroRNAs (miRNAs) have important roles in regulation of cell growth, proliferation, metabolism, and migration. Since, deregulation of miR-409 has been reported in a wide range of cancers, in the present review, we investigated the molecular mechanisms of miR-409 during tumor progression and invasion. It has been shown that miR-409 functions as a tumor suppressor in different tumor types. MiR-409 can reduce tumor cell proliferation, growth, and migration by regulation of signaling pathways, cellular metabolism, transcription factors, and cellular adhesion. This review can be an effective step in introducing miR-409 as a non-invasive marker in cancer patients.
Collapse
Affiliation(s)
- Yasamin Rajabloo
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanieh Latifi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Stevens SA, Sunilkumar S, Subrahmanian SM, Toro AL, Cavus O, Omorogbe EV, Bradley EA, Dennis MD. REDD1 Deletion Suppresses NF-κB Signaling in Cardiomyocytes and Prevents Deficits in Cardiac Function in Diabetic Mice. Int J Mol Sci 2024; 25:6461. [PMID: 38928166 PMCID: PMC11204184 DOI: 10.3390/ijms25126461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/06/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Activation of the transcription factor NF-κB in cardiomyocytes has been implicated in the development of cardiac function deficits caused by diabetes. NF-κB controls the expression of an array of pro-inflammatory cytokines and chemokines. We recently discovered that the stress response protein regulated in development and DNA damage response 1 (REDD1) was required for increased pro-inflammatory cytokine expression in the hearts of diabetic mice. The studies herein were designed to extend the prior report by investigating the role of REDD1 in NF-κB signaling in cardiomyocytes. REDD1 genetic deletion suppressed NF-κB signaling and nuclear localization of the transcription factor in human AC16 cardiomyocyte cultures exposed to TNFα or hyperglycemic conditions. A similar suppressive effect on NF-κB activation and pro-inflammatory cytokine expression was also seen in cardiomyocytes by knocking down the expression of GSK3β. NF-κB activity was restored in REDD1-deficient cardiomyocytes exposed to hyperglycemic conditions by expression of a constitutively active GSK3β variant. In the hearts of diabetic mice, REDD1 was required for reduced inhibitory phosphorylation of GSK3β at S9 and upregulation of IL-1β and CCL2. Diabetic REDD1+/+ mice developed systolic functional deficits evidenced by reduced ejection fraction. By contrast, REDD1-/- mice did not exhibit a diabetes-induced deficit in ejection fraction and left ventricular chamber dilatation was reduced in diabetic REDD1-/- mice, as compared to diabetic REDD1+/+ mice. Overall, the results support a role for REDD1 in promoting GSK3β-dependent NF-κB signaling in cardiomyocytes and in the development of cardiac function deficits in diabetic mice.
Collapse
Affiliation(s)
- Shaunaci A. Stevens
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Siddharth Sunilkumar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Sandeep M. Subrahmanian
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Allyson L. Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Omer Cavus
- Division of Cardiovascular Medicine, Penn State Health Heart and Vascular Institute, Hershey S. Milton Medical Center, Hershey, PA 17033, USA
| | - Efosa V. Omorogbe
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Elisa A. Bradley
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
- Division of Cardiovascular Medicine, Penn State Health Heart and Vascular Institute, Hershey S. Milton Medical Center, Hershey, PA 17033, USA
| | - Michael D. Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
4
|
Liu M, Zhao J, Lu Y, Chen Z, Feng X, Pan G. Gab1 Overexpression Attenuates Susceptibility to Ventricular Arrhythmias in Pressure Overloaded Heart Failure Mouse Hearts. Cardiovasc Drugs Ther 2024; 38:253-262. [PMID: 36374360 DOI: 10.1007/s10557-022-07394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Grb2 associated binding protein 1 (Gab1) is an adaptor protein that is important for intracellular signal transduction which involved in several pathological process. However, the role of Gab1 in pressure overload-induced ventricular arrhythmias (VAs) remain poorly understood. In the current study, we aimed to test the role of Gab1 in VA susceptibility induced by pressure overload. METHODS We overexpressed Gab1 in the hearts using an adeno-associated virus 9 (AAV9) system through tail vein injection. Aortic banding (AB) surgery was performed in C57BL6/J mice to induce heart failure (HF). Four weeks following AB, histology, echocardiography, and biochemical analysis were conducted to investigate cardiac structural remodeling and electrophysiological studies were performed to check the electrical remodeling. Western blot analysis was used to explore the underlying mechanisms. RESULTS The mRNA and protein expression were downregulated in AB hearts compared to sham hearts. Gab1 overexpression significantly reversed AB-induced cardiac structural remodeling including ameliorated AB-induced cardiac dysfunction, cardiac fibrosis, and inflammatory response. Moreover, Gab1 overexpression also markedly alleviated AB-induced electrical remodeling including ion channel alterations and VA susceptibility. Mechanistically, we found that TLR4/MyD88/NF-κB contributes to the cardio protective effect of Gab1 overexpression on AB-induced VAs. CONCLUSIONS Our study manifested that Gab1 may serve as a promising anti-arrhythmic target via inhibiting TLR4/MyD88/NF-κB signaling pathway induced by AB.
Collapse
Affiliation(s)
- Mingxin Liu
- Department of Cardiology, Yueyang Central Hospital, No.39 Dongmaoling Road, Yueyang 414000, Hunan, China.
| | - Jianhua Zhao
- Department of Cardiology, Yueyang Central Hospital, No.39 Dongmaoling Road, Yueyang 414000, Hunan, China
| | - Yonghua Lu
- Department of Cardiology, Yueyang Central Hospital, No.39 Dongmaoling Road, Yueyang 414000, Hunan, China
| | - Zhi Chen
- Department of Cardiology, Yueyang Central Hospital, No.39 Dongmaoling Road, Yueyang 414000, Hunan, China
| | - Xiaojian Feng
- Department of Cardiology, Yueyang Central Hospital, No.39 Dongmaoling Road, Yueyang 414000, Hunan, China
| | - Gang Pan
- Department of Cardiology, Yueyang Central Hospital, No.39 Dongmaoling Road, Yueyang 414000, Hunan, China.
| |
Collapse
|
5
|
Hu Z. Exploring the mechanism of curcumin in the treatment of doxorubicin-induced cardiotoxicity based on network pharmacology and molecular docking technology. Medicine (Baltimore) 2024; 103:e36593. [PMID: 38363942 PMCID: PMC10869047 DOI: 10.1097/md.0000000000036593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/21/2023] [Indexed: 02/18/2024] Open
Abstract
Doxorubicin (DOX) is one of the most effective chemotherapeutic agents. However, the nonselective effect leads to serious cardiotoxicity risk in clinical use. Curcumin is a well-known dietary polyphenol that showed a protective effect against the cardiotoxic effect of DOX. This study aimed to assess the role of curcumin in protection against DOX-induced cardiotoxicity. Potential compound and disease targets were obtained from relevant databases, and common targets were screened. Protein-protein interaction (PPI) was used to predict the core targets. Gene ontology (GO) bioprocess analysis and Kyoto encyclopedia of genes and genome enrichment analysis enriched the possible biological processes (BP), cellular components, molecular function, and signaling pathways involved. Finally, the binding of curcumin to target proteins was evaluated through molecular docking. The docking score verified the reliability of the prediction results. In total, 205 curcumin and 700 disease targets were identified. A topological analysis of the PPI network revealed 10 core targets including TP53, tumor necrosis factor-alpha (TNF), AKT1, vascular endothelial growth factor A (VEGFA), prostaglandin-endoperoxide synthase 2 (PTGS2), signal transducer and activator of the transcription 3 (STAT3), HIF1A, MYC, epidermal growth factor receptor (EGFR), and CASP3 (Caspase-3). Furthermore, the enrichment analyses indicated that the effects of curcumin were mediated by genes related to oxidation, inflammation, toxification, cell proliferation, migration, apoptosis, wounding, metabolism, proteolysis, and the signaling pathway of calcium (Ca2+). Molecular docking showed that curcumin could bind with the target proteins with strong molecular force, exhibiting good docking activity. Curcumin has a multi-cardioprotective effect by modulating the core targets' expression in DOX-induced cardiotoxicity. This study elucidated the key target proteins and provided a theoretical basis for further exploring curcumin in the prevention and treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhen Hu
- Department of Electrocardiography, Wuhan No.1 Hospital, Wuhan, China
| |
Collapse
|
6
|
Su X, Xue H, Lou Y, Lv X, Mi X, Lu J, Chen X. Investigation of the Potential Mechanism of Compound Dragon's Blood Capsule against Myocardial Ischemia Based on Network Pharmacology. Comb Chem High Throughput Screen 2024; 27:2940-2950. [PMID: 38231051 DOI: 10.2174/0113862073264485240102064653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Dragon's blood is widely consumed in China, Vietnam and Laos to promote blood circulation. A Compound Dragon's blood capsule (CDC) is a patented medicine composed of dragon's blood, notoginseng, and borneol. This combination is purported to stabilize coronary heart disease and myocardial ischemia. However, the possible mechanisms and the characterization of its drug targets' relevance at the systemic level remain unclear. AIM The present study aims to reveal the potential mechanisms of CDC's anti-myocardial ischemia effect. MATERIALS AND METHODS The potential mechanisms were investigated by network pharmacology and qRT-PCR was used to verify the expression levels of key genes of PI3k-Akt pathway. RESULTS S1PR2 and AGTR1 were the common targets, which involved 6 biological processes annotated by KEGG and GO analysis. The qRT-PCR results showed a remarkable increase in the expression of Pi3k, Pdk1, Akt, Mdm2, Bcl2, and mTOR. Results also showed a decline in the expression of P53 and Casp3 after CDC intervention. CONCLUSION CDC has a significant anti-myocardial ischemia effect through the PI3k/Akt pathway, which demonstrates that CDC is a suitable adjuvant to treat CHD and provides a theoretical basis for its further clinical application.
Collapse
Affiliation(s)
- Xin Su
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong, 666100, China
| | - Hongwei Xue
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100094, China
| | - Yang Lou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100094, China
| | - Xinkai Lv
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100094, China
| | - Xiao Mi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100094, China
| | - Juan Lu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100094, China
| | - Xi Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100094, China
| |
Collapse
|
7
|
Zhao Y, Li X, Nan J. Systematic assessment of the ecotoxicological effects and mechanisms of biochar-derived dissolved organic matter (DOM) on the earthworm Eisenia fetida. ENVIRONMENTAL RESEARCH 2023; 236:116855. [PMID: 37567380 DOI: 10.1016/j.envres.2023.116855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
Biochar-derived dissolved organic matter (DOM) contains toxic substances that are first released into the soil after biochar application. However, the ecological risks of biochar-derived DOM on soil invertebrate earthworms are unclear. Therefore, this study investigated the ecological risks and toxic mechanisms of sewage sludge biochar (SSB)-derived DOM on the earthworm Eisenia fetida (E. fetida) via microcosm experiments. DOM exposure induced earthworm death, growth inhibition, and cocoon decline. Moreover, DOM, especially the 10% DOM300 (derived from SSB prepared at 300 °C) treatments, disrupted the antioxidant defense response and lysosomal stability in earthworms. Integrated biomarker response v2 (IBRv2) analysis was performed to assess the comprehensive toxicity of DOM in E. fetida, and the results revealed that DOM300 might exert more hazardous effects on earthworms than DOM500 (prepared at 500 °C) and DOM700 (prepared at 700 °C), as revealed by increases in the IBRv2 value of 3.48-18.21. Transcriptome analysis revealed that 10% DOM300 exposure significantly disrupted carbohydrate and protein digestion and absorption and induced endocrine disorder. Interestingly, 10% DOM300 exposure also significantly downregulated the expression of genes involved in signaling pathways, e.g., the P13K-AKT, cGMP-PKG, and ErbB signaling pathways, which are related to cell growth, survival, and metabolism, suggesting that DOM300 might induce neurotoxicity in E. fetida. Altogether, these results may contribute to a better understanding of the toxicity and defense mechanisms of biochar-derived DOM on earthworms, especially during long-term applications, and thus provide guidelines for using biochar as a soil amendment.
Collapse
Affiliation(s)
- Yue Zhao
- School of Environment, Harbin Institute of Technology, Harbin, 150090, China
| | - Xin Li
- School of Environment, Harbin Institute of Technology, Harbin, 150090, China.
| | - Jun Nan
- School of Environment, Harbin Institute of Technology, Harbin, 150090, China
| |
Collapse
|
8
|
Ren Z, Luo S, Cui J, Tang Y, Huang H, Ding G. Research Progress of Maternal Metabolism on Cardiac Development and Function in Offspring. Nutrients 2023; 15:3388. [PMID: 37571325 PMCID: PMC10420869 DOI: 10.3390/nu15153388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
The developmental origin of health and disease (DOHaD) hypothesis refers to the adverse effects of suboptimal developmental environments during embryonic and early fetal stages on the long-term health of offspring. Intrauterine metabolic perturbations can profoundly impact organogenesis in offspring, particularly affecting cardiac development and giving rise to potential structural and functional abnormalities. In this discussion, we contemplate the existing understanding regarding the impact of maternal metabolic disorders, such as obesity, diabetes, or undernutrition, on the developmental and functional aspects of the offspring's heart. This influence has the potential to contribute to the susceptibility of offspring to cardiovascular health issues. Alteration in the nutritional milieu can influence mitochondrial function in the developing hearts of offspring, while also serving as signaling molecules that directly modulate gene expression. Moreover, metabolic disorders can exert influence on cardiac development-related genes epigenetically through DNA methylation, levels of histone modifications, microRNA expression, and other factors. However, the comprehensive understanding of the mechanistic underpinnings of these phenomena remains incomplete. Further investigations in this domain hold profound clinical significance, as they can contribute to the enhancement of public health and the prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Zhuoran Ren
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Sisi Luo
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- Shanghai First Maternity and Infant Hospital, Shanghai 200126, China
| | - Jiajun Cui
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Yunhui Tang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| |
Collapse
|
9
|
Chen X, Li Y, Zhang Z, Chen L, Liu Y, Huang S, Zhang X. Xianling Gubao attenuates high glucose-induced bone metabolism disorder in MG63 osteoblast-like cells. PLoS One 2022; 17:e0276328. [PMID: 36548302 PMCID: PMC9778583 DOI: 10.1371/journal.pone.0276328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
Diabetes mellitus (DM) patients are prone to osteoporosis, and high glucose (HG) can affect bone metabolism. In the present study, we investigated the protective effects of traditional Chinese herbal formulation Xianling Gubao (XLGB) on HG-treated MG63 osteoblast-like cells. MG63 cells were incubated with control (mannitol), HG (20 mM glucose) or HG + XLGB (20 mM glucose+200 mg/L XLGB) mediums. Cell proliferation, apoptosis, migration and invasion were examined using CCK8, colony-formation, flow cytometry, Hoechst/PI staining, wound-healing and transwell assays, respectively. ELISA, RT-PCR and western blot analysis were used to detect the levels of osteogenesis differentiation-associated markers such as ALP, OCN, OPN, RUNX2, OPG, and OPGL in MG63 cells. The levels of the PI3K/Akt signaling pathway related proteins, cell cycle-related proteins, and mitochondrial apoptosis-related proteins were detected using western blot analysis. In HG-treated MG63 cells, XLGB significantly attenuated the suppression on the proliferation, migration and invasion of MG63 cells caused by HG. HG downregulated the activation of the PI3K/Akt signaling pathway and the expressions of cell cycle-related proteins, while XLGB reversed the inhibition of HG on MG63 cells. Moreover, XLGB significantly reduced the promotion on the apoptosis of MG63 cells induced by HG, the expressions of mitochondrial apoptosis-related proteins were suppressed by XLGB treatment. In addition, the expressions of osteogenesis differentiation-associated proteins were also rescued by XLGB in HG-treated MG63 cells. Our data suggest that XLGB rescues the MG63 osteoblasts against the effect of HG. The potential therapeutic mechanism of XLGB partially attributes to inhibiting the osteoblast apoptosis and promoting the bone formation of osteoblasts.
Collapse
Affiliation(s)
- Xinlong Chen
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yan Li
- Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Jinan, China
| | - Zhongwen Zhang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, China
| | - Liping Chen
- Department of Endocrinology and Metabology, Weifang Medical University, Shandong Provincial Qianfoshan Hospital, Weifang, China
| | - Yaqian Liu
- Department of Endocrinology and Metabology, Weifang Medical University, Shandong Provincial Qianfoshan Hospital, Weifang, China
| | - Shuhong Huang
- Institute of Basic Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Xiaoqian Zhang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, China
- * E-mail:
| |
Collapse
|
10
|
Gab1 Overexpression Alleviates Doxorubicin-Induced Cardiac Oxidative Stress, Inflammation, and Apoptosis Through PI3K/Akt Signaling Pathway. J Cardiovasc Pharmacol 2022; 80:804-812. [PMID: 35856909 DOI: 10.1097/fjc.0000000000001333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/21/2022] [Indexed: 12/13/2022]
Abstract
ABSTRACT Grb2-associated binding protein 1 (Gab1), an intracellular scaffolding adaptor, was involved in several cardiovascular diseases. However, the role of Gab1 in doxorubicin (DOX)-induced cardiotoxicity remains largely unknown. The present study investigated whether Gab1 protected against DOX-induced cardiotoxicity and the underlying mechanism. We overexpressed Gab1 in the hearts using an adeno-associated virus 9 system through tail vein injection. C57BL/6 mice were subjected to DOX (15 mg/kg/d, i.p.) to generate DOX-induced cardiotoxicity. Echocardiography, histological analysis, immunofluorescence and enzyme-linked immunosorbent assay (ELISA) kits, Western blotting, and quantitative real-time polymerase chain reaction (PCR) evaluated DOX-induced cardiotoxicity and the underlying mechanisms. Myocardial Gab1 protein and messenger RNA (mRNA) levels were markedly decreased in DOX-administered mice. Overexpression of Gab1 in myocardium significantly improved cardiac function and attenuated cardiac oxidative stress, inflammatory response, and apoptosis induced by DOX. Mechanistically, we found that PI3K/Akt signaling pathway was downregulated after DOX treatment, and Gab1 overexpression activated PI3K/Akt signaling pathway, whereas PI3K/Akt signaling pathway inhibition abolished the beneficial effect of Gab1 overexpression in the heart. Collectively, our results indicated that Gab1 is essential for cardioprotection against DOX-induced oxidative stress, inflammatory response, and apoptosis by mediating PI3K/Akt signaling pathway. And cardiac gene therapy with Gab1 provides a novel therapeutic strategy against DOX-induced cardiotoxicity.
Collapse
|
11
|
Guan T, Zheng Y, Jin S, Wang S, Hu M, Liu X, Huang S, Liu Y. Troxerutin alleviates kidney injury in rats via PI3K/AKT pathway by enhancing MAP4 expression. FOOD & NUTRITION RESEARCH 2022; 66:8469. [PMID: 35844954 PMCID: PMC9252313 DOI: 10.29219/fnr.v66.8469] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/07/2022] [Accepted: 02/18/2022] [Indexed: 01/21/2023]
Abstract
Background Troxerutin is a flavonoid compound and possesses potential anti-cancer, antioxidant, and anti-inflammatory activities. Besides, cisplatin is one of the most widely used therapeutic agents, but the clinical uses of cisplatin are often associated with multiple side effects, among which nephrotoxicity is more common. Objective and design This study explored the protective effects of troxerutin (150 mg kg−1 day−1 for 14 days) against cisplatin-induced kidney injury and the potential mechanism using Wistar rats as an experimental mammalian model. Results We discovered that troxerutin could significantly alleviate cisplatin-induced renal dysfunction, such as increased levels of blood urea nitrogen and creatinine (P < 0.01), as well as improved abnormal renal tissue microstructure and ultrastructure. Additionally, troxerutin significantly decreased malondialdehyde (MDA), hydrogen peroxide (H2O2), NO, inducible nitric oxide synthase (iNOS) levels (P < 0.01), p-NF-κB p65/NF-κB p65, TNF-α, Pro-IL-1β, IL-6, B cell lymphoma-2 (Bcl-2)/Bcl-xl associated death promoter (Bad), Cytochrome C (Cyt C), Cleaved-caspase 9, Cleaved-caspase 3, and Cleaved-caspase 8 protein levels (P < 0.01) in the kidney tissues of cisplatin-treated rats; and increased superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), total antioxidant capacity (T-AOC) activities (P < 0.01), IL-10, Bcl-2 protein levels (P < 0.01). Conclusion These results suggested that the underlying mechanism might be attributed to the regulation of Phosphoinositide 3 kinase/Protein kinase B (PI3K/AKT) pathway via enhancing MAP4 expression to attenuate cellular apoptosis, alleviating oxidative stress and inflammatory response.
Collapse
Affiliation(s)
- Tongxu Guan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Yingce Zheng
- College of Life Science, Northeast Agricultural University, Harbin, P. R. China
| | - Shengzi Jin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Shuang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Mengxin Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Xingyao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Siqi Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Yun Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, P. R. China
- Yun Liu, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China, Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| |
Collapse
|
12
|
Fangchinoline induces gallbladder cancer cell apoptosis by suppressing PI3K/Akt/XIAP axis. PLoS One 2022; 17:e0266738. [PMID: 35446864 PMCID: PMC9022853 DOI: 10.1371/journal.pone.0266738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/25/2022] [Indexed: 11/19/2022] Open
Abstract
Gallbladder cancer (GBC) is the most common biliary tract malignancy with a dismal prognosis. The development of new drugs may help to improve prognosis. This study found that fangchinoline, a bisbenzylisoquinoline alkaloids, inhibited the proliferation and clone formation of GBC cells in a dose-dependent manner. Moreover, Hoechst staining, TUNEL assays, and flow cytometry demonstrated that fangchinoline effectively induced apoptosis in GBC cells. Further studies found that an anti-apoptotic pathway, the PI3K/Akt/XIAP axis, was significantly inhibited in GBC cells after treating with fangchinoline. Finally, we confirmed that fangchinoline restrained xenograft tumor growth in vivo. Our findings indicate that fangchinoline can be considered a potential drug for GBC treatment.
Collapse
|
13
|
Liu ZQ, Cheng M, Fu F, Li R, Han J, Yang X, Deng Q, Li LS, Lei TY, Li DZ, Liao C. Identification of differential microRNAs and messenger RNAs resulting from ASXL transcriptional regulator 3 knockdown during during heart development. Bioengineered 2022; 13:9948-9961. [PMID: 35435106 PMCID: PMC9161854 DOI: 10.1080/21655979.2022.2062525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 11/30/2022] Open
Abstract
Congenital heart disease (CHD) is the most common birth defect. Although ASXL transcriptional regulator 3 (ASXL3) has been reported to cause hereditary CHD, ASXL3-mediated mechanisms in heart development remain unclear. In this study, we used dimethyl sulfoxide (DMSO) to induce differentiation in P19 cells, observed cell morphology using light microscopy after ASXL3 knockdown, and determined the levels of associated myocardial cell markers using reverse transcription-quantitative polymerase chain reaction and western blotting. Subsequently, we used microRNA sequencing, messenger RNA (mRNA) sequencing, and bioinformatics to initially identify the possible mechanisms through which ASXL3-related microRNAs and mRNAs affect heart development. The results indicated that DMSO induced P19 cell differentiation, which could be inhibited by ASXL3 knockdown. We screened 1214 and 1652 differentially expressed microRNAs and mRNAs, respectively, through ASXL3 knockdown and sequencing; these differentially expressed miRNAs were largely enriched in PI3K-Akt, mitogen-activated protein kinase, and Rap1 signaling pathways. Additionally, 11 miRNAs associated with heart development were selected through a literature review. Our analysis indicated the involvement of mmu-miR-323-3p in P19 cell differentiation through the PI3K-Akt pathway. In conclusion, ASXL3 may be involved in the regulation of heart development. This comprehensive study of differentially expressed microRNAs and mRNAs through ASXL3 knockdown in P19 cells provides new insights that may aid the prevention and treatment of CHD.
Collapse
Affiliation(s)
- Ze-Qun Liu
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Mi Cheng
- Department of Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Fang Fu
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Ru Li
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Jin Han
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Xin Yang
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Qiong Deng
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Lu-Shan Li
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Ting-Ying Lei
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Dong-Zhi Li
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| | - Can Liao
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, GuangzhouGuangdong, China
| |
Collapse
|
14
|
Lee J, Gong YX, Jeong H, Seo H, Xie DP, Sun HN, Kwon T. Pharmacological effects of Picrasma quassioides (D. Don) Benn for inflammation, cancer and neuroprotection (Review). Exp Ther Med 2021; 22:1357. [PMID: 34659503 PMCID: PMC8515544 DOI: 10.3892/etm.2021.10792] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Picrasma quassioides (D. Don) Benn is an Asian shrub with a considerable history of traditional medicinal use. P. quassioides and its extracts exhibit good therapeutic properties against several diseases, including anti-inflammatory, antibacterial and anticancer effects. However, the composition of compounds contained in P. quassioides is complex; although various studies have examined mixtures or individual compounds extracted from it, studies on the application of P. quassioides extracts remain limited. In the present review, the structures and functions of the compounds identified from P. quassioides and their utility in anti-inflammatory, anticancer and neuroprotectant therapies was discussed. The present review provided up-to-date information on pharmacological activities and clinical applications for P. quassioides extracts.
Collapse
Affiliation(s)
- Jaihyung Lee
- Epigenetics Drug Discovery Center, Hwalmyeong Convalescence Hospital, Gapyeong, Gyeonggi 12458, Republic of Korea
- Korean Convergence Medicine Center, Hwalmyeong Hospital of Korean Medicine, Seoul 03790, Republic of Korea
| | - Yi-Xi Gong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hyunjeong Jeong
- Epigenetics Drug Discovery Center, Hwalmyeong Convalescence Hospital, Gapyeong, Gyeonggi 12458, Republic of Korea
- Korean Convergence Medicine Center, Hwalmyeong Hospital of Korean Medicine, Seoul 03790, Republic of Korea
| | - Hoyoung Seo
- Epigenetics Drug Discovery Center, Hwalmyeong Convalescence Hospital, Gapyeong, Gyeonggi 12458, Republic of Korea
- Korean Convergence Medicine Center, Hwalmyeong Hospital of Korean Medicine, Seoul 03790, Republic of Korea
| | - Dan-Ping Xie
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hu-Nan Sun
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk 56216, Republic of Korea
| |
Collapse
|
15
|
Zhang B, Zhang CY, Zhang XL, Sun GB, Sun XB. Guan Xin Dan Shen formulation protects db/db mice against diabetic cardiomyopathy via activation of Nrf2 signaling. Mol Med Rep 2021; 24:531. [PMID: 34036388 PMCID: PMC8170264 DOI: 10.3892/mmr.2021.12170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
Guan Xin Dan Shen formulation (GXDSF) is a widely used treatment for the management of coronary heart disease in China and is composed of three primary components: Dalbergiae odoriferae Lignum, Salviae miltiorrhizae Radix et Rhizoma and Panax notoginseng Radix et Rhizoma. However, the potential use of GXDSF for the management of diabetic cardiomyopathy (DCM) has not been previously assessed. The present study aimed to assess the effects of GXDSF on DCM, as well as the underlying mechanism. In the present study, db/db mice were used. Following treatment with GXDSF for 10 weeks, fasting blood glucose, insulin sensitivity, serum lipid levels and cardiac enzyme levels were detected. Cardiac pathological alterations and cardiac function were assessed by performing hematoxylin and eosin staining and echocardiograms, respectively. TUNEL assays were conducted to assess cardiomyocyte apoptosis. Additionally, reverse transcription‑quantitative PCR and western blotting were performed to evaluate the expression of apoptosis‑associated genes and proteins, respectively. In the model group, the db/db mice displayed obesity, hyperlipidemia and hyperglycemia, accompanied by noticeable myocardial hypertrophy and diastolic dysfunction. Following treatment with GXDSF for 10 weeks, serum triglyceride levels were lower and insulin sensitivity was enhanced in db/db mice compared with the model group, which indicated improvement in condition. Cardiac hypertrophy and dysfunction were also improved in db/db mice following treatment with GXDSF, resulting in significantly increased left ventricular ejection fraction and fractional shortening compared with the model group. Following treatment with metformin or GXDSF, model‑induced increases in levels of myocardial enzymes were decreased in the moderate and high dose groups. Moreover, the results indicated that, compared with the model group, GXDSF significantly inhibited cardiomyocyte apoptosis in diabetic heart tissues by increasing Bcl‑2 expression and decreasing the expression levels of Bax, cleaved caspase‑3 and cleaved caspase‑9. Mechanistically, GXDSF enhanced Akt phosphorylation, which upregulated antioxidant enzymes mediated by nuclear factor erythroid 2‑related factor 2 (Nrf2) signaling. Collectively, the results of the present study indicated that GXDSF attenuated cardiac dysfunction and inhibited cardiomyocyte apoptosis in diabetic mice via activation of Akt/Nrf2 signaling. Therefore, GXDSF may serve as a potential therapeutic agent for the management of DCM.
Collapse
Affiliation(s)
- Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China
| | - Chen-Yang Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China
| | - Xue-Lian Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China
| | - Gui-Bo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China
| | - Xiao-Bo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China
| |
Collapse
|
16
|
Selig JI, Boulgaropoulos J, Niazy N, Ouwens DM, Preuß K, Horn P, Westenfeld R, Lichtenberg A, Akhyari P, Barth M. Crosstalk of Diabetic Conditions with Static Versus Dynamic Flow Environment-Impact on Aortic Valve Remodeling. Int J Mol Sci 2021; 22:ijms22136976. [PMID: 34203572 PMCID: PMC8268732 DOI: 10.3390/ijms22136976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 01/12/2023] Open
Abstract
Type 2 diabetes mellitus (T2D) is one of the prominent risk factors for the development and progression of calcific aortic valve disease. Nevertheless, little is known about molecular mechanisms of how T2D affects aortic valve (AV) remodeling. In this study, the influence of hyperinsulinemia and hyperglycemia on degenerative processes in valvular tissue is analyzed in intact AV exposed to an either static or dynamic 3D environment, respectively. The complex native dynamic environment of AV is simulated using a software-governed bioreactor system with controlled pulsatile flow. Dynamic cultivation resulted in significantly stronger fibrosis in AV tissue compared to static cultivation, while hyperinsulinemia and hyperglycemia had no impact on fibrosis. The expression of key differentiation markers and proteoglycans were altered by diabetic conditions in an environment-dependent manner. Furthermore, hyperinsulinemia and hyperglycemia affect insulin-signaling pathways. Western blot analysis showed increased phosphorylation level of protein kinase B (AKT) after acute insulin stimulation, which was lost in AV under hyperinsulinemia, indicating acquired insulin resistance of the AV tissue in response to elevated insulin levels. These data underline a complex interplay of diabetic conditions on one hand and biomechanical 3D environment on the other hand that possesses an impact on AV tissue remodeling.
Collapse
Affiliation(s)
- Jessica I. Selig
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| | - Joana Boulgaropoulos
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| | - Naima Niazy
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| | - D. Margriet Ouwens
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Auf’m Hennekamp 65, 40225 Düsseldorf, Germany;
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, Neuherberg, 85764 München, Germany
- Department of Endocrinology, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Karlheinz Preuß
- Faculty of Biotechnology, Bioprocessing, Modulation and Simulation, University of Applied Sciences Mannheim, Paul-Wittsack-Straße 10, 68163 Mannheim, Germany;
| | - Patrick Horn
- Department of Cardiology, Pneumology and Angiology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (P.H.); (R.W.)
| | - Ralf Westenfeld
- Department of Cardiology, Pneumology and Angiology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (P.H.); (R.W.)
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
- Correspondence:
| | - Mareike Barth
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| |
Collapse
|
17
|
Mibefradil Alleviates High-Glucose-induced Cardiac Hypertrophy by Inhibiting PI3K/Akt/mTOR-mediated Autophagy. J Cardiovasc Pharmacol 2021; 76:246-254. [PMID: 32433360 DOI: 10.1097/fjc.0000000000000844] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cardiac hypertrophy causes heart failure and is associated with hyperglycemia in patients with diabetes mellitus. Mibefradil, which acts as a T-type calcium channel blocker, exerts beneficial effects in patients with heart failure. In this study, we explored the effects and mechanism of mibefradil on high-glucose-induced cardiac hypertrophy in H9c2 cells. H9c2 cells were incubated in a high-glucose medium and then treated with different concentrations of mibefradil in the presence or absence of the Akt inhibitor MK2206 or mTOR inhibitor rapamycin. Cell size was evaluated through immunofluorescence, and mRNA expression of cardiac hypertrophy markers (atrial natriuretic peptide, brain natriuretic peptide, and β-myosin heavy chain) was assessed by using quantitative real-time polymerase chain reaction. Changes in the expression of p-PI3K, p-Akt, and p-mTOR were evaluated using Western blotting, and autophagosome formation was detected using transmission electron microscopy. Our results indicate that mibefradil reduced the size of H9c2 cells, decreased mRNA expression of atrial natriuretic peptide, brain natriuretic peptide, and β-myosin heavy chain, and decreased the level of autophagic flux. However, MK2206 and rapamycin induced autophagy and reversed the effects of mibefradil on high-glucose-induced H9c2 cells. In conclusion, mibefradil ameliorated high-glucose-induced cardiac hypertrophy by activating the PI3K/Akt/mTOR pathway and inhibiting excessive autophagy. Our study shows that mibefradil can be used therapeutically to ameliorate cardiac hypertrophy in patients with diabetes mellitus.
Collapse
|
18
|
Fang B, Wen S, Li Y, Bai F, Wei Y, Xiong Y, Huang Q, Lin X. Prediction and verification of target of helenalin against hepatic stellate cell activation based on miR-200a-mediated PI3K/Akt and NF-κB pathways. Int Immunopharmacol 2021; 92:107208. [PMID: 33444919 DOI: 10.1016/j.intimp.2020.107208] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/18/2020] [Accepted: 11/12/2020] [Indexed: 01/22/2023]
Abstract
Hepatic stellate cell (HSC) activation is a crucial event in the progress of liver fibrosis. In this study, the target of helenalin was firstly predicted by bioinformatics analysis, and then the prediction was verified by various experiments. HSC-T6 cells were activated by interleukin-1 beta (IL-1β) and then treated with helenalin. Moreover, HSC-T6 cells were transfected with miR-200a mimic or inhibitor, and the effect of helenalin on the miR-200a-mediated PI3K/Akt and NF-κB signaling pathways was investigated. The bioinformatics analysis indicated that miR-200a might regulate the PI3K/Akt pathway, NF-κB activation, Bcl-2 family and Caspases, ultimately affecting cell survival and apoptosis. Interestingly, the molecular docking demonstrated that the target of helenalin might be miR-200a-mediated the PI3K/Akt and NF-κB pathways. Moreover, the experiments showed that helenalin administration led to the inactivation of HSC-T6 cells, as evidenced by the inhibition of cell proliferation, α-SMA expression and collagen production. The mechanism studies showed that helenalin reduced collagen accumulation by restoring the balance of MMPs/TIMPs. Moreover, helenalin markedly suppressed HSC activation by inhibiting the PI3K/Akt pathway and alleviated inflammatory response by blocking the NF-κB signal transduction. Further study indicated that helenalin up-regulated miR-200a expression, thus leading to the inhibition of the PI3K/Akt and NF-κB signaling pathways. In conclusion, helenalin inhibits HSC activation via inhibiting the miR-200a-mediated PI3K/Akt and NF-κB pathways, and it may be developed as a potential medicine for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Bin Fang
- Guangxi Medical University, Nanning 530021, China
| | - Shujuan Wen
- Guangxi Medical University, Nanning 530021, China
| | - Yan Li
- Guangxi Medical University, Nanning 530021, China
| | - Facheng Bai
- Guangxi Medical University, Nanning 530021, China
| | - Yuanyuan Wei
- Guangxi Medical University, Nanning 530021, China
| | - Yuhua Xiong
- Guangxi Medical University, Nanning 530021, China
| | - Quanfang Huang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, China.
| | - Xing Lin
- Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
19
|
Upregulation of miRNA-23a-3p rescues high glucose-induced cell apoptosis and proliferation inhibition in cardiomyocytes. In Vitro Cell Dev Biol Anim 2020; 56:866-877. [PMID: 33197036 PMCID: PMC7723946 DOI: 10.1007/s11626-020-00518-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
Maternal hyperglycemia potentially inhibits the development of the fetal heart by suppressing cardiomyocyte proliferation and promoting apoptosis. Different studies have indicated that miRNAs are key regulators of cardiomyocyte proliferation, differentiation, and apoptosis and play a protective role in a variety of cardiovascular diseases. However, the biological function of miRNA-23a in hyperglycemia-related cardiomyocyte injury is not fully understood. The present study investigated the effect of miRNA-23a-3p on cell proliferation and apoptosis in a myocardial injury model induced by high glucose. H9c2 cardiomyocytes were exposed to high glucose to establish an in vitro myocardial injury model and then transfected with miRNA-23a-3p mimics. After miRNA-23a-3p transfection, lens-free microscopy was used to dynamically monitor cell numbers and confluence and calculate the cell cycle duration. CCK-8 and EdU incorporation assays were performed to detect cell proliferation. Flow cytometry was used to measured cell apoptosis. Upregulation of miRNA-23a-3p significantly alleviated high glucose-induced cell apoptosis and cell proliferation inhibition (p < 0.01 and p < 0.0001, respectively). The cell cycle of the miRNA-23a-3p mimics group was significantly shorter than that of the negative control group (p < 0.01). The expression of cell cycle–activating and apoptosis inhibition-associated factors Ccna2, Ccne1, and Bcl-2 was downregulated by high glucose and upregulated by miRNA-23a-3p overexpression in high glucose-injured H9c2 cells. miRNA-23a-3p mimics transfection before high glucose treatment had a significantly greater benefit than transfection after high glucose treatment (p < 0.0001), and the rescue effect of miRNA-23a-3p increased as the concentration increased. This study suggests that miRNA-23a-3p exerted a dose- and time-dependent protective effect on high glucose-induced H9c2 cardiomyocyte injury.
Collapse
|
20
|
Cui Y, Chen R, Ma L, Yang W, Chen M, Zhang Y, Yu S, Dong W, Zeng W, Lan X, Pan C. miR-205 Expression Elevated With EDS Treatment and Induced Leydig Cell Apoptosis by Targeting RAP2B via the PI3K/AKT Signaling Pathway. Front Cell Dev Biol 2020; 8:448. [PMID: 32596241 PMCID: PMC7300349 DOI: 10.3389/fcell.2020.00448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/13/2020] [Indexed: 12/19/2022] Open
Abstract
The adult Leydig cells (ALCs), originated from stem Leydig cells (SLCs), can secrete testosterone which is essential for germ cell development and sexual behavior maintenance. As a synthetic compound, ethane dimethane sulfonate (EDS), a well-known alkylating agent, has been reported to specifically ablate ALCs. In this study, EDS was verified to ablate differentiated pig LCs by experiments. Subsequently, the primary isolated pig LCs (containing SLCs and differentiated LCs) and EDS-treated LCs (almost exclusively SLCs) were collected for RNA-seq 4,904 genes and 15 miRNAs were differently expressed between the two groups. Down-regulated genes in the EDS-treated group were mainly related to steroid hormone biosynthesis. The highest up-regulation miRNAs was miR-205 after EDS treatment. Additionally, miR-205 was expressed more highly in pig SLCs clones compared with differentiated LCs. Through qRT-PCR, western blot (WB), TUNEL, EDU and flow cytometry, miR-205 was found to induce cell apoptosis, but did not affect proliferation or differentiation in both TM3 and GC-1spg mouse cell lines. Through luciferase reporter assays and WB, RAP2B was identified as a target gene of miR-205. Besides, overexpression of miR-205 inhibited the expressions of PI3K, Akt and p-AKT. All these findings were helpful for elucidating the regulation mechanism in pig LCs.
Collapse
Affiliation(s)
- Yang Cui
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Rui Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Lin Ma
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Wenjing Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Mingyue Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Yanghai Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Shuai Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Wuzi Dong
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Wenxian Zeng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| | - Chuanying Pan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Xianyang, China
| |
Collapse
|
21
|
Effects of Hydroalcoholic Leaf Extract of Avicennia marina on Apoptotic, Inflammatory, Oxidative Stress, and Lipid Peroxidation Indices and Liver Histology of Type 1 Diabetic Rats. HEPATITIS MONTHLY 2020. [DOI: 10.5812/hepatmon.99454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
22
|
Chen SH, Liu XN, Peng Y. MicroRNA-351 eases insulin resistance and liver gluconeogenesis via the PI3K/AKT pathway by inhibiting FLOT2 in mice of gestational diabetes mellitus. J Cell Mol Med 2019; 23:5895-5906. [PMID: 31287224 PMCID: PMC6714143 DOI: 10.1111/jcmm.14079] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/10/2018] [Accepted: 11/13/2018] [Indexed: 12/29/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is known as different degree glucose intolerance that is initially identified during pregnancy. MicroRNAs (miRs) may be a potential candidate for treatment of GDM. Herein, we suggested that miR‐351 could be an inhibitor in the progression of GDM via the phosphoinositide 3‐kinase/protein kinase B (PI3K/AKT) pathway. Microarray analysis was used to identify differentially expressed genes and predict miRs regulating flotillin 2 (FLOT2). Target relationship between miR‐351 and FLOT2 was verified. Gestational diabetes mellitus mice were treated with a series of mimic, inhibitor and small interfering RNA to explore the effect of miR‐351 on insulin resistance (IR), cell apoptosis in pancreatic tissues and liver gluconeogenesis through evaluating GDM‐related biochemical indexes, as well as expression of miR‐351, FLOT2, PI3K/AKT pathway‐, IR‐ and liver gluconeogenesis‐related genes. MiR‐351 and FLOT2 were reported to be involved in GDM. FLOT2 was the target gene of miR‐351. Gestational diabetes mellitus mice exhibited IR and liver gluconeogenesis, up‐regulated FLOT2, activated PI3K/AKT pathway and down‐regulated miR‐351 in liver tissues. Additionally, miR‐351 overexpression and FLOT2 silencing decreased the levels of FLOT2, phosphoenolpyruvate carboxykinase, glucose‐6‐phosphatase, fasting blood glucose, fasting insulin, total cholesterol, triglyceride, glyeosylated haemoglobin and homeostasis model of assessment for IR index (HOMA‐IR), extent of PI3K and AKT phosphorylation, yet increased the levels of HOMA for islet β‐cell function, HOMA for insulin sensitivity index and glucose transporter 2 expression, indicating reduced cell apoptosis in pancreatic tissues and alleviated IR and liver gluconeogenesis. Our results reveal that up‐regulation of miR‐351 protects against IR and liver gluconeogenesis by repressing the PI3K/AKT pathway through regulating FLOT2 in GDM mice, which identifies miR‐351 as a potential therapeutic target for the clinical management of GDM.
Collapse
Affiliation(s)
- Shu-Hong Chen
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong Province, P.R. China
| | - Xiao-Nan Liu
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong Province, P.R. China
| | - Yan Peng
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong Province, P.R. China
| |
Collapse
|
23
|
Su D, Zhao J, Hu S, Guan L, Li Q, Shi C, Ma X, Gou J, Zhou Y. GSK3β and MCL-1 mediate cardiomyocyte apoptosis in response to high glucose. Histochem Cell Biol 2019; 152:217-225. [DOI: 10.1007/s00418-019-01798-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2019] [Indexed: 12/15/2022]
|
24
|
Selig JI, Ouwens DM, Raschke S, Thoresen GH, Fischer JW, Lichtenberg A, Akhyari P, Barth M. Impact of hyperinsulinemia and hyperglycemia on valvular interstitial cells - A link between aortic heart valve degeneration and type 2 diabetes. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2526-2537. [PMID: 31152868 DOI: 10.1016/j.bbadis.2019.05.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 04/27/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes is a known risk factor for cardiovascular diseases and is associated with an increased risk to develop aortic heart valve degeneration. Nevertheless, molecular mechanisms leading to the pathogenesis of valve degeneration in the context of diabetes are still not clear. Hence, we hypothesized that classical key factors of type 2 diabetes, hyperinsulinemia and hyperglycemia, may affect signaling, metabolism and degenerative processes of valvular interstitial cells (VIC), the main cell type of heart valves. Therefore, VIC were derived from sheep and were treated with hyperinsulinemia, hyperglycemia and the combination of both. The presence of insulin receptors was shown and insulin led to increased proliferation of the cells, whereas hyperglycemia alone showed no effect. Disturbed insulin response was shown by impaired insulin signaling, i.e. by decreased phosphorylation of Akt/GSK-3α/β pathway. Analysis of glucose transporter expression revealed absence of glucose transporter 4 with glucose transporter 1 being the predominantly expressed transporter. Glucose uptake was not impaired by disturbed insulin response, but was increased by hyperinsulinemia and was decreased by hyperglycemia. Analyses of glycolysis and mitochondrial respiration revealed that VIC react with increased activity to hyperinsulinemia or hyperglycemia, but not to the combination of both. VIC do not show morphological changes and do not acquire an osteogenic phenotype by hyperinsulinemia or hyperglycemia. However, the treatment leads to increased collagen type 1 and decreased α-smooth muscle actin expression. This work implicates a possible role of diabetes in early phases of the degeneration of aortic heart valves.
Collapse
Affiliation(s)
- Jessica I Selig
- Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.
| | - D Margriet Ouwens
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Department of Endocrinology, Ghent University Hospital, Ghent, Belgium.
| | - Silja Raschke
- Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - G Hege Thoresen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Jens W Fischer
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.
| | - Artur Lichtenberg
- Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.
| | - Payam Akhyari
- Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.
| | - Mareike Barth
- Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
25
|
Expression of Immune Regulatory Genes in the Porcine Internal Genital Tract Is Differentially Triggered by Spermatozoa and Seminal Plasma. Int J Mol Sci 2019; 20:ijms20030513. [PMID: 30691059 PMCID: PMC6387272 DOI: 10.3390/ijms20030513] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 01/08/2023] Open
Abstract
Mating or cervical deposition of spermatozoa or seminal plasma (SP) modifies the expression of genes affecting local immune defense processes at the oviductal sperm reservoir in animals with internal fertilization, frequently by down-regulation. Such responses may occur alongside sperm transport to or even beyond the reservoir. Here, immune-related gene expression was explored with cDNA microarrays on porcine cervix-to-infundibulum tissues, pre-/peri-ovulation. Samples were collected 24 h post-mating or cervical deposition of sperm-peak spermatozoa or SP (from the sperm-peak fraction or the whole ejaculate). All treatments of this interventional study affected gene expression. The concerted action of spermatozoa and SP down-regulated chemokine and cytokine (P00031), interferon-gamma signaling (P00035), and JAK/STAT (P00038) pathways in segments up to the sperm reservoir (utero-tubal junction (UTJ)/isthmus). Spermatozoa in the vanguard sperm-peak fraction (P1-AI), uniquely displayed an up-regulatory effect on these pathways in the ampulla and infundibulum. Sperm-free SP, on the other hand, did not lead to major effects on gene expression, despite the clinical notion that SP mitigates reactivity by the female immune system after mating or artificial insemination.
Collapse
|
26
|
Sultan S. The effect of maternal type 2 diabetes on fetal endothelial gene expression and function. Acta Diabetol 2019; 56:73-85. [PMID: 30167870 DOI: 10.1007/s00592-018-1207-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 08/05/2018] [Indexed: 01/12/2023]
Abstract
AIMS Maternal type 2 diabetes (T2D) can result in adverse pathological outcomes to both the mother and fetus. The present study aimed to investigate the pathological effects of maternal T2D on the gene expression patterns and functions of fetal human umbilical vein endothelial cells (HUVECs), a representative of fetal vascular cells. METHODS Cell proliferation, apoptosis, mitochondrial ROS production and cell cycle were measured using flowcytometry. Genome-wide expression was measured using Affymetrix microarray. Gene expression of CCND2, STAT1, ITGB8, ALDH2, and ADAMTS5 was measured using real-time PCR. RESULTS HUVECs derived from T2D mothers (T2D-HUVECs) showed elevated levels of mitochondrial superoxide anions, reduced cell proliferation, and increased apoptosis rates relative to HUVECs derived from healthy control mothers (C.HUVECs). In addition , T2D-HUVECs showed a decreased proportion of cells in G0/G1 and cell cycle arrest at the S phases relative to controls. Interestingly, microarray experiments revealed significant differences in genome-wide expression profiles between T2D-HUVECs and C.HUVECs. In particular, the analysis identified 90 upregulated genes and 42 downregulated genes. The upregulated genes CCND2, STAT1, ITGB8, ALDH2, and ADAMTS5 were validated as potential biomarkers for fetal endothelial dysfunction. Functional network analysis revealed that these genes are the important players that participate in the pathogenesis of endothelial dysfunction, which in turn influences the inflammatory response, cellular movement, and cardiovascular system development and function. CONCLUSION Sustained alterations in the overall function of T2D-HUVEC and gene expression profiles provided insights into the role of maternal T2D on the pathophysiology of the fetal endothelial dysfunction.
Collapse
Affiliation(s)
- Samar Sultan
- Medical Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
27
|
Song D, Liu X, Diao Y, Sun Y, Gao G, Zhang T, Chen K, Pei L. Hydrogen‑rich solution against myocardial injury and aquaporin expression via the PI3K/Akt signaling pathway during cardiopulmonary bypass in rats. Mol Med Rep 2018; 18:1925-1938. [PMID: 29956781 PMCID: PMC6072160 DOI: 10.3892/mmr.2018.9198] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/05/2018] [Indexed: 12/19/2022] Open
Abstract
Myocardial ischemia, hypoxia and reperfusion injury are induced by aortic occlusion, cardiac arrest and resuscitation during cardiopulmonary bypass (CPB), which can severely affect cardiac function. The aim of the present study was to investigate the effects of hydrogen-rich solution (HRS) and aquaporin (AQP) on cardiopulmonary bypass (CPB)-induced myocardial injury, and determine the mechanism of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. Sprague Dawley rats were divided into a sham operation group, a CPB surgery group and a HRS group. A CPB model was established, and the hemodynamic parameters were determined at the termination of CPB. The myocardial tissues were observed by hematoxylin and eosin, and Masson staining. The levels of myocardial injury markers [adult cardiac troponin I (cTnI), lactate dehydrogenase (LDH), creatine kinase MB (CK-MB) and brain natriuretic peptide (BNP)], inflammatory factors [interleukin (IL)-1β, IL-6 and tumor necrosis factor-α (TNF-α)] and oxidative stress indicators [superoxide dismutase (SOD), malondialdehyde (MDA) and myeloperoxidase (MPO)] were determined by ELISA. Furthermore, H9C2 cells were treated with HRS following hypoxia/reoxygenation. Cell viability and cell apoptosis were investigated. The expression of apoptosis regulator Bcl-2 (Bcl-2), apoptosis regulator Bax (Bax), caspase 3, AQP-1, AQP-4, phosphorylated (p)-Akt, heme oxygenase 1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) were investigated using western blotting and quantitative-polymerase chain reaction of tissues and cells. Following CPB, myocardial cell arrangement was disordered, myocardial injury markers (cTnI, LDH, CK-MB and BNP), inflammatory cytokines (IL-1β, IL-6 and TNF-α) and MDA levels were significantly increased compared with the sham group; whereas the SOD levels were significantly downregulated following CPB compared with the sham group. HRS attenuated myocardial injury, reduced the expression levels of cTnI, LDH, CK-MB, BNP, IL-1β, IL-6, TNF-α, MDA and MPO, and increased SOD release. Levels of Bcl-2, AQP-1, AQP-4, p-Akt, HO-1 and Nrf2 were significantly increased following HRS; whereas Bax and caspase-3 expression levels were significantly reduced following CPB. HRS treatment significantly increased the viability of myocardial cells, reduced the rate of myocardial cell apoptosis and the release of MDA and LDH compared with the CPB group. A PI3K inhibitor (LY294002) was revealed to reverse the protective effect of HRS treatment. HRS was demonstrated to attenuate CPB-induced myocardial injury, suppress AQP-1 and AQP-4 expression following CPB treatment and protect myocardial cells via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Dandan Song
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110016, P.R. China
| | - Xuelei Liu
- Department of Clinical Laboratory, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yugang Diao
- Department of Anesthesiology, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yingjie Sun
- Department of Anesthesiology, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Guangjie Gao
- Department of Anesthesiology, The 463rd Hospital of People's Liberation Army China, Shenyang, Liaoning 110012, P.R. China
| | - Tiezheng Zhang
- Department of Anesthesiology, General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Keyan Chen
- Department of Laboratory Animal Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Ling Pei
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
28
|
Xu L, Li J, Kuang Z, Kuang Y, Wu H. Knockdown of Gab1 Inhibits Cellular Proliferation, Migration, and Invasion in Human Oral Squamous Carcinoma Cells. Oncol Res 2018; 26:617-624. [PMID: 28893350 PMCID: PMC7844679 DOI: 10.3727/096504017x15043589260618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Grb2-associated binder 1 (Gab1) is often aberrant in cancerous cells and tissues, whose alteration is responsible for aggressive phenotypes. In this study, we examined the Gab1 expression in human oral squamous cell carcinoma (OSCC) tissues and investigated the cellular and molecular effect of Gab1 on migration, invasion, and cell growth of the OSCC cell lines SCC15 and SCC25. We found that Gab1 was overexpressed in OSCC tissues and cells, which is related to the protein levels of various molecules associated with cellular proliferation, migration, and invasion. Functional assays identified that Gab1 overexpression promoted cell proliferation and invasion of OSCC cells and inhibited cell apoptosis in the SCC15 and SCC25 cell lines. On the other hand, Gab1 silencing affected the proliferation and invasion of OSCC cells and induced cell apoptosis. Western blot assay identified that Gab1 overexpression suppressed the expression of Cdc20 homolog 1 (Cdh1) and then promoted cell invasion in OSCC cells. Furthermore, Gab1-mediated Cdh1 downregulation was significantly reversed when the cells were subjected to an inhibitor of p-Akt. In conclusion, these results suggested that Gab1 induced malignant progression of OSCC cells probably via activation of the Akt/Cdh1 signaling pathway. Thus, Gab1 may be a potential therapeutic target in the treatment of OSCC patients.
Collapse
Affiliation(s)
- Luyong Xu
- Department of Stomatology, Rizhao People’s Hospital, Jining Medical University, Rizhao, P.R. China
| | - Jie Li
- Department of Stomatology, Rizhao People’s Hospital, Jining Medical University, Rizhao, P.R. China
| | - Zheng Kuang
- Department of Stomatology, Rizhao People’s Hospital, Jining Medical University, Rizhao, P.R. China
| | - Yan Kuang
- Department of Stomatology, Rizhao People’s Hospital, Jining Medical University, Rizhao, P.R. China
| | - Hong Wu
- Department of Stomatology, Rizhao People’s Hospital, Jining Medical University, Rizhao, P.R. China
| |
Collapse
|