1
|
Varanda RV, Kumari J, van Rheden REM, Cuijpers VMJI, Bloemen M, Göllesch F, Von den Hoff JW, Henneman S, Xie R, Wagener FADTG, Suttorp CM. Survival of periodontal ligament myofibroblasts after short-term mechanical strain in rats and in vitro: Could myofibroblasts contribute to orthodontic relapse? Arch Oral Biol 2025; 172:106173. [PMID: 39778331 DOI: 10.1016/j.archoralbio.2025.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/16/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
OBJECTIVES To investigate in vivo whether myofibroblasts formed in the PDL after exposure to short-term high experimental orthodontic forces in rats survive. To study in vitro whether human PDL fibroblasts can differentiate into myofibroblasts and survive when chemical or mechanical stimuli are removed. DESIGN Nine 6-week-old male Wistar rats were used in this experiment. Rat molars were exposed to high but rapidly decreasing experimental orthodontic forces by applying a rubber band and analyzed for the presence of myofibroblasts using ASMA staining. In vitro, human periodontal ligament (PDL) fibroblasts were exposed to transforming growth factor β1 (TGFβ1) and/or mechanical stress and monitored for myofibroblast formation and survival after these stimuli were abrogated. RESULTS In vivo exposure to orthodontic forces strongly induced myofibroblast formation in the stretched regions of the PDL. Furthermore, many PDL myofibroblasts remained present 6 days after exposure to these short-term high orthodontic forces. Human PDL fibroblasts were shown to differentiate into myofibroblasts after 2 days of TGFβ1 exposure and survive for at least 2 more days after removing chemical stimuli (TGFβ1) or mechanical strain. Under in vitro conditions, both TGFβ1 and mechanical strain for 3 days promoted (myo)fibroblast formation, and these cells persisted for 3 more days after the removal of both stimuli. CONCLUSIONS PDL myofibroblasts survive after the removal of mechanical strain in vivo and in vitro. This supports the hypothesis that myofibroblasts, which form in response to mechanical strain and chemical cues in the periodontal ligament (PDL), play a role in relapse following orthodontic tooth movement.
Collapse
Affiliation(s)
- Raquel Veloso Varanda
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands
| | - Jyoti Kumari
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands; Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, the Netherlands
| | - René E M van Rheden
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands
| | - Vincent M J I Cuijpers
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands
| | - Marjon Bloemen
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands
| | - Fleur Göllesch
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands
| | - Johannes W Von den Hoff
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands
| | - Sjoerd Henneman
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands
| | - Rui Xie
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands
| | - Frank A D T G Wagener
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands.
| | - C Maarten Suttorp
- Department of Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud university medical center, Philips van Leydenlaan 25, Nijmegen 6525 EX, the Netherlands.
| |
Collapse
|
2
|
Thai BS, Chia LY, Nguyen ATN, Qin C, Ritchie RH, Hutchinson DS, Kompa A, White PJ, May LT. Targeting G protein-coupled receptors for heart failure treatment. Br J Pharmacol 2024; 181:2270-2286. [PMID: 37095602 DOI: 10.1111/bph.16099] [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: 10/26/2022] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 04/26/2023] Open
Abstract
Heart failure remains a leading cause of morbidity and mortality worldwide. Current treatment for patients with heart failure include drugs targeting G protein-coupled receptors such as β-adrenoceptor antagonists (β-blockers) and angiotensin II type 1 receptor antagonists (or angiotensin II receptor blockers). However, many patients progress to advanced heart failure with persistent symptoms, despite treatment with available therapeutics that have been shown to reduce mortality and mortality. GPCR targets currently being explored for the development of novel heart failure therapeutics include adenosine receptor, formyl peptide receptor, relaxin/insulin-like family peptide receptor, vasopressin receptor, endothelin receptor and the glucagon-like peptide 1 receptor. Many GPCR drug candidates are limited by insufficient efficacy and/or dose-limiting unwanted effects. Understanding the current challenges hindering successful clinical translation and the potential to overcome existing limitations will facilitate the future development of novel heart failure therapeutics. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Bui San Thai
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ling Yeong Chia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Anh T N Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Chengxue Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Andrew Kompa
- Department Medicine and Radiology, University of Melbourne, St Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
3
|
Wei F, Lin K, Ruan B, Wang C, Yang L, Wang H, Wang Y. Epigallocatechin gallate protects MC3T3-E1 cells from cadmium-induced apoptosis and dysfunction via modulating PI3K/AKT/mTOR and Nrf2/HO-1 pathways. PeerJ 2024; 12:e17488. [PMID: 38827303 PMCID: PMC11141548 DOI: 10.7717/peerj.17488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
Epigallocatechin gallate (EGCG), an active constituent of tea, is recognized for its anticancer and anti-inflammatory properties. However, the specific mechanism by which EGCG protects osteoblasts from cadmium-induced damage remains incompletely understood. Here, the action of EGCG was investigated by exposing MC3T3-E1 osteoblasts to EGCG and CdCl2 and examining their growth, apoptosis, and differentiation. It was found that EGCG promoted the viability of cadmium-exposed MC3T3-E1 cells, mitigated apoptosis, and promoted both maturation and mineralization. Additionally, CdCl2 has been reported to inhibit both the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and nuclear factor erythroid 2-related factor 2/heme oxygenase-1(Nrf2/HO-1) signaling pathways. EGCG treatment attenuated cadmium-induced apoptosis in osteoblasts and restored their function by upregulating both signaling pathways. The findings provide compelling evidence for EGCG's role in attenuating cadmium-induced osteoblast apoptosis and dysfunction through activating the PI3K/AKT/mTOR and Nrf2/HO-1 pathways. This suggests the potential of using EGCG for treating cadmium-induced osteoblast dysfunction.
Collapse
Affiliation(s)
- Fanhao Wei
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Kai Lin
- Nanjing University Medical School, Nanjing, China
| | - Binjia Ruan
- Nanjing University Medical School, Nanjing, China
| | | | - Lixun Yang
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Hongwei Wang
- Nanjing University Medical School, Nanjing, China
| | - Yongxiang Wang
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
| |
Collapse
|
4
|
Zizi V, Becatti M, Bani D, Nistri S. Serelaxin Protects H9c2 Cardiac Myoblasts against Hypoxia and Reoxygenation-Induced Damage through Activation of AMP Kinase/Sirtuin1: Further Insight into the Molecular Mechanisms of the Cardioprotection of This Hormone. Antioxidants (Basel) 2024; 13:163. [PMID: 38397761 PMCID: PMC10886064 DOI: 10.3390/antiox13020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Serelaxin (RLX), namely the human recombinant Relaxin-2 hormone, protects the heart from ischemia/reperfusion (I/R)-induced damage due to its anti-inflammatory, anti-apoptotic and antioxidant properties. RLX acts by binding to its specific RXFP1 receptor whereby it regulates multiple transduction pathways. In this in vitro study, we offer the first evidence for the involvement of the AMP kinase/Sirtuin1 (AMPK/SIRT1) pathway in the protection by RLX against hypoxia/reoxygenation (H/R)-induced damage in H9c2 cells. The treatment of the H/R-exposed cells with RLX (17 nmol L-1) enhanced SIRT1 expression and activity. The inhibition of SIRT1 signaling with EX527 (10 µmol L-1) reduced the beneficial effect of the hormone on mitochondrial efficiency and cell apoptosis. Moreover, RLX upregulated the AMPK pathway, as shown by the increase in the expression of phospho-AMPK-activated protein. Finally, AMPK pathway inhibition by Compound C (10 and 20 μmol L-1) abrogated the increase in SIRT1 expression induced by RLX, thus suggesting the involvement of the AMPK pathway in this effect of RLX. These results strengthen the concept that RLX exerts its cardioprotective effects against H/R-induced injury through multiple pathways which also include AMPK/SIRT1. These new findings support the use of RLX or RLX-derived molecules as a promising therapeutic for those diseases in which I/R and oxidative stress play a pathogenic role.
Collapse
Affiliation(s)
- Virginia Zizi
- Department of Experimental & Clinical Medicine, Research Unit of Histology & Embryology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (V.Z.); (D.B.)
| | - Matteo Becatti
- Department of Experimental & Clinical Biomedical Sciences “Mario Serio”, Section of Biochemical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Florence, Italy;
| | - Daniele Bani
- Department of Experimental & Clinical Medicine, Research Unit of Histology & Embryology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (V.Z.); (D.B.)
| | - Silvia Nistri
- Department of Experimental & Clinical Medicine, Research Unit of Histology & Embryology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy; (V.Z.); (D.B.)
| |
Collapse
|
5
|
Cao HY, Sun SF, Yi C, Yang CY, Chen KL, Zhang XW, Liu YB. Muyocoxanthones O-S: Undescribed xanthones with antioxidative damage bioactivity to cardiomyocytes from the endophytic fungus Muyocopron laterale. PHYTOCHEMISTRY 2023; 209:113625. [PMID: 36858338 DOI: 10.1016/j.phytochem.2023.113625] [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: 11/23/2022] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
The metabolites from the endophytic fungus Muyocopron laterale hosted in the medicinal plant Tylophora ovata were investigated, and five undescribed xanthones, muyocoxanthones O-S, along with seven known compounds were isolated. Their structures were elucidated by HR-ESI-MS, NMR, and ECD calculations. Compounds were evaluated for their anti-cardiomyocyte oxidative damage activity using a model of oxidative damage induced by cell hypoxia incubation. Muyocoxanthones O-Q and blennolide L exhibited moderate activity against oxidative damage to cardiomyocytes with relative viabilities of 62.4, 54.8, 60.3 and 54.9%, respectively.
Collapse
Affiliation(s)
- Hai-Yan Cao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100050, China
| | - Sen-Feng Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Cheng Yi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Chen-Yu Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ke-Liang Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xiao-Wei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yun-Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
6
|
Wu M, Lai H, Peng W, Zhou X, Zhu L, Tu H, Yuan K, Yang Z. Monotropein: A comprehensive review of biosynthesis, physicochemical properties, pharmacokinetics, and pharmacology. Front Pharmacol 2023; 14:1109940. [PMID: 36937894 PMCID: PMC10017856 DOI: 10.3389/fphar.2023.1109940] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Monotropein, a principal natural compound in iridoid glycosides extracted from Morindae officinalis radix, has potent pharmacological activities. To understand and utilize monotropein, we systematically summarized the studies on monotropein, including its biosynthetic pathway, physicochemical properties, pharmacokinetics, and pharmacology. Interestingly, we found that the multiple bioactivities of monotropein, such as anti-osteoporosis, anti-inflammation, anti-oxidation, anti-nociception, and hepatic or renal protection, are closely associated with its capability of downregulating the nuclear factor-κB signaling pathway, inhibiting the mitogen-activated protein kinase signaling pathway, attenuating the activation of nuclear factor E2-related factor 2/heme oxygenase-1 signaling pathway, and regulating the mammalian target of rapamycin/autophagy signaling pathway. However, the clinically therapeutic effects and the potential problems need to be addressed. This review highlights the current research progress on monotropein, which provides a reference for further investigation of monotropein.
Collapse
Affiliation(s)
- Mingquan Wu
- Department of Pharmacy, Sichuan Orthopedic Hospital, Chengdu, Sichuan, China
- *Correspondence: Mingquan Wu, ; Zhirui Yang,
| | - Huabing Lai
- Department of Rehabilitation and Prosthetic Orthopedics Center, Sichuan Orthopedic Hospital, Chengdu, Sichuan, China
| | - Wei Peng
- Department of Pharmacy, Sichuan Orthopedic Hospital, Chengdu, Sichuan, China
| | - Xu Zhou
- Department of Pharmacy, Sichuan Orthopedic Hospital, Chengdu, Sichuan, China
| | - Liyang Zhu
- Department of Pharmacy, Sichuan Orthopedic Hospital, Chengdu, Sichuan, China
| | - He Tu
- Department of Pharmacy, Sichuan Orthopedic Hospital, Chengdu, Sichuan, China
| | - Kezhu Yuan
- Department of Scientific Research, Sichuan Orthopedic Hospital, Chengdu, Sichuan, China
| | - Zhirui Yang
- Department of Nuclear Medicine, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
- *Correspondence: Mingquan Wu, ; Zhirui Yang,
| |
Collapse
|
7
|
Relaxin inhibits 177Lu-EDTMP associated cell death in osteosarcoma cells through notch-1 pathway. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2022; 72:575-585. [PMID: 36651368 DOI: 10.2478/acph-2022-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/20/2022] [Indexed: 01/25/2023]
Abstract
177Lu-EDTMP (Ethylenediamine tetramethylene phosphonic acid) is the most used radioactive agent for pain palliation in bone cancer patients. The present study aims to study the impact of relaxin-2 on the 177Lu-EDTMP associated cell toxicity and death in osteosarcoma cells. MG63 and Saos-2 cells were cultured with 177Lu-EDTMP (37 MBq) for 24 h with and without pretreatment of recombinant relaxin 2 (RLXH2) for 12 and 24 h. 177Lu-EDTMP associated cellular deterioration and death was determined by LDH, MTT, and trypan blue dye assays. ELISA-based kit was used to determine apoptotic DNA fragmentation. Western blotting was used to determine expression levels of apoptotic-related signalling pathway proteins like bcl2, poly(ADP-ribose) polymerase (PARP), and MAPK (mitogen-activated protein kinase). Our results found that RLXH2 counters 177Lu-EDTMP associated cellular toxicity. Similarly, RLXH2 was able to counter 177Lu-EDTMP induced cell death in a concentration and time--dependent manner. Furthermore, it was found that RLXH2 treatment prevents apoptosis in 177Lu-EDTMP challenged cells through activation of the notch-1 pathway in a concentration- and time-dependent manner. We reported that RLXH2 significantly declined cellular toxicity and apoptosis associated with 177Lu-EDTMP in MG63 and Saos-2 cells through the notch-1 pathway.
Collapse
|
8
|
Wang F, Gong Y, Chen T, Li B, Zhang W, Yin L, Zhao H, Tang Y, Wang X, Huang C. Maresin1 ameliorates ventricular remodelling and arrhythmia in mice models of myocardial infarction via NRF2/HO-1 and TLR4/NF-kB signalling. Int Immunopharmacol 2022; 113:109369. [DOI: 10.1016/j.intimp.2022.109369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/04/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
|
9
|
Qian J, Wan W, Fan M. HMOX1 silencing prevents doxorubicin-induced cardiomyocyte injury, mitochondrial dysfunction, and ferroptosis by downregulating CTGF. Gen Thorac Cardiovasc Surg 2022; 71:280-290. [PMID: 36008747 DOI: 10.1007/s11748-022-01867-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/14/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Doxorubicin is a type of effective antitumor drug but can contribute to cardiomyocyte injuries. We aimed to dissect the mechanism of the HMOX1/CTGF axis in DOX-induced cardiomyocyte injury, mitochondrial dysfunction, and ferroptosis. METHODS Bioinformatics analysis was conducted to retrieve differentially expressed genes in a DOX-induced mouse model. Mouse cardiomyocytes, HL-1 cells, were induced with l µM DOX, after which gain- or loss-of-function assays were applied. CCK-8, fluorescent probe assay, flow cytometry, and corresponding kits were employed to detect cell viability, ROS levels, mitochondrial membrane potential and cell apoptosis, and GSH and Fe2+ contents, respectively. qRT-PCR or Western blot assay was adopted to test HMOX1, CTGF, BCL-2, Caspase3, Cleaved-Caspase3, and GPX4 expression. RESULTS Bioinformatics analysis showed that HMOX1 and CTGF were highly expressed in DOX-induced mice and correlated with each other. Also, HMOX1 and CTGF expression was high in HL-1 cells after DOX treatment, along with an obvious decrease in cell viability and GSH and GPX4 expression, an increase in ROS levels, apoptosis, and Fe2+ contents, and mitochondrial membrane potential dysfunction or loss. HMOX1 or CTGF silencing diminished cell apoptosis, Cleaved-Caspase3 expression, Fe2+ contents, and ROS levels, enhanced cell viability and the expression of GSH, GPX4, and BCL-2, and recovered mitochondrial membrane potential in DOX-induced HL-1 cells. Nevertheless, the effects of HMOX1 silencing on the viability, apoptosis, ferroptosis, and mitochondrial dysfunction of DOX-induced HL-1 cells were counteracted by CTGF overexpression. CONCLUSIONS In conclusion, HMOX1 silencing decreased CTGF expression to alleviate DOX-induced injury, mitochondrial dysfunction, and ferroptosis of mouse cardiomyocytes.
Collapse
Affiliation(s)
- Jia Qian
- Department of Heart Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No. 110, Ganhe Road, Hongkou District, Shanghai, 200437, People's Republic of China
| | - Wenting Wan
- Department of Heart Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No. 110, Ganhe Road, Hongkou District, Shanghai, 200437, People's Republic of China
| | - Min Fan
- Department of Heart Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No. 110, Ganhe Road, Hongkou District, Shanghai, 200437, People's Republic of China.
| |
Collapse
|
10
|
Chunduri P, Patel SA, Levick SP. Relaxin/serelaxin for cardiac dysfunction and heart failure in hypertension. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:183-211. [PMID: 35659372 DOI: 10.1016/bs.apha.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The pregnancy related hormone relaxin is produced throughout the reproductive system. However, relaxin also has important cardiovascular effects as part of the adaptation that the cardiovascular system undergoes in response to the extra demands of pregnancy. These effects are primarily mediated by the relaxin family peptide receptor 1, which is one of four known relaxin receptors. The effects of relaxin on the cardiovascular system during pregnancy, as well as its anti-fibrotic and anti-inflammatory properties, have led to extensive studies into the potential of relaxin therapy as an approach to treat heart failure. Cardiomyocytes, cardiac fibroblasts, and endothelial cells all possess relaxin family peptide receptor 1, allowing for direct effects of therapeutic relaxin on the heart. Many pre-clinical animal studies have demonstrated a beneficial effect of exogenous relaxin on adverse cardiac remodeling including inflammation, fibrosis, cardiomyocyte hypertrophy and apoptosis, as well as effects on cardiac contractile function. Despite this, clinical studies have yielded disappointing results for the synthetic seralaxin, even though seralaxin was well tolerated. This article will provide background on relaxin in the context of normal physiology, as well as the role of relaxin in pregnancy-related adaptations of the cardiovascular system. We will also present evidence from pre-clinical animal studies that demonstrate the potential benefits of relaxin therapy, as well as discussing the results from clinical trials. Finally, we will discuss possible reasons for the failure of these clinical trials as well as steps being taken to potentially improve relaxin therapy for heart failure.
Collapse
Affiliation(s)
- Prasad Chunduri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Shrey A Patel
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Scott P Levick
- Physiology and Pharmacology, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
11
|
Wang L, Liu Y, Zhang X, Ye Y, Xiong X, Zhang S, Gu L, Jian Z, Wang H. Endoplasmic Reticulum Stress and the Unfolded Protein Response in Cerebral Ischemia/Reperfusion Injury. Front Cell Neurosci 2022; 16:864426. [PMID: 35602556 PMCID: PMC9114642 DOI: 10.3389/fncel.2022.864426] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is an acute cerebrovascular disease characterized by sudden interruption of blood flow in a certain part of the brain, leading to serious disability and death. At present, treatment methods for ischemic stroke are limited to thrombolysis or thrombus removal, but the treatment window is very narrow. However, recovery of cerebral blood circulation further causes cerebral ischemia/reperfusion injury (CIRI). The endoplasmic reticulum (ER) plays an important role in protein secretion, membrane protein folding, transportation, and maintenance of intracellular calcium homeostasis. Endoplasmic reticulum stress (ERS) plays a crucial role in cerebral ischemia pathophysiology. Mild ERS helps improve cell tolerance and restore cell homeostasis; however, excessive or long-term ERS causes apoptotic pathway activation. Specifically, the protein kinase R-like endoplasmic reticulum kinase (PERK), activating transcription factor 6 (ATF6), and inositol-requiring enzyme 1 (IRE1) pathways are significantly activated following initiation of the unfolded protein response (UPR). CIRI-induced apoptosis leads to nerve cell death, which ultimately aggravates neurological deficits in patients. Therefore, it is necessary and important to comprehensively explore the mechanism of ERS in CIRI to identify methods for preserving brain cells and neuronal function after ischemia.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xu Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingze Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shudi Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
- Zhihong Jian,
| | - Hongfa Wang
- Rehabilitation Medicine Center, Department of Anesthesiology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Hongfa Wang,
| |
Collapse
|
12
|
6-Gingerol exerts a protective effect against hypoxic injury through the p38/Nrf2/HO-1 and p38/NF-κB pathway in H9c2 cells. J Nutr Biochem 2022; 104:108975. [PMID: 35245652 DOI: 10.1016/j.jnutbio.2022.108975] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 10/02/2021] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
Abstract
Ginger, one of the most widely consumed condiment for various foods and beverages, has many pharmacological effects. 6-gingerol, a naturally occurring phenol, is one of the major pungent constituents of ginger. The purpose of this study was to characterize the effect of 6-gingerol on the p38/Nrf2/HO-1 and p38/NF-κB signaling pathway, as a possible means of combating hypoxia-related oxidative stress. H9c2 cells were chemically induced with CoCl2 to mimic hypoxia-associated cellular damage. Cardiomyocyte injury was assessed by lactate dehydrogenase and creatine kinase. Reactive oxygen species production was assessed by 2',7'-dichlorodihydrofluorescein diacetate. The antioxidative property of 6-gingerol was measured by estimating the activities of superoxide dismutase, catalase, glutathione and glutathione disulfide. Apoptosis was detected by flow cytometry after Annexin V-FITC-propidium iodide double staining. Western blotting was used to evaluate levels of p-p38, p38, cytoplasm p65, nuclear p65, total p65, nuclear Nrf2, total Nrf2, Keap1, HIF-1α, and HO-1. 6-gingerol was able to counter hypoxia-induced cardiomyocyte injury as evidenced by inhibiting the levels of oxidative stress indexes and increasing the percentage of apoptosis. Furthermore, 6-gingerol was able to down-regulate p-p38/p38, nuclear p65, total p65 and Keap1 expression induced by CoCl2 stimulation and increased cytoplasm p65, nuclear Nrf2, total Nrf2, HO-1, and HIF-1α expression. However, treatment with specific Nrf2 inhibitor blunted the activation of Nrf2 signaling and removed the protective effects of 6-gingerol. These experiments provide evidence that 6-gingerol exerts cytoprotective effects, which may be associated with the regulation of oxidative stress and apoptosis, potentially through activating the Nrf2 pathway and inhibiting the p38/NF-κB pathways.
Collapse
|
13
|
Ding H, Chen W, Chen X. Serum miR-96-5p is a novel and non-invasive marker of acute myocardial infarction associated with coronary artery disease. Bioengineered 2022; 13:3930-3943. [PMID: 35109756 PMCID: PMC8973839 DOI: 10.1080/21655979.2022.2031392] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute myocardial infarction (AMI) is a severe cardiovascular disease. AMI associated with coronary artery disease (AMI-CAD) is a subtype of AMI, composed of AMI patients caused by CAD. This study aimed to evaluate the diagnostic value of miR-96-5p in AMI induced by coronary artery disease. Expression of miR-96-5p and BCL2L13 was evaluated by serum samples and cells utilizing Western blot and RT-qPCR assays. The diagnostic value of miR-96-5p in AMI-CAD was analyzed with a receiver operating characteristic (ROC) curves. The correlation between miR-96-5p and BCL2L13 was examined by Spearman's correlation analysis. The level of oxidative stress and apoptosis were estimated via relative commercial kit, flow cytometry apoptosis assay and TUNEL staining assay. Our study discovered that miR-96-5p was down-regulated while BCL2L13 was up-regulated in patients with AMI-CAD. miR-96-5p was a potential diagnostic parameter, which may help distinguish AMI-CAD patients from healthy controls. In vitro experiments, miR-96-5p expression was down regulated while BCL2L13 was up-regulated in hypoxic cardiomyocytes. After confirming the targeted link of miR-96-5p to BCL2L13 using luciferase reporter and RNA pull down assays, we discovered that miR-96-5p overexpression may restore oxidative stress and cell apoptosis induced by hypoxia treatment in H9c2 cells; meanwhile, co-transfection with BCL2L13 overexpressing plasmid might partly countervail the ameliorative effects of miR-96-5p on oxidative stress and apoptosis. Collectively, miR-96-5p may function as a potential diagnostic biomarker for AMI-CAD patients, and the up-regulation of miR-96-5p would ameliorate AMI-associated cardiomyocytes injury by targeting BCL2L13, hence contributing to the clinical treatment of AMI-CAD.
Collapse
Affiliation(s)
- Hui Ding
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Southeast University, Nanjing, People's Republic of China.,Department of Cardiovascular Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Southeast University, Nanjing, People's Republic of China.,Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
14
|
Samuel CS, Bennett RG. Relaxin as an anti-fibrotic treatment: Perspectives, challenges and future directions. Biochem Pharmacol 2021; 197:114884. [PMID: 34968489 DOI: 10.1016/j.bcp.2021.114884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Fibrosis refers to the scarring and hardening of tissues, which results from a failed immune system-coordinated wound healing response to chronic organ injury and which manifests from the aberrant accumulation of various extracellular matrix components (ECM), primarily collagen. Despite being a hallmark of prolonged tissue damage and related dysfunction, and commonly associated with high morbidity and mortality, there are currently no effective cures for its regression. An emerging therapy that meets several criteria of an effective anti-fibrotic treatment, is the recombinant drug-based form of the human hormone, relaxin (also referred to as serelaxin, which is bioactive in several other species). This review outlines the broad anti-fibrotic and related organ-protective roles of relaxin, mainly from studies conducted in preclinical models of ageing and fibrotic disease, including its ability to ameliorate several aspects of fibrosis progression and maturation, from immune cell infiltration, pro-inflammatory and pro-fibrotic cytokine secretion, oxidative stress, organ hypertrophy, cell apoptosis, myofibroblast differentiation and ECM production, to its ability to facilitate established ECM degradation. Studies that have compared and/or combined these therapeutic effects of relaxin with current standard of care medication have also been discussed, along with the main challenges that have hindered the translation of the anti-fibrotic efficacy of relaxin to the clinic. The review then outlines the future directions as to where scientists and several pharmaceutical companies that have recognized the therapeutic potential of relaxin are working towards, to progress its development as a treatment for human patients suffering from various fibrotic diseases.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Robert G Bennett
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; Department of Internal Medicine, Division of Diabetes, Endocrinology & Metabolism, University of Nebraska Medical Center, Omaha, NE 68198-4130, USA.
| |
Collapse
|
15
|
Nesfatin-1 protects H9c2 cardiomyocytes against cobalt chloride-induced hypoxic injury by modulating the MAPK and Notch1 signaling pathways. ACTA ACUST UNITED AC 2021; 28:21. [PMID: 34517917 PMCID: PMC8436528 DOI: 10.1186/s40709-021-00147-4] [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: 11/14/2020] [Accepted: 06/30/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND This study aimed to explore the effect of nesfatin-1 on cobalt chloride (CoCl2)-induced hypoxic injury in cardiomyocyte H9c2 cells. METHODS H9c2 cardiomyocytes were induced by different concentrations of CoCl2 to mimic the hypoxia condition. Cell viability was detected by MTT assay. Cell apoptosis was detected by TUNEL staining and flow cytometry. ROS production was detected using the fluorescence probe DCFH-DA. The mitochondrial membrane potential (MMP) was detected using the TMRE method. The levels of released lactate dehydrogenase (LDH), malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT) were detected using the commercial kits. The protein levels of MAPK signaling members (p-JNK1/2, p-ERK1/2, and p-p38) and Notch1 signaling members (Notch1, Hes 1, and Jagged 1) were detected by Western blot. RESULTS CoCl2 significantly promoted cell apoptosis, increased LDH leakage, MDA concentration, and decreased cell viability, SOD activity, GSH production, and CAT activity. CoCl2-induced hypoxic injury in H9c2 cells was partially restored by nesfatin-1 treatment. Moreover, nesfatin-1 treatment attenuated CoCl2-induced increase in ROS production and mitochondrial dysfunction, decreased mitochondrial membrane potential, Bax/Bcl-2 imbalance, as well as c-caspase-9 and c-caspase-3 levels. Moreover, nesfatin-1 treatment inhibited the activation of MAPK and Notch1 signaling pathways. CONCLUSIONS Nesfatin-1 could effectively protect H9c2 cells against CoCl2-induced hypoxic injury by blocking MAPK and Notch1 signaling pathways, suggesting that nesfatin-1 might be a promising therapeutic agent for hypoxic cardiac injury.
Collapse
|
16
|
Liu XJ, Lv YF, Cui WZ, Li Y, Liu Y, Xue YT, Dong F. Icariin inhibits hypoxia/reoxygenation-induced ferroptosis of cardiomyocytes via regulation of the Nrf2/HO-1 signaling pathway. FEBS Open Bio 2021; 11:2966-2976. [PMID: 34407320 PMCID: PMC8564343 DOI: 10.1002/2211-5463.13276] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/25/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
Myocardial infarction (MI) is caused by the formation of plaques in the arterial walls, leading to a decrease of blood flow to the heart and myocardium injury as a result of hypoxia. Ferroptosis is a crucial event in myocardial injury, and icariin (ICA) exerts protective effects against myocardial injury. Here, we investigated the protective mechanism of ICA in hypoxia/reoxygenation (H/R)-induced ferroptosis of cardiomyocytes. H9C2 cells were subjected to H/R induction. The content of lactate dehydrogenase and the levels of oxidative stress and intracellular ferrous ion Fe2+ were measured. The levels of ferroptosis markers (ACSL4 and GPX4) were detected. H/R-induced H9C2 cells were cultured with ICA in the presence or absence of ferroptosis inducer (erastin). Znpp (an HO-1 inhibitor) was added to ICA-treated H/R cells to verify the role of the Nrf2/HO-1 pathway. H/R-induced H9C2 cells showed reduced viability, enhanced oxidative stress and lactate dehydrogenase content, increased levels of Fe2+ and ACSL4, and decreased levels of GPX4. ICA inhibited H/R-induced ferroptosis and oxidative stress in cardiomyocytes. Erastin treatment reversed the inhibitory effect of ICA on ferroptosis in H/R cells. The expression of Nrf2 and HO-1 in H/R-induced H9C2 cells was reduced, whereas ICA treatment reversed this trend. Inhibition of the Nrf2/HO-1 pathway reversed the protective effect of ICA on H/R-induced ferroptosis. Collectively, our results suggest that ICA attenuates H/R-induced ferroptosis of cardiomyocytes by activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Xiu-Juan Liu
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Yan-Fei Lv
- Department of Rehabilitation Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Wen-Zhu Cui
- Department of cardiovascular diseases, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Yang Liu
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Yi-Tao Xue
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Feng Dong
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| |
Collapse
|
17
|
Yu J, Li W, Xiao X, Huang Q, Yu J, Yang Y, Han T, Zhang D, Niu X. (-)-Epicatechin gallate blocks the development of atherosclerosis by regulating oxidative stress in vivo and in vitro. Food Funct 2021; 12:8715-8727. [PMID: 34365492 DOI: 10.1039/d1fo00846c] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
(-)-Epicatechin gallate (ECG), as a compound in green tea extract polyphenols, has specific therapeutic effects against oxidative stress. Oxidative stress exists throughout the pathological development of atherosclerosis. In this study, two atherosclerosis models, oxidized low-density lipoprotein (ox-LDL)-induced vascular smooth muscle cells (VSMCs) and high fat diet (HFD)-induced ApoE-/- mice, were applied to explore the mechanism of ECG intervention on AS. In vivo and in vitro studies showed that ECG reduced the level of MDA and increased the activity of SOD, which are oxidative stress factors. ECG also improved HFD-induced disorder of lipid factor expression in the serum of ApoE-/- mice and alleviated oxidative stress by enhancing the antioxidant activity. The potential mechanism was supposed to be the inhibition of the phosphorylation of p65 by ECG in the NF-κB pathway in the aorta, thereby blocking the expression of inflammatory mediators. In addition, ECG increased the stability of atherosclerosis plaques by reducing the expression of MMP-2 and ICAM-1 in atherosclerosis diseased tissues. ECG reduced lipid accumulation in the aorta and its roots and also plaque neoplasia. Western blotting experiments indicated that ECG increased the nuclear transfer of Nrf2 and the expression of heme oxygenase 1 (HO-1) was increased. These results demonstrated that ECG significantly reduced the formation of aortic plaque in ApoE-/- mice which was possibly triggered by the inhibition of hyperlipidemia and oxidative stress that exhibited the anti-atherosclerotic potential.
Collapse
Affiliation(s)
- Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Xin Xiao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, P.R. China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, P.R. China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| |
Collapse
|
18
|
Bian R, Gong J, Li J, Li P. Relaxin increased blood pressure and sympathetic activity in paraventricular nucleus of hypertensive rats via enhancing oxidative stress. Peptides 2021; 141:170550. [PMID: 33839220 DOI: 10.1016/j.peptides.2021.170550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 02/08/2023]
Abstract
Relaxin, an ovarian polypeptide hormone, is found in the hypothalamic paraventricular nucleus (PVN) which is an important central integrative site for the control of blood pressure and sympathetic outflow. The aim of this study was to determine if superoxide anions modulate the effects of relaxin in the PVN. Experiments were performed in normotensive Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs). Relaxin mRNA and protein, and its receptor, relaxin family peptide receptor 1 (RXFP1) levels in PVN were 3.24, 3.17, and 3.64 times higher in SHRs than in WKY rats, respectively. Microinjection of relaxin-2 into the PVN dose-dependently increased mean arterial pressure (MAP), renal sympathetic nerve activity (RSNA) and heart rate (HR) in both WKY rats and SHRs, although the effects on MAP (16.87 ± 1.99 vs. 8.97 ± 1.48 mm Hg in 100 nmol), RSNA (22.60 ± 2.15 vs. 11.77 ± 1.43 % in 100 nmol) and HR (22.85 ± 3.13 vs. 12.62 ± 2.83 beats/min in 100 nmol) were greater in SHRs. Oxidative stress level was enhanced after relaxin-2 microinjection into the PVN. Pretreatment with superoxide anion scavengers or NADPH oxidase inhibitor blocked, and superoxide dismutase inhibitor potentiated the effects of relaxin-2 on MAP, RSNA and HR. RXFP1 knockdown significantly attenuated the blood pressure of SHRs, and inhibited the increases of atrial natriuretic peptide, brain natriuretic peptide, collagen I, collagen III and fibronectin in the heart of SHRs. These results demonstrated that relaxin is expressed in the PVN, and contributes to hypertension and sympathetic overdrive via oxidative stress. Down-regulation of RXFP1 in the PVN could attenuate hypertension and cardiac remodeling.
Collapse
Affiliation(s)
- Rong Bian
- Center of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juexiao Gong
- Department of Cardiology, the Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianan Li
- Center of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
19
|
Aloin antagonizes stimulated ischemia/reperfusion-induced damage and inflammatory response in cardiomyocytes by activating the Nrf2/HO-1 defense pathway. Cell Tissue Res 2021; 384:735-744. [PMID: 33502605 DOI: 10.1007/s00441-020-03345-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022]
Abstract
Myocardial ischemia/reperfusion injury (I/RI) frequently incurs in acute myocardial infarction with high morbidity and mortality worldwide and is characterized with cardiomyocyte apoptosis and inflammatory response. Aloin is a major anthraquinone from Aloe species and fulfills pleiotropic protective functions in several disease models including hepatic injury. Nevertheless, the potential of aloin in MI/RI remains elusive. Intriguingly, aloin had modest cytotoxicity in H9c2 cardiomyocytes. Importantly, aloin dose-dependently ameliorated cell viability that was inhibited in response to simulated ischemia/reperfusion (SI/R) stimulation. Moreover, the enhanced apoptosis in cells under SI/R conditions were reduced after aloin treatment, concomitant with the decrease in pro-apoptotic Bax protein levels and increase in anti-apoptotic Bcl-2 protein expression. Of interest, aloin administration attenuated SI/R-induced oxidant stress by decreasing reactive oxygen species (ROS) production, lactate dehydrogenase (LDH), and malondialdehyde (MDA) release and increasing activity of anti-oxidant stress enzyme superoxide dismutase (SOD). Additionally, the elevated pro-inflammatory cytokine levels were counteracted after aloin treatment in cells under SI/R conditions, including TNF-α, IL-6, and IL-1β. Mechanically, aloin further enforced the activation of the NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling. Noticeably, blockage of this pathway by si-Nrf2 transfection blunted aloin-mediated cardioprotective efficacy against SI/R-evoked oxidative stress injury and inflammatory response. Thus, these findings corroborate that aloin may antagonize SI/R-induced cardiomyocyte injury by attenuating excessive oxidative stress and inflammation, thereby endorsing its potential as a promising therapeutic agent against myocardial infarction.
Collapse
|
20
|
Abstract
Heart failure is a worldwide pandemic influencing 26 million individuals worldwide and is expanding. Imbalanced redox homeostasis in cardiac cells alters the structure and function of the cells, which leads to contractile dysfunction, myocardial hypertrophy, and fibrosis in chronic heart failure. Various targets and agents acting on these such as siRNA, miRNA, interleukin-1, opioids, vasodilators, and SGLT2 inhibitors are being evaluated for heart failure, and nuclear factor erythroid 2-related factor 2 (NRF2) is one of them. NRF2 is a master transcription factor which is expressed in most of the tissues and exhibits a major role in amplification of the antioxidant pathways associated with the enzymes present in myocardium. Increased ROS generation and PI3K-Akt signaling can activate the receptor NRF2. Various in vitro and in vivo and few clinical studies suggested NRF2 may possess a potential for targeting oxidative stress-induced cardiovascular diseases including heart failures. All these studies collectively propose that upregulation of NRF2 will attenuate the increase in hemodynamic stress and provide beneficial role in cardiovascular diseases. The current review shall familiarize readers about the regulations and functions of NRF2. We have also discussed the current evidences suggesting beneficial role of NRF2 activators in heart failure. Graphical abstract.
Collapse
|
21
|
Nistri S, Fiorillo C, Becatti M, Bani D. Human Relaxin-2 (Serelaxin) Attenuates Oxidative Stress in Cardiac Muscle Cells Exposed In Vitro to Hypoxia-Reoxygenation. Evidence for the Involvement of Reduced Glutathione Up-Regulation. Antioxidants (Basel) 2020; 9:antiox9090774. [PMID: 32825567 PMCID: PMC7555919 DOI: 10.3390/antiox9090774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023] Open
Abstract
Serelaxin (RLX) designates the pharmaceutical form of the human natural hormone relaxin-2 that has been shown to markedly reduce tissue and cell damage induced by hypoxia and reoxygenation (HR). The evidence that RLX exerts similar protective effects on different organs and cells at relatively low, nanomolar concentrations suggests that it specifically targets a common pathogenic mechanism of HR-induced damage, namely oxidative stress. In this study we offer experimental evidence that RLX (17 nmol L-1), added to the medium of HR-exposed H9c2 rat cardiac muscle cells, significantly reduces cell oxidative damage, mitochondrial dysfunction and apoptosis. These effects appear to rely on the up-regulation of the cellular availability of reduced glutathione (GSH), a ubiquitous endogenous antioxidant metabolite. Conversely, superoxide dismutase activity was not influenced by RLX, which, however, was not endowed with chemical antioxidant properties. Taken together, these findings verify the major pharmacological role of RLX in the protection against HR-induced oxidative stress, and shed first light on its mechanisms of action.
Collapse
Affiliation(s)
- Silvia Nistri
- Department of Experimental & Clinical Medicine, Research Unit of Histology & Embryology, University of Florence, viale G. Pieraccini 6, 50139 Florence, Italy;
| | - Claudia Fiorillo
- Department of, Experimental & Clinical Biomedical Sciences “Mario Serio”, Section of Biochemical Sciences, University of Florence, viale G.B. Morgagni 50, 50134 Florence, Italy;
| | - Matteo Becatti
- Department of, Experimental & Clinical Biomedical Sciences “Mario Serio”, Section of Biochemical Sciences, University of Florence, viale G.B. Morgagni 50, 50134 Florence, Italy;
- Correspondence: (M.B.); (D.B.); Tel.: +39-055-2751-261 (M.B.); +39-055-2758-153 (D.B.)
| | - Daniele Bani
- Department of Experimental & Clinical Medicine, Research Unit of Histology & Embryology, University of Florence, viale G. Pieraccini 6, 50139 Florence, Italy;
- Correspondence: (M.B.); (D.B.); Tel.: +39-055-2751-261 (M.B.); +39-055-2758-153 (D.B.)
| |
Collapse
|
22
|
Li B, Nasser M, Masood M, Adlat S, Huang Y, Yang B, Luo C, Jiang N. Efficiency of Traditional Chinese medicine targeting the Nrf2/HO-1 signaling pathway. Biomed Pharmacother 2020; 126:110074. [DOI: 10.1016/j.biopha.2020.110074] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 02/09/2023] Open
|
23
|
Yang R, Song C, Chen J, Zhou L, Jiang X, Cao X, Sun Y, Zhang Q. Limonin ameliorates acetaminophen-induced hepatotoxicity by activating Nrf2 antioxidative pathway and inhibiting NF-κB inflammatory response via upregulating Sirt1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 69:153211. [PMID: 32259676 DOI: 10.1016/j.phymed.2020.153211] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/06/2020] [Accepted: 03/19/2020] [Indexed: 05/21/2023]
Abstract
BACKGROUND Limonin, a bioactive compound from citrus plants, exerts antioxidant activities, however its therapeutic potential in acetaminophen (APAP)-induced hepatotoxicity remains unclear. PURPOSE Our study aims to investigate the protective effect of limonin on APAP-induced hepatotoxicity and illuminate the underlying mechanisms. STUDY design In vitro, we chose L-02 cells to establish in vitro APAP-induced liver injury model. L-02 cells were treated with APAP (7.5 mM) for 24 h after pre-incubation with limonin (10, 25, 50 μM) or NAC (250 μM) for 2 h. In vivo, we used C57BL/6 mice as an in vivo APAP-induced liver injury model. C57BL/6 mice with pre-treatment of limonin (40, 80 mg/kg) or NAC (150 mg/kg) for 1 h, were given with a single dose of APAP (300 mg/kg). METHODS After pre-incubation with limonin (10, 25, 50 μM) for 2 h, L-02 cells were treated with APAP (7.5 mM) for 24 h.The experiments in vitro included MTT assay, Annexin V/PI staining, measurement of reactive oxygen species (ROS), quantitative real-time PCR analysis, Western blot analysis, immunofluorescence microscopy and analysis of LDH activity. Transfection of Nrf2 or Sirt1 siRNA was also conducted in vitro. In vivo, C57BL/6 mice with pre-treatment of limonin (40, 80 mg/kg) or NAC (150 mg/kg) for 1 h, were given with a single dose of APAP (300 mg/kg). Mice were sacrificed at 4, 12 h after APAP poisoning, and analysis of ALT and AST in serum, GSH level in liver tissues, liver histological observation and immunohistochemistry were performed. RESULTS Limonin increased the cell viability and alleviated APAP-induced apoptosis in hepatocytes. Limonin also inhibited APAP-induced mitochondrial-mediated apoptosis by decreasing the ratio of Bax/Bcl-2, recovery of mitochondrial membrane potential (MMP), inhibiting ROS production and cleavage of caspase-3 in L-02 cells. Moreover, limonin induced activation of Nrf2 and increased protein expression and mRNA levels of its downstream targets, including HO-1, NQO1 and GCLC/GCLM. The inhibition of limonin on apoptosis and promotion on Nrf2 antioxidative pathway were lessened after the application of Nrf2 siRNA. In addition, limonin inhibited NF-κB transcriptional activation, NF-κB-regulated genes and protein expression of inflammatory related proteins iNOS and COX2. Furthermore, limonin increased the protein expression of Sirt1. Sirt1 siRNA transfection confirmed that limonin activated Nrf2 antioxidative pathway and inhibited NF-κB inflammatory response by upregulating Sirt1. Finally, we established APAP-induced liver injury in vivo and demonstrated that limonin alleviated APAP-induced hepatotoxicity by activating Nrf2 antioxidative signals and inhibiting NF-κB inflammatory response via upregulating Sirt1. CONCLUSION In summary, this study documented that limonin mitigated APAP-induced hepatotoxicity by activating Nrf2 antioxidative pathway and inhibiting NF-κB inflammatory response via upregulating Sirt1, and demonstrated that limonin had therapeutic promise in APAP-induced liver injury.
Collapse
Affiliation(s)
- Runyu Yang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Changqin Song
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Jiaxi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Lvqi Zhou
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Xiubo Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Xiaomei Cao
- Department of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yang Sun
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China; Department of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China; Department of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| |
Collapse
|
24
|
Lu D, Zhang Y, Xue W, Sun J, Yang C, Lin C, Li Y, Liu T. Shenxiong Glucose Injection Protects H9c2 Cells From CoCl 2-Induced Oxidative Damage via Antioxidant and Antiapoptotic Pathways. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20920054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular disease has become one of the main diseases that endanger humans, and oxidative damage plays an important role in this. Shenxiong glucose injection (SGI) is a common clinical treatment in China for the treatment of this condition. To understand further the protective effects and related mechanisms of SGI on cardiovascular diseases, H9c2 cells were treated with SGI at different concentrations (0.5%, 1%, 2% [v/v]) before hypoxic damage was induced by treatment with CoCl2). In CoCl2-induced H9c2 cells, SGI treatment increased cell viability and the activity of superoxide dismutase, glutathione peroxidase, catalase, elevated mitochondrial membrane potential, and decreased the rate of cellular apoptosis, lactic dehydrogenase release, and the content of malondialdehyde and reactive oxygen species, while also upregulating Bcl-2 expression and downregulating Bax, Cyt-c, and cleaved caspase-3 expression. Together, these results suggested that SGI has a protective effect on CoCl2-induced damage, and its mechanism may be related to increased antioxidant and antiapoptosis capacity in H9c2 cells. This study provides the basis for further research and potential practical applications of SGI.
Collapse
Affiliation(s)
- Dingyan Lu
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
- School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Yubin Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
- School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Weina Xue
- School of Medicine and Health Management, Guizhou Medical University, Guiyang, Guizhou, P.R. China
| | - Jia Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Chang Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Changhu Lin
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Yongjun Li
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| |
Collapse
|
25
|
Silencing of TXNIP Alleviated Oxidative Stress Injury by Regulating MAPK-Nrf2 Axis in Ischemic Stroke. Neurochem Res 2019; 45:428-436. [PMID: 31858374 DOI: 10.1007/s11064-019-02933-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/30/2019] [Accepted: 12/13/2019] [Indexed: 12/21/2022]
Abstract
Ischemic stroke is a life-threatening cerebrovascular thrombotic disease, oxidative stress is considered to be a critical factor to stroke pathophysiology. This study aimed to investigate the underlying molecular mechanism and propose the potential therapeutic strategy for ischemic stroke. Bioinformatics analysis based on a public microarray profile (GSE 61616) of ischemic stroke rats was performed as a pilot research. Oxidative stress was enriched as a significantly gene ontology item, and thioredoxin-interacting protein (TXNIP) and MAPK signaling were identified as the hub gene and pathway, respectively. The experiments in middle cerebral artery occlusion rats demonstrated that ischemia induced the activation of oxidative stress. The expressions of TXNIP, p-p38, p-JNK, p-ERK were significantly increased while Nrf2 and HO-1 expressions were decreased after stroke. Rescue assays were conducted in primary cultured neurons to explore the accurate interrelations among these factors. The results indicated that MAPK specific inhibitor and siRNA-TXNIP significantly alleviated the oxidative stress injury induced by oxygen-glucose deprivation. In addition, knocking down of TXNIP inhibited the activation of MAPK pathway and promoted Nrf2 pathway. Taken together, these findings indicated that TXNIP aggravated the oxidative stress injury by regulating MAPK-Nrf2 axis in ischemic stroke. Silencing of TXNIP seems a promising therapeutic strategy to alleviate ischemic stroke.
Collapse
|
26
|
Xu Y, Guan J, Xu J, Chen S, Sun G. Z-Guggulsterone attenuates glucocorticoid-induced osteoporosis through activation of Nrf2/HO-1 signaling. Life Sci 2019; 224:58-66. [DOI: 10.1016/j.lfs.2019.03.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/13/2022]
|