1
|
Wei H, Guo X, Yan J, Tian X, Yang W, Cui K, Wang L, Guo B. Neuregulin-4 alleviates isoproterenol (ISO)-induced cardial remodeling by inhibiting inflammation and apoptosis via AMPK/NF-κB pathway. Int Immunopharmacol 2024; 143:113301. [PMID: 39418729 DOI: 10.1016/j.intimp.2024.113301] [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: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
Cardiac remodeling refers to the abnormal changes in cardiac structure and function caused by various pathological conditions. It is an inevitable pathological process in the occurrence and development of heart failure and is related to a variety of cardiovascular diseases. Inflammation and apoptosis are critical pathological processes involved in cardiac remodeling. Neuregulin 4 (Nrg 4) is an adipokine produced primarily by brown adipose tissue that may play a protective role in a variety of inflammatory diseases. The aim of this study was to investigate whether Nrg4 can delay the progression of cardiac remodeling by regulating AMPK/NF-κB pathway, inhibiting inflammation and apoptosis. In our study, we established a model of cardiac remodeling in mice after 14 days of isoproterenol (ISO) intervention, and then gave Nrg4 treatment for another 4 weeks. The cardiac function, the degree of myocardial hypertrophy and myocardial fibrosis of the mice were observed. At the same time, the levels of apoptosis-related proteins (Bax,Bcl-2,Caspase-3), IL-6,IL-Iβ and TNF-α, as well as the activation level of AMPK/NF-κB signaling pathway were evaluated.Nrg4 alleviated ISO-induced cardiac dysfunction, cardiac hypertrophy and fibrosis in mice. Nrg4 also attenuated ISO-induced apoptosis and reduces levels of inflammatory factors to protect ISO-induced myocardial damage. At the same time, the effect of Nrg4 on AMPK/NF-κB pathway was measured in vivo and in vitro. The administration of an AMPK inhibitor was found to reverse the anti-hypertrophy, anti-inflammatory, and anti-apoptotic effects of Nrg4. Our findings suggest that Nrg4 may play a protective role in cardiac remodeling by inhibiting inflammation and apoptosis via AMPK/NF-κB pathway.
Collapse
Affiliation(s)
- Huiqing Wei
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Xiaohua Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Jie Yan
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Xiaochao Tian
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Wenhui Yang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Kun Cui
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Lijie Wang
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China
| | - Bingyan Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Heping West Road No. 215, Shijiazhuang 050000, China; Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
2
|
Asiwe JN, Ajayi AM, Ben-Azu B, Fasanmade AA. Vincristine attenuates isoprenaline-induced cardiac hypertrophy in male Wistar rats via suppression of ROS/NO/NF-қB signalling pathways. Microvasc Res 2024; 155:104710. [PMID: 38880384 DOI: 10.1016/j.mvr.2024.104710] [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: 04/19/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Vincristine (VCR), a vinca alkaloid with anti-tumor and anti-oxidant properties, is acclaimed to possess cardioprotective action. However, the molecular mechanism underlying this protective effect remains unknown. This study investigated the effects of VCR on isoprenaline (ISO), a beta-adrenergic receptor agonist, induced cardiac hypertrophy in male Wistar rats. Animals were pre-treated with ISO (1 mg/kg) intraperitoneally for 14 days before VCR (25 μg/kg) intraperitoneal injection from days 1 to 28. Thereafter, mechanical, and electrical activities of the hearts of the rats were measured using a non-invasive blood pressure monitor and an electrocardiograph, respectively. After which, the heart was homogenized, and supernatants were assayed for contractile proteins: endothelin-1, cardiac troponin-1, angiotensin-II, and creatine kinase-MB, with markers of oxidative/nitrergic stress (SOD, CAT, MDA, GSH, and NO), inflammation (TNF-a and IL-6, NF-kB), and caspase-3 indicative of VCR reduced elevated blood pressure and reversed the abnormal electrocardiogram. ISO-induced increased endothelin-1, cardiac troponin-1, angiotensin-II, and creatine phosphokinase-MB, which were reversed by VCR. ISO also increased TNF-α, IL-6, NF-kB expression with increased caspase-3-mediated apoptosis in the heart. However, VCR reduced ISO-induced inflammation and apoptosis, with improved endogenous antioxidant agents (GSH, SOD, CAT) relative to ISO controls. Moreso, VCR, protected against ISO-induced histoarchitectural degeneration of cardiac myofibre. The result of this study revealed that VCR treatment significantly reverses ISO-induced cardiac hypertrophic phenotypes, via mechanisms connected to improved levels of proteins involved in excitation-contraction, and suppression of oxido-inflammatory and apoptotic pathways.
Collapse
Affiliation(s)
- Jerome Ndudi Asiwe
- Department of Physiology, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria; Department of Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria.
| | - Abayomi M Ajayi
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Nigeria
| | - Benneth Ben-Azu
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | | |
Collapse
|
3
|
Liang Q, Liu X, Peng X, Luo T, Su Y, Xu X, Xie H, Gao H, Chen Z, Xie C. Salvianolic acid B in fibrosis treatment: a comprehensive review. Front Pharmacol 2024; 15:1442181. [PMID: 39139645 PMCID: PMC11319160 DOI: 10.3389/fphar.2024.1442181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Fibrosis is a public health issue of great concern characterized by the excessive deposition of extracellular matrix, leading to the destruction of parenchymal tissue and organ dysfunction that places a heavy burden on the global healthcare system due to its high incidence, disability, and mortality. Salvianolic acid B (SalB) has positively affected various human diseases, including fibrosis. In this review, we concentrate on the anti-fibrotic effects of SalB from a molecular perspective while providing information on the safety, adverse effects, and drug interactions of SalB. Additionally, we discuss the innovative SalB formulations, which give some references for further investigation and therapeutic use of SalB's anti-fibrotic qualities. Even with the encouraging preclinical data, additional research is required before relevant clinical trials can be conducted. Therefore, we conclude with recommendations for future studies. It is hoped that this review will provide comprehensive new perspectives on future research and product development related to SalB treatment of fibrosis and promote the efficient development of this field.
Collapse
Affiliation(s)
- Qingzhi Liang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaoqin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xi Peng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ting Luo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi Su
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hongyan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hong Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhengtao Chen
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Fang R, Zhou R, Ju D, Li M, Wang H, Pan L, Wang X, Han M, Yu Y. Zhen-wu-tang protects against myocardial fibrosis by inhibiting M1 macrophage polarization via the TLR4/NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155719. [PMID: 38763013 DOI: 10.1016/j.phymed.2024.155719] [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: 01/20/2024] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Myocardial fibrosis is a risk factor that contributes to the increase in the incidence of cardiovascular disease and death, posing a significant threat to human health. Zhen-wu-tang (ZWT) is a classical Chinese medicinal recipe that has been extensively used to manage cardiovascular disorders throughout history. However, the fundamental processes involved in its effects were not clear. OBJECTIVE This study examined the therapeutic effects of ZWT on myocardial fibrosis induced by isoproterenol (ISO) in mice, the effect of regulation and underlying mechanism on the polarization of M1 macrophage. METHODS In vivo, a myocardial fibrosis mouse model was induced via intraperitoneal infusion of isoproterenol (ISO). ZWT or captopril (CAP) was administered intragastrically for 30 days. Cardiac function was evaluated by electrocardiogram (ECG) and echocardiography. By analysing myocardial fibrosis pathomorphologically and identifying fibrosis-related indicators, the protective effect of the ZWT on the heart was evaluated. A model of macrophage polarization was established in vitro by activating RAW264.7 cells with lipopolysaccharide (LPS). The regulatory effects of ZWT on macrophage polarization and the signalling pathways involved were examined by immunofluorescence staining, Western blotting (WB), quantitative real-time PCR (qRT-PCR) and siRNA transfection. RESULTS ZWT improved cardiac function; reduced fibrotic deposition in cardiac tissues; decreased α-SMA, collagen I, and collagen III levels; and inhibited myocardial fibrosis in mice with ISO-induced myocardial fibrosis. Furthermore, the results showed that ZWT could suppress M1 macrophage polarization by downregulating the expression of CD86 and iNOS in vitro and in vivo. Finally, the results confirmed that ZWT could significantly reduce TLR4/NF-κB signalling pathway activation. CONCLUSION ZWT showed therapeutic effects on ISO-induced myocardial fibrosis mice, and reduced M1 macrophages polarization through inhibiting TLR4/NF-κB pathway, suggesting that ZWT is a promising drug for myocardial fibrosis treatment.
Collapse
Affiliation(s)
- Rong Fang
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712083, PR China
| | - Rui Zhou
- Co-Construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi and Education Ministry, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang 712083, PR China
| | - Di Ju
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712083, PR China
| | - Mi Li
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712083, PR China
| | - Haifang Wang
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712083, PR China
| | - Liangliang Pan
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712083, PR China
| | - Xueqing Wang
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712083, PR China
| | - Man Han
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712083, PR China.
| | - Yuanwang Yu
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712083, PR China.
| |
Collapse
|
5
|
Alhusaini AM, Alghibiwi HK, Sarawi WS, Alsaab JS, Alshehri SM, Alqahtani QH, Alshanwani AR, Aljassas EA, Alsultan EN, Hasan IH. Resveratrol-Based Liposomes Improve Cardiac Remodeling Induced by Isoproterenol Partially by Modulating MEF2, Cytochrome C and S100A1 Expression. Dose Response 2024; 22:15593258241247980. [PMID: 38645382 PMCID: PMC11027597 DOI: 10.1177/15593258241247980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/28/2024] [Indexed: 04/23/2024] Open
Abstract
Isoproterenol (ISO), a chemically synthesized catecholamine, belongs to β-adrenoceptor agonist used to treat bradycardia. The β-adrenergic agonist is an essential regulator of myocardial metabolism and contractility; however, excessive exposure to ISO can initiate oxidative stress and inflammation. This study aims to investigate the molecular mechanisms underlying ISO-induced cardiac remodeling, the protective efficacy of resveratrol (RSVR), and its liposomal formulation (L-RSVR) against such cardiac change. Wistar albino rats were evenly divided into 4 groups. Control group, ISO group received ISO (50 mg/kg, s.c.) twice a week for 2 weeks, and RSVR- and L-RSVR-treated groups in which rats received either RSVR or L-RSVR (20 mg/kg/day, p.o.) along with ISO for 2 weeks. ISO caused a significant elevation of the expression levels of BAX and MEF2 mRNA, S100A1 and cytochrome C proteins, as well as DNA fragmentation in cardiac tissue compared to the control group. Treatment with either RSVR or L-RSVR for 14 days significantly ameliorated the damage induced by ISO, as evidenced by the improvement of all measured parameters. The present study shows that L-RSVR provides better cardio-protection against ISO-induced cardiac injury in rats, most likely through modulation of cardiac S100A1 protein expression and inhibition of inflammation and apoptosis.
Collapse
Affiliation(s)
- Ahlam M. Alhusaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hanan K. Alghibiwi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wedad S. Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Juman S. Alsaab
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Samiyah M. Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Qamraa H. Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Aliah R. Alshanwani
- Department of Physiology, College of Medicine and King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia
| | - Ebtesam A. Aljassas
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ebtesam N. Alsultan
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Iman H. Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Brockmueller A, Sajeev A, Koklesova L, Samuel SM, Kubatka P, Büsselberg D, Kunnumakkara AB, Shakibaei M. Resveratrol as sensitizer in colorectal cancer plasticity. Cancer Metastasis Rev 2024; 43:55-85. [PMID: 37507626 PMCID: PMC11016130 DOI: 10.1007/s10555-023-10126-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
Despite tremendous medical treatment successes, colorectal cancer (CRC) remains a leading cause of cancer deaths worldwide. Chemotherapy as monotherapy can lead to significant side effects and chemoresistance that can be linked to several resistance-activating biological processes, including an increase in inflammation, cellular plasticity, multidrug resistance (MDR), inhibition of the sentinel gene p53, and apoptosis. As a consequence, tumor cells can escape the effectiveness of chemotherapeutic agents. This underscores the need for cross-target therapeutic approaches that are not only pharmacologically safe but also modulate multiple potent signaling pathways and sensitize cancer cells to overcome resistance to standard drugs. In recent years, scientists have been searching for natural compounds that can be used as chemosensitizers in addition to conventional medications for the synergistic treatment of CRC. Resveratrol, a natural polyphenolic phytoalexin found in various fruits and vegetables such as peanuts, berries, and red grapes, is one of the most effective natural chemopreventive agents. Abundant in vitro and in vivo studies have shown that resveratrol, in interaction with standard drugs, is an effective chemosensitizer for CRC cells to chemotherapeutic agents and thus prevents drug resistance by modulating multiple pathways, including transcription factors, epithelial-to-mesenchymal transition-plasticity, proliferation, metastasis, angiogenesis, cell cycle, and apoptosis. The ability of resveratrol to modify multiple subcellular pathways that may suppress cancer cell plasticity and reversal of chemoresistance are critical parameters for understanding its anti-cancer effects. In this review, we focus on the chemosensitizing properties of resveratrol in CRC and, thus, its potential importance as an additive to ongoing treatments.
Collapse
Affiliation(s)
- Aranka Brockmueller
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstr. 11, D-80336, Munich, Germany
| | - Anjana Sajeev
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, 781039, India
| | - Lenka Koklesova
- Clinic of Gynecology and Obstetrics, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 03601, Martin, Slovakia
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar (Medbay), Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 03601, Martin, Slovakia
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar (Medbay), Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, 781039, India
| | - Mehdi Shakibaei
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstr. 11, D-80336, Munich, Germany.
| |
Collapse
|
7
|
Duraisamy P, Angusamy A, Ravi S, Krishnan M, Martin LC, Manikandan B, Sundaram J, Ramar M. Phytol from Scoparia dulcis prevents NF-κB-mediated inflammatory responses during macrophage polarization. 3 Biotech 2024; 14:80. [PMID: 38375513 PMCID: PMC10874368 DOI: 10.1007/s13205-024-03924-9] [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: 09/28/2023] [Accepted: 01/07/2024] [Indexed: 02/21/2024] Open
Abstract
Macrophages are primary immune cells that mediate a wide range of inflammatory diseases through their polarization potential. In this study, phytol isolated from Scoparia dulcis has been explored against 7-ketocholesterol and bacterial lipopolysaccharide-induced macrophage polarization in IC-21 cells. Isolated phytol has been characterized using GC-MS, TLC, HPTLC, FTIR, 1H-NMR, and HPLC analyses. The immunomodulatory effects of viable concentrations of phytol were tested on oxidative stress, arginase activity, nuclear and mitochondrial membrane potentials in IC-21 cells in addition to the modulation of calcium and lipids. Further, gene and protein expression of atherogenic markers were studied. Results showed that the isolated phytol at a viable concentration of 400 µg/ml effectively reduced the production of nitric oxide, superoxide anion (ROS generation), calcium and lipid accumulation, stabilized nuclear and mitochondrial membranes, and increased arginase activity. The atherogenic markers including iNOS, COX-2, IL-6, IL-1β, MMP-9, CD36, and NF-κB were significantly downregulated at the levels of gene and protein expression, while macrophage surface and nuclear receptor markers (CD206, CD163, and PPAR-γ) were significantly upregulated by phytol pre-treatment in macrophages. Therefore, the present pharmacognostic study supports the role of phytol isolated from Scoparia dulcis in preventing M2-M1 macrophage polarization under inflammatory conditions, making it a promising compound. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-03924-9.
Collapse
Affiliation(s)
| | - Annapoorani Angusamy
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| | - Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| | | | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni’s College for Women, Chennai, 600015 India
| | - Janarthanan Sundaram
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600025 India
| |
Collapse
|
8
|
Liu C, Cheng T, Wang Y, Li G, Wang Y, Tian W, Feng L, Zhang S, Xu Y, Gao Y, Li J, Liu J, Cui J, Yan J, Cao L, Pan Z, Qi Z, Yang L. Syringaresinol Alleviates Early Diabetic Retinopathy by Downregulating HIF-1α/VEGF via Activating Nrf2 Antioxidant Pathway. Mol Nutr Food Res 2024; 68:e2200771. [PMID: 38356045 DOI: 10.1002/mnfr.202200771] [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: 11/07/2022] [Revised: 09/10/2023] [Indexed: 02/16/2024]
Abstract
SCOPE Early diabetic retinopathy (DR) is characterized by chronic inflammation, excessive oxidative stress, and retinal microvascular damage. Syringaresinol (SYR), as a natural polyphenolic compound, has been proved to inhibit many disease progression due to its antiinflammatory and antioxidant properties. The present study focuses on exploring the effect of SYR on hyperglycemia-induced early DR as well as the underlying mechanisms. METHODS AND RESULTS Wild-type (WT) and nuclear factor erythroid 2-related factor 2 (Nrf2)-knockout C57BL/6 mice of type 1 diabetes and high glucose (HG)-induced RF/6A cells are used as in vivo and in vitro models, respectively. This study finds that SYR protects the retinal structure and function in diabetic mice and reduces the permeability and apoptosis of HG-treated RF/6A cells. Meanwhile, SYR distinctly mitigates inflammation and oxidative stress in vivo and vitro. The retinal microvascular damages are suppressed by SYR via downregulating hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway. Whereas, SYR-provided protective effects are diminished in Nrf2-knockout mice, indicating that SYR improves DR progression by activating Nrf2. Similarly, SYR cannot exert protective effects against HG-induced oxidative stress and endothelial injury in small interfering RNA (siRNA)-Nrf2-transfected RF/6A cells. CONCLUSION In summary, SYR suppresses oxidative stress via activating Nrf2 antioxidant pathway, which ameliorates retinal microvascular damage by downregulating HIF-1α/VEGF, thereby alleviating early DR progression.
Collapse
Affiliation(s)
- Chang Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
- Nankai University Eye Institute, Tianjin, 300071, China
| | - Tianwei Cheng
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Yufei Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Guangru Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Yachen Wang
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology Tianjin Medical University, Tianjin, 300020, China
| | - Wencong Tian
- Department of General Surgery, Tianjin Union Medical Center, Tianjin, 300122, China
| | - Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Shengzheng Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Jianlin Cui
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
| | - Lei Cao
- Department of General Surgery, Tianjin Union Medical Center, Tianjin, 300122, China
| | - Zhongjie Pan
- Tianjin Union Medical Center, Tianjin, 300122, China
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
- Nankai University Eye Institute, Tianjin, 300071, China
- Tianjin Union Medical Center, Tianjin, 300122, China
- Xinjiang Production and Construction Corps Hospital, Xinjiang, 830002, China
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University Tianjin, Tianjin, 300071, China
- Tianjin Union Medical Center, Tianjin, 300122, China
| |
Collapse
|
9
|
Xu WL, Zhou PP, Yu X, Tian T, Bao JJ, Ni CR, Zha M, Wu X, Yu JY. Myricetin induces M2 macrophage polarization to alleviate renal tubulointerstitial fibrosis in diabetic nephropathy via PI3K/Akt pathway. World J Diabetes 2024; 15:105-125. [PMID: 38313853 PMCID: PMC10835493 DOI: 10.4239/wjd.v15.i1.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/28/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Development of end-stage renal disease is predominantly attributed to diabetic nephropathy (DN). Previous studies have indicated that myricetin possesses the potential to mitigate the pathological alterations observed in renal tissue. Nevertheless, the precise molecular mechanism through which myricetin influences the progression of DN remains uncertain. AIM To investigate the effects of myricetin on DN and explore its potential therapeutic mechanism. METHODS Db/db mice were administered myricetin intragastrically on a daily basis at doses of 50 mg/kg or 100 mg/kg for a duration of 12 wk. Subsequently, blood and urine indexes were assessed, along with examination of renal tissue pathology. Kidney morphology and fibrosis were evaluated using various staining techniques including hematoxylin and eosin, periodic acid-Schiff, Masson's trichrome, and Sirius-red. Additionally, high-glucose culturing was conducted on the RAW 264.7 cell line, treated with 25 mM myricetin or co-administered with the PI3K/Akt inhibitor LY294002 for a period of 24 h. In both in vivo and in vitro settings, quantification of inflammation factor levels was conducted using western blotting, real-time qPCR and ELISA. RESULTS In db/db mice, administration of myricetin led to a mitigating effect on DN-induced renal dysfunction and fibrosis. Notably, we observed a significant reduction in expressions of the kidney injury markers kidney injury molecule-1 and neutrophil gelatinase associated lipocalin, along with a decrease in expressions of inflammatory cytokine-related factors. Furthermore, myricetin treatment effectively inhibited the up-regulation of tumor necrosis factor-alpha, interleukin-6, and interluekin-1β induced by high glucose in RAW 264.7 cells. Additionally, myricetin modulated the M1-type polarization of the RAW 264.7 cells. Molecular docking and bioinformatic analyses revealed Akt as the target of myricetin. The protective effect of myricetin was nullified upon blocking the polarization of RAW 264.7 via inhibition of PI3K/Akt activation using LY294002. CONCLUSION This study demonstrated that myricetin effectively mitigates kidney injury in DN mice through the regulation of macrophage polarization via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Wei-Long Xu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Pei-Pei Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Xu Yu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Ting Tian
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Jin-Jing Bao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Chang-Rong Ni
- Department of Pharmacy, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Min Zha
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Xiao Wu
- Department of Pneumology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Jiang-Yi Yu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| |
Collapse
|
10
|
Geng N, Fu J, Lv Z, Li J, Kong Y, Qu L, Guo Z, Zhao J, Zhu L, Wang F, Zhao C, Liu S, Hu Z, Li N. M1 polarization of chicken macrophage HD11 can be activated by duck Tembusu virus via MyD88-NF-κB-mediated signaling pathway. Vet Microbiol 2023; 285:109867. [PMID: 37639898 DOI: 10.1016/j.vetmic.2023.109867] [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: 06/05/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Duck Tembusu virus (DTMUV) has caused significant economic losses to the global duck industry since its outbreak in 2010. The macrophages act as the key immune cell, and its polarization in different functional states is very important for host's immune responses and microbial infections. Avian macrophages are the main target cells of DTMUV, its polarization induced by DTMUV and the underlying mechanisms were explored in this study. Through quantitative real-time PCR, nitrite assay, and flow cytometry analysis, we found that DTMUV caused severe inflammatory responses in chicken macrophage line HD11 by reprogramming the expression of M1- and M2-associated genes, leading to the polarization of HD11 macrophage to M1-type. In term of mechanism, transcriptomics was performed to analyze the M1-type polarization triggered by DTMUV, it was found that most differential genes were implicated in biological processes, and DTMUV infection significantly activated innate immune signaling pathways, including cytokine-cytokine receptor interaction, MAPK signaling pathway. Moreover, transcription factors NF-κB and AP1 also be activated after viral infection. However, further validation analysis by inhibitors and siRNAs of NF-κB and AP1 showed that NF-κB molecule was essential for DTMUV-induced M1 polarization in HD11 cell, but not AP1. Additionally, the inhibiting assays targeting MyD88 and TRIF molecules were conducted to determine their effect on NF-κB and M1-associated genes upregulated by DTMUV. The results showed that although the inhibition of both MyD88 and TRIF significantly downregulated the mRNA level of NF-κB, but the expression of M1-associated genes such as CD86 was lower in MyD88 inhibition group than in the other group, indicating that the role of MyD88 in mediating M1 polarization induced by DTMUV was more important. Overall, these results demonstrated that DTMUV infection induces M1-type polarization in chicken macrophage HD11 through MyD88-NF-κB signaling pathways. This finding will lay the foundation for further study the pathogenesis of DTMUV, and provide new insights into the prevention and control of this disease.
Collapse
Affiliation(s)
- Ningwei Geng
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Ji Fu
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Zehao Lv
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Jing Li
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Yuxin Kong
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Lei Qu
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Zhiyun Guo
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Jun Zhao
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Liya Zhu
- Animal Husbandry and Veterinary Service Centre of Linshu, Linyi, 276700 Shandong Province, China
| | - Feng Wang
- Taian City Research Center of Animal Disease Control and Prevention, 8 Hushan East Road, Taian City, 271000 Shandong Province, China
| | - Cui Zhao
- Taian City Research Center of Animal Disease Control and Prevention, 8 Hushan East Road, Taian City, 271000 Shandong Province, China
| | - Sidang Liu
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China
| | - Zhiyong Hu
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China.
| | - Ning Li
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Road, Taian City, 271018 Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Taian City, 271018 Shandong Province, China.
| |
Collapse
|
11
|
Ravi S, Duraisamy P, Krishnan M, Martin LC, Manikandan B, Ramar M. Sitosterol-rich Digera muricata against 7-ketocholesterol and lipopolysaccharide-mediated atherogenic responses by modulating NF-ΚB/iNOS signalling pathway in macrophages. 3 Biotech 2023; 13:331. [PMID: 37670802 PMCID: PMC10475456 DOI: 10.1007/s13205-023-03741-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023] Open
Abstract
Digera muricata L., commonly known as Tartara, is an edible herb used as traditional medicine in many countries of Africa and Asia. This study aimed to elucidate the effect of a phytosterol-rich extract of D. muricata on 7-ketocholesterol-mediated atherosclerosis in macrophages. The extract was examined by phytochemical analyses, GC-MS, TLC, DPPH scavenging and hRBC membrane stabilization assays. Macrophage polarization was studied with experimental groups framed based on alamar blue cell viability and griess assays. Regulations of arginase enzyme activity, ROS generation, mitochondrial membrane potential, cell membrane integrity, pinocytosis, lipid uptake and peroxidation, as well as, intracellular calcium deposition were determined. In addition, expressions of atherogenic mediators were analysed using PCR, ELISA and immunocytochemistry techniques. Diverse phytochemicals with higher free radical scavenging activity and anti-inflammatory potential have been detected in the D. muricata. Co-treatment with D. muricata markedly reduced the atherogenic responses induced by 7KCh in the presence of LPS such as ROS, especially, NO and O2- along with lipid peroxidation. Furthermore, D. muricata significantly normalized mitochondrial membrane potential, cell membrane integrity, pinocytic activity, intracellular lipid accumulation and calcium deposition. These results provided us with the potentiality of D. muricata in ameliorating atherogenesis. Additionally, it decreased the expression of pro-atherogenic mediators (iNOS, COX-2, MMP9, IL-6, IL-1β, CD36, CD163 and TGFβ1) and increased anti-atherogenic mediators (MRC1 and PPARγ) with high cellular expressions of NF-κB and iNOS. Results showed the potential of sitosterol-rich D. muricata as a versatile biomedical therapeutic agent against abnormal macrophage polarization and its associated pathologies.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| | | | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni’s College for Women, Chennai, 600 015 India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| |
Collapse
|
12
|
Qian J, Liang S, Wang Q, Xu J, Huang W, Wu G, Liang G. Toll-like receptor-2 in cardiomyocytes and macrophages mediates isoproterenol-induced cardiac inflammation and remodeling. FASEB J 2023; 37:e22740. [PMID: 36583707 DOI: 10.1096/fj.202201345r] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/28/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022]
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality worldwide. Activation of the innate immune system initiates an inflammatory response during cardiac remodeling induced by isoproterenol (ISO). Here, we investigated whether Toll-like receptor-2 (TLR2) mediates ISO-induced inflammation, hypertrophy, and fibrosis. TLR2 was found to be increased in the heart tissues of mouse with HF under ISO challenge. Further, cardiomyocytes and macrophages were identified as the main cellular sources of the increased TLR2 levels in the model under ISO stimulation. The effect of TLR2 deficiency on ISO-induced cardiac remodeling was determined using TLR2 knockout mice and bone marrow transplantation models. In vitro studies involving ISO-treated cultured cardiomyocytes and macrophages showed that TLR2 knockdown significantly decreased ISO-induced cell inflammation and remodeling via MAPKs/NF-κB signaling. Mechanistically, ISO significantly increased the TLR2-MyD88 interaction in the above cells in a TLR1-dependent manner. Finally, DAMPs, such as HSP70 and fibronectin 1 (FN1), were found to be released from the cells under ISO stimulation, which further activated TLR1/2-Myd88 signaling and subsequently activated pro-inflammatory cytokine expression and cardiac remodeling. In summary, our findings suggest that TLR2 may be a target for the alleviation of chronic adrenergic stimulation-associated HF. In addition, this paper points out the possibility of TLR2 as a new target for heart failure under ISO stimulation.
Collapse
Affiliation(s)
- Jinfu Qian
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shiqi Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qinyan Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiachen Xu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Obeidat HM, Althunibat OY, Alfwuaires MA, Aladaileh SH, Algefare AI, Almuqati AF, Alasmari F, Aldal’in HK, Alanezi AA, Alsuwayt B, Abukhalil MH. Cardioprotective Effect of Taxifolin against Isoproterenol-Induced Cardiac Injury through Decreasing Oxidative Stress, Inflammation, and Cell Death, and Activating Nrf2/HO-1 in Mice. Biomolecules 2022; 12:1546. [PMID: 36358896 PMCID: PMC9687704 DOI: 10.3390/biom12111546] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 09/02/2023] Open
Abstract
Oxidative stress and inflammation are key components in cardiovascular diseases and heart dysfunction. Herein, we evaluated the protective effects of (+)-taxifolin (TAX), a potent flavonoid with significant antioxidant and anti-inflammatory actions, on myocardial oxidative tissue injury, inflammation, and cell death, using a mouse model of isoproterenol (ISO)-induced acute myocardial injury. Mice were given TAX (25 and 50 mg/kg, orally) for 14 days before receiving two subsequent injections of ISO (100 mg/kg, s.c.) at an interval of 24 h on the 15th and 16th days. The ISO-induced cardiac tissue injury was evidenced by increased serum creatine kinase-MB (CK-MB), cardiac troponin I (cTnI), and lactate dehydrogenase (LDH), along with several histopathological changes. The ISO also induced increased malondialdehyde (MDA) with concomitant declined myocardial glutathione level and antioxidant enzymes activities. Moreover, ISO-induced heart injury was accompained with elevated cardiac NF-κB p65, TNF-α, IL-1β, Bax, and caspase-3, as well as decreased Bcl-2, Nrf2, and HO-1. Remarkably, TAX reduced the severity of cardiac injury, oxidative stress, inflammation, and cell death, while enhancing antioxidants, Bcl-2, and Nrf2/HO-1 signaling in ISO-injected mice. In conclusion, TAX protects against ISO-induced acute myocardial injury via activating the Nrf2/HO-1 signaling pathway and attenuating the oxidative tissue injury and key regulators of inflammatory response and apoptosis. Thus, our findings imply that TAX may constitute a new cardioprotective therapy against acute MI, which undoubtedly deserves further exploration in upcoming human trials.
Collapse
Affiliation(s)
- Heba M. Obeidat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an 71111, Jordan
| | - Osama Y. Althunibat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an 71111, Jordan
| | - Manal A. Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Saleem H. Aladaileh
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an 71111, Jordan
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| | - Abdulmohsen I. Algefare
- Department of Biological Sciences, Faculty of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Afaf F. Almuqati
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hammad Khalifeh Aldal’in
- Department of Medical Support, Al-Karak University College, Al-Balqa Applied University, Al-Karak 19117, Jordan
| | - Abdulkareem A. Alanezi
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| | - Bader Alsuwayt
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia
| | - Mohammad H. Abukhalil
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma’an 71111, Jordan
- Department of Biology, College of Science, Al-Hussein Bin Talal University, Ma’an 71111, Jordan
| |
Collapse
|
14
|
Zhang W, Zhang R, Chang Z, Wang X. Resveratrol activates CD8+ T cells through IL-18 bystander activation in lung adenocarcinoma. Front Pharmacol 2022; 13:1031438. [PMCID: PMC9630476 DOI: 10.3389/fphar.2022.1031438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Resveratrol, a natural product, has demonstrated anti-tumor effects in various kinds of tumor types, including colon, breast, and pancreatic cancers. Most research has focused on the inhibitory effects of resveratrol on tumor cells themselves rather than resveratrol’s effects on tumor immunology. In this study, we found that resveratrol inhibited the growth of lung adenocarcinoma in a subcutaneous tumor model by using the β-cyclodextrin-resveratrol inclusion complex. After resveratrol treatment, the proportion of M2-like tumor-associated macrophages (TAMs) was reduced and tumor-infiltrating CD8T cells showed significantly increased activation. The results of co-culture and antibody neutralization experiments suggested that macrophage-derived IL-18 may be a key cytokine in the resveratrol anti-tumor effect of CD8T cell activation. The results of this study demonstrate a novel view of the mechanisms of resveratrol tumor suppression. This natural product could reprogram TAMs and CD8T effector cells for tumor treatment.
Collapse
Affiliation(s)
- Wei Zhang
- Emergency and Disaster Medical Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Ruohao Zhang
- School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Zhiguang Chang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Zhiguang Chang, ; Xiaobo Wang,
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- *Correspondence: Zhiguang Chang, ; Xiaobo Wang,
| |
Collapse
|
15
|
Fan S, Hu Y, You Y, Xue W, Chai R, Zhang X, Shou X, Shi J. Role of resveratrol in inhibiting pathological cardiac remodeling. Front Pharmacol 2022; 13:924473. [PMID: 36120366 PMCID: PMC9475218 DOI: 10.3389/fphar.2022.924473] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/04/2022] [Indexed: 12/05/2022] Open
Abstract
Cardiovascular disease is a group of diseases with high mortality in clinic, including hypertension, coronary heart disease, cardiomyopathy, heart valve disease, heart failure, to name a few. In the development of cardiovascular diseases, pathological cardiac remodeling is the most common cardiac pathological change, which often becomes a domino to accelerate the deterioration of the disease. Therefore, inhibiting pathological cardiac remodeling may delay the occurrence and development of cardiovascular diseases and provide patients with greater long-term benefits. Resveratrol is a non-flavonoid polyphenol compound. It mainly exists in grapes, berries, peanuts and red wine, and has cardiovascular protective effects, such as anti-oxidation, inhibiting inflammatory reaction, antithrombotic, dilating blood vessels, inhibiting apoptosis and delaying atherosclerosis. At present, the research of resveratrol has made rich progress. This review aims to summarize the possible mechanism of resveratrol against pathological cardiac remodeling, in order to provide some help for the in-depth exploration of the mechanism of inhibiting pathological cardiac remodeling and the development and research of drug targets.
Collapse
Affiliation(s)
- Shaowei Fan
- Department of cardiological medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Yuanhui Hu
- Department of cardiological medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- *Correspondence: Yuanhui Hu,
| | - Yaping You
- Department of cardiological medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Wenjing Xue
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Ruoning Chai
- Department of cardiological medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Xuesong Zhang
- Department of cardiological medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Xintian Shou
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Jingjing Shi
- Department of cardiological medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
16
|
Guo S, Yang Y, Qian W, Yao Y, Zhou G, Shen L, Zhou J. MicroRNA-384-5p protects against cardiac hypertrophy via the ALPK3 signaling pathway. J Biochem Mol Toxicol 2022; 36:e23093. [PMID: 35510648 DOI: 10.1002/jbt.23093] [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: 04/03/2021] [Revised: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/06/2022]
Abstract
Heart failure is a condition caused by a variety of pathophysiological factors. One important pathological change of chronic heart failure is myocardial hypertrophy. In recent years, several studies have found that dysregulated microRNAs are involved in regulating the pathological process of heart failure. In this study, cardiac hypertrophy models were constructed using isoproterenol (ISO)-/angiotensin-II (Ang-II) to explore the role of miR-384-5p in cardiac hypertrophy and its molecular mechanism in vivo and in vitro. Echocardiography, invasive pressure-volume analysis and hematoxylin-eosin staining were used to explore cardiac structure and function. ALPK3 mRNA and protein expression were detected using quantitative reverse transcription polymerase chain reaction (RT-qPCR) and western blot analysis and miR-384-5p expression were assessed via RT-qPCR. Our findings determined that miR-384-5p was notably decreased in cardiac hypertrophic tissues and cells, and overexpression of miR-384-5p could ameliorate pressure overload. Furthermore, ALPK3 was determined to downregulate the ALPK3 expression to aggravate cardiomyocyte hypertrophy. Our findings provided a potential therapeutic target for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Suxia Guo
- The Department of Cardiology, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Yanhua Yang
- The Department of Cardiology, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Weichun Qian
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yongzhao Yao
- The Department of Cardiology, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Guoxiang Zhou
- The Department of Cardiology, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Lihan Shen
- The Department of Cardiology, Dongguan People's Hospital, Dongguan, Guangdong, China
| | - Jianping Zhou
- The Department of Thoracic, Dongguan People's Hospital, Dongguan, Guangdong, China
| |
Collapse
|
17
|
Small molecule QF84139 ameliorates cardiac hypertrophy via activating the AMPK signaling pathway. Acta Pharmacol Sin 2022; 43:588-601. [PMID: 33967278 PMCID: PMC8888632 DOI: 10.1038/s41401-021-00678-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/02/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac hypertrophy is a common adaptive response to a variety of stimuli, but prolonged hypertrophy leads to heart failure. Hence, discovery of agents treating cardiac hypertrophy is urgently needed. In the present study, we investigated the effects of QF84139, a newly synthesized pyrazine derivative, on cardiac hypertrophy and the underlying mechanisms. In neonatal rat cardiomyocytes (NRCMs), pretreatment with QF84139 (1-10 μM) concentration-dependently inhibited phenylephrine-induced hypertrophic responses characterized by fetal genes reactivation, increased ANP protein level and enlarged cardiomyocytes. In adult male mice, administration of QF84139 (5-90 mg·kg-1·d-1, i.p., for 2 weeks) dose-dependently reversed transverse aortic constriction (TAC)-induced cardiac hypertrophy displayed by cardiomyocyte size, left ventricular mass, heart weights, and reactivation of fetal genes. We further revealed that QF84139 selectively activated the AMPK signaling pathway without affecting the phosphorylation of CaMKIIδ, ERK1/2, AKT, PKCε, and P38 kinases in phenylephrine-treated NRCMs and in the hearts of TAC-treated mice. In NRCMs, QF84139 did not show additive effects with metformin on the AMPK activation, whereas the anti-hypertrophic effect of QF84139 was abolished by an AMPK inhibitor Compound C or knockdown of AMPKα2. In AMPKα2-deficient mice, the anti-hypertrophic effect of QF84139 was also vanished. These results demonstrate that QF84139 attenuates the PE- and TAC-induced cardiac hypertrophy via activating the AMPK signaling. This structurally novel compound would be a promising lead compound for developing effective agents for the treatment of cardiac hypertrophy.
Collapse
|
18
|
Shi J, Zhang Y, Zhang X, Chen R, Wei J, Hou J, Wang B, Lai H, Huang Y. Remodeling immune microenvironment in periodontitis using resveratrol liposomes as an antibiotic-free therapeutic strategy. J Nanobiotechnology 2021; 19:429. [PMID: 34930286 PMCID: PMC8686397 DOI: 10.1186/s12951-021-01175-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Periodontitis is a complicated inflammatory disease that damages the tooth-supporting tissues, with limited pharmacotherapy available. Macrophage-targeting therapy is promising for inflammatory diseases. Resveratrol (RSV), a nonflavonoid polyphenol, is known for its anti-inflammatory and immunomodulatory effects. However, its medical application is limited by its poor stability and water-solubility, as well as its low bioavailability. RESULT A therapeutic resveratrol-loaded liposomal system (Lipo-RSV) was developed to treat periodontitis. The physical properties of Lipo-RSV and its ability to regulate macrophages were investigated. The results showed that Lipo-RSV had good biocompatibility and could re-educate the inflammatory macrophages from M1- to M2-like phenotype through activating p-STAT3 and downregulating p-STAT1. Besides, the Lipo-RSV could scavenge ROS and inhibit the NF-κB signal and inflammasomes, thereby reducing the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α. CONCLUSION These results revealed that Lipo-RSV could be a potential therapeutic system for the antibiotic-free treatment for periodontal diseases.
Collapse
Affiliation(s)
- Junyu Shi
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yi Zhang
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai, 201203, China
| | - Xiaomeng Zhang
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Ruiying Chen
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Jianxu Wei
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Jiazhen Hou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai, 201203, China
| | - Bing Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai, 201203, China
| | - Hongchang Lai
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineCollege of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, 528437, China.
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai, 201203, China.
- Taizhou University, School of Advanced Study, Institute of Natural Medicine and Health Product, Taizhou, 318000, China.
| |
Collapse
|
19
|
Feng L, Li G, An J, Liu C, Zhu X, Xu Y, Gao Y, Li J, Liu J, Yan J, Wang Y, Ren J, Yang L, Qi Z. Exercise Training Protects Against Heart Failure Via Expansion of Myeloid-Derived Suppressor Cells Through Regulating IL-10/STAT3/S100A9 Pathway. Circ Heart Fail 2021; 15:e008550. [PMID: 34911348 DOI: 10.1161/circheartfailure.121.008550] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Exercise training (ET) has a protective effect on the progression of heart failure, however, the specific mechanism has not been fully explored. Myeloid-derived suppressor cells (MDSCs) are a group of myeloid-derived immunosuppressive cells, which showed a protective effect in the progression of heart failure. Thus, we hypothesized that the protective effect of ET on heart failure may be related to the infiltration of MDSCs. METHODS C57BL/6 mice were made to run on a treadmill 6× a week for 4 weeks followed by isoproterenol injection from third week. Heart function was evaluated by echocardiography and the proportion of MDSCs was detected by flow cytometry. Hypertrophic markers, cardiac fibrosis, and inflammatory factors were detected by real-time PCR, ELISA, histological staining, and Western blot. RESULTS ET treatment in isoproterenol-induced heart failure mice (n=7) enhanced cardiac function (57% increase in FS%, P=0.002) and improved morphological changes compared with isoproterenol mice (n=17). ET further caused 79% increasing in cardiac MDSCs in isoproterenol mice (P<0.001). In addition, depletion of MDSCs by 5-Fluorouracil blunted the cardio-protective effect of ET. T-cell proliferation assay showed that ET did not affect the suppressive activity of MDSCs. Furthermore, we found that ET activated the secretion of IL (interleukin)-10 by macrophages in isoproterenol mice. MDSCs expansion and cardio protection was not present in tamoxifen-inducible macrophage-specific IL-10 knockout mice. Western blot results confirmed that IL-10 regulated the differentiation of MDSCs through the translocation of p-STAT3 (signal transducer and activator of transcription 3)/S100A9 (S100 calcium-binding protein A9) to the nucleus. CONCLUSIONS ET could increase MDSCs by stimulating the secretion of IL-10 from macrophage, which was through IL-10/STAT3/S100A9 signaling pathway, thereby achieving the role of heart protection.
Collapse
Affiliation(s)
- Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Guangru Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Jiale An
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Chang Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Xiaolong Zhu
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, China. (X.Z., J.R.).,Department of Cardiovascular Surgery, Tianjin Chest Hospital, China (X.Z.)
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.).,Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, China (Y.G., L.Y., Z.Q.)
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Yachen Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.)
| | - Jiling Ren
- Department of Pathogen Biology, Basic Medical College, Tianjin Medical University, China. (X.Z., J.R.).,Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China. (J.R.)
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.).,Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, China (Y.G., L.Y., Z.Q.)
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China (L.F., G.L., J.A., C.L., Y.X., Y.G., J. Li, J. Liu, J.Y., Y.W., L.Y., Z.Q.).,Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, China (Y.G., L.Y., Z.Q.).,School of Tropical Medicine and Laboratory Medicine, Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, China (Z.Q.)
| |
Collapse
|
20
|
An J, Feng L, Ren J, Li Y, Li G, Liu C, Yao Y, Yao Y, Jiang Z, Gao Y, Xu Y, Wang Y, Li J, Liu J, Cao L, Qi Z, Yang L. Chronic stress promotes breast carcinoma metastasis by accumulating myeloid-derived suppressor cells through activating β-adrenergic signaling. Oncoimmunology 2021; 10:2004659. [PMID: 34858728 PMCID: PMC8632282 DOI: 10.1080/2162402x.2021.2004659] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Numerous studies have found that chronic stress could promote tumor progression and this may be related to inhibtion of immune system. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells with immunosuppressive activity. MDSCs may represent a key link between chronic stress and tumor progression. However, the role of stress-induced MDSCs in breast cancer progression is unclear. The present study showed that pre-exposure of chronic stress could lead to MDSCs elevation and facilitated breast cancer metastasis in tumor-bearing mice. Adoptive transfer of MDSCs could significantly increase lung metastatic foci. In contrast, lung metastasis could be alleviated by depleting endogenous MDSCs with Gr-1 antibody. The concentration of norepinephrine in serum and the expression of tyrosine hydroxylase in bone marrow could be significantly elevated by chronic stress. Moreover, propranolol, an inhibitor of β-adrenergic signaling, could inhibit breast carcinoma metastasis and prevent the expansion of chronic stress-induced MDSCs. Further study revealed that the expressions of IL-6 and JAK/STAT3 signaling pathways were upregulated by chronic stress in mice, and this upregulation could be inhibited by propranolol. Blocking the IL-6 signal or inhibiting the activation of the JAK/STAT3 signaling pathway could reduce tumor growth and metastasis by attenuating the accumulation of MDSCs in vivo. Besides, propranolol inhibited the expression of IL-6 in supernatant of 4T1 cells induced by isoproterenol and reduced the proportion of inducible MDSCs in vitro. Taken together, these data indicated that chronic stress may accumulate MDSCs via activation of β-adrenergic signaling and IL-6/STAT3 pathway, thereby promoting breast carcinoma metastasis.
Collapse
Affiliation(s)
- Jiale An
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jiling Ren
- Department of Pathogen Biology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Yafei Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Guangru Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Chang Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yong Yao
- Department of Nuclear Medicine, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, Guangdong, China
| | - Ye Yao
- Department of Pathogen Biology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Zecheng Jiang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yachen Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Lei Cao
- Department of General Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
21
|
Wang L, Lu Q, Gao W, Yu S. Recent advancement on development of drug-induced macrophage polarization in control of human diseases. Life Sci 2021; 284:119914. [PMID: 34453949 DOI: 10.1016/j.lfs.2021.119914] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Macrophages, an important part of human immune system, possess a high plasticity and heterogeneity (macrophage polarization) as classically activated macrophages (M1) and alternatively activated macrophages (M2), which exert pro-inflammatory/anti-tumor and anti-inflammatory/pro-tumor effects, respectively. Thus, drug development in induction of macrophage polarization could be used to treat different human diseases. This review summarizes the recent advancement on modulation of macrophage polarization and its related molecular mechanisms induced by a number of agents. Research on the anti-inflammatory drugs to regulate the macrophage polarization accounts for a large proportion in the field and types of diseases investigated could include atherosclerosis, enteritis, nephritis, and the nervous system and skeletal diseases, while study of the anti-tumor agents to modify macrophage polarization is a novel area of research. Future study of the molecular mechanisms by which the different agents regulate the macrophage polarization could lead to an effective control of various human diseases, including inflammation and cancers.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China; School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qi Lu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacy, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, China
| | - Wenwen Gao
- Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China
| | - Shuwen Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacy, Qilu Hospital of Shandong University, Clinical Trial Center, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
22
|
Gong Z, Ye Q, Wu JW, Zhou JL, Kong XY, Ma LK. UCHL1 inhibition attenuates cardiac fibrosis via modulation of nuclear factor-κB signaling in fibroblasts. Eur J Pharmacol 2021; 900:174045. [PMID: 33745956 DOI: 10.1016/j.ejphar.2021.174045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/17/2021] [Accepted: 03/16/2021] [Indexed: 01/05/2023]
Abstract
The ubiquitin-proteasome system (UPS) plays an essential role in cellular homeostasis and myocardial function. Ubiquitin carboxy-terminal hydrolase 1 (UCHL1) is involved in cardiac remodeling, but its underlying mechanisms are largely unknown. Here, we observed that the UCHL1 was significantly up-regulated in angiotensin II-infused heart and primary cardiac fibroblast (CF). Systemic administration of the UCHL1 inhibitor LDN57444 significantly ameliorated cardiac fibrosis and improved cardiac function induced by angiotensin II. Also, LDN57444 inhibited CF cell proliferation as well as attenuated collagen I, and CTGF gene expression in the presence of Ang II. Mechanistically, UCHL1 promotes angiotensin II-induced fibrotic responses by way of activating nuclear factor kappa B (NF-κB) signaling. Moreover, suppression of the NF-κB pathway interfered with UCHL1 overexpression-mediated fibrotic responses. Besides, the chromatin immunoprecipitation assay demonstrated that NF-κB can bind to the UCHL1 promoter and trigger its transcription in cardiac fibroblasts. These findings suggest that UCHL1 positively regulates cardiac fibrosis by modulating NF-κB signaling pathway and identify UCHL1 could be a new treatment strategy for cardiac fibrosis.
Collapse
Affiliation(s)
- Zheng Gong
- Provincial Hospital of Anhui Medical University, Hefei, 230000, Anhui, PR China
| | - Qing Ye
- The First Hospital of University of Science and Technology of China, Hefei, 230000, Anhui, PR China
| | - Jia-Wei Wu
- The First Hospital of University of Science and Technology of China, Hefei, 230000, Anhui, PR China
| | - Jun-Ling Zhou
- The First Hospital of University of Science and Technology of China, Hefei, 230000, Anhui, PR China
| | - Xiang-Yong Kong
- The First Hospital of University of Science and Technology of China, Hefei, 230000, Anhui, PR China
| | - Li-Kun Ma
- Provincial Hospital of Anhui Medical University, Hefei, 230000, Anhui, PR China; The First Hospital of University of Science and Technology of China, Hefei, 230000, Anhui, PR China.
| |
Collapse
|
23
|
Protective Role of Polyphenols in Heart Failure: Molecular Targets and Cellular Mechanisms Underlying Their Therapeutic Potential. Int J Mol Sci 2021; 22:ijms22041668. [PMID: 33562294 PMCID: PMC7914665 DOI: 10.3390/ijms22041668] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a leading cause of death in the United States, with a 5-year mortality rate of 50% despite modern pharmacological therapies. Plant-based diets are comprised of a diverse polyphenol profile, which lends to their association with reduced cardiovascular disease risk. Whether a polyphenol-rich diet can slow the progression of or reverse HF in humans is not known. To date, in vitro and in vivo studies have reported on the protective role of polyphenols in HF. In this review, we will discuss the major mechanisms by which polyphenols mitigate HF in vitro and in vivo, including (1) reduced cardiac inflammation and oxidative stress, (2) reduced mitochondrial dysfunction, (3) improved Ca2+ homeostasis, (4) increased survival signaling, and (5) increased sirtuin 1 activity.
Collapse
|
24
|
The Interplay Between Antioxidants and the Immune System: A Promising Field, Still Looking for Answers. Nutrients 2020; 12:nu12061550. [PMID: 32466603 PMCID: PMC7352790 DOI: 10.3390/nu12061550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
|