1
|
Zhang R, Li W, Yang J, Fan X, Fan H, Li W. The Role of Luteolin in Inhibiting Prostaglandin-Endoperoxide Synthase 2 to Relieve Neointimal Hyperplasia in Arteriovenous Fistula. Adv Biol (Weinh) 2025; 9:e2400437. [PMID: 39960128 DOI: 10.1002/adbi.202400437] [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: 07/26/2024] [Revised: 01/24/2025] [Indexed: 04/17/2025]
Abstract
This study aims to investigate the role and mechanism of luteolin in inflammation and phenotypic switch of vascular smooth muscle cells (VSMCs) in an arteriovenous fistula (AVF) model, for providing a potential agent for the prevention and therapy of AVF neointimal hyperplasia. In vivo, an AVF model is created in Sprague Dawley rats. In vitro, rat VSMCs are treated with platelet-derived growth factor-BB (PDGF-BB) to induce the phenotypic switch of VSMCs. Histological AVF changes are analyzed using hematoxylin-eosin. Western blot and quantitative real-time polymerase chain reaction (qRT-PCR) are utilized to detect prostaglandin-endoperoxide synthase 2 (PTGS2) expression. In vivo, luteolin inhibits neointima formation and reduces vimentin, α-SMA, MCP-1, MMP-9, TNF-α, and IL-6 levels. In vitro, under PDGF-BB treatment, luteolin inhibits proliferation and migration and reduces TNF-α, vimentin, α-SMA, MCP-1, MMP-9, and IL-6 levels in VSMCs. In rat AVF tissues, PTGS2 expression is increased. Luteolin inhibits PTGS2 expression in vivo and in vitro. PTGS2 overexpression reverses the role of luteolin in extracellular matrix protein expression, proliferation, inflammation, and migration in VSMCs treated with PDGF-BB. Altogether, in the AVF, luteolin inhibits proliferation, migration, the phenotypic switch of VSMCs, neointima formation, and the inflammatory response through inhibiting PTGS2 expression.
Collapse
Affiliation(s)
- Ruibin Zhang
- The First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, P. R. China
- Department of Nephrology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, P. R. China
| | - Wei Li
- Department of hand and foot, Gaomi People's Hospital, Gaomi, Shandong, 261500, P. R. China
| | - Jihua Yang
- Department of Ultrasound, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, P. R. China
| | - Xiujie Fan
- Department of Medical Experimental Diagnosis Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, P. R. China
| | - Huili Fan
- Department of Nephrology, Jinxiang Affiliated Hospital of Jining Medical University, Jining, Shandong, 272200, P. R. China
| | - Wei Li
- Department of Nephrology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, Shandong, 250014, P. R. China
| |
Collapse
|
2
|
Dabravolski SA, Churov AV, Sukhorukov VN, Kovyanova TI, Beloyartsev DF, Lyapina IN, Orekhov AN. The role of lipase maturation factor 1 in hypertriglyceridaemia and atherosclerosis: An update. SAGE Open Med 2024; 12:20503121241289828. [PMID: 39483624 PMCID: PMC11526315 DOI: 10.1177/20503121241289828] [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: 06/08/2024] [Accepted: 09/18/2024] [Indexed: 11/03/2024] Open
Abstract
Lipase maturation factor 1 is an endoplasmic reticulum-resident transmembrane protein, which acts as a critical chaperone necessary for the folding, dimerisation, and secretion of lipases. In this review, we summarise data about the recently revealed role of lipase maturation factor 1 in endoplasmic reticulum redox homeostasis, its novel interaction partners among oxidoreductases and lectin chaperones, and the identification of fibronectin and the low-density lipoprotein receptor as novel non-lipase client proteins of lipase maturation factor 1. Additionally, the role of lipase maturation factor 1-derived circular RNA in atherosclerosis progression via the miR-125a-3p/vascular endothelial growth factor A\Fibroblast Growth Factor 1 axis is discussed. Finally, we focus on the causative role of lipase maturation factor 1 variants in the development of hypertriglyceridaemia - a type of dyslipidaemia that significantly contributes to the development of atherosclerosis and other cardiovascular diseases via different mechanisms.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Karmiel, Israel
| | - Alexey V Churov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | | | - Tatiana I Kovyanova
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Institute for Atherosclerosis Research, Moscow, Russia
| | | | - Irina N Lyapina
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | | |
Collapse
|
3
|
Sadek KM, Shib NA, Taher ES, Rashed F, Shukry M, Atia GA, Taymour N, El-Nablaway M, Ibrahim AM, Ramadan MM, Abdelkader A, Abdo M, Imbrea I, Pet E, Ali LS, Abdeen A. Harnessing the power of bee venom for therapeutic and regenerative medical applications: an updated review. Front Pharmacol 2024; 15:1412245. [PMID: 39092234 PMCID: PMC11291246 DOI: 10.3389/fphar.2024.1412245] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/24/2024] [Indexed: 08/04/2024] Open
Abstract
Honeybees have been helpful insects since ancient centuries, and this benefit is not limited to being a honey producer only. After the bee stings a person, pain, and swelling occur in this place, due to the effects of bee venom (BV). This is not a poison in the total sense of the word because it has many benefits, and this is due to its composition being rich in proteins, peptides, enzymes, and other types of molecules in low concentrations that show promise in the treatment of numerous diseases and conditions. BV has also demonstrated positive effects against various cancers, antimicrobial activity, and wound healing versus the human immunodeficiency virus (HIV). Even though topical BV therapy is used to varying degrees among countries, localized swelling or itching are common side effects that may occur in some patients. This review provides an in-depth analysis of the complex chemical composition of BV, highlighting the diverse range of bioactive compounds and their therapeutic applications, which extend beyond the well-known anti-inflammatory and pain-relieving effects, showcasing the versatility of BV in modern medicine. A specific search strategy was followed across various databases; Web of sciences, Scopus, Medline, and Google Scholar including in vitro and in vivo clinical studies.to outline an overview of BV composition, methods to use, preparation requirements, and Individual consumption contraindications. Furthermore, this review addresses safety concerns and emerging approaches, such as the use of nanoparticles, to mitigate adverse effects, demonstrating a balanced and holistic perspective. Importantly, the review also incorporates historical context and traditional uses, as well as a unique focus on veterinary applications, setting it apart from previous works and providing a valuable resource for researchers and practitioners in the field.
Collapse
Affiliation(s)
- Kadry M. Sadek
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Naira A. Shib
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Ehab S. Taher
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
| | - Fatema Rashed
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Gamal A. Atia
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Noha Taymour
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammad El-Nablaway
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ateya M. Ibrahim
- Department of Administration and Nursing Education, College of Nursing, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Family and Community Health Nursing, Faculty of Nursing, Port Said University, Port Said, Egypt
| | - Mahmoud M. Ramadan
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Afaf Abdelkader
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Mohamed Abdo
- Department of Animal Histology and Anatomy, School of Veterinary Medicine, Badr University in Cairo (BUC), Badr City, Egypt
- Department of Anatomy and Embryology, Faculty Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Ilinca Imbrea
- Department of Forestry, Faculty of Engineering and Applied Technologies, University of Life Sciences “King Mihai I” from Timisoara, Timisoara, Romania
| | - Elena Pet
- Department of Management and Rural Development, Faculty of Management and Rural Tourism, University of Life Sciences “King Mihai I” from Timisoara, Timisoara, Romania
| | - Lashin S. Ali
- Department of Basic Medical Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| |
Collapse
|
4
|
Wang P, Xu X, Gu G, Guo Q, Rao Y, Yang K, Xi T, Yuan Y, Chen S, Qi X. Inhibition effect of copper-bearing metals on arterial neointimal hyperplasia via the AKT/Nrf2/ARE pathway in vitro and in vivo. Regen Biomater 2024; 11:rbae042. [PMID: 39027361 PMCID: PMC11256920 DOI: 10.1093/rb/rbae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 07/20/2024] Open
Abstract
In-stent restenosis can be caused by the activation, proliferation and migration of vascular smooth muscle cells (VSMCs), which affects long-term efficacy of interventional therapy. Copper (Cu) has been proved to accelerate the endothelialization and reduce thrombosis formation, but little is known about its inhibition effect on the excessive proliferation of VSMCs. In this study, 316L-Cu stainless steel and L605-Cu cobalt-based alloy with varying Cu content were fabricated and their effects on surface property, blood compatibility and VSMCs were studied in vitro and in vivo. CCK-8 assay and EdU assay indicated that the Cu-bearing metals had obvious inhibitory effect on proliferation of VSMCs. Blood clotting and hemolysis tests showed that the Cu-bearing metals had good blood compatibility. The inhibition effect of the Cu-bearing metals on migration of cells was detected by Transwell assay. Further studies showed that Cu-bearing metals significantly decreased the mRNA expressions of bFGF, PDGF-B, HGF, Nrf2, GCLC, GCLM, NQO1 and HO1. The phosphorylation of AKT and Nrf2 protein expressions in VSMCs were significantly decreased by Cu-bearing metals. Furthermore, it was also found that SC79 and TBHQ treatments could recover the protein expressions of phospho-AKT and Nrf2, and their downstream proteins as well. Moreover, 316L-Cu stent proved its inhibitory action on the proliferation of VSMCs in vivo. In sum, the results demonstrated that the Cu-bearing metals possessed apparent inhibitory effect on proliferation and migration of VSMCs via regulating the AKT/Nrf2/ARE pathway, showing the Cu-bearing metals as promising stent materials for long-term efficacy of implantation.
Collapse
Affiliation(s)
- Peng Wang
- Department of Interventional Therapy, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaohe Xu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Guisong Gu
- Shi-Changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Qianwen Guo
- Department of Interventional Therapy, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yanzhi Rao
- Department of Interventional Therapy, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ke Yang
- Shi-Changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Tong Xi
- Shi-Changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Yonghui Yuan
- Liaoning Cancer Hospital & Institute, Clinical Research Center for Malignant Tumor of Liaoning Province, Cancer Hospital of China Medical University, Shenyang 110042, China
| | - Shanshan Chen
- Shi-Changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Xun Qi
- Department of Interventional Therapy, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Diagnostic Imaging and Interventional Radiology of Liaoning Province, Department of Radiology, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
5
|
Bindlish A, Sawal A. Bee Sting Venom as a Viable Therapy for Breast Cancer: A Review Article. Cureus 2024; 16:e54855. [PMID: 38533165 PMCID: PMC10964279 DOI: 10.7759/cureus.54855] [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: 08/13/2023] [Accepted: 02/25/2024] [Indexed: 03/28/2024] Open
Abstract
Breast cancer is a kind of aggressive cancer that significantly affects women worldwide, thus making research on alternative and new therapies necessary. The potential impact of bee venom on breast cancer is the main subject of this analysis of this research article. Bee venom has drawn the attention of the world with the help of its constituent ingredients, namely the bioactive compounds, enzymes, and complex blend of proteins. They have a particularly varied chemical makeup and proven anti-cancer capabilities. This is a detailed review demonstrating the components of bee venom and their individual functions in fighting cancer, as well as the results of previously conducted in-vitro and in-vivo research. As described later, bee venom has given positive results in triggering apoptosis, preventing cell migration, inhibiting metastasis and invasion, and suppressing the existing breast cancer cells. It is found to have worked better along with the already existing chemotherapy treatments. These results were also proved with the help of various animal studies that showed reduced tumor development, reduced metastasis, and improved therapeutic effectiveness. Furthermore, certain studies and case reports from all over the world have exhibited consistent results in females affected by breast cancer. This study found that people receiving chemotherapy experienced improved health outcomes and reduced discomfort, with fewer negative side effects. It is important to conduct extensive research on the safety and effectiveness of this treatment because it is yet to be approved. The ideal dosage and administration methods must be explored in clinical trials. Moreover, it is imperative to evaluate the results of any combined treatments with current medications. There should be constant monitoring to prevent any potential side effects. Other important things like allergic reactions and hidden concerns should also be considered.
Collapse
Affiliation(s)
- Aabhas Bindlish
- Anatomy, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Anupama Sawal
- Anatomy, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
6
|
Therapeutic Use of Bee Venom and Potential Applications in Veterinary Medicine. Vet Sci 2023; 10:vetsci10020119. [PMID: 36851423 PMCID: PMC9965945 DOI: 10.3390/vetsci10020119] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
Apitherapy is a branch of alternative medicine that consists of the treatment of diseases through products collected, processed, and secreted by bees, specifically pollen, propolis, honey, royal jelly, and bee venom. In traditional medicine, the virtues of honey and propolis have been well-known for centuries. The same, however, cannot be said for venom. The use of bee venom is particularly relevant for many therapeutic aspects. In recent decades, scientific studies have confirmed and enabled us to understand its properties. Bee venom has anti-inflammatory, antioxidant, central nervous system inhibiting, radioprotective, antibacterial, antiviral, and antifungal properties, among others. Numerous studies have often been summarised in reviews of the scientific literature that have focused on the results obtained with mouse models and their subsequent transposition to the human patient. In contrast, few reviews of scientific work on the use of bee venom in veterinary medicine exist. This review aims to take stock of the research achievements in this particular discipline, with a view to a recapitulation and stabilisation in the different research fields.
Collapse
|
7
|
Subedi U, Manikandan S, Bhattarai S, Sharma P, Sharma S, Sun H, Miriyala S, Panchatcharam M. The Autotaxin-LPA Axis Emerges as a Novel Regulator of Smooth Muscle Cell Phenotypic Modulation during Intimal Hyperplasia. Int J Mol Sci 2023; 24:2913. [PMID: 36769255 PMCID: PMC9917461 DOI: 10.3390/ijms24032913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Neointimal hyperplasia is characterized by a loss of the contractile phenotype of vascular smooth muscle cells (VSMCs). Our group has recently shown that VSMC proliferation and migration are mediated by lysophosphatidic acid (LPA) during restenosis, but the role of autotaxin (ATX; lysophospholipase D), which produces LPA, remains unclear. Endothelial denudation of the mouse carotid artery was performed to induce neointimal hyperplasia, and the extent of damage caused by the ATX-LPA axis was assessed in VSMCs. We observed the upregulation of ATX activity (p < 0.0002) in the injured carotid artery using an AR2 probe fluorescence assay. Further, the tissue carotid LPA levels were elevated 2.7-fold in carotid vessels, augmenting neointimal hyperplasia. We used an electrical cell-substrate impedance sensor (ECIS) to measure VSMC proliferation and migration. Treatment with an ATX inhibitor (PF8380) or LPA receptor inhibitor (Ki16425) attenuated VSMC proliferation (extracellular signal-regulated kinases) activity and migration in response to recombinant ATX. Indeed, PF8380 treatment rescued the aggravated post-wire injury neointima formation of carotid arteries. The upregulation of ATX following vessel injury leads to LPA production in VSMCs, favoring restenosis. Our observations suggest that inhibition of the ATX-LPA axis could be therapeutically targeted in restenosis to minimize VSMC phenotypic modulation and inflammation after vascular injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sumitra Miriyala
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences-Shreveport, Shreveport, LA 71130, USA
| | - Manikandan Panchatcharam
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences-Shreveport, Shreveport, LA 71130, USA
| |
Collapse
|
8
|
Park J, Jang KM, Park KK. Effects of Apamin on MPP +-Induced Calcium Overload and Neurotoxicity by Targeting CaMKII/ERK/p65/STAT3 Signaling Pathways in Dopaminergic Neuronal Cells. Int J Mol Sci 2022; 23:15255. [PMID: 36499581 PMCID: PMC9736188 DOI: 10.3390/ijms232315255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/19/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD), a neurodegenerative disorder, is characterized by the loss of dopaminergic (DA) neurons. The pathogenesis of PD is associated with several factors including oxidative stress, inflammation, and mitochondrial dysfunction. Ca2+ signaling plays a vital role in neuronal signaling and altered Ca2+ homeostasis has been implicated in many neuronal diseases including PD. Recently, we reported that apamin (APM), a selective antagonist of the small-conductivity Ca2+-activated K+ (SK) channel, suppresses neuroinflammatory response. However, the mechanism(s) underlying the vulnerability of DA neurons were not fully understood. In this study, we investigated whether APM affected 1-methyl-4-phenyl pyridinium (MPP+)-mediated neurotoxicity in SH-SY5Y cells and rat embryo primary mesencephalic neurons. We found that APM decreased Ca2+ overload arising from MPP+-induced neurotoxicity response through downregulating the level of CaMKII, phosphorylation of ERK, and translocation of nuclear factor NFκB/signal transducer and activator of transcription (STAT)3. Furthermore, we showed that the correlation of MPP+-mediated Ca2+ overload and ERK/NFκB/STAT3 in the neurotoxicity responses, and dopaminergic neuronal cells loss, was verified through inhibitors. Our findings showed that APM might prevent loss of DA neurons via inhibition of Ca2+-overload-mediated signaling pathway and provide insights regarding the potential use of APM in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Jihyun Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Kyung Mi Jang
- Department of Pediatrics, College of Medicine, Yeungnam University, Daegu 42415, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| |
Collapse
|
9
|
Asma ST, Bobiş O, Bonta V, Acaroz U, Shah SRA, Istanbullugil FR, Arslan-Acaroz D. General Nutritional Profile of Bee Products and Their Potential Antiviral Properties against Mammalian Viruses. Nutrients 2022; 14:nu14173579. [PMID: 36079835 PMCID: PMC9460612 DOI: 10.3390/nu14173579] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/19/2022] Open
Abstract
Bee products have been extensively employed in traditional therapeutic practices to treat several diseases and microbial infections. Numerous bioactive components of bee products have exhibited several antibacterial, antifungal, antiviral, anticancer, antiprotozoal, hepatoprotective, and immunomodulatory properties. Apitherapy is a form of alternative medicine that uses the bioactive properties of bee products to prevent and/or treat different diseases. This review aims to provide an elaborated vision of the antiviral activities of bee products with recent advances in research. Since ancient times, bee products have been well known for their several medicinal properties. The antiviral and immunomodulatory effects of bee products and their bioactive components are emerging as a promising alternative therapy against several viral infections. Numerous studies have been performed, but many clinical trials should be conducted to evaluate the potential of apitherapy against pathogenic viruses. In that direction, here, we review and highlight the potential roles of bee products as apitherapeutics in combating numerous viral infections. Available studies validate the effectiveness of bee products in virus inhibition. With such significant antiviral potential, bee products and their bioactive components/extracts can be effectively employed as an alternative strategy to improve human health from individual to communal levels as well.
Collapse
Affiliation(s)
- Syeda Tasmia Asma
- Department of Food Hygiene and Technology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar 03200, Turkey
| | - Otilia Bobiş
- Department of Beekeeping and Sericulture, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, 400372 Cluj-Napoca, Romania
- Correspondence: (O.B.); (U.A.)
| | - Victoriţa Bonta
- Department of Beekeeping and Sericulture, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, 400372 Cluj-Napoca, Romania
| | - Ulas Acaroz
- Department of Food Hygiene and Technology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar 03200, Turkey
- Correspondence: (O.B.); (U.A.)
| | - Syed Rizwan Ali Shah
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar 03200, Turkey
| | - Fatih Ramazan Istanbullugil
- Department of Food Hygiene and Technology, Faculty of Veterinary Medicine, Kyrgyz-Turkish Manas University, Bishkek KG-720038, Kyrgyzstan
| | - Damla Arslan-Acaroz
- Department of Biochemistry, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar 03200, Turkey
| |
Collapse
|
10
|
Badr-Eldin SM, Aldawsari HM, Fahmy UA, Ahmed OAA, Alhakamy NA, Al-hejaili OD, Alhassan AA, Ammari GA, Alhazmi SI, Alawadi RM, Bakhaidar R, Alamoudi AJ, Neamatallah T, Tima S. Optimized Apamin-Mediated Nano-Lipidic Carrier Potentially Enhances the Cytotoxicity of Ellagic Acid against Human Breast Cancer Cells. Int J Mol Sci 2022; 23:9440. [PMID: 36012704 PMCID: PMC9408819 DOI: 10.3390/ijms23169440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/03/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
Ellagic acid has recently attracted increasing attention regarding its role in the prevention and treatment of cancer. Surface functionalized nanocarriers have been recently studied for enhancing cancer cells' penetration and achieving better tumor-targeted delivery of active ingredients. Therefore, the present work aimed at investigating the potential of APA-functionalized emulsomes (EGA-EML-APA) for enhancing cytototoxic activity of EGA against human breast cancer cells. Phospholipon® 90 G: cholesterol molar ratio (PC: CH; X1, mole/mole), Phospholipon® 90 G: Tristearin weight ratio (PC: TS; X2, w/w) and apamin molar concentration (APA conc.; X3, mM) were considered as independent variables, while vesicle size (VS, Y1, nm) and zeta potential (ZP, Y2, mV) were studied as responses. The optimized formulation with minimized vs. and maximized absolute ZP was predicted successfully utilizing a numerical technique. EGA-EML-APA exhibited a significant cytotoxic effect with an IC50 value of 5.472 ± 0.21 µg/mL compared to the obtained value from the free drug 9.09 ± 0.34 µg/mL. Cell cycle profile showed that the optimized formulation arrested MCF-7 cells at G2/M and S phases. In addition, it showed a significant apoptotic activity against MCF-7 cells by upregulating the expression of p53, bax and casp3 and downregulating bcl2. Furthermore, NF-κB activity was abolished while the expression of TNfα was increased confirming the significant apoptotic effect of EGA-EML-APA. In conclusion, apamin-functionalized emulsomes have been successfully proposed as a potential anti-breast cancer formulation.
Collapse
Affiliation(s)
- Shaimaa M. Badr-Eldin
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hibah M. Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Usama A. Fahmy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Osama A. A. Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Omar D. Al-hejaili
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Alhanoof A. Alhassan
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ghadeer A. Ammari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shouq I. Alhazmi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Raghad M. Alawadi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rana Bakhaidar
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulmohsen J. Alamoudi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Thikryat Neamatallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
11
|
Alamoudi AA, Alharbi AS, Abdel-Naim AB, Badr-Eldin SM, Awan ZA, Okbazghi SZ, Ahmed OAA, Alhakamy NA, Fahmy UA, Esmat A. Novel Nanoconjugate of Apamin and Ceftriaxone for Management of Diabetic Wounds. Life (Basel) 2022; 12:1096. [PMID: 35888184 PMCID: PMC9323216 DOI: 10.3390/life12071096] [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: 06/16/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetic hyperglycemia delays wound healing, leading to serious consequences. Topical antibiotics can reduce the risk of a wound infection during healing; nevertheless, the microbial fight against antibiotics brings about public health challenges. Anti-microbial peptides (AMPs) belong to a novel class of drug that is used to prevent and treat systemic and topical infections. The aim of the current work was to achieve better wound healing in diabetic rats by conjugating the anti-microbial peptide "apamin" (APA) with the broad-spectrum antibiotic "ceftriaxone" (CTX) to form a nanocomplex. The CTX-APA nanoconjugate formulation was optimized using a Box-Behnken design. The optimized CTX-APA nanoconjugate formulation was evaluated for its size and zeta potential, and was then examined using transmission electron microscopy (TEM). The CTX-APA nanoconjugate was loaded onto a hydroxypropyl methylcellulose (2% w/v)-based hydrogel. It was observed that the application of the CTX-APA nanocomplex on the wounded skin of diabetic rats accelerated the regeneration of the epithelium, granulation tissue formation, epidermal proliferation, and keratinization. The nanocomplex was capable of significantly reducing the expression of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), while increasing the expression of transforming growth factor beta-1 (TGF-β1) as well as the angiogenic markers: hypoxia-inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF). Conclusively, the application of an ion-paired CTX-APA nanocomplex enhances wound healing in diabetic rats.
Collapse
Affiliation(s)
- Abdullah A. Alamoudi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.A.); (A.S.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (U.A.F.)
| | - Awaad S. Alharbi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.A.); (A.S.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (U.A.F.)
- Alrass General Hospital, Ministry of Health, Qassim Region, Ar Rass 58883, Saudi Arabia
| | - Ashraf B. Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Shaimaa M. Badr-Eldin
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.A.); (A.S.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (U.A.F.)
- Department of Pharmaceutics, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Zuhier A. Awan
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Solomon Z. Okbazghi
- Global Analytical and Pharmaceutical Development, Alexion Pharmaceuticals, New Haven, CT 06510, USA;
| | - Osama A. A. Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.A.); (A.S.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (U.A.F.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.A.); (A.S.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (U.A.F.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Usama A. Fahmy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.A.); (A.S.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (U.A.F.)
| | - Ahmed Esmat
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
12
|
Evening Primrose Extracts Inhibit PDGF-BB-Induced Vascular Smooth Muscle Cell Proliferation and Migration by Regulating Cell-Cycle-Related Proteins. Curr Issues Mol Biol 2022; 44:1928-1940. [PMID: 35678660 PMCID: PMC9164085 DOI: 10.3390/cimb44050131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
The proliferation and migration of vascular smooth muscle cells (VSMCs) are important factors in the occurrence of cardiovascular diseases, such as blood flow abnormalities, stroke and atherosclerosis. Evening primrose, known as Oenothera biennis, is a plant native to Korea that exerts physiological activities, such as antioxidant effects, the inhibition of lipid accumulation and the prevention of muscle atrophy. However, the function of evening primrose stem (EVP) in the regulation of VSMC proliferation and migration and the underlying mechanisms have not been identified. In this study, the effect of EVP on the platelet-derived growth factor (PDGF)-induced proliferation and migration of VSMCs was investigated. The results show that PDGF-BB-induced proliferation of VSMCs was inhibited by EVP at concentrations of 25, 50 or 100 μg/mL in a concentration-dependent manner, and a migration assay showed that EVP inhibited cell migration. Cell cycle analysis was performed to confirm the mechanism by which cell proliferation and migration was inhibited. The results indicate that proteins involved in the cell cycle, such as cyclin, CDK and phosphorylated Rb, were downregulated by EVP at concentrations of 100 μg/mL, thereby increasing the proportion of cells in the G0/G1 phase and inhibiting cell cycle progression. In the PDGF receptor (PDGFR) signaling pathway, phosphorylation of the PDGFR was inhibited by EVP at concentrations of 100 μg/mL, and PLCγ phosphorylation was also decreased. The PDGF-BB-induced effect of EVP on the proliferation of VSMCs involved the inhibition of Akt phosphorylation and the reduction in the phosphorylation of MAPK proteins such as ERK, P38 and JNK. In conclusion, the results demonstrate that EVP inhibited PDGF-BB-induced VSMC proliferation and migration by regulating cell-cycle-related proteins.
Collapse
|
13
|
Gwon MG, An HJ, Gu H, Kim YA, Han SM, Park KK. Apamin inhibits renal fibrosis via suppressing TGF-β1 and STAT3 signaling in vivo and in vitro. J Mol Med (Berl) 2021; 99:1265-1277. [PMID: 34031696 DOI: 10.1007/s00109-021-02087-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Renal fibrosis is a progressive and chronic process that influences kidneys with chronic kidney disease (CKD), irrespective of cause, leading to irreversible failure of renal function and end-stage kidney disease. Among the signaling related to renal fibrosis, transforming growth factor-β1 (TGF-β1) signaling is a major pathway that induces the activation of myofibroblasts and the production of extracellular matrix (ECM) molecules. Apamin, a component of bee venom (BV), has been studied in relation to various diseases. However, the effect of apamin on renal interstitial fibrosis has not been investigated. The aim of this study was to estimate the beneficial effect of apamin in unilateral ureteral obstruction (UUO)-induced renal fibrosis and TGF-β1-induced renal fibroblast activation. This study revealed that obstructive kidney injury induced an inflammatory response, tubular atrophy, and ECM accumulation. However, apamin treatment suppressed the increased expression of fibrotic-related genes, including α-SMA, vimentin, and fibronectin. Administration of apamin also attenuated the renal tubular cells injury and tubular atrophy. In addition, apamin attenuated fibroblast activation, ECM synthesis, and inflammatory cytokines such as TNF-α, IL-1β, and IL-6 by suppressing the TGF-β1-canonical and non-canonical signaling pathways. This study showed that apamin inhibits UUO-induced renal fibrosis in vivo and TGF-β1-induced renal fibroblasts activation in vitro. Apamin inhibited the inflammatory response, tubular atrophy, ECM accumulation, fibroblast activation, and renal interstitial fibrosis through suppression of TGF-β1/Smad2/3 and STAT3 signaling pathways. These results suggest that apamin might be a potential therapeutic agent for renal fibrosis. KEY MESSAGES: UUO injury can induce renal dysfunction; however, apamin administration prevents renal failure in UUO mice. Apamin inhibited renal inflammatory response and ECM deposition in UUO-injured mice. Apamin suppressed the activation of myofibroblasts in vivo and in vitro. Apamin has the anti-fibrotic effect on renal fibrosis via regulation of TGF-β1 canonical and non-canonical signaling.
Collapse
Affiliation(s)
- Mi-Gyeong Gwon
- Department of Pathology, School of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, 42472, Daegu, Republic of Korea
| | - Hyun-Jin An
- Department of Pathology, School of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, 42472, Daegu, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, School of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, 42472, Daegu, Republic of Korea
| | - Young-Ah Kim
- Department of Pathology, School of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, 42472, Daegu, Republic of Korea
| | - Sang Mi Han
- National Academy of Agricultural Science, Jeonjusi, Jeonbuk, 54875, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, 42472, Daegu, Republic of Korea.
| |
Collapse
|
14
|
Yang Y, Mao W, Wang L, Lu L, Pang Y. Circular RNA circLMF1 regulates PDGF-BB-induced proliferation and migration of human aortic smooth muscle cells by regulating the miR-125a-3p/VEGFA or FGF1 axis. Clin Hemorheol Microcirc 2021; 80:167-183. [PMID: 34092624 DOI: 10.3233/ch-211166] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Atherosclerosis is a major cause of cardiovascular disease, in which vascular smooth muscle cells (VSMCs) proliferation and migration play a vital role. Circular RNAs (circRNAs) have been reported to be correlated with the VSMCs function. Therefore, this study is designed to explore the role and mechanism of circRNA lipase maturation factor 1 (circLMF1) in Human aortic VSMCs (HASMCs). The microarray was used for detecting the expression of circLMF1 in proliferative and quiescent HASMCs. Levels of circLMF1, microRNA-125a-3p (miR-125a-3p), vascular endothelial growth factor A (VEGFA), and fibroblast growth factor 1 (FGF1) were determined by real-time quantitative polymerase chain reaction (RT-qPCR). Cell viability, cell cycle progression, and migration were assessed by Cell Counting Kit-8 (CCK-8), flow cytometry, wound healing, and transwell assays, respectively. Western blot assay determined proliferating cell nuclear antigen (PCNA), Cyclin D1, matrix metalloproteinase (MMP2), osteopontin (OPN), VEGFA, and FGF1 protein levels. The possible interactions between miR-125a-3p and circLMF1, and miR-125a-3p and VEGFA or FGF1 were predicted by circbank or targetscan, and then verified by a dual-luciferase reporter, RNA Immunoprecipitation (RIP), RNA pull-down assays. CircLMF1, VEGFA, and FGF1 were increased, and miR-125a-3p was decreased in platelet-derived growth factor-BB (PDGF-BB)-inducted HASMCs. Functionally, circLMF1 knockdown hindered cell viability, cell cycle progression, and migration in PDGF-BB-treated HASMCs. Mechanically, circLMF1 could regulate VEGFA or FGF1 expression through sponging miR-125a-3p. Our findings revealed that circLMF1 deficiency could inhibit cell viability, cell cycle progression, and migration of PDGF-BB stimulated atherosclerosis model partly through the miR-125a-3p/VEGFA or FGF1 axis, suggesting that targeting circLMF1 can be a feasible therapeutic strategy for atherosclerosis.
Collapse
Affiliation(s)
- Yanping Yang
- Department of Cardiac Surgery, The Cardio-Cerebro Vascular Disease Specialist Hospital of Qinghai Province, Xining City, China
| | - Wenkai Mao
- Department of Cardiac Surgery, The Cardio-Cerebro Vascular Disease Specialist Hospital of Qinghai Province, Xining City, China
| | - Liming Wang
- Department of Cardiac Surgery, The Cardio-Cerebro Vascular Disease Specialist Hospital of Qinghai Province, Xining City, China
| | - Lin Lu
- Department of Cardiac Surgery, The Cardio-Cerebro Vascular Disease Specialist Hospital of Qinghai Province, Xining City, China
| | - Yunfeng Pang
- Department of Cardiac Surgery, The Cardio-Cerebro Vascular Disease Specialist Hospital of Qinghai Province, Xining City, China
| |
Collapse
|
15
|
Chen X, Wang D, Qian L. LncRNA Fetal-Lethal Noncoding Developmental Regulatory RNA (FENDRR) Suppresses Cell Proliferation and Promotes Apoptosis in Platelet Derived Growth Factor BB/Tumor Necrosis Factor α Induced Vascular Smooth Muscle Cells. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Atherosclerosis is one of the primary causes that lead to cardiovascular disease. LncRNAs have been regarded as key modulators in many pathological processes. The study aims to identify the regulatory role of LncRNA fetal-lethal noncoding developmental regulatory RNA (FENDRR) in atherosclerosis.
Cell viability proliferation, cell cycle and cell apoptosis were evaluated by Cell Counting Kit-8 (CCK-8) assay flow cytometric analysis and western blot analysis. Quantitative real-time PCR (qRT-PCR) was carried out to determine FENDRR expression in PDGF-BB/TNF-α induced VSMCs.
Levels of TNF-α, IL-1, IL-6, MCP-1 and ICAM-1 were investigated by enzyme-linked immunosorbent assay (ELISA). The results showed that cell viability was enhanced and FENDRR expression was downregulated after VSMCs were induced by platelet derived growth factor BB (PDGF-BB) or
tumor necrosis factor a (TNF-α). Cell proliferation was inhibited by FENDRR overexpression in a time-dependent manner in PDGF-BB or TNF-α induced VSMCs. Moreover, FENDRR overexpression blocked cell cycle, suppressed the generations of TNF-α, IL-1, IL-6,
MCP-1 and ICAM-1, and facilitated cell apoptosis in VSMCs induced by PDGF-BB or TNF-α. These findings indicate the functional role of LncRNA FENDRR in atherosclerosis that attenuates cell proliferation and accelerates cell apoptosis.
Collapse
Affiliation(s)
- Xiaofang Chen
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Dongjin Wang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Lingmei Qian
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
16
|
Liu Y, Liu P, Song Y, Li S, Shi Y, Quan K, Yu G, Li P, An Q, Zhu W. A heparin-rosuvastatin-loaded P(LLA-CL) nanofiber-covered stent inhibits inflammatory smooth-muscle cell viability to reduce in-stent stenosis and thrombosis. J Nanobiotechnology 2021; 19:123. [PMID: 33926468 PMCID: PMC8086342 DOI: 10.1186/s12951-021-00867-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/20/2021] [Indexed: 11/10/2022] Open
Abstract
Background An endovascular covered-stent has unique advantages in treating complex intracranial aneurysms; however, in-stent stenosis and late thrombosis have become the main factors affecting the efficacy of covered-stent treatment. Smooth-muscle-cell phenotypic modulation plays an important role in late in-stent stenosis and thrombosis. Here, we determined the efficacy of using covered stents loaded with drugs to inhibit smooth-muscle-cell phenotypic modulation and potentially lower the incidence of long-term complications. Methods Nanofiber-covered stents were prepared using coaxial electrospinning, with the core solution prepared with 15% heparin and 20 µM rosuvastatin solution (400: 100 µL), and the shell solution prepared with 120 mg/mL hexafluoroisopropanol. We established a rabbit carotid-artery aneurysm model, which was treated with covered stents. Angiography and histology were performed to evaluate the therapeutic efficacy and incidence rate of in-stent stenosis and thrombosis. Phenotype, function, and inflammatory factors of smooth-muscle cells were studied to explore the mechanism of rosuvastatin action in smooth-muscle cells. Result Heparin–rosuvastatin-loaded nanofiber scaffold mats inhibited the proliferation of synthetic smooth-muscle cells, and the nanofiber-covered stent effectively treated aneurysms in the absence of notable in-stent stenosis. Additionally, in vitro experiments showed that rosuvastatin inhibited the smooth-muscle-cell phenotypic modulation of platelet-derived growth factor-BB induction and decreased synthetic smooth-muscle-cell viability, as well as secretion of inflammatory cytokines. Conclusion Rosuvastatin inhibited the abnormal proliferation of synthetic smooth-muscle cells, and heparin–rosuvastatin-loaded covered stents reduced the incidence of stenosis and late thrombosis, thereby improving the healing rates of stents used for aneurysm treatment. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00867-8.
Collapse
Affiliation(s)
- Yingjun Liu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Yaying Song
- Department of Neurology, Renji Hospital of Shanghai Jiao Tong University, Shanghai, China.,Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Sichen Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Yuan Shi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Kai Quan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Guo Yu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
| | - Peiliang Li
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China. .,Neurosurgical Institute of Fudan University, Shanghai, China. .,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China. .,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
| | - Qingzhu An
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China. .,Neurosurgical Institute of Fudan University, Shanghai, China. .,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China. .,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China. .,Neurosurgical Institute of Fudan University, Shanghai, China. .,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China. .,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
| |
Collapse
|
17
|
Tuo H, Yao B, He B, Yu S, Li D, Li W, Jin L. Silence of Insulin-Like Growth Factor 2 mRNA-Binding Protein 1 Prevents Vascular Smooth Muscle Cells Proliferation via Nuclear Factor of Activated T Cells Isoform-3/Ca 2+/Calmodulin Pathway. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Increased proliferation of vascular smooth muscle cells (VSMCs) contributes to the pathogenesis of atherosclerosis (AS), and the insulin like growth factor 2 (IGF2) is involved in AS through effects on VSMCs growth and migration. The IGF2 mRNA-binding protein 1(IGF2BP1) is a secreted
protein that can bind to IGF2 and regulate its localization, however, whether IGF2BP1 could regulate VSMCs proliferation remains to be elucidated. This study aimed to investigate the role of IGF2BP1 in VSMCs proliferation and uncover the potential mechanism. Primary human aortic VSMCs that
transfected with or without shRNA-IGF2BP1 were stimulated by platelet-derived growth factor-BB (PDGF-BB), and then cell proliferation, intracellular Ca2+ level, cell apoptosis and the expression of IGF2BP1, calmodulin (CaM) and cell cycle-related proteins were detected. RNA pull
down assay was used to determine the interaction between IGF2BP1 and nuclear factor of activated T cells isoform-3 (NFATc3). We found that PDGF-BB promoted cell proliferation and enhanced IGF2BP1 protein expression in a concentration-dependent manner. The 10 μg/L PDGF-BB significantly increased
intracellular Ca2+ level, NFATc3, CaM and calcineurin A protein expression, TUNEL-positive cells, and the expression of cell cycle-related proteins cyclin D1/E1/B1. However, knockdown of IGF2BP1 significantly blunted all these effects induced by PDGF-BB. In addition, IGF2BP1 could
bind to NFATc3 RNA. Collectively, knockdown of IGF2BP1 could inhibit PDGFBB- induced VSMCs proliferation via targeting NFATc3/Ca2+/calmodulin pathway and disturbing the effect of NFATc3/ on cell cycle.
Collapse
Affiliation(s)
- Hu Tuo
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Baozhen Yao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Bing He
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Shiqian Yu
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Danni Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Wenjing Li
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| | - Lin Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, RP China
| |
Collapse
|
18
|
Ishii S, Ashino T, Fujimori H, Numazawa S. Reactive sulfur species inhibit the migration of PDGF-treated vascular smooth muscle cells by blocking the reactive oxygen species-regulated Akt signaling pathway. Free Radic Res 2021; 55:186-197. [PMID: 33641584 DOI: 10.1080/10715762.2021.1887485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Vascular smooth muscle cell (VSMC) migration contributes to vascular remodeling after injury, whereas oxidative stress generated through dysfunctional redox homeostasis induces hypermigration, leading to arteriosclerosis. Platelet-derived growth factor (PDGF)-induced reactive oxygen species (ROS) serve as intracellular signaling molecules in VSMCs. Reactive sulfur species (RSS) may serve as a biological defense system because of the antioxidative properties of highly nucleophilic sulfane sulfur. However, insufficient information is available on its function in PDGF-induced VSMC migration. Here we show that PDGF significantly increased the levels of intracellular sulfane sulfur and that intracellular sulfane sulfur donors, donor 5a and Na2S4, inhibited the increase in ROS levels in PDGF-treated VSMCs and inhibited their migration. Consistent with the migration results, sulfane sulfur donors inhibited Akt phosphorylation, a downstream signaling molecule in the PDGF cascade, without affecting the autophosphorylation of PDGF receptor-β. Further, sulfane sulfur donors inhibited vinculin and paxillin recruitment to the leading edge of VSMCs in response to PDGF to decrease focal adhesion formation. These findings suggest that RSS are required for PDGF-stimulated VSMC migration through the regulation of the ROS-regulated Akt pathway, which may contribute to focal adhesion formation. Our findings provide insight into RSS as novel regulators of vascular redox homeostasis.
Collapse
Affiliation(s)
- Shunichi Ishii
- Division of Toxicology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan.,Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Takashi Ashino
- Division of Toxicology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan.,Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Hiroki Fujimori
- Division of Toxicology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan.,Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Satoshi Numazawa
- Division of Toxicology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan.,Pharmacological Research Center, Showa University, Tokyo, Japan
| |
Collapse
|
19
|
Fan K, Ruan X, Wang L, Lu W, Shi Q, Xu Y. Circ_0004872 promotes platelet-derived growth factor-BB-induced proliferation, migration and dedifferentiation in HA-VSMCs via miR-513a-5p/TXNIP axis. Vascul Pharmacol 2021; 140:106842. [PMID: 33592319 DOI: 10.1016/j.vph.2021.106842] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/21/2021] [Accepted: 02/08/2021] [Indexed: 01/22/2023]
Abstract
The proliferation, migration and dedifferentiation of vascular smooth muscle cells (VSMCs) exert crucial roles in atherosclerosis (AS) progression. The aim of our study was to explore the influences of circular RNA 0004872 (circ_0004872) in platelet-derived growth factor-BB (PDGF-BB)-induced AS cell model and investigate the underlying mechanisms. Real-time quantitative polymerase chain reaction (RT-qPCR) was implemented for the expression detection of circ_0004872, mitogen-activated protein kinase 1 (MAPK1) messenger RNA (mRNA), microRNA-513a-5p (miR-513a-5p) and thioredoxin interacting protein (TXNIP). Cell proliferation was analyzed via Cell Counting Kit 8 (CCK8) assay. Cell migration was assessed via wound healing assay and transwell migration assay. Western blot assay was used to measure the expression of alpha smooth muscle actin (α-SMA), osteopontin (OPN), calponin and TXNIP. Dual-luciferase reporter assay and RNA-pull down assay were used for confirmation of interaction between miR-513a-5p and circ_0004872 or TXNIP. Circ_0004872 expression was elevated in PDGF-BB-induced human aortic vascular smooth muscle cells (HA-VSMCs) and carotid plaque tissues. Circ_0004872 silencing alleviated PDGF-BB-induced proliferation, migration and dedifferentiation in HA-VSMCs. MiR-513a-5p bound to circ_0004872, and circ_0004872 knockdown-induced effects in PDGF-BB-treated HA-VSMCs were largely attenuated by the silencing of miR-513a-5p. MiR-513a-5p bound to the 3' untranslated region (3'UTR) of TXNIP, and miR-513a-5p overexpression-mediated effects were counteracted by the transfection of pcDNA-TXNIP in PDGF-BB-induced HA-VSMCs. TXNIP was modulated by circ_0004872/miR-513a-5p signaling cascade in HA-VSMCs. Circ_0004872 accelerated PDGF-BB-induced proliferation, migration and dedifferentiation in HA-VSMCs through enhancing TXNIP level via sponging miR-513a-5p.
Collapse
Affiliation(s)
- Kaikai Fan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Xinhua Ruan
- Department of Cardiovascular Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Leilei Wang
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Wanli Lu
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospitale, Tianjin, China
| | - Qiangwei Shi
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yawei Xu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Gao C, Zhang K, Liang F, Ma W, Jiang X, Wang H, Zhan H, Sonkoly E, Hu H, Zhao Z. Inhibition of the Ras/ERK1/2 pathway contributes to the protective effect of ginsenoside Re against intimal hyperplasia. Food Funct 2021; 12:6755-6765. [PMID: 34116563 DOI: 10.1039/d1fo00015b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neointimal hyperplasia is the major cause of carotid stenosis after vascular injury, which restricts the long-term efficacy of endovascular treatment and endarterectomy in preventing stenosis. Ginsenoside Re (Re) is a major active ingredient of ginseng having multifaceted pharmacological effects on the cardiovascular system, and is a potential treatment for restenosis. In this study, we demonstrated that Re treatment significantly inhibited vascular injury-induced neointimal thickening, reduced the intimal area and intima/media (I/M) ratio, increased the lumen area, and inhibited pro-inflammatory cytokines. In cultured A7R5 cells, Re inhibited LPS-induced proliferation and migration as evidenced by suppressed colony formation and shortened migration distance, accompanied by the downregulated expression of pro-inflammatory cytokines. Re promoted VSMC apoptosis induced by balloon injury in vivo and LPS challenge in vitro. Moreover, Re inhibited autophagy in VSMCs evoked by balloon injury and LPS as supported by reduced LC3II and increased p62 expressions. Suppression of autophagy with the specific autophagy inhibitor spautin-1 efficiently inhibited LPS-induced cell proliferation and inflammation and promoted caspase-3/7 activities. Mechanistically, we found that Re attenuated Ras/ERK1/2 expression in VSMCs in vivo and in vitro. The MEK1/2 inhibitor PD98059 showed similar effects to Re on cell proliferation, migration, apoptosis, and the levels of autophagy and cytokines. In conclusion, we provided significant evidence that Re inhibited vascular injury-induced neointimal thickening probably by promoting VSMC apoptosis and inhibiting autophagy via suppression of the Ras/MEK/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Chenying Gao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bee Venom in Wound Healing. MOLECULES (BASEL, SWITZERLAND) 2020; 26:molecules26010148. [PMID: 33396220 PMCID: PMC7795515 DOI: 10.3390/molecules26010148] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/20/2020] [Accepted: 12/24/2020] [Indexed: 01/05/2023]
Abstract
Bee venom (BV), also known as api-toxin, is widely used in the treatment of different inflammatory diseases such as rheumatoid arthritis or multiple sclerosis. It is also known that BV can improve the wound healing process. BV plays a crucial role in the modulation of the different phases of wound repair. It possesses anti-inflammatory, antioxidant, antifungal, antiviral, antimicrobial and analgesic properties, all of which have a positive impact on the wound healing process. The mentioned process consists of four phases, i.e., hemostasis, inflammation, proliferation and remodeling. The impaired wound healing process constitutes a significant problem especially in diabetic patients, due to hypoxia state. It had been found that BV accelerated the wound healing in diabetic patients as well as in laboratory animals by impairing the caspase-3, caspase-8 and caspase-9 activity. Moreover, the activity of BV in wound healing is associated with regulating the expression of transforming growth factor (TGF-β1), vascular endothelial growth factor and increased collagen type I. BV stimulates the proliferation and migration of human epidermal keratinocytes and fibroblasts. In combination with polyvinyl alcohol and chitosan, BV significantly accelerates the wound healing process, increasing the hydroxyproline and glutathione and lowering the IL-6 level in wound tissues. The effect of BV on the wounds has been proved by numerous studies, which revealed that BV in the wound healing process brings about a curative effect and could be applied as a new potential treatment for wound repair. However, therapy with bee venom may induce allergic reactions, so it is necessary to assess the existence of the patient’s hypersensitivity to apitoxin before treatment.
Collapse
|
22
|
Park J, Jang KM, Park KK. Apamin Suppresses LPS-Induced Neuroinflammatory Responses by Regulating SK Channels and TLR4-Mediated Signaling Pathways. Int J Mol Sci 2020; 21:4319. [PMID: 32560481 PMCID: PMC7352249 DOI: 10.3390/ijms21124319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/11/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation plays a vital role in neurodegenerative conditions. Microglia are a key component of the neuroinflammatory response. There is a growing interest in developing drugs to target microglia and thereby control neuroinflammatory processes. Apamin (APM) is a specifically selective antagonist of small conductance calcium-activated potassium (SK) channels. However, its effect on neuroinflammation is largely unknown. We examine the effects of APM on lipopolysaccharide (LPS)-stimulated BV2 and rat primary microglial cells. Regarding the molecular mechanism by which APM significantly inhibits proinflammatory cytokine production and microglial cell activation, we found that APM does so by reducing the expression of phosphorylated CaMKII and toll-like receptor (TLR4). In particular, APM potently suppressed the translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/signal transducer and activator of transcription (STAT)3 and phosphorylated mitogen-activated protein kinases (MAPK)-extracellular signal-regulated kinase (ERK). In addition, the correlation of NF-κB/STAT3 and MAPK-ERK in the neuroinflammatory response was verified through inhibitors. The literature and our findings suggest that APM is a promising candidate for an anti-neuroinflammatory agent and can potentially be used for the prevention and treatment of various neurological disorders.
Collapse
Affiliation(s)
- Jihyun Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| | - Kyung Mi Jang
- Department of Pediatrics, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| |
Collapse
|
23
|
Activation of Inward Rectifier K + Channel 2.1 by PDGF-BB in Rat Vascular Smooth Muscle Cells through Protein Kinase A. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4370832. [PMID: 32461988 PMCID: PMC7212311 DOI: 10.1155/2020/4370832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 03/19/2020] [Indexed: 11/17/2022]
Abstract
Platelet-derived growth factor-BB (PDGF-BB) can induce the proliferation, migration, and phenotypic modulation of vascular smooth muscle cells (VSMCs). We used patch clamp methods to study the effects of PDGF-BB on inward rectifier K+ channel 2.1 (Kir2.1) channels in rat thoracic aorta VSMCs (RASMCs). PDGF-BB (25 ng/mL) increased Kir2.x currents (−11.81 ± 2.47 pA/pF, P < 0.05 vs. CON, n = 10). Ba2+(50 μM) decreased Kir2.x currents (−2.13 ± 0.23 pA/pF, P < 0.05 vs. CON, n = 10), which were promoted by PDGF-BB (−6.98 ± 1.03 pA/pF). PDGF-BB specifically activates Kir2.1 but not Kir2.2 and Kir2.3 channels in HEK-293 cells. The PDGF-BB-induced stimulation of Kir2.1 currents was blocked by the PDGF-BB receptor β (PDGF-BBRβ) inhibitor AG1295 and was not affected by the PDGF-BBRα inhibitor AG1296. The PDGF-BB-induced stimulation of Kir2.1 currents was blocked by the protein kinase A inhibitor Rp-8-CPT-cAMPs; however, the antagonist of protein kinase B (GSK690693) had marginal effects on current activity. The PDGF-BB-induced stimulation of Kir2.1 currents was enhanced by forskolin, an adenylyl cyclase (AC) activator, and was blocked by the AC inhibitor SQ22536. We conclude that PDGF-BB increases Kir2.1 currents via PDGF-BBRβ through activation of cAMP-PKA signaling in RASMCs.
Collapse
|
24
|
Gu H, Han SM, Park KK. Therapeutic Effects of Apamin as a Bee Venom Component for Non-Neoplastic Disease. Toxins (Basel) 2020; 12:195. [PMID: 32204567 PMCID: PMC7150898 DOI: 10.3390/toxins12030195] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023] Open
Abstract
Bee venom is a natural toxin produced by honeybees and plays an important role in defending bee colonies. Bee venom has several kinds of peptides, including melittin, apamin, adolapamine, and mast cell degranulation peptides. Apamin accounts for about 2%-3% dry weight of bee venom and is a peptide neurotoxin that contains 18 amino acid residues that are tightly crosslinked by two disulfide bonds. It is well known for its pharmacological functions, which irreversibly block Ca2+-activated K+ (SK) channels. Apamin regulates gene expression in various signal transduction pathways involved in cell development. The aim of this study was to review the current understanding of apamin in the treatment of apoptosis, fibrosis, and central nervous system diseases, which are the pathological processes of various diseases. Apamin's potential therapeutic and pharmacological applications are also discussed.
Collapse
Affiliation(s)
- Hyemin Gu
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| | - Sang Mi Han
- National Academy of Agricultural Science, Jeonjusi, Jeonbuk 54875, Korea;
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| |
Collapse
|
25
|
Su B, Zhang X, Luo G. Homeobox B5 suppression attenuates proliferation and elevates apoptosis in hepatoma cell lines through ERK/MDM2 signalling. Clin Exp Pharmacol Physiol 2020; 47:1058-1066. [PMID: 32037602 DOI: 10.1111/1440-1681.13278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022]
Abstract
Homeobox B5 (HOXB5), a member of the HOX gene family, is an important gene in tumourigenesis. However, its role in hepatocellular carcinoma (HCC) cell proliferation and apoptosis remains unclear. In this study, we investigated the role and regulation mechanism of HOXB5 in HCC cell lines Hep3B and LM6. The data indicated high expression of HOXB5 in HCC tissues and cell lines. In HCC cells, inhibition of HOXB5 by transfection with HOXB5 siRNA significantly constrained cell viability, and Bcl-2 levels, and it increased cell apoptosis, cytochrome c levels, BAX levels, and caspase-3 activity. On the contrary, HOXB5 overexpression increased proliferation and Bcl-2 levels but inhibited BAX levels and caspase-3 activity in these cells. HOXB5 downregulation attenuated activation of extracellular signal-regulated kinase (ERK) and expression of the murine double minute 2 (MDM2) oncogene. Incubation with the ERK activator, phorbol 12-myristate 13-acetate (40 μmol/L), for 12 hours reversed the effects of HOXB5 inhibition on MDM2 expression, cell proliferation, and apoptosis in HCC cells. Overall, this study demonstrated that HOXB5 inhibition regulated MDM2 expression by controlling ERK activation and that it modulated proliferation and apoptosis in HCC cells.
Collapse
Affiliation(s)
- Baowei Su
- Department of Hepatobiliary Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Xueli Zhang
- Department of Hepatobiliary Surgery, Liaocheng People's Hospital, Liaocheng, China
| | - Guang Luo
- Department of Radiology, Qingdao No. 6 People's Hospital, Qingdao, China
| |
Collapse
|
26
|
Wang YT, Chen J, Li X, Umetani M, Chen Y, Li PL, Zhang Y. Contribution of transcription factor EB to adipoRon-induced inhibition of arterial smooth muscle cell proliferation and migration. Am J Physiol Cell Physiol 2019; 317:C1034-C1047. [PMID: 31483704 PMCID: PMC6879882 DOI: 10.1152/ajpcell.00294.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/11/2022]
Abstract
Abnormal vascular smooth muscle cell (SMC) dedifferentiation with increased proliferation and migration during pathological vascular remodeling is associated with vascular disorders, such as atherosclerosis and in-stent restenosis. AdipoRon, a selective agonist of adiponectin receptor, has been shown to protect against vascular remodeling by preventing SMC dedifferentiation. However, the molecular mechanisms that mediate adipoRon-induced SMC differentiation are not well understood. The present study aimed to elucidate the role of transcription factor EB (TFEB), a master regulator of autophagy, in mediating adipoRon's effect on SMCs. In cultured arterial SMCs, adipoRon dose-dependently increased TFEB activation, which is accompanied by upregulated transcription of genes involved in autophagy pathway and enhanced autophagic flux. In parallel, adipoRon suppressed serum-induced cell proliferation and caused cell cycle arrest. Moreover, adipoRon inhibited SMC migration as characterized by wound-healing retardation, F-actin reorganization, and matrix metalloproteinase-9 downregulation. These inhibitory effects of adipoRon on proliferation and migration were attenuated by TFEB gene silencing. Mechanistically, activation of TFEB by adipoRon is dependent on intracellular calcium, but it is not associated with changes in AMPK, ERK1/2, Akt, or molecular target of rapamycin complex 1 activation. Using ex vivo aortic explants, we demonstrated that adipoRon inhibited sprouts that had outgrown from aortic rings, whereas lentiviral TFEB shRNA transduction significantly reversed this effect of adipoRon on aortic rings. Taken together, our results indicate that adipoRon activates TFEB signaling that helps maintain the quiescent and differentiated status of arterial SMCs, preventing abnormal SMC dedifferentiation. This study provides novel mechanistic insights into understanding the therapeutic effects of adipoRon on TFEB signaling and pathological vascular remodeling.
Collapse
Affiliation(s)
- Yun-Ting Wang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Jiajie Chen
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Michihisa Umetani
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Yang Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
27
|
Wang YT, Li X, Chen J, McConnell BK, Chen L, Li PL, Chen Y, Zhang Y. Activation of TFEB ameliorates dedifferentiation of arterial smooth muscle cells and neointima formation in mice with high-fat diet. Cell Death Dis 2019; 10:676. [PMID: 31515484 PMCID: PMC6742653 DOI: 10.1038/s41419-019-1931-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/09/2019] [Accepted: 08/26/2019] [Indexed: 12/30/2022]
Abstract
Autophagy is recently implicated in regulating vascular smooth muscle cell (SMC) homeostasis and in the pathogenesis of vascular remodeling. Transcription factor EB (TFEB) is a master regulator of autophagy signaling pathways. However, the molecular mechanisms and functional roles of TFEB in SMC homeostasis have not been elucidated. Here, we surveyed the ability of TFEB to regulate autophagy pathway in SMCs, and whether pharmacological activation of TFEB favors SMC homeostasis preventing dedifferentiation and pathogenic vascular remodeling. In primary cultured SMCs, TFEB activator trehalose induced nuclear translocation of TFEB and upregulation of TFEB-controlled autophagy genes leading to enhanced autophagy signaling. Moreover, trehalose suppressed serum-induced SMC dedifferentiation to synthetic phenotypes as characterized by inhibited proliferation and migration. These effects of trehalose were mimicked by ectopic upregulation of TFEB and inhibited by TFEB gene silencing. In animal experiments, partial ligation of carotid arteries induced downregulation of TFEB pathway in the media layer of these arteries. Such TFEB suppression was correlated with increased SMC dedifferentiation and aggravated high-fat diet (HFD)-induced neointima formation. Treatment of mice with trehalose reversed this TFEB pathway suppression, and prevented SMC dedifferentiation and HFD-induced neointima formation. In conclusion, our findings have identified TFEB as a novel positive regulator for autophagy pathway and cellular homeostasis in SMCs. Our data suggest that suppression of TFEB may be an initiating mechanism that promotes SMC dedifferentiation leading to accelerated neointima formation in vascular disorders associated with metabolic stress, whereas trehalose reverses these changes. These findings warrant further evaluation of trehalose in the clinical settings.
Collapse
Affiliation(s)
- Yun-Ting Wang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Jiajie Chen
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Bradley K McConnell
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Li Chen
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yang Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA.
| |
Collapse
|
28
|
MicroRNA-125a-3p affects smooth muscle cell function in vascular stenosis. J Mol Cell Cardiol 2019; 136:85-94. [PMID: 31499051 DOI: 10.1016/j.yjmcc.2019.08.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 08/04/2019] [Accepted: 08/31/2019] [Indexed: 11/21/2022]
Abstract
AIMS Many studies have indicated that microRNAs are closely related to the process of peripheral arterial disease (PAD). Previously, we found that microRNA-125a-3p (miR-125a-3p) in restenotic arteries after interventional therapy of lower extremity vessels was notably decreased compared with that of normal control arteries. However, its role in the development of vascular stenosis is not yet clearly understood. The purpose of this study was to investigate the expression, regulatory mechanism and function of miR-125a-3p in the process of vascular stenosis. METHODS AND RESULTS Quantitative reverse-transcription polymerase chain reaction assays indicated that miR-125a-3p in restenotic arteries after interventional therapy was significantly lower than that in normal control arteries. Immunofluorescence and in situ hybridization co-staining assays in arterial sections demonstrated that miR-125a-3p was mainly expressed in the medial smooth muscle layer. Transfection of miR-125a-3p mimics into cultured vascular smooth muscle cells (VSMCs) effectively inhibited cell proliferation and migration. Then, western blot and luciferase activity assays showed that recombinant human mitogen-activated protein kinase 1 (MAPK1) was a functional target of miR-125a-3p and was involved in miR-125a-3p-mediated cell effects. Finally, the lentiviral infection of miR-125a-3p in balloon-injured rat carotid vascular walls showed that miR-125a-3p overexpression significantly reduced the probability of neointimal membrane production. CONCLUSIONS miR-125a-3p can effectively inhibit the function of VSMCs and the occurrence of vascular stenosis by targeting MAPK1. This study introduces a new molecular mechanism of PAD. We show that regulation of the miR-125a-3p level has the potential to provide a new treatment for PAD and other proliferative vascular diseases.
Collapse
|
29
|
Bee Venom: Overview of Main Compounds and Bioactivities for Therapeutic Interests. Molecules 2019; 24:molecules24162997. [PMID: 31430861 PMCID: PMC6720840 DOI: 10.3390/molecules24162997] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 11/17/2022] Open
Abstract
Apitherapy is an alternate therapy that relies on the usage of honeybee products, most importantly bee venom for the treatment of many human diseases. The venom can be introduced into the human body by manual injection or by direct bee stings. Bee venom contains several active molecules such as peptides and enzymes that have advantageous potential in treating inflammation and central nervous system diseases, such as Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis. Moreover, bee venom has shown promising benefits against different types of cancer as well as anti-viral activity, even against the challenging human immunodeficiency virus (HIV). Many studies described biological activities of bee venom components and launched preclinical trials to improve the potential use of apitoxin and its constituents as the next generation of drugs. The aim of this review is to summarize the main compounds of bee venom, their primary biological properties, mechanisms of action, and their therapeutic values in alternative therapy strategies.
Collapse
|
30
|
Zhang W, Liu D, Han X, Ren J, Zhou P, Ding P. MicroRNA-451 inhibits vascular smooth muscle cell migration and intimal hyperplasia after vascular injury via Ywhaz/p38 MAPK pathway. Exp Cell Res 2019; 379:214-224. [PMID: 30930138 DOI: 10.1016/j.yexcr.2019.03.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 01/09/2023]
Abstract
Increasing evidence has indicated that intimal hyperplasia is a common event in the pathophysiology of many vascular diseases including atherosclerosis (AS). Recently, deregulated microRNAs (miRNAs) have been reported to be associated with the pathophysiology of AS. However, the biological function and regulatory mechanisms of miRNAs in intimal hyperplasia in AS remain largely unclear. The aim of this study was to investigate the effects of miRNAs on intimal hyperplasia and reveal the underlying mechanisms of their effects. Firstly, the model of rat vascular injury was successfully constructed in vivo. Then, the miRNAs expression profiles were analyzed by miRNA microarray. It was observed that miR-451 was significantly downregulated in injury carotid arteries. Subsequently, we investigated miR-451 function and found that upregulation of miR-451 by agomir-451 improves intimal thickening in rats following vascular injury. It was also observed that miR-451 was downregulated in the VSMCs following platelet-derived growth factor type BB (PDGF-BB) stimulation. The upregulation of miR-451 attenuated PDGF-BB-induced VSMCs injury, as evidenced by inhibition of proliferation, invasion and migration. Besides, overexpression of miR-451 blocked the activation of p38 MAPK signaling pathway in PDGF-BB treated VSMCs, as demonstrated by the downregulation of phosphorylated (p-) p38. In addition, Ywhaz, a positive regulator of p38 MAPK signaling pathway, was found to be a direct target of miR-451 in the VSMCs and this was validated using a luciferase reporter assay. Overexpression of Ywhaz partially abolished the inhibitory effects of miR-451 overexpression on PDGF-BB induced VSMCs injury. Collectively, these findings indicated that miR-451 protected intimal hyperplasia and PDGF-BB-induced VSMCs injury by Ywhaz/p38 MAPK pathway, and miR-451 may be considered as a potential therapeutic target in the treatment of AS.
Collapse
Affiliation(s)
- Wenguang Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dongmei Liu
- Department of Radiation Oncology, Henan Province Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Jianzhuang Ren
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Pengli Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Pengxu Ding
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
31
|
Adela R, Reddy PNC, Ghosh TS, Aggarwal S, Yadav AK, Das B, Banerjee SK. Serum protein signature of coronary artery disease in type 2 diabetes mellitus. J Transl Med 2019; 17:17. [PMID: 30674322 PMCID: PMC6345069 DOI: 10.1186/s12967-018-1755-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Abstract
Background Coronary artery disease (CAD) is the leading cause of morbidity and mortality in patients with type 2 diabetes mellitus (T2DM). The purpose of the present study was to discriminate the Indian CAD patients with or without T2DM by using multiple pathophysiological biomarkers. Methods Using sensitive multiplex protein assays, we assessed 46 protein markers including cytokines/chemokines, metabolic hormones, adipokines and apolipoproteins for evaluating different pathophysiological conditions of control, T2DM, CAD and T2DM with CAD patients (T2DM_CAD). Network analysis was performed to create protein-protein interaction networks by using significantly (p < 0.05) altered protein markers in each disease using STRING 10.5 database. We used two supervised analysis methods i.e., between class analysis (BCA) and principal component analysis (PCA) to reveals distinct biomarkers profiles. Further, random forest classification (RF) was used to classify the diseases by the panel of markers. Results Our two supervised analysis methods BCA and PCA revealed a distinct biomarker profiles and high degree of variability in the marker profiles for T2DM_CAD and CAD. Thereafter, the present study identified multiple potential biomarkers to differentiate T2DM, CAD, and T2DM_CAD patients based on their relative abundance in serum. RF classified T2DM based on the abundance patterns of nine markers i.e., IL-1β, GM-CSF, glucagon, PAI-I, rantes, IP-10, resistin, GIP and Apo-B; CAD by 14 markers i.e., resistin, PDGF-BB, PAI-1, lipocalin-2, leptin, IL-13, eotaxin, GM-CSF, Apo-E, ghrelin, adipsin, GIP, Apo-CII and IP-10; and T2DM _CAD by 12 markers i.e., insulin, resistin, PAI-1, adiponectin, lipocalin-2, GM-CSF, adipsin, leptin, Apo-AII, rantes, IL-6 and ghrelin with respect to the control subjects. Using network analysis, we have identified several cellular network proteins like PTPN1, AKT1, INSR, LEPR, IRS1, IRS2, IL1R2, IL6R, PCSK9 and MYD88, which are responsible for regulating inflammation, insulin resistance, and atherosclerosis. Conclusion We have identified three distinct sets of serum markers for diabetes, CAD and diabetes associated with CAD in Indian patients using nonparametric-based machine learning approach. These multiple marker classifiers may be useful for monitoring progression from a healthy person to T2DM and T2DM to T2DM_CAD. However, these findings need to be further confirmed in the future studies with large number of samples. Electronic supplementary material The online version of this article (10.1186/s12967-018-1755-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ramu Adela
- Drug Discovery Research Center, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India.,Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, NIPER, Guwahati, Assam, India
| | | | - Tarini Shankar Ghosh
- Centre for Human Microbial Ecology, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Suruchi Aggarwal
- Drug Discovery Research Center, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Amit Kumar Yadav
- Drug Discovery Research Center, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Bhabatosh Das
- Centre for Human Microbial Ecology, Translational Health Science and Technology Institute, Faridabad, 121001, India
| | - Sanjay K Banerjee
- Drug Discovery Research Center, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India.
| |
Collapse
|
32
|
Abd El-Wahed AA, Khalifa SA, Sheikh BY, Farag MA, Saeed A, Larik FA, Koca-Caliskan U, AlAjmi MF, Hassan M, Wahabi HA, Hegazy MEF, Algethami AF, Büttner S, El-Seedi HR. Bee Venom Composition: From Chemistry to Biological Activity. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2019:459-484. [DOI: 10.1016/b978-0-444-64181-6.00013-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
33
|
Contribution of p62/SQSTM1 to PDGF-BB-induced myofibroblast-like phenotypic transition in vascular smooth muscle cells lacking Smpd1 gene. Cell Death Dis 2018; 9:1145. [PMID: 30451833 PMCID: PMC6242941 DOI: 10.1038/s41419-018-1197-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 12/17/2022]
Abstract
Accumulating evidence indicates a critical role of autophagy in regulating vascular smooth muscle cell (SMC) homeostasis in atherogenesis. However, little is known about the modulatory role of autophagy in PDGF-BB-induced SMC transition towards the synthetic phenotype and extracellular matrix remodeling. We recently demonstrated that acid sphingomyelinase (ASM, encoded by Smpd1 gene) controls autophagy maturation in coronary arterial SMCs. Here, we demonstrate that PDGF-BB stimulation causes a myofibroblast-like non-canonical synthetic phenotype transition in Smpd1−/− SMCs. These non-canonical phenotypic changes induced by PDGF-BB in Smpd1−/− SMCs were characterized by increased expression of fibroblast-specific protein (FSP-1), massive deposition of collagen type I, decreased cell size, elevated inflammatory status with enhanced cytokine release and adhesion molecule expression. Mechanistically, PDGF-BB induces prolonged Akt activation that causes decreased autophagosome biogenesis and thereby exaggerates p62/SQSTM1 accumulation in Smpd1−/− SMCs. More importantly, Akt inhibition or p62/SQSTM1 gene silencing attenuates PDGF-BB-induced phenotypic changes in Smpd1−/− SMCs. This first demonstration of a p62/SQSTM1-dependent myofibroblast-like phenotypic transition in Smpd1−/− SMCs suggests that ASM-mediated autophagy pathway contributes to maintaining the arterial smooth muscle homeostasis in situation of vascular remodeling during atherosclerosis.
Collapse
|
34
|
Li DJ, Tong J, Zeng FY, Guo M, Li YH, Wang H, Wang P. Nicotinic ACh receptor α7 inhibits PDGF-induced migration of vascular smooth muscle cells by activating mitochondrial deacetylase sirtuin 3. Br J Pharmacol 2018; 176:4388-4401. [PMID: 30270436 DOI: 10.1111/bph.14506] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 09/07/2018] [Accepted: 09/13/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE PDGF-BB is an angiogenic factor involved in cardiovascular diseases. Here, we investigated the possible effects of activation of the nicotinic ACh receptor α7 subtype (α7nAChR) on PDGF-BB-induced proliferation and migration in vascular smooth muscle cells (VSMCs). EXPERIMENTAL APPROACH PNU-282987, a selective α7nAChR pharmacological agonist, was used to activate α7nAChR. The influences of α7nAChR activation on PDGF-BB-induced proliferation and migration, as well as the phosphorylation of focal adhesion kinase (FAK)/Src, a pro-migration signalling pathway, were determined in VSMCs. A variety of biochemical assays were applied to explore the underlying molecular mechanisms. KEY RESULTS PDGF-BB induced pronounced migration and proliferation of VSMCs. Activation of α7nAChRs by PNU-282987 blocked PDGF-BB-induced migration but not proliferation in wild-type (WT) VSMCs, whereas this effect was absent in α7nAChR-knockout VSMCs. Accordingly, PNU-282987 attenuated PDGF-BB-induced phosphorylation of FAKTyr397 and SrcTyr416 in WT VSMCs. Mechanistically, PNU-282987 suppressed PDGF-BB-induced oxidative stress, as demonstrated by the alterations in ROS, H2 O2 content, superoxide anion and total antioxidant activity. A sirtuin 3 (SIRT3) inhibitor 3-(1H-1,2,3-triazol-4-yl) pyridine or shRNA-mediated SIRT3 knockdown abolished the inhibitory effect of PNU-282987. PNU-282987 did not modulate SIRT3 protein expression but enhanced mitochondrial SIRT3 deacetylase activity. In line with this action, PNU-282987 enhanced the deacetylation of mitochondrial FoxO3. Lastly, PNU-282987 corrected the PDGF-BB-induced mitochondrial dysfunction by increasing mitochondrial citrate synthase activity, ATP content and nicotinamide adenine dinucleotide pool. CONCLUSIONS Pharmacological activation of α7nAChRs inhibits PDGF-BB-induced VSMC migration by activating the mitochondrial deacetylase SIRT3, implying an important role for α7nAChRs in mitochondria biology and PDGF-related diseases. LINKED ARTICLES This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.
Collapse
Affiliation(s)
- Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China.,Ninghai First Hospital, Zhejiang, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China.,Ninghai First Hospital, Zhejiang, China
| | - Fei-Yan Zeng
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China.,Ninghai First Hospital, Zhejiang, China
| | - Mengqi Guo
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China
| | - Yong-Hua Li
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China
| | - Pei Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China.,Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
35
|
Kudryashova TV, Shen Y, Pena A, Cronin E, Okorie E, Goncharov DA, Goncharova EA. Inhibitory Antibodies against Activin A and TGF-β Reduce Self-Supported, but Not Soluble Factors-Induced Growth of Human Pulmonary Arterial Vascular Smooth Muscle Cells in Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19102957. [PMID: 30274147 PMCID: PMC6212879 DOI: 10.3390/ijms19102957] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
Increased growth and proliferation of distal pulmonary artery vascular smooth muscle cells (PAVSMC) is an important pathological component of pulmonary arterial hypertension (PAH). Transforming Growth Factor-β (TGF-β) superfamily plays a critical role in PAH, but relative impacts of self-secreted Activin A, Gremlin1, and TGF-β on PAH PAVSMC growth and proliferation are not studied. Here we report that hyper-proliferative human PAH PAVSMC have elevated secretion of TGF-β1 and, to a lesser extent, Activin A, but not Gremlin 1, and significantly reduced Ser465/467-Smad2 and Ser423/425-Smad3 phosphorylation compared to controls. Media, conditioned by PAH PAVSMC, markedly increased Ser465/467-Smad2, Ser423/425-Smad3, and Ser463/465-Smad1/5 phosphorylation, up-regulated Akt, ERK1/2, and p38 MAPK, and induced significant proliferation of non-diseased PAVSMC. Inhibitory anti-Activin A antibody reduced PAH PAVSMC growth without affecting canonical (Smads) or non-canonical (Akt, ERK1/2, p38 MAPK) effectors. Inhibitory anti-TGF-β antibody significantly reduced P-Smad3, P-ERK1/2 and proliferation of PAH PAVSMC, while anti-Gremlin 1 had no anti-proliferative effect. PDGF-BB diminished inhibitory effects of anti-Activin A and anti-TGF-β antibodies. None of the antibodies affected growth and proliferation of non-diseased PAVSMC induced by PAH PAVSMC-secreted factors. Together, these data demonstrate that human PAH PAVSMC have secretory, proliferative phenotype that could be targeted by anti-Activin A and anti-TGF-β antibodies; potential cross-talk with PDGF-BB should be considered while developing therapeutic interventions.
Collapse
Affiliation(s)
- Tatiana V Kudryashova
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
| | - Yuanjun Shen
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
| | - Andressa Pena
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
| | - Emily Cronin
- Division of Mathematics and Sciences, Walsh University, North Canton, OH 44720, USA.
| | - Evelyn Okorie
- Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Dmitry A Goncharov
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
| | - Elena A Goncharova
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA.
- Division of Pulmonary, Allergy and Critical Care, University of Pittsburgh Department of Medicine, Pittsburgh, PA 15213, USA.
- University of Pittsburgh Department of Bioengineering, Pittsburgh, PA 15213, USA.
| |
Collapse
|
36
|
Zhang S, Liu Y, Ye Y, Wang XR, Lin LT, Xiao LY, Zhou P, Shi GX, Liu CZ. Bee venom therapy: Potential mechanisms and therapeutic applications. Toxicon 2018; 148:64-73. [PMID: 29654868 DOI: 10.1016/j.toxicon.2018.04.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/15/2018] [Accepted: 04/10/2018] [Indexed: 01/09/2023]
Abstract
Bee venom is a very complex mixture of natural products extracted from honey bee which contains various pharmaceutical properties such as peptides, enzymes, biologically active amines and nonpeptide components. The use of bee venom into the specific points is so called bee venom therapy, which is widely used as a complementary and alternative therapy for 3000 years. A growing number of evidence has demonstrated the anti-inflammation, the anti-apoptosis, the anti-fibrosis and the anti-arthrosclerosis effects of bee venom therapy. With these pharmaceutical characteristics, bee venom therapy has also been used as the therapeutic method in treating rheumatoid arthritis, amyotrophic lateral sclerosis, Parkinson's disease, Alzheimer's disease, liver fibrosis, atherosclerosis, pain and others. Although widely used, several cases still reported that bee venom therapy might cause some adverse effects, such as local itching or swelling. In this review, we summarize its potential mechanisms, therapeutic applications, and discuss its existing problems.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Yi Liu
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Yang Ye
- Department of Acupuncture and Moxibustion, Dongfang Hospital, Beijing University of Chinese Medicine, No. 6 Fangxingyuan 1st Block, Fengtai District, Beijing, 100078, China
| | - Xue-Rui Wang
- Department of Acupuncture and Moxibustion, Dongfang Hospital, Beijing University of Chinese Medicine, No. 6 Fangxingyuan 1st Block, Fengtai District, Beijing, 100078, China
| | - Li-Ting Lin
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Ling-Yong Xiao
- Department of Acupuncture and Moxibustion, Dongfang Hospital, Beijing University of Chinese Medicine, No. 6 Fangxingyuan 1st Block, Fengtai District, Beijing, 100078, China
| | - Ping Zhou
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Guang-Xia Shi
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Cun-Zhi Liu
- Department of Acupuncture and Moxibustion, Dongfang Hospital, Beijing University of Chinese Medicine, No. 6 Fangxingyuan 1st Block, Fengtai District, Beijing, 100078, China.
| |
Collapse
|
37
|
Dong X, Hu H, Fang Z, Cui J, Liu F. CTRP6 inhibits PDGF-BB-induced vascular smooth muscle cell proliferation and migration. Biomed Pharmacother 2018; 103:844-850. [PMID: 29710500 DOI: 10.1016/j.biopha.2018.04.112] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/09/2018] [Accepted: 04/16/2018] [Indexed: 10/17/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration play critical roles in the development and progression of atherosclerosis. C1q/tumor necrosis factor-related protein 6 (CTRP6), a member of CTRPs family, was involved in cardiovascular diseases, inflammatory reaction and adipogenesis. However, the role of CTRP6 in VSMCs remains largely unknown. The purpose of this study is to investigate the effects of CTRP6 on VSMC proliferation and migration and explore the possible mechanism. Our results indicated that CTRP6 expression was dramatically down-regulated in human atherosclerotic tissues and in cultured VSMCs stimulated by platelet-derived growth factor-BB (PDGF-BB). In addition, CTRP6 overexpression significantly inhibited the proliferation and migration of VSMCs exposed to PDGF-BB, as well as increased expression of α-SMA and SM22α in PDGF-BB-stimulated VSMCs. Furthermore, CTRP6 overexpression efficiently prevented the activation of PI3K/Akt/mTOR in VSMCs in response to PDGF-BB. In conclusion, these findings showed that CTRP6 inhibits PDGF-BB-induced VSMC proliferation and migration, at least in part, through suppressing the PI3K/Akt/mTOR signaling pathway. Therefore, CTRP6 may be a potential target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xunzhong Dong
- Department of Vascular Surgery, The Affiliated Provincial Hospital of Anhui Medical University, Hefei, 230001, Anhui Province, China; Department of Vascular Surgery, The People's Hospital of Bozhou, Clinical College of Anhui Medical University, Bozhou, 236800, Anhui Province, China
| | - Hejie Hu
- Department of Vascular Surgery, The Affiliated Provincial Hospital of Anhui Medical University, Hefei, 230001, Anhui Province, China.
| | - Zhengdong Fang
- Department of Vascular Surgery, The Affiliated Provincial Hospital of Anhui Medical University, Hefei, 230001, Anhui Province, China
| | - Jian Cui
- Department of Vascular Surgery, The People's Hospital of Bozhou, Clinical College of Anhui Medical University, Bozhou, 236800, Anhui Province, China
| | - Fangxin Liu
- Department of Ultrasound, The People's Hospital of Bozhou, Clinical College of Anhui Medical University, Bozhou, 236800, Anhui Province, China
| |
Collapse
|
38
|
Lee J, Lee CY, Seo HH, Bazarragchaa B, Batdelger G, Choi S, Hwang KC, Lee S, Lim S. Extract of Oxytropis pseudoglandulosa inhibits vascular smooth muscle cell proliferation and migration via suppression of ERK1/2 and Akt signaling pathways1. Clin Hemorheol Microcirc 2018; 69:277-287. [PMID: 29660921 DOI: 10.3233/ch-189126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Excessive vascular smooth muscle cell (VSMC) proliferation and migration accelerate the development of occlusive vascular disease. Therefore, finding a means to control the aberrant proliferation and migration of VSMCs has own clinical significance. In the present study, we examined the feasibility of using extract from medicinal plant Oxytropis pseudoglandulosa (OG) to control pathologic proliferation and migration of VSMCs, which never have been tested. Our data indicate that the extract of OG significantly suppressed proliferation and migration of VSMCs without cytotoxic effect, suggesting the OG extract may be an alternative agent to effectively control the aberrant VSMC proliferation and migration without any serious adverse effect. These data suggest that the extract of OG may be a potent therapeutic agent for the treatment of occlusive vascular disease and warrant further studies to identify the major acting ingredient and to validate in vivo efficacy.
Collapse
Affiliation(s)
- Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, Seoul, Korea
| | - Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | | | - Gantuya Batdelger
- Institute of General and Experimental Biology, Mongolian Academy of Sciences (MAS), Ulaanbaatar, Mongolia
| | - Sangho Choi
- International Biological Material Research Center (IBMRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Korea
| |
Collapse
|
39
|
Uhrin P, Wang D, Mocan A, Waltenberger B, Breuss JM, Tewari D, Mihaly-Bison J, Huminiecki Ł, Starzyński RR, Tzvetkov NT, Horbańczuk J, Atanasov AG. Vascular smooth muscle cell proliferation as a therapeutic target. Part 2: Natural products inhibiting proliferation. Biotechnol Adv 2018; 36:1608-1621. [PMID: 29678389 DOI: 10.1016/j.biotechadv.2018.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Many natural products have been so far tested regarding their potency to inhibit vascular smooth muscle cell proliferation, a process involved in atherosclerosis, pulmonary hypertension and restenosis. Compounds studied in vitro and in vivo as VSMC proliferation inhibitors include, for example indirubin-3'-monoxime, resveratrol, hyperoside, plumericin, pelargonidin, zerumbone and apamin. Moreover, taxol and rapamycin, the most prominent compounds applied in drug-eluting stents to counteract restenosis, are natural products. Numerous studies show that natural products have proven to yield effective inhibitors of vascular smooth muscle cell proliferation and ongoing research effort might result in the discovery of further clinically relevant compounds.
Collapse
Affiliation(s)
- Pavel Uhrin
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, Vienna 1090, Austria.
| | - Dongdong Wang
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Magdalenka 05552, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, Vienna 1090, Austria; Institute of Clinical Chemistry, University Hospital Zurich, Wagistrasse 14, Schlieren 8952, Switzerland
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Hatieganu" University of Medicine and Pharmacy, Strada Victor Babeş 8, Cluj-Napoca 400012, Romania; ICHAT and Institute for Life Sciences, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, Calea Mănăştur 3-5, Cluj-Napoca 400372, Romania
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, Innsbruck 6020, Austria
| | - Johannes M Breuss
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, Vienna 1090, Austria
| | - Devesh Tewari
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Nainital, Uttarakhand 263136, India
| | - Judit Mihaly-Bison
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, Vienna 1090, Austria
| | - Łukasz Huminiecki
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Magdalenka 05552, Poland
| | - Rafał R Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Magdalenka 05552, Poland
| | - Nikolay T Tzvetkov
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, Bonn 53121, Germany; NTZ Lab Ltd., Krasno Selo 198, Sofia 1618, Bulgaria
| | - Jarosław Horbańczuk
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Magdalenka 05552, Poland
| | - Atanas G Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Magdalenka 05552, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, Vienna 1090, Austria.
| |
Collapse
|
40
|
Tong L, Qi G. Crocin prevents platelet‑derived growth factor BB‑induced vascular smooth muscle cells proliferation and phenotypic switch. Mol Med Rep 2018; 17:7595-7602. [PMID: 29620234 PMCID: PMC5983945 DOI: 10.3892/mmr.2018.8854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/29/2018] [Indexed: 12/18/2022] Open
Abstract
The phenotypic switch of vascular smooth muscle cells (VSMCs) is a major initiating factor for atherosclerotic cardiovascular diseases. Platelet-derived growth factor-BB (PDGF-BB) initiates a number of biological processes that contribute to VSMC proliferation and phenotypic switch. Crocin, a component of saffron, has been reported to inhibit atheromatous plaque formation. However, the effects of crocin on PDGF-BB-induced VSMC proliferation and phenotypic switch remain unclear. The aim of the present study was to investigate the role of crocin on PDGF-BB-induced VSMCs proliferation and phenotypic switch and its underlying mechanisms. Cell proliferation and markers of VSMCs phenotypic switch were measured using a Cell Counting Kit-8 assay and western blot analysis, respectively. The signaling pathways involved in the effects of crocin on VSMCs were validated by western blot analysis with or without the use of specific pathway inhibitors. Crocin significantly inhibited PDGF-BB-induced VSMCs proliferation compared with the PDGF-BB only group (P<0.05). In addition, crocin significantly abrogated the PDGF-BB-induced increase in contractile protein α-smooth muscle actin, calponin and decrease in synthetic proteins osteopontin (OPN) in a concentration dependent manner (P<0.05). In addition, crocin slowed PDGF-BB-induced Janus kinase (JAK)-signal transducer and activator of transcription 3 (STAT3) and extracellular signal-regulated kinase (ERK)/Kruppel-like factor 4 (KLF4) signaling activation in VSMCs. By applying the JAK inhibitor (AG490) and ERK1/2 inhibitor (U0126), the results suggested that the crocin inhibited PDGF-BB-induced VSMCs phenotypic switch through the JAK/STAT3 and ERK/KLF4 signaling pathways. These results suggested that crocin may effectively prevent PDGF-BB-induced VSMCs proliferation and phenotypic switch and may be a promising candidate for the therapy of atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Lijian Tong
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Guoxian Qi
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
41
|
Zhang X, Zhao F, Zhao JF, Fu HY, Huang XJ, Lv BD. PDGF-mediated PI3K/AKT/β-catenin signaling regulates gap junctions in corpus cavernosum smooth muscle cells. Exp Cell Res 2017; 362:252-259. [PMID: 29174980 DOI: 10.1016/j.yexcr.2017.11.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/17/2017] [Accepted: 11/18/2017] [Indexed: 02/06/2023]
Abstract
Erectile dysfunction (ED) is the most common sexual disorder that men report to healthcare providers. Gap junctions (GJs) are thought to be responsible for synchronous shrinkage of corpus cavernosum smooth muscle cells (CCSMCs), and play thus an important role in the maintenance of an erection. Hypoxia has been suggested as a pathological mechanism underlying ED. Here we demonstrate that hypoxia increased the expression of platelet-derived growth factor (PDGF) and the main GJ component connexin (Cx)43 in CCSMCs. Inhibiting PDGF receptor (PDGFR) activity decreased Cx43 expression. Treatment with different concentrations of PDGF increased the levels of phosphorylated protein kinase B (AKT), β-catenin, and Cx43, whereas inhibition of PDGFR or activation of phosphatidylinositol 3 kinase (PI3K)/AKT signaling altered β-catenin and Cx43 expression. Meanwhile, silencing β-catenin resulted in the downregulation of Cx43. These results demonstrate that PDGF secretion by CCSMCs and vascular endothelial cells is enhanced under hypoxic conditions, leading to increased Cx43 expression through PI3K/AKT/β-catenin signaling and ultimately affecting GJ function in ED. Thus, targeting this pathway is a potential therapeutic strategy for the treatment of ED.
Collapse
Affiliation(s)
- Xiang Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fan Zhao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jian-Feng Zhao
- Department of Urology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui-Ying Fu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Andrology Laboratory on Integration of Chinese and Western Medicine, Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine, Hangzhou, China
| | - Xiao-Jun Huang
- Department of Urology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo-Dong Lv
- Department of Urology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China; Andrology Laboratory on Integration of Chinese and Western Medicine, Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine, Hangzhou, China.
| |
Collapse
|
42
|
Ham O, Lee SY, Song BW, Lee CY, Lee J, Seo HH, Kim SW, Lim S, Kim IK, Lee S, Hwang KC. Small molecule-mediated induction of miR-9 suppressed vascular smooth muscle cell proliferation and neointima formation after balloon injury. Oncotarget 2017; 8:93360-93372. [PMID: 29212155 PMCID: PMC5706801 DOI: 10.18632/oncotarget.21382] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 09/04/2017] [Indexed: 01/04/2023] Open
Abstract
Pathologic proliferation and migration of vascular smooth muscle cells (VSMCs) exacerbate cardiovascular disease. MicroRNAs (miRNAs), as endogenous inhibitors of protein synthesis, are expected to modulate pathologic proliferation of VSMCs. Here we report that both platelet-derived growth factor receptor (PDGFR) targeting miR-9 and a small molecule that increases miR-9 can inhibit the serum-induced proliferation of VSMCs. First, based on miRNA-target prediction databases and empirical data, we have selected miR-9 as a potent anti-proliferative miRNA in VSMCs. Further examination indicated that miR-9 directly targets PDGFR disrupting downstream signaling cascades, and this resulted in inhibition of VSMC proliferation and migration. Exogenous delivery of miR-9 inhibited VSMC proliferation in vitro, and a small molecule that increased miR-9 expression also inhibited neointima formation following balloon injury in vivo. We provide evidence of miRNA-mediated modulation of VSMC proliferation and further demonstrate that small molecule-mediated regulation of miRNA targeting a key regulator of VSMC proliferation is a viable therapeutic strategy for treating vascular disease involving pathologic VSMC proliferation such as restenosis.
Collapse
Affiliation(s)
- Onju Ham
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se-Yeon Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byeong-Wook Song
- EIT/LOFUS R&D Center, International St. Mary’s Hospital, Incheon, Republic of Korea
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| | - Soyeon Lim
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| |
Collapse
|
43
|
Kim WH, An HJ, Kim JY, Gwon MG, Gu H, Lee SJ, Park JY, Park KD, Han SM, Kim MK, Park KK. Apamin inhibits TNF-α- and IFN-γ-induced inflammatory cytokines and chemokines via suppressions of NF-κB signaling pathway and STAT in human keratinocytes. Pharmacol Rep 2017; 69:1030-1035. [PMID: 28958612 DOI: 10.1016/j.pharep.2017.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/26/2017] [Accepted: 04/11/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is identified by an increase in infiltrations of several inflammatory cells including type 2 helper (Th2) lymphocytes. Th2-related chemokines such as thymus and activation-regulated chemokine (TARC/CCL17) and macrophage-derived chemokine (MDC/CCL22), and pro-inflammatory cytokines including interleukin (IL)-1β and IL-6 are considered to play a crucial role in AD. Tumor necrosis factor (TNF)-α- and interferon (IFN)-γ induce the inflammatory condition through production of TARC, MDC, IL-1β and IL-6, and activations of related transcription factors, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator of transcription (STAT) in keratinocytes. Apamin, a peptide component of bee venom, has been reported its beneficial activities in various diseases. However, anti-inflammatory effects of apamin on inflammatory condition in keratinocytes have not been explored. Therefore, the present study aimed to demonstrate the anti-inflammatory effect of apamin on TNF-α- and IFN-γ-induced inflammatory condition in keratinocytes. METHODS HaCaT was used as human keratinocytes cell line. Cell Counting Kit-8 was performed to measure a cytotoxicity of apamin. The effects of apamin on TNF-α-/IFN-γ-induced inflammatory condition were determined by real-time PCR and Western blot analysis. Further, NF-κB signaling pathways, STAT1, and STAT3 were analyzed by Western blot and immunofluorescence. RESULTS Apamin ameliorated the inflammatory condition through suppression of Th2-related chemokines and pro-inflammatory cytokines. Further, apamin down-regulated the activations of NF-κB signaling pathways and STATs in HaCaT cells. CONCLUSIONS These results suggest that apamin has therapeutic effect on AD through improvement of inflammatory condition.
Collapse
Affiliation(s)
- Woon-Hae Kim
- Department of Pathology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| | - Hyun-Jin An
- Department of Pathology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| | - Jung-Yeon Kim
- Department of Pathology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| | - Mi-Gyeong Gwon
- Department of Pathology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| | - Hyemin Gu
- Department of Pathology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| | - Sun-Jae Lee
- Department of Pathology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| | - Ji Y Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| | - Kyung-Duck Park
- Department of Dermatology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| | - Sang-Mi Han
- Department of Agricultural Biology, National Academy of Agricultural Science, Rural Development Administration, 300, Nongsaengmyeong-ro, Wansan-gu, Jeonju-si, Jeollabuk-do 54875, Republic of Korea.
| | - Min-Kyung Kim
- Department of Pathology, College of Medicine, Dongguk University, 123, Dongdae-ro, Gyeongju-si, Gyeongsangbuk-do 38066, Republic of Korea.
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, 33, Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| |
Collapse
|
44
|
Peng L, Huang X, Jin X, Jing Z, Yang L, Zhou Y, Ren J, Zhao Y. Wedelolactone, a plant coumarin, prevents vascular smooth muscle cell proliferation and injury-induced neointimal hyperplasia through Akt and AMPK signaling. Exp Gerontol 2017. [PMID: 28634089 DOI: 10.1016/j.exger.2017.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Wedelolactone (WDL) is a natural compound derived from Chinese herbal medicine Eclipta prostrate L, and has been reported to exhibit various effects potentially beneficial for human health. However, the possible preventive effects of WDL toward vascular remodeling and mechanisms involved have not been investigated to date. In this study, we investigated the effects of WDL on proliferation induced by platelet-derived growth factor (PDGF) in primary rat aortic smooth muscle cells (VSMCs) and on neointimal hyperplasia resulted from balloon injury in rats. WDL exhibited strong inhibitory effects against PDGF-induced VSMC proliferation. Cell cycle analysis revealed that WDL induced G0/G1 arrest and prevented cell cycle from entering S phase. Immunoblot analysis suggested that the cell cycle arrest induced by WDL was through Akt suppression and adenosine 5'-monophosphate-activated protein kinase (AMPK) activation, with a subsequent cyclin-dependent kinase inhibitor p21 induction and cyclin D1 inhibition. We also observed that WDL notably reduced neointima-to-media area ratio of balloon-injured rat common carotid arteries (CCAs) in comparison with those untreated balloon-injured CCAs. The regulation of WDL on protein expressions of Akt, AMPK and cyclin D1 in vivo were also consistent with that in vitro. Taken together, our results suggest WDL exhibits potential preventive effects toward vascular remodeling and neointimal hyperplasia through the reduction of VSMC proliferation via inhibition of Akt and activation of AMPK.
Collapse
Affiliation(s)
- Lu Peng
- Medical College, Xiamen University, Xiamen 361000, China
| | - Xuefeng Huang
- Zhongshan Hospital, Xiamen University, Xiamen 361000, China
| | - Xin Jin
- Medical College, Xiamen University, Xiamen 361000, China
| | - Zuo Jing
- Medical College, Xiamen University, Xiamen 361000, China
| | - Lichao Yang
- Medical College, Xiamen University, Xiamen 361000, China
| | - Yu Zhou
- Medical College, Xiamen University, Xiamen 361000, China
| | - Jie Ren
- Medical College, Xiamen University, Xiamen 361000, China
| | - Yun Zhao
- Medical College, Xiamen University, Xiamen 361000, China; Fujian Provincial Key Laboratory of Regenerative Medicine, Xiamen 361000, China.
| |
Collapse
|
45
|
Kim JY, An HJ, Kim WH, Park YY, Park KD, Park KK. Apamin suppresses biliary fibrosis and activation of hepatic stellate cells. Int J Mol Med 2017; 39:1188-1194. [PMID: 28405682 PMCID: PMC5403474 DOI: 10.3892/ijmm.2017.2922] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/07/2017] [Indexed: 12/16/2022] Open
Abstract
Cholestatic liver disease is characterized by the progressive destruction of biliary epithelial cells (BECs) followed by fibrosis, cirrhosis and liver failure. Activated hepatic stellate cells (HSCs) and portal fibroblasts are the major cellular effectors of enhanced collagen deposition in biliary fibrosis. Apamin, an 18 amino acid peptide neurotoxin found in apitoxin (bee venom), is known to block Ca2+-activated K+ channels and prevent carbon tetrachloride-induced liver fibrosis. In the present study, we aimed to ascertain whether apamin inhibits biliary fibrosis and the proliferation of HSCs. Cholestatic liver fibrosis was established in mouse models with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) feeding. Cellular assays were performed on HSC-T6 cells (rat immortalized HSCs). DDC feeding led to increased hepatic damage and proinflammtory cytokine levels. Notably, apamin treatment resulted in decreased liver injury and proinflammatory cytokine levels. Moreover, apamin suppressed the deposition of collagen, proliferation of BECs and expression of fibrogenic genes in the DDC-fed mice. In HSCs, apamin suppressed activation of HSCs by inhibiting the Smad signaling pathway. These data suggest that apamin may be a potential therapeutic target in cholestatic liver disease.
Collapse
Affiliation(s)
| | | | | | | | - Kyung Duck Park
- Department of Dermatology, College of Medicine, Catholic University of Daegu, Daegu 705-718, Republic of Korea
| | | |
Collapse
|
46
|
Seo HH, Kim SW, Lee CY, Lim KH, Lee J, Lim S, Lee S, Hwang KC. 7-cyclopentyl-5-(4-phenoxyphenyl)-7H-pyrrolo[2,3-d] pyrimidin-4-ylamine inhibits the proliferation and migration of vascular smooth muscle cells by suppressing ERK and Akt pathways. Eur J Pharmacol 2017; 798:35-42. [PMID: 28185804 DOI: 10.1016/j.ejphar.2017.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 01/31/2017] [Accepted: 02/02/2017] [Indexed: 11/26/2022]
Abstract
Excessive vascular smooth muscle cell (VSMC) proliferation and migration after vascular injury significantly contributes to the development of occlusive vascular disease. Therefore, inhibiting the proliferation and migration of VSMCs is a validated therapeutic modality for occlusive vascular disease such as atherosclerosis and restenosis. In the present study, we screened chemical compounds for their anti-proliferative effects on VSMCs using multiple approaches, such as MTT assays, wound healing assays, and trans-well migration assays. Our data indicate that 7-cyclopentyl-5-(4-phenoxyphenyl)-7H-pyrrolo[2,3-d] pyrimidin-4-ylamine, a lymphocyte-specific protein tyrosine kinase (Lck) inhibitor, significantly inhibited both VSMC proliferation and migration. 7-cyclopentyl-5-(4-phenoxyphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-ylamine suppresses VSMC proliferation and migration via down-regulating the protein kinase B (Akt) and extracellular signal regulated kinase (ERK) pathways, and it significantly decreased the expression of proliferating cell nuclear antigen (PCNA) and cyclin D1 and, the phosphorylation of retinoblastoma protein (pRb). Additionally, 7-cyclopentyl-5-(4-phenoxyphenyl)-7H-pyrrolo[2,3-d] pyrimidin-4-ylamine suppressed the migration of VSMCs from endothelium-removed aortic rings, as well as neointima formation following rat carotid balloon injury. The present study identified 7-cyclopentyl-5-(4-phenoxyphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-ylamine as a potent VSMC proliferation and migration inhibitor and warrants further studies to elucidate its more detailed molecular mechanisms, such as its primary target, and to further validate its in vivo efficacy as a therapeutic agent for pathologic vascular conditions, such as restenosis and atherosclerosis.
Collapse
Affiliation(s)
- Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, Korea
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, Seoul, Korea
| | - Kyu Hee Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonbuk National University, Jeonju, Jeollabuk-Do, Korea
| | - Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, Korea.
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, Korea.
| |
Collapse
|
47
|
Wang HJ, Zhou Y, Liu RM, Qin YS, Cen YH, Hu LY, Wang SM, Hu ZJ. IP-10/CXCR3 Axis Promotes the Proliferation of Vascular Smooth Muscle Cells through ERK1/2/CREB Signaling Pathway. Cell Biochem Biophys 2017; 75:139-147. [PMID: 28111710 DOI: 10.1007/s12013-017-0782-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/11/2017] [Indexed: 10/20/2022]
Abstract
Excessive proliferation of vascular smooth muscle cells is one of the main pathological processes leading to atherosclerosis and intimal hyperplasia after vascular interventional therapy. Our previous study has shown that interferon-γ inducible protein-10 contributes to the proliferation of vascular smooth muscle cell. However, the underlying mechanisms remain unclear. Extracellular signal-regulated kinase 1/2, serine/threonine kinase Akt, and cAMP response element binding protein are signaling pathways, which are considered to play important roles in the processes of vascular smooth muscle cell proliferation. Moreover, chemokine receptor 3 and Toll-like receptor 4 are potential receptors of inducible protein-10 in this process. In the present study, IP-10 was found to directly induce vascular smooth muscle cell proliferation, and exposure to inducible protein-10 activated extracellular signal-regulated kinase 1/2, serine/threonine kinase, and cAMP response element binding protein signaling. Inhibitor of extracellular signal-regulated kinase 1/2, rather than inhibitor of serine/threonine kinase, inhibited the phosphorylation of cAMP response element binding protein and reduced inducible protein-10-stimulated vascular smooth muscle cell proliferation. Knockdown of cAMP response element binding protein by siRNA inhibited inducible protein-10-induced vascular smooth muscle cell proliferation. Moreover, anti-CXCR3 IgG, instead of anti-Toll-like receptor 4 IgG, reduced inducible protein-10-induced vascular smooth muscle cell proliferation and inducible protein-10-stimulated extracellular signal-regulated kinase 1/2 and cAMP response element binding protein activation. Together, these results indicate that inducible protein-10 promotes vascular smooth muscle cell proliferation via chemokine receptor 3 and activation of extracellular signal-regulated kinase 1/2 inducible protein-10-induced vascular smooth muscle cell proliferation. These data provide important targets for future studies to modulate atherosclerosis and restenosis after vascular interventional therapy.
Collapse
Affiliation(s)
- Hui-Jin Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.,Department of General Surgery, Huadu District People's Hospital, Southern Medical University, Guanghzou, 510800, China
| | - Yu Zhou
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Rui-Ming Liu
- Laboratory of Department of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuan-Sen Qin
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying-Huan Cen
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ling-Yu Hu
- Department of Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, 510080, China
| | - Shen-Ming Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zuo-Jun Hu
- Department of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
48
|
Voos P, Yazar M, Lautenschläger R, Rauh O, Moroni A, Thiel G. The small neurotoxin apamin blocks not only small conductance Ca 2+ activated K + channels (SK type) but also the voltage dependent Kv1.3 channel. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:517-523. [PMID: 28108814 DOI: 10.1007/s00249-016-1196-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/12/2016] [Accepted: 12/30/2016] [Indexed: 11/29/2022]
Abstract
Apamin is frequently used as a specific blocker of small-conductance Ca2+-activated (SK type) K+ channels. Here we show that the small neurotoxin is not as specific as anticipated. It is also a high-affinity inhibitor with an IC50 of 13 nM of the Kv1.3 channel; it blocks the latter with potency similar to the Kv1.3 blocker PAP-1. Since SK type channels and Kv1.3 channels are frequently coexpressed in different tissues such as cells of the immune system, apamin must be used with caution as a pharmacological tool.
Collapse
Affiliation(s)
- Patrick Voos
- Department of Biology, Plant Membrane Biophysics, Technische Universität Darmstadt, Schnittspahnstrasse 3, 64287, Darmstadt, Germany
| | - Mehtap Yazar
- Department of Biology, Plant Membrane Biophysics, Technische Universität Darmstadt, Schnittspahnstrasse 3, 64287, Darmstadt, Germany
| | - René Lautenschläger
- Department of Biology, Plant Membrane Biophysics, Technische Universität Darmstadt, Schnittspahnstrasse 3, 64287, Darmstadt, Germany
| | - Oliver Rauh
- Department of Biology, Plant Membrane Biophysics, Technische Universität Darmstadt, Schnittspahnstrasse 3, 64287, Darmstadt, Germany
| | - Anna Moroni
- Department of Biology and CNR IBF-Mi, Università degli Studi di Milano, Milan, Italy
| | - Gerhard Thiel
- Department of Biology, Plant Membrane Biophysics, Technische Universität Darmstadt, Schnittspahnstrasse 3, 64287, Darmstadt, Germany.
| |
Collapse
|
49
|
Pak SC. An Introduction to the Toxins Special Issue on "Bee and Wasp Venoms: Biological Characteristics and Therapeutic Application". Toxins (Basel) 2016; 8:toxins8110315. [PMID: 27801836 PMCID: PMC5127112 DOI: 10.3390/toxins8110315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 10/19/2016] [Indexed: 02/07/2023] Open
Affiliation(s)
- Sok Cheon Pak
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia.
| |
Collapse
|
50
|
Chaisakul J, Hodgson WC, Kuruppu S, Prasongsook N. Effects of Animal Venoms and Toxins on Hallmarks of Cancer. J Cancer 2016; 7:1571-8. [PMID: 27471574 PMCID: PMC4964142 DOI: 10.7150/jca.15309] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/18/2016] [Indexed: 12/20/2022] Open
Abstract
Animal venoms are a cocktail of proteins and peptides, targeting vital physiological processes. Venoms have evolved to assist in the capture and digestion of prey. Key venom components often include neurotoxins, myotoxins, cardiotoxins, hematoxins and catalytic enzymes. The pharmacological activities of venom components have been investigated as a source of potential therapeutic agents. Interestingly, a number of animal toxins display profound anticancer effects. These include toxins purified from snake, bee and scorpion venoms effecting cancer cell proliferation, migration, invasion, apoptotic activity and neovascularization. Indeed, the mechanism behind the anticancer effect of certain toxins is similar to that of agents currently used in chemotherapy. For example, Lebein is a snake venom disintegrin which generates anti-angiogenic effects by inhibiting vascular endothelial growth factors (VEGF). In this review article, we highlight the biological activities of animal toxins on the multiple steps of tumour formation or hallmarks of cancer. We also discuss recent progress in the discovery of lead compounds for anticancer drug development from venom components.
Collapse
Affiliation(s)
- Janeyuth Chaisakul
- 1. Department of Pharmacology, Phramongkutklao College of Medicine, Bangkok 10400, Thailand
| | - Wayne C Hodgson
- 2. Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Sanjaya Kuruppu
- 2. Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia.; 3. Department of Biochemistry & Molecular Biology, Biomedical Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Naiyarat Prasongsook
- 4. Division of Medical Oncology, Department of Medicine, Phramongkutklao Hospital, Bangkok 10400, Thailand
| |
Collapse
|