1
|
Singh MT, Thaggikuppe Krishnamurthy P, Magham SV. Harnessing the synergistic potential of NK1R antagonists and selective COX-2 inhibitors for simultaneous targeting of TNBC cells and cancer stem cells. J Drug Target 2024; 32:258-269. [PMID: 38252517 DOI: 10.1080/1061186x.2024.2309568] [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: 09/26/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2024]
Abstract
Triple-negative breast cancer (TNBC) lacks the expression of oestrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), rendering it unresponsive to endocrine therapy and HER2 targeted treatments. Though certain chemotherapeutics targeting the cell cycle have shown efficacy to a certain extent, the presence of chemotherapy-resistant cancer stem cells (CSCs) presents a significant challenge in tackling TNBC. Multiple lines of evidence suggest the upregulation of neuropeptide Substance P (SP), its NK-1 receptor (NK1R) and the Cyclooxygenase-2 (COX-2) enzyme in TNBC patients. Upregulation of the SP/NK1R system and COX-2 influences major signalling pathways involved in cell proliferation, growth, survival, angiogenesis, inflammation, metastasis and stem cell activity. The simultaneous activation and crosstalk between the pathways activated by SP/NK1R and COX-2 consequently increase the levels of key regulators of self-renewal pathways in CSCs, promoting stemness. The combination therapy with NK1R antagonists and COX-2 inhibitors can simultaneously target TNBC cells and CSCs, thereby enhancing treatment efficacy and reducing the risk of recurrence and relapse. This review discusses the rationale for combining NK1R antagonists and COX-2 inhibitors for the better management of TNBC and a novel strategy to deliver drug cargo precisely to the tumour site to address the challenges associated with off-target binding.
Collapse
Affiliation(s)
- Madhu Tanya Singh
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Sai Varshini Magham
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| |
Collapse
|
2
|
Ko KR, Han SH, Choi S, An HJ, Kwak EB, Jeong Y, Baek M, Lee J, Choi J, Kim IS, Lee S. Substance P Inhibitor Promotes Tendon Healing in a Collagenase-Induced Rat Model of Tendinopathy. Am J Sports Med 2022; 50:3681-3689. [PMID: 36197354 DOI: 10.1177/03635465221126175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The substance P-neurokinin 1 receptor pathway has been proposed as a therapeutic target for tendinopathy. However, there is a lack of evidence regarding its practical applications. PURPOSE To investigate the therapeutic effects of substance P inhibitor (SPI) on inflamed tenocytes in vitro and in a collagenase-induced rat model of tendinopathy in vivo. STUDY DESIGN Controlled laboratory study. METHODS We analyzed the mRNA levels of inflammatory (cyclooxygenase [COX]-2 and interleukin [IL]-6) and tenogenic (Mohawk and scleraxis [SCX]) markers using reverse transcription quantitative polymerase chain reaction to demonstrate the effects of SPI on lipopolysaccharide-treated (inflamed) tenocytes. A collagenase-induced rat model of tendinopathy was created by injecting 20 µL of collagenase into the Achilles tendon. A behavior test using an incapacitance apparatus was performed to detect changes in postural equilibrium. The tendon specimens were obtained, and their gross findings were examined. The tensile strength was measured, and histopathological evaluation was performed (hematoxylin and eosin, alcian blue, and immunohistochemical staining). RESULTS The mRNA levels of COX-2, IL-6, Mohawk, and SCX differed significantly between inflamed tenocytes and those treated with SPI. SPI improved the weight burden in a rat model of tendinopathy in a behavioral test. The specimens of the SPI group showed a normal tendon-like appearance. In the biomechanical test, the tensile strength of the SPI group was significantly greater than that of the tendinopathy group. In the histopathological evaluation, the degree of collagen matrix breakdown was mild in the SPI group. In alcian blue staining, only small focal depositions of proteoglycans and glycosaminoglycans were observed in the SPI group. The SPI group showed decreased expression of IL-6 and neurokinin 1 receptor. CONCLUSION This study suggests that SPI has therapeutic effects on tendon healing and restoration in a collagenase-induced rat model of tendinopathy. CLINICAL RELEVANCE SPI is a promising agent for tendinopathy in humans.
Collapse
Affiliation(s)
- Kyung Rae Ko
- Department of Orthopaedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soo-Hong Han
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, Republic of Korea
| | - Sujin Choi
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, Republic of Korea
| | - Hyun-Ju An
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, Republic of Korea
| | - Eun-Bee Kwak
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, Republic of Korea
| | - Yunhui Jeong
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, Republic of Korea
| | - Minjung Baek
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, Republic of Korea
| | - Jusung Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, Republic of Korea
| | - Junwon Choi
- Department of Molecular Science and Technology, Ajou University, Suwon-si, Republic of Korea
| | - Il-Su Kim
- Department of Orthopaedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si, Republic of Korea
| |
Collapse
|
3
|
Li Q, Jiang B, Zhang Z, Huang Y, Xu Z, Chen X, Huang Y, Jian J. SP protects Nile tilapia (Oreochromis niloticus) against acute Streptococcus agalatiae infection. FISH & SHELLFISH IMMUNOLOGY 2022; 123:218-228. [PMID: 35257891 DOI: 10.1016/j.fsi.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/16/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
Substance P (SP) is a neuropeptide that involves in a wide variety of physiological and pathological events, mainly exerts its roles by neurokinin 1 receptor (NK1R), also modulates immune function. However, the roles of SP during immune response to acute bacterial infection of Nile tilapia (Oreochromis niloticus) remain unclear. In this study, the gene of SP precursor (tachykinin precursor 1, TAC1) and the gene of SP receptor (NK1R) from Nile tilapia were identified, and the roles of SP during an acute bacterial infection in a warm water environment were investigated. On-TAC1(Oreochromis niloticus-TAC1) contains conservative SP & NKA peptide sequences and On-NK1R contains seven conservative transmembrane domains. Their transcriptional levels were most abundant in brain and the On-TAC1 transcripts can be induced in the tilapia challenged with Streptococcus agalactiae. Furthermore, the experimental results revealed that On-SP could promote pyroptosis, suppress inflammation, and improve survival rate during acute bacterial infection. The present data lays a theoretical foundation to further elucidate the mechanism of SP protecting fish against pathogens.
Collapse
Affiliation(s)
- Qi Li
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
| | - Baijian Jiang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
| | - Zhiqiang Zhang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
| | - Yongxiong Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
| | - Zhou Xu
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
| | - Xinjin Chen
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
| | - Yu Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animal, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| |
Collapse
|
4
|
Kim D, Park D, Kim TH, Chung JJ, Jung Y, Kim SH. Substance P/Heparin-Conjugated PLCL Mitigate Acute Gliosis on Neural Implants and Improve Neuronal Regeneration via Recruitment of Neural Stem Cells. Adv Healthc Mater 2021; 10:e2100107. [PMID: 34227258 DOI: 10.1002/adhm.202100107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/03/2021] [Indexed: 12/15/2022]
Abstract
The inflammatory host tissue response, characterized by gliosis and neuronal death at the neural interface, limits signal transmission and longevity of the neural probe. Substance P induces an anti-inflammatory response and neuronal regeneration and recruits endogenous stem cells. Heparin prevents nonspecific protein adsorption, suppresses the inflammatory response, and is beneficial to neuronal behavior. Poly(l-lactide-co-ε-caprolactone) (PLCL) is a soft and flexible polymer, and PLCL covalently conjugated with biomolecules has been widely used in tissue engineering. Coatings of heparin-conjugated PLCL (Hep-PLCL), substance P-conjugated PLCL (SP-PLCL), and heparin/substance P-conjugated PLCL (Hep/SP-PLCL) reduced the adhesion of astrocytes and fibroblasts and improved neuronal adhesion and neurite development compared to bare glass. The effects of these coatings are evaluated using immunohistochemistry analysis after implantation of coated stainless steel probes in rat brain for 1 week. In particular, Hep/SP-PLCL coating reduced the activation of microglia and astrocytes, the neuronal degeneration caused by inflammation, and indicated a potential for neuronal regeneration at the tissue-device interface. Suppression of the acute host tissue response by coating Hep/SP-PLCL could lead to improved functionality of the neural prosthesis.
Collapse
Affiliation(s)
- Donghak Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145 Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - DoYeun Park
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - Tae Hee Kim
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - Justin J. Chung
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| | - Soo Hyun Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145 Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
- Biomaterials Research Center Korea Institute of Science and Technology (KIST) 5, Hwarang‐ro 14‐gil, Seongbuk‐gu Seoul 02792 Republic of Korea
| |
Collapse
|
5
|
Su G, Sun G, Liu H, Shu L, Zhang W, Liang Z. Prokineticin 2 relieves hypoxia/reoxygenation-induced injury through activation of Akt/mTOR pathway in H9c2 cardiomyocytes. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:345-352. [PMID: 31899964 DOI: 10.1080/21691401.2019.1709850] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Prokineticin 2 (PK2) was reported to be decreased in the hearts of end-state heart failure patients. Our study aimed to explore the effects of PK2 on hypoxia/reoxygenation (H/R) injury and the underlying mechanism. H9c2 cardiomyocytes were treated with 5 nM PK2 in the presence or absence of 5 mM dual phosphatidylinositol 3-kinase (PI3K)/the mammalian target of rapamycin (mTOR) inhibitor (BEZ235) for 24 h and then subjected to H/R treatment. Cell viability and lactate dehydrogenase (LDH) release were evaluated by CCK-8 and LDH release assays, respectively. Apoptosis was determined by flow cytometry analysis. Oxidative stress was assessed by measuring superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) activities and malondialdehyde (MDA) content. Results showed that H/R treatment decreased PK2 expression and inactivated the Akt/mTOR pathway in H9c2 cardiomyocytes. PK2 treatment activated the Akt/mTOR pathway in H/R-exposed H9c2 cardiomyocytes. H/R stimulation suppressed cell viability, increased LDH release, induced apoptosis and oxidative stress in H9c2 cardiomyocytes, while these effects were neutralised by treatment with PK2. However, the inhibitory effects of PK2 on H/R-induced injury in H9c2 cardiomyocytes were abolished by the addition of BEZ235. In conclusion, PK2 relieved H/R-induced injury in H9c2 cardiomyocytes by activation of the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Gang Su
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangli Sun
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hai Liu
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liliang Shu
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiwei Zhang
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenxing Liang
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Hu YH, Sun J, Zhang J, Hua FZ, Liu Q, Liang YP. Long non-coding RNA ROR sponges miR-138 to aggravate hypoxia/reoxygenation-induced cardiomyocyte apoptosis via upregulating Mst1. Exp Mol Pathol 2020; 114:104430. [PMID: 32240614 DOI: 10.1016/j.yexmp.2020.104430] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 12/21/2019] [Accepted: 03/28/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hypoxia/reoxygenation (H/R) injury of cardiomyocytes causes an irreversible damage to heart and largely results in acute myocardial infarction. Study has indicated lncRNA ROR aggravates myocardial ischemia/reperfusion (I/R) injury. Also, lncRNA ROR sponges miR-138 to promote osteogenesis. MiR-138 involves in hypoxic pulmonary vascular remodelling by targeting Mst1. However, the interaction between lncRNA ROR, miR-138 and Mst1 involved in myocardial H/R injury is still unknown. METHODS H9C2 cells were used to establish H/R injury model. The expression levels of lncRNA ROR and miR-138 were modified by transfection with the miR-138 mimics or lncRNA ROR overexpression plasmid. MTT and flow cytometry analysis were performed to detect cell proliferation and apoptosis. Dual luciferase reporter assay was used to determine interaction between lncRNA ROR and miR-138 or miR-138 and Mst1. Expression levels of lncRNA ROR, miR-138, Mst1 and apoptosis-related markers were determined by qRT-PCR or western blotting. RESULTS LncRNA ROR was significantly up-regulated, while miR-138 was obviously down-regulated in H/R-induced injury of H9C2 cells. Furthermore, miR-138 overexpression alleviated cardiac cell apoptosis induced by H/R injury. Mst1 was revealed to be a target of miR-138 and negatively regulated by miR-138. Mst1 overexpression reversed the protective effects of miR-138 on H/R injury of H9C2 cells. LncRNA ROR was identified as a sponge for miR-138. MiR-138 could protect H9C2 cells form H/R injury induced by lncRNA ROR overexpression. CONCLUSION Our study provides that lncRNA ROR sponges miR-138 to aggravate H/R-induced myocardial cell injury by upregulating the expression of Mst1.
Collapse
Affiliation(s)
- Yan-Hui Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Jing Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Fu-Zhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Qin Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Ying-Ping Liang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
7
|
The Guizhi Gancao Decoction Attenuates Myocardial Ischemia-Reperfusion Injury by Suppressing Inflammation and Cardiomyocyte Apoptosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1947465. [PMID: 30800167 PMCID: PMC6360628 DOI: 10.1155/2019/1947465] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 12/10/2018] [Accepted: 12/20/2018] [Indexed: 12/25/2022]
Abstract
Guizhi Gancao Decoction (GGD) is a well-known traditional Chinese herbal medicine for the treatment of various cardiovascular diseases, such as myocardial ischemia-reperfusion (I/R) injury and arrhythmia. However, the mechanism by which GGD contributes to the amelioration of cardiac injury remains unclear. The aim of this study was to investigate the potential protective role of GGD against myocardial I/R injury and its possible mechanism. Consistent with the effect of the positive drug (Trimetazidine, TMZ), we subsequently validated that GGD could ameliorate myocardial I/R injury as evidenced by histopathological examination and triphenyltetrazolium chloride (TTC) staining. Moreover, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay demonstrated that GGD suppressed myocardial apoptosis, which may be related to the upregulation of Bcl-2, PPARα, and PPARγ and downregulation of Bax, caspase-3, and caspase-9. Pretreatment with GGD attenuated the levels of proinflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin- (IL-) 6, and IL-1β in serum by inhibiting Toll-like receptor 4 (TLR4)/NF-κB signaling pathway. These results indicated that GGD exhibits cardioprotective effects on myocardial I/R injury through inhibition of the TLR4/NF-κB signaling pathway, which led to reduced inflammatory response and the subsequent cardiomyocyte apoptosis.
Collapse
|
8
|
Levick SP. Understanding the Complex Roles of Substance P in the Diseased Heart. Heart Lung Circ 2018; 27:1394-1397. [PMID: 30262153 DOI: 10.1016/j.hlc.2018.08.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 08/30/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Scott P Levick
- Kolling Institute for Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Shou X, Zhou R, Zhu L, Ren A, Wang L, Wang Y, Zhou J, Liu X, Wang B. Emodin, A Chinese Herbal Medicine, Inhibits Reoxygenation-Induced Injury in Cultured Human Aortic Endothelial Cells by Regulating the Peroxisome Proliferator-Activated Receptor-γ (PPAR-γ) and Endothelial Nitric Oxide Synthase (eNOS) Signaling Pathway. Med Sci Monit 2018; 24:643-651. [PMID: 29386501 PMCID: PMC5804301 DOI: 10.12659/msm.908237] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Ischemia-reperfusion injury is associated with vascular dysfunction. The aim of this study was to investigate the role of emodin, a Chinese herbal medicine, in hypoxia-reoxygenation injury in cultured human aortic endothelial cells (HAECs) and its effects on the expression of the peroxisome proliferator-activated receptor-γ (PPAR-γ) and endothelial nitric oxide synthase (eNOS) signaling pathway. Material/Methods An in vitro hypoxia-reoxygenation model used cultured human aortic endothelial cells (HAECs). A colorimetric method evaluated the activity of peroxisome proliferator-activated receptor-γ (PPAR-γ). Phosphorylation of PPAR-γ and endothelial nitric oxide synthase (eNOS) were measured by Western blotting. Expression of inflammatory cytokines, tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-8 were evaluated by enzyme-linked immunosorbent assay (ELISA) and Western blotting. Nitric oxide (NO) production was detected by diaminofluorescein-FM diacetate (DAF-FM DA) fluorescence. Immunoprecipitation was used to evaluate the molecular coupling of heat shock protein (HSP)90 and eNOS. Results Hypoxia-reoxygenation injury of HAECs reduced the activity and phosphorylation of PPAR-γ, and eNOS, NO production, and HSP90/eNOS molecular coupling in a time-dependent manner. Hypoxia-reoxygenation increased the levels of inflammatory cytokines TNF-α, IL-6, and IL-8 in a time-dependent manner. Emodin treatment recovered PPAR-γ activity and phosphorylation, eNOS phosphorylation, and HSP90/eNOS coupling in HAECS in a concentration-dependent manner, which was reversed by the PPAR-γ inhibitor GW9662, and the eNOS inhibitor, L-NAME. The recovery of HSP90/eNOS coupling by emodin was impaired by GW9662 treatment. Conclusions An in vitro hypoxia-reoxygenation (ischemia-reperfusion injury) model of induction of endothelial cell inflammatory mediators showed that emodin recovered the PPAR-γ and eNOS pathway activity.
Collapse
Affiliation(s)
- Xiaoling Shou
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Rongfang Zhou
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Liyue Zhu
- Rehabilitation Center, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Aihua Ren
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Lei Wang
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Yan Wang
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Jianmei Zhou
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Xinwen Liu
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Bozhong Wang
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|