1
|
Khoi CS, Lin TY, Chiang CK. Targeting Insulin Resistance, Reactive Oxygen Species, Inflammation, Programmed Cell Death, ER Stress, and Mitochondrial Dysfunction for the Therapeutic Prevention of Free Fatty Acid-Induced Vascular Endothelial Lipotoxicity. Antioxidants (Basel) 2024; 13:1486. [PMID: 39765815 PMCID: PMC11673094 DOI: 10.3390/antiox13121486] [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: 10/31/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Excessive intake of free fatty acids (FFAs), especially saturated fatty acids, can lead to atherosclerosis and increase the incidence of cardiovascular diseases. FFAs also contribute to obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Palmitic acid (PA) is human plasma's most abundant saturated fatty acid. It is often used to study the toxicity caused by free fatty acids in different organs, including vascular lipotoxicity. Fatty acid overload induces endothelial dysfunction through various molecular mechanisms. Endothelial dysfunction alters vascular homeostasis by reducing vasodilation and increasing proinflammatory and prothrombotic states. It is also linked to atherosclerosis, which leads to coronary artery disease, peripheral artery disease, and stroke. In this review, we summarize the latest studies, revealing the molecular mechanism of free fatty acid-induced vascular dysfunction, targeting insulin resistance, reactive oxygen species, inflammation, programmed cell death, ER stress, and mitochondrial dysfunction. Meanwhile, this review provides new strategies and perspectives for preventing and reducing the impact of cardiovascular diseases on human health through the relevant targeting molecular mechanism.
Collapse
Affiliation(s)
- Chong-Sun Khoi
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City 220216, Taiwan;
- Graduate School of Biotechnology and Bioengineering, College of Engineering, Yuan Ze University, Taoyuan City 320315, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City 220216, Taiwan;
- Department of Mechanical Engineering, College of Engineering, Yuan Ze University, Taoyuan City 320315, Taiwan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei City 10617, Taiwan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei City 10617, Taiwan
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei City 100229, Taiwan
| |
Collapse
|
2
|
Park KH, Kwon HJ, Jeong EO, Koh HS, Lim J. The Potential Therapeutic Effects of Tadalafil on the Endothelium in a Subarachnoid Hemorrhage Animal Model: Insights from Immunohistochemical Staining. Curr Issues Mol Biol 2024; 46:9555-9564. [PMID: 39329919 PMCID: PMC11430039 DOI: 10.3390/cimb46090567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
This study investigated the potential of phosphodiesterase type 5 (PDE-5) inhibitors, specifically tadalafil, in preventing the delayed cerebral ischemia (DCI) post-rupture of cerebral aneurysms. A total of 19 rabbits were used in this study, divided into different treatment groups, including nimodipine alone, tadalafil alone, and a combination of nimodipine and tadalafil. Both nimodipine and tadalafil showed some impact on reducing endothelial apoptosis in the basilar arteries, although the effects were not statistically significant. Notably, the nimodipine group exhibited significantly lower levels of Bax in the small arterioles compared to the SAH group. These findings suggest that while tadalafil may not directly prevent endothelial cell death like nimodipine, its neuroprotective properties hint at its potential utility in DCI treatment. Further research involving a broader range of apoptosis-related proteins is recommended to enhance our understanding in this area.
Collapse
Affiliation(s)
- Kwang Hyon Park
- Department of Neurosurgery, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea;
| | - Hyon-Jo Kwon
- Department of Neurosurgery, Chungnam National University School of Medicine, Daejeon 305764, Republic of Korea; (H.-J.K.); (H.-S.K.)
- Department of Neurosurgery, Chungnam National University Hospital, Daejeon 34134, Republic of Korea;
| | - Eun-Oh Jeong
- Department of Neurosurgery, Chungnam National University Hospital, Daejeon 34134, Republic of Korea;
| | - Hyeon-Song Koh
- Department of Neurosurgery, Chungnam National University School of Medicine, Daejeon 305764, Republic of Korea; (H.-J.K.); (H.-S.K.)
- Department of Neurosurgery, Chungnam National University Hospital, Daejeon 34134, Republic of Korea;
| | - Jeongwook Lim
- Department of Neurosurgery, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea;
- Department of Neurosurgery, Chungnam National University School of Medicine, Daejeon 305764, Republic of Korea; (H.-J.K.); (H.-S.K.)
| |
Collapse
|
3
|
Gong WY, Xu B, Liu L, Li ST. Dezocine relieves the postoperative hyperalgesia in rats through suppressing the hyper-action of Akt1/GSK-3β pathway. Exp Brain Res 2022; 240:1435-1444. [PMID: 35333956 DOI: 10.1007/s00221-022-06341-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/24/2022] [Indexed: 11/28/2022]
Abstract
The relieving role of dezocine in pain after surgery was previously reported, while the potential mechanism was not completely clear. Therefore, the current research probed into the regulatory mechanism of dezocine in pain after surgery. A postoperative pain model was established by performing plantar incision surgery on the juvenile Sprague-Dawley rats. After the rats were treated with dezocine or SC79 (Akt1 activator), the paw withdrawal threshold and paw withdrawal latency of rats were detected to evaluate the mechanical allodynia and thermal hyperalgesia. After the plantar tissue, dorsal root ganglions, and spinal cord of rats were collected, the expressions of Akt1, p-Akt1, GSK-3β, and p-GSK-3β in the tissues were determined by western blot to evaluate the activation state of the Akt1/GSK-3β pathway. After surgery, the paw withdrawal threshold and paw withdrawal latency of rats were lessened, whereas the ratios of p-Akt1/Akt1 and p-GSK-3β/GSK-3β were augmented in rat plantar tissue, dorsal root ganglions, and spinal cord. After treatment with dezocine alone, the paw withdrawal threshold and paw withdrawal latency of postoperative rats were elevated, but ratios of p-Akt1/Akt1 and p-GSK-3β/GSK-3β were reduced. After co-treatment with dezocine and SC79, SC79 reversed the effects of dezocine on elevating the paw withdrawal threshold and paw withdrawal latency, and reducing the ratios of p-Akt1/Akt1 and p-GSK-3β/GSK-3β in postoperative rats. Dezocine ameliorated the postoperative hyperalgesia in rats via repressing the hyper-action of Akt1/GSK-3β pathway.
Collapse
Affiliation(s)
- Wen-Yi Gong
- Department of Anesthesiology, Shanghai General Hospital of Nanjing Medical University, No.100, Haining Road, Hongkou District, Shanghai, 200080, People's Republic of China.,Department of Anesthesiology, Wusong Hospital, No.101, North Tongtai Road, Baoshan District, Shanghai, 200940, People's Republic of China
| | - Bing Xu
- Department of Anesthesiology, Wusong Hospital, No.101, North Tongtai Road, Baoshan District, Shanghai, 200940, People's Republic of China
| | - Li Liu
- Department of Anesthesiology, Wusong Hospital, No.101, North Tongtai Road, Baoshan District, Shanghai, 200940, People's Republic of China
| | - Shi-Tong Li
- Department of Anesthesiology, Shanghai General Hospital of Nanjing Medical University, No.100, Haining Road, Hongkou District, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
4
|
Tang W, Luo L, Hu B, Zheng M. Butorphanol alleviates lipopolysaccharide-induced inflammation and apoptosis of cardiomyocytes via activation of the κ-opioid receptor. Exp Ther Med 2021; 22:1248. [PMID: 34539844 PMCID: PMC8438658 DOI: 10.3892/etm.2021.10683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/16/2021] [Indexed: 12/25/2022] Open
Abstract
Sepsis-induced myocardial dysfunction is a leading cause of the high mortality rates associated with sepsis. The aim of the present study was to investigate the effect of butorphanol on sepsis-induced cardiomyocyte dysfunction. Lipopolysaccharide (LPS) was used to induce H9C2 cardiomyocytes to establish an in vitro sepsis model. The effect of butorphanol on the viability of LPS-induced H9C2 cells was analyzed using a Cell Counting Kit-8 assay. The levels of tumor necrosis factor-α, interleukin (IL)-1β and IL-6 were detected using ELISA. Western blotting was used to analyze the expression levels of inflammation-and apoptosis-related proteins. Cell apoptosis was measured using a TUNEL assay. The expression levels of κ-opioid receptor (KOR) were analyzed using reverse transcription-quantitative PCR analysis and western blotting. Following LPS induction, the levels of inflammatory cytokines and proapoptotic proteins were found to be upregulated in H9C2 cells, while butorphanol treatment downregulated these levels. The expression levels of KOR were also upregulated following butorphanol treatment in LPS-induced H9C2 cells. Addition of the KOR inhibitor, nor-binaltorphimine, alleviated the inhibitory effects of butorphanol on inflammation and apoptosis in LPS-induced H9C2 cells. In conclusion, the findings of the present study provided evidence indicating that butorphanol may alleviate LPS-induced inflammation and apoptosis in cardiomyocytes by upregulating KOR expression, which may provide a novel insight into the potential therapeutic effects of butorphanol and its underlying mechanism of action.
Collapse
Affiliation(s)
- Weiqing Tang
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Liu Luo
- Department of Anesthesiology, The Affiliated Zhuzhou Hospital of Xiangya School of Medicine, Central South University, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Baoji Hu
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Mingzhi Zheng
- Department of Anesthesiology, The Affiliated Zhuzhou Hospital of Xiangya School of Medicine, Central South University, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| |
Collapse
|
5
|
The expression of kappa-opioid receptor promotes the migration of breast cancer cells in vitro. BMC Anesthesiol 2021; 21:210. [PMID: 34461834 PMCID: PMC8404350 DOI: 10.1186/s12871-021-01429-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 08/18/2021] [Indexed: 12/17/2022] Open
Abstract
Background Opioid receptors are implicated in cell proliferation and cancer migration. However, the effects and underlying mechanisms of opioid receptor κ (OPRK1) in breast cancer remain unknown. Methods Small interfering RNA (siRNAs) was used to knockdown the expression of OPRK1. Western blot was used to determine the protein expression and reverse transcription-quantitative PCR (RT-qPCR) determined the genes transcription. Cell viability was detected by MTT assay and cell death rates were determined by Annexin V/PI and flow cytometry. Cell migration and invasion were detected by wound healing analysis and transwell assay, respectively. Results Our research demonstrated that OPRK1 was overexpressed in breast cancer cells compared with the normal human mammary epithelial cells. OPRK1 knockdown could inhibited cell viability and migration in cancer cells, accompanied with the decreased proteins and genes expression of N-cadherin, Snail, MMP2 and Vimentin, while the E-cadherin expression was increased. Additionally, OPRK1 knockdown also promoted PI3K/AKT signaling inactivation. Activation of AKT reversed the OPRK1 knockdown-induced cell viability inhibition and migration suppression, while inhibition of AKT reduced cell viability and promoted cell death. Conclusions Our findings illustrated the role of OPRK1 played on promoting migration in vitro, and we also provided the therapeutic research of OPRK1 knockdown combined with AKT inhibition.
Collapse
|
6
|
Zhang S, Yang F, Shi R, Liu C, Zhao L, Gu X, Liu Y, Fu F, Feng N, Liu Y, Jia M, Fan R, Yang L, Li J, Li J, Pei J. Activation of κ-opioid receptor inhibits inflammatory response induced by sodium palmitate in human umbilical vein endothelial cells. Cytokine 2021; 146:155659. [PMID: 34332276 DOI: 10.1016/j.cyto.2021.155659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/11/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The current study aims to investigate the effect of κ-opioid receptor (κ-OR) activation on sodium palmitate (SP)-induced human umbilical vein endothelial cells (HUVECs) inflammatory response and elucidate the underlying mechanisms. METHODS A hyperlipidemic cell model was established and treated with κ-OR agonist (U50,488H), and antagonist (norbinaltorphimine, nor-BNI), or inhibitors targeting PI3K, Akt or eNOS (LY294002, MK2206-2HCl or L-NAME, respectively). Furthermore, the expression levels of NLRP3, caspase-1, p-Akt, Akt, p-eNOS, and total eNOS were evaluated. Additionally, the production of reactive oxygen species, and levels of inflammatory factors, such as TNF-α, IL-1β, IL-6, IL-1 and adhesion molecules, such as ICAM-1, VCAM-1, P-selectin, and E-selectin were determined. The adherence rates of the neutrophils and monocytes were assessed as well. RESULTS The SP-induced hyperlipidemic cell model demonstrated increased expression of NLRP3 and caspase-1 proteins (P < 0.05) and elevated ROS levels (P < 0.01), and decreased phosphorylated-Akt and phosphorylated-eNOS expression (P < 0.05). In addition, SP significantly increased TNF-α, IL-1β, IL-6, ICAM-1, VCAM-1, P-selectin, and E-selectin levels (P < 0.01), decreased IL-10 levels (P < 0.01), and increased the adhesion rates of monocytes and neutrophils (P < 0.01). The SP-induced inflammatory response in HUVECs was ameliorated by κ-OR agonist, U50,488H. However, the protective effect of U50,488H was abolished by κ-OR antagonist, nor-BNI, and inhibitors of PI3K, Akt and eNOS. CONCLUSION Our findings suggest that κ-OR activation inhibits SP-induced inflammation by activating the PI3K/Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Shumiao Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Fan Yang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Rui Shi
- School of Life Sciences, Northwest University, No.1 North Taibai Road, Xi'an 710069, Shaanxi Province, People's Republic of China
| | - Chaoyang Liu
- School of Life Sciences, Northwest University, No.1 North Taibai Road, Xi'an 710069, Shaanxi Province, People's Republic of China
| | - Lei Zhao
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an 710033, Shaanxi Province, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Yinji Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Na Feng
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Yali Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Min Jia
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Rong Fan
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Lu Yang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Jun Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Juan Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China.
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China; School of Life Sciences, Northwest University, No.1 North Taibai Road, Xi'an 710069, Shaanxi Province, People's Republic of China.
| |
Collapse
|
7
|
Abstract
This paper is the forty-second consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2019 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
8
|
Niu J, Wu C, Zhang M, Yang Z, Liu Z, Fu F, Li J, Feng N, Gu X, Zhang S, Liu Y, Fan R, Li J, Pei J. κ-opioid receptor stimulation alleviates rat vascular smooth muscle cell calcification via PFKFB3-lactate signaling. Aging (Albany NY) 2021; 13:14355-14371. [PMID: 34016793 PMCID: PMC8202865 DOI: 10.18632/aging.203050] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/31/2021] [Indexed: 12/15/2022]
Abstract
In the present study, the effects and mechanism of action of U50,488H (a selective κ-opioid receptor agonist) on calcification of rat vascular smooth muscle cells (VSMCs) induced by β-glycerophosphate (β-GP) were investigated. VSMCs were isolated and cultured in traditional FBS-based media. A calcification model was established in VSMCs under hyperphosphatemia and intracellular calcium contents. Alkaline phosphatase (ALP), lactate dehydrogenase (LDH), and lactate were detected in cell culture supernatants before and after treatment. Alizarin red staining was used to detect the degree of calcification of VSMCs. Expression levels of key molecules of osteogenic markers, fructose-2,6-biphosphatase 3 (PFKFB3), and proline hydroxylase 2 (PHD2), were determined using western blotting. Further, vascular calcification was induced by vitamin D3 plus nicotine in rats and isolated thoracic aortas, calcium concentration was assessed in rat aortic rings in vitro. We demonstrated that U50,488H inhibited VSMC calcification in a concentration-dependent manner. Moreover, U50,488H significantly inhibited osteogenic differentiation and ALP activity in VSMCs pretreated with β-GP. Further studies confirmed that PFKFB3 expression, LDH level, and lactate content significantly increased during calcification of VSMCs; U50,488H reversed these changes. PHD2 expression showed the opposite trend compared to PFKFB3 expression. nor-BNI or 3-PO abolished U50,488H protective effects. Besides, U50,488H inhibited VSMC calcification in rat aortic rings ex vivo. Collectively, our experiments show that κ-opioid receptor activation inhibits VSMC calcification by reducing PFKFB3 expression and lactate content, providing a potential drug target and strategy for the clinical treatment of vascular calcification.
Collapse
Affiliation(s)
- Jin Niu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
- Department of Healthcare of 940 Hospital, Joint Logistics Support Force of PLA, Lanzhou 730000, Gansu Province, China
| | - Chen Wu
- Department of Neurology, Xinjiang Military General Hospital, Urumqi 830000, Xinjiang Province, China
| | - Min Zhang
- Department of College of Life Sciences, Northwest University, Xi'an 710032, Shaanxi Province, China
| | - Zhen Yang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Zhenhua Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Jun Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Na Feng
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Shumiao Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Yali Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Rong Fan
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Juan Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
9
|
Wang XL, Zeng S, Li XX, Zhao Y, Wang XH, Li T, Liu S. The Protective Effects of Butorphanol on Pulmonary Function of Patients with Obesity Undergoing Laparoscopic Bariatric Surgery: a Double-Blind Randomized Controlled Trial. Obes Surg 2021; 30:3919-3929. [PMID: 32535786 DOI: 10.1007/s11695-020-04755-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Obesity is a risk factor for postoperative pulmonary complications (PPCs). Recent studies have reported the pulmonary protective role of the kappa opioid receptor (KOR). Butorphanol is a narcotic with strong KOR agonist action, and the role in pulmonary protection is uncertain. Here, we hypothesized that butorphanol exerts protective effects on pulmonary function in patients with obesity undergoing laparoscopic bariatric surgery. METHODS Patients with a body mass index ≥ 30 kg/m2 scheduled for laparoscopic bariatric surgery were randomized to receive butorphanol or normal saline. Butorphanol was administered as an initial loading dose of 10 μg/kg at 5 min before induction followed by 5 μg/(kg h) during surgery. The primary outcome was arterial-alveolar oxygen tension ratio (a/A ratio). Secondary outcomes included other pulmonary variables, biomarkers reflecting pulmonary injury, and incidence of PPCs within 7 days after surgery. RESULTS Patients in the butorphanol group had a significantly higher a/A ratio at 1 h after the operation began (68 ± 7 vs. 55 ± 8, P < 0.001), end of the operation (73 ± 8 vs. 59 ± 7, P < 0.001), and 1 h after extubation (83 ± 9 vs. 70 ± 5, P < 0.001) compared with those in the control group. In addition, in the butorphanol group, dead space to tidal volume ratios were significantly lower than those in the control group at the same time points (all P < 0.001). In the control group, the levels of biomarkers reflecting pulmonary injury were significantly higher than those in the butorphanol group at 3 h, 6 h, 12 h, and 24 h postoperatively (P < 0.001). The incidence of PPCs was similar in both groups. CONCLUSION Butorphanol administration protected pulmonary function by improving oxygenation and reducing dead space ventilation in patients with obesity undergoing laparoscopic bariatric surgery. Butorphanol may therefore provide clinical benefits in patients with obesity.
Collapse
Affiliation(s)
- Xiu-Li Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Si Zeng
- Department of Anesthesiology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiao-Xiao Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ye Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xing-He Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tong Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Su Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou, 221000, Jiangsu, China.
| |
Collapse
|
10
|
Chen R, Chen T, Wang T, Dai X, Meng K, Zhang S, Jiang D, Wang Y, Zhou K, Geng T, Xu J, Wang Y. Tongmai Yangxin pill reduces myocardial no-reflow by regulating apoptosis and activating PI3K/Akt/eNOS pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 261:113069. [PMID: 32619593 DOI: 10.1016/j.jep.2020.113069] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/15/2020] [Accepted: 05/31/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tongmai Yangxin pill (TMYX) is derived from the Zhigancao decoction recorded in Shang han lun by Zhang Zhongjing during the Han dynasty and was further improved by Professor Ruan Shiyi, a cardiovascular expert at Tianjin University of Traditional Chinese Medicine. TMYX is used for the clinical treatment of chest pain, heartache, and qi-yin-deficiency coronary heart disease and can improve vascular endothelial function in patients with angina pectoris or coronary heart disease by up-regulating nitric oxide activity and then regulating vascular tension. Whether TMYX can further improve myocardial no-reflow by up-regulating NO activity and then dilating blood vessels remains unclear. AIM OF THE STUDY This study aimed to reveal whether TMYX can further improve myocardial NR by up-regulating NO activity and then dilating blood vessels. The mechanism underlying PI3K/Akt/eNOS pathway activation and apoptosis regulation is also explored. MATERIALS AND METHODS The left anterior descending coronary arteries of healthy adult male SD rats were ligated to establish a NR model. The rats were assigned to 14 groups: control, sham, NR, TMYX (4.0 g/kg), sodium nitroprusside (SNP), Tongxinluo capsule (TXL), PI3K blocker (LY), TMYX + LY, SNP + LY, TXL + LY, eNOS blocker (L-NAME), TMYX + L-NAME, SNP + L-NAME, and TXL + L-NAME groups. Cardiac function was measured through echocardiography. Thioflavin S, Evans Blue, and TTC staining were adopted to evaluate NR and ischemic areas. Cell inflammation degree and edema were assessed by hematoxylin-eosin staining. Automated biochemical analyzer and kit were used to detect the activities of myocardial oxidants, including reactive oxygen species, super oxide dismutase, malonaldehyde, and NO. The expression levels of genes and proteins in the PI3K/Akt/eNOS signaling pathway and apoptosis were detected via real-time fluorescence quantitative PCR and Western blot analysis, respectively. A microvascular tension sensor was adopted to detect coronary artery diastolic function in vitro. RESULTS TMYX reduced NR and ischemic areas; suppressed LV-mass; enhanced EF, FS, LVOT peak, and LVSV; and improved cardiac structure and function. Moreover, it decreased creatine kinase (CK), CK-MB, and lactic dehydrogenase activities. TMYX increased NO and super oxide dismutase activities; inhibited malonaldehyde activity; reduced muscle fiber swelling and inflammatory cell infiltration; and improved vasodilation in vitro. In the NR myocardium, TMYX stimulated myocardial PI3K activities and PI3K (Tyr458) phosphorylation and enhanced Akt activities and Akt phosphorylation at Tyr315. TMYX increased the activities of eNOS and the phosphorylation of eNOS at Ser1177 in the NR myocardium and attenuated cardiomyocyte apoptosis by increasing the expression of Bcl-2 and decreasing that of caspase-3 and Bax. All these effects of TMYX were abolished by the specific inhibitors of PI3K (LY) and eNOS (L-NAME). CONCLUSIONS TMYX attenuates myocardial NR after ischemia and reperfusion by activating the PI3K/Akt/eNOS pathway and regulating apoptosis, further up-regulating NO activity and relaxing coronary microvessels.
Collapse
Affiliation(s)
- Rui Chen
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Ting Chen
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Tianqi Wang
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Xiangdong Dai
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Ke Meng
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Shuying Zhang
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Di Jiang
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Yanyan Wang
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Kun Zhou
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Tong Geng
- Tianjin Zhongxin Pharmaceutical Group Co. Ltd. Research Institute Branch, Tianjin, 300457, China.
| | - Jinpeng Xu
- Tianjin Zhongxin Pharmaceutical Group Co. Ltd, Drug Marketing Co., Ltd, Tianjin, 300193, China.
| | - Yi Wang
- Institute of traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
11
|
Ortiz MI, Cariño-Cortés R, Castañeda-Hernández G. Participation of the opioid receptor - nitric oxide - cGMP - K + channel pathway in the peripheral antinociceptive effect of nalbuphine and buprenorphine in rats. Can J Physiol Pharmacol 2020; 98:753-762. [PMID: 33095677 DOI: 10.1139/cjpp-2020-0104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The aim of this study was to examine if the peripheral antinociceptive effects of the opioid agonist/antagonist nalbuphine and buprenorphine involve the sequential participation of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) synthesis followed by K+ channel opening in the formalin test. Wistar rats (180-220 g) were injected in the dorsal surface of the right hind paw with formalin (1%). Rats received a subcutaneous (s.c.) injection into the dorsal surface of the paw of vehicles or increasing doses of nalbuphine (50-200 μg/paw) or buprenorphine (1-5 μg/paw) 20 min before formalin injection into the paw. Nalbuphine antinociception was reversed by the s.c. injection into the paw of the inhibitor of NO synthesis (NG-nitro-l-arginine methyl ester (L-NAME)), by the inhibitor of guanylyl cyclase (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ)), by the Kir6.1-2, ATP-sensitive K+ channel inhibitors (glibenclamide and glipizide), by the KCa2.1-3, small conductance Ca2+-activated K+ channel blocker (apamin), by the KCa1.1, large conductance Ca2+-activated K+ channel blocker (charybdotoxin), and by the KV, voltage-dependent K+ channel inhibitors (4-aminopyridine (4-AP) and tetraethylammonium chloride (TEA)). The antinociceptive effect produced by buprenorphine was blocked by the s.c. injection of 4-AP and TEA but not by L-NAME, ODQ, glibenclamide, glipizide, apamin, or charybdotoxin. The present results provide evidence for differences in peripheral mechanisms of action between these opioid drugs.
Collapse
Affiliation(s)
- Mario I Ortiz
- Área Académica de Medicina del Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Hidalgo, Mexico
| | - Raquel Cariño-Cortés
- Área Académica de Medicina del Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, Hidalgo, Mexico
| | - Gilberto Castañeda-Hernández
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
12
|
Guo X, Liu X, Wang J, Fu X, Yao J, Zhang X, Jackson S, Li J, Zhang W, Sun D. Pigment epithelium-derived factor (PEDF) ameliorates arsenic-induced vascular endothelial dysfunction in rats and toxicity in endothelial EA.hy926 cells. ENVIRONMENTAL RESEARCH 2020; 186:109506. [PMID: 32315827 DOI: 10.1016/j.envres.2020.109506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/28/2020] [Accepted: 04/08/2020] [Indexed: 06/11/2023]
Abstract
Although the harmful effects of arsenic exposure on the cardiovascular system have received great attention, there is still no effective treatment. Vascular endothelial dysfunction (VED) is the initial step of cardiovascular diseases, where pigment epithelium-derived factor (PEDF) plays an important role in maintaining endothelial function. Here, we explored the protective role of PEDF in VED induced by arsenic, and its underlying molecular mechanism, designing an in vivo rat model of arsenic exposure recovery and in vitro endothelial EA. hy926 cell-based assays. The edema of aortic endothelial cells in rats significantly improved during recovery from arsenite exposure compared with rats exposed to 10 and 50 mg/L arsenite continuously. In addition, serum levels of nitric oxide (NO), von Willebrand factor, and nitric oxide synthase (inducible and total activities) in rats, which were greatly affected by arsenite exposure, returned to levels similar to those in the control group after recovery with distilled water. The recovery from arsenite exposure was associated with increased levels of PEDF; decreased protein levels of Fas, FasL, P53, and phospho-p38; and inhibited apoptosis in aortic endothelial cells in vivo. Recombinant human PEDF treatment (100 nM) prevented the toxic effects of arsenite (50 μM) on endothelial cells in vitro by increasing NO content, decreasing reactive oxygen species (ROS) levels, and inhibiting apoptosis, as well as increasing cell viability and decreasing levels of P53 and phospho-p38. Our findings suggest that PEDF protects endothelial cells from arsenic-induced VED by increasing NO release and inhibiting apoptosis, where P53 and p38MAPK are its main targets.
Collapse
Affiliation(s)
- Xiangnan Guo
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Harbin Medical University Cancer Hospital, China
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
| | - Jingqiu Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
| | - Xiaoyan Fu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
| | - Jinyin Yao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
| | - Xiaodan Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
| | - Sira Jackson
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
| | - Jinyu Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China
| | - Wei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China.
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China.
| |
Collapse
|