201
|
Ji L, Song T, Ge C, Wu Q, Ma L, Chen X, Chen T, Chen Q, Chen Z, Chen W. Identification of bioactive compounds and potential mechanisms of scutellariae radix-coptidis rhizoma in the treatment of atherosclerosis by integrating network pharmacology and experimental validation. Biomed Pharmacother 2023; 165:115210. [PMID: 37499457 DOI: 10.1016/j.biopha.2023.115210] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
OBJECTIVE This study aims at investigating the potential targets and functional mechanisms of Scutellariae Radix-Coptidis Rhizoma (QLYD) against atherosclerosis (AS) through network pharmacology, molecular docking, bioinformatic analysis and experimental validation. METHODS The compositions of QLYD were collected from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and literature, where the main active components of QLYD and corresponding targets were identified. The potential therapeutic targets of AS were excavated using the OMIM database, DrugBank database, DisGeNET database, CTD database and GEO datasets. The protein-protein interaction (PPI) network of common targets was constructed and visualized by Cytoscape 3.7.2 software. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analysis were performed to analyze the function of core targets in the PPI network. Molecular docking was carried out using AutoDockTools, AutoDock Vina, and PyMOL software to verify the correlation between the main components of QLYD and the core targets. Mouse AS model was established and the results of network pharmacology were verified by in vivo experiments. RESULTS Totally 49 active components and 225 corresponding targets of QLYD were obtained, where 68 common targets were identified by intersecting with AS-related targets. Five hub genes including IL6, VEGFA, AKT1, TNF, and IL1B were screened from the PPI network. GO functional analysis reported that these targets had associations mainly with cellular response to oxidative stress, regulation of inflammatory response, epithelial cell apoptotic process, and blood coagulation. KEGG pathway analysis demonstrated that these targets were correlated to AGE-RAGE signaling pathway in diabetic complications, TNF signaling pathway, IL-17 signaling pathway, MAPK signaling pathway, and NF-kappa B signaling pathway. Results of molecular docking indicated good binding affinity of QLYD to FOS, AKT1, and TNF. Animal experiments showed that QLYD could inhibit inflammation, improve blood lipid levels and reduce plaque area in AS mice to prevent and treat AS. CONCLUSION QLYD may exert anti-inflammatory and anti-oxidative stress effects through multi-component, multi-target and multi-pathway to treat AS.
Collapse
Affiliation(s)
- Lingyun Ji
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Ting Song
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250011, China
| | - Chunlei Ge
- Department of Respiratory Medicine, Linyi Tradition Chinese Medical Hospital, Linyi, Shandong Province 276600, China
| | - Qiaolan Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Lanying Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Xiubao Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250011, China
| | - Ting Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Qian Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Zetao Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250011, China; Subject of Integrated Chinese and Western Medicine,Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China.
| | - Weida Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250011, China.
| |
Collapse
|
202
|
Zhang K, Zhang J, Kan C, Tian H, Ma Y, Huang N, Han F, Hou N, Sun X. Role of dysfunctional peri-organ adipose tissue in metabolic disease. Biochimie 2023; 212:12-20. [PMID: 37019205 DOI: 10.1016/j.biochi.2023.03.015] [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: 12/04/2022] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023]
Abstract
Metabolic disease is a complex disorder defined by a group with interrelated factors. There is growing evidence that obesity can lead to a variety of metabolic diseases, including diabetes and cardiovascular disease. Excessive adipose tissue (AT) deposition and ectopic accumulation can lead to increased peri-organ AT thickness. Dysregulation of peri-organ (perivascular, perirenal, and epicardial) AT is strongly associated with metabolic disease and its complications. The mechanisms include secretion of cytokines, activation of immunocytes, infiltration of inflammatory cells, involvement of stromal cells, and abnormal miRNA expression. This review discusses the associations and mechanisms by which various types of peri-organ AT affect metabolic diseases while addressing it as a potential future treatment strategy.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongzhan Tian
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yanhui Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Na Huang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China; Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China; Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China.
| |
Collapse
|
203
|
Zhang T, Deng W, Deng Y, Liu Y, Xiao S, Luo Y, Xiang W, He Q. Mechanisms of ferroptosis regulating oxidative stress and energy metabolism in myocardial ischemia-reperfusion injury and a novel perspective of natural plant active ingredients for its treatment. Biomed Pharmacother 2023; 165:114706. [PMID: 37400352 DOI: 10.1016/j.biopha.2023.114706] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 07/05/2023] Open
Abstract
Acute myocardial infarction remains the leading cause of death in humans. Timely restoration of blood perfusion to ischemic myocardium remains the most effective strategy in the treatment of acute myocardial infarction, which can significantly reduce morbidity and mortality. However, after restoration of blood flow and reperfusion, myocardial injury will aggravate and induce apoptosis of cardiomyocytes, a process called myocardial ischemia-reperfusion injury. Studies have shown that the loss and death of cardiomyocytes caused by oxidative stress, iron load, increased lipid peroxidation, inflammation and mitochondrial dysfunction, etc., are involved in myocardial ischemia-reperfusion injury. In recent years, with the in-depth research on the pathology of myocardial ischemia-reperfusion injury, people have gradually realized that there is a new form of cell death in the pathological process of myocardial ischemia-reperfusion injury, namely ferroptosis. A number of studies have found that in the myocardial tissue of patients with acute myocardial infarction, there are pathological changes closely related to ferroptosis, such as iron metabolism disorder, lipid peroxidation, and increased reactive oxygen species free radicals. Natural plant products such as resveratrol, baicalin, cyanidin-3-O-glucoside, naringenin, and astragaloside IV can also exert therapeutic effects by correcting the imbalance of these ferroptosis-related factors and expression levels. Combining with our previous studies, this review summarizes the regulatory mechanism of natural plant products intervening ferroptosis in myocardial ischemia-reperfusion injury in recent years, in order to provide reference information for the development of targeted ferroptosis inhibitor drugs for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Tianqing Zhang
- Department of Cardiology, The First People's Hospital of Changde City, Changde 415003, Hunan, China
| | - Wenxu Deng
- The Central Hospital of Hengyang, Hengyang, Hunan 421001, China
| | - Ying Deng
- People's Hospital of Ningxiang City, Ningxiang, Hunan, China
| | - Yao Liu
- The Second Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medcial School, University of South China, Hunan 421001, China.
| | - Sijie Xiao
- Department of Ultrasound, The First People's Hospital of Changde City, Changde 415003, China
| | - Yanfang Luo
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang Xiang
- Department of Immunology and Rheumatology, The First People's Hospital of Changde City, Changde 415003, China
| | - Qi He
- People's Hospital of Ningxiang City, Ningxiang, Hunan, China
| |
Collapse
|
204
|
Li Y, Zhu Y, Hu F, Liu L, Shen G, Tu Q. Procyanidin B2 regulates the Sirt1/Nrf2 signaling pathway to improve random-pattern skin flap survival. Phytother Res 2023; 37:3913-3925. [PMID: 37128130 DOI: 10.1002/ptr.7847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 05/03/2023]
Abstract
Random-pattern skin flaps have been widely used in the reconstruction of damaged tissues. Ischemia-reperfusion injury occurring in the distal regions of the flap is a common issue, which often leads to flap necrosis and restricts its clinical applications. Procyanidin B2 (PB2), a naturally occurring flavonoid in large quantities in various fruits, has been demonstrated to exhibit several significant pharmacological properties. However, the effect of PB2 on flap viability is not clearly known. Here, using Western blot analysis, immunohistochemistry, and immunofluorescence staining, we observed that PB2 significantly reduced oxidative stress and inflammation and enhanced angiogenesis. Mechanically, we provided evidence for the first time that the beneficial effects of PB2 occur through the activation of the Sirt1/Nrf2 signaling pathway. Moreover, co-administration of PB2 and EX527, a selective inhibitor of Sirt1, resulted in down-regulation of the expression of Sirt1, Nrf2, and downstream antioxidants. In summary, our study showed that PB2 might be a novel therapeutic strategy for improving the survival of random-pattern skin flaps.
Collapse
Affiliation(s)
- Yao Li
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an, China
- Nanjing Medical University, Nanjing, China
| | - Yurun Zhu
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an, China
- Nanjing Medical University, Nanjing, China
| | - Fei Hu
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Lue Liu
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an, China
| | - Guangjie Shen
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an, China
| | - Qiming Tu
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an, China
| |
Collapse
|
205
|
Fehsel K. Why Is Iron Deficiency/Anemia Linked to Alzheimer's Disease and Its Comorbidities, and How Is It Prevented? Biomedicines 2023; 11:2421. [PMID: 37760862 PMCID: PMC10526115 DOI: 10.3390/biomedicines11092421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Impaired iron metabolism has been increasingly observed in many diseases, but a deeper, mechanistic understanding of the cellular impact of altered iron metabolism is still lacking. In addition, deficits in neuronal energy metabolism due to reduced glucose import were described for Alzheimer's disease (AD) and its comorbidities like obesity, depression, cardiovascular disease, and type 2 diabetes mellitus. The aim of this review is to present the molecular link between both observations. Insufficient cellular glucose uptake triggers increased ferritin expression, leading to depletion of the cellular free iron pool and stabilization of the hypoxia-induced factor (HIF) 1α. This transcription factor induces the expression of the glucose transporters (Glut) 1 and 3 and shifts the cellular metabolism towards glycolysis. If this first line of defense is not adequate for sufficient glucose supply, further reduction of the intracellular iron pool affects the enzymes of the mitochondrial electron transport chain and activates the AMP-activated kinase (AMPK). This enzyme triggers the translocation of Glut4 to the plasma membrane as well as the autophagic recycling of cell components in order to mobilize energy resources. Moreover, AMPK activates the autophagic process of ferritinophagy, which provides free iron urgently needed as a cofactor for the synthesis of heme- and iron-sulfur proteins. Excessive activation of this pathway ends in ferroptosis, a special iron-dependent form of cell death, while hampered AMPK activation steadily reduces the iron pools, leading to hypoferremia with iron sequestration in the spleen and liver. Long-lasting iron depletion affects erythropoiesis and results in anemia of chronic disease, a common condition in patients with AD and its comorbidities. Instead of iron supplementation, drugs, diet, or phytochemicals that improve energy supply and cellular glucose uptake should be administered to counteract hypoferremia and anemia of chronic disease.
Collapse
Affiliation(s)
- Karin Fehsel
- Neurobiochemical Research Unit, Department of Psychiatry, Medical Faculty, Heinrich-Heine-University, 240629 Düsseldorf, Germany
| |
Collapse
|
206
|
Szekeres R, Priksz D, Kiss R, Romanescu DD, Bombicz M, Varga B, Gesztelyi R, Szilagyi A, Takacs B, Tarjanyi V, Pelles-Tasko B, Forgacs I, Remenyik J, Szilvassy Z, Juhasz B. Therapeutic Aspects of Prunus cerasus Extract in a Rabbit Model of Atherosclerosis-Associated Diastolic Dysfunction. Int J Mol Sci 2023; 24:13253. [PMID: 37686067 PMCID: PMC10488229 DOI: 10.3390/ijms241713253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
This study evaluates the potential therapeutic effects of anthocyanin-rich Prunus cerasus (sour cherry) extract (PCE) on atherosclerosis-associated cardiac dysfunction, described by the impairment of the NO-PKG (nitric oxide-protein kinase G) pathway and the antioxidant capacity. Initially, a rabbit model of atherosclerotic cardiovascular disease was established by administering a cholesterol-rich diet, enabling the examination of the impact of 9 g/kg PCE on the pre-existing compromised cardiovascular condition. After that, the animals were divided into four groups for 12 weeks: the (1) untreated control group; (2) PCE-administered healthy rabbits; (3) hypercholesterolemic (HC) group kept on an atherogenic diet; and (4) PCE-treated HC group. Dyslipidemia, impaired endothelial function, and signs of diastolic dysfunction were evident in hypercholesterolemic rabbits, accompanied by a reduced cardiac expression of eNOS (endothelial nitric oxide synthase), PKG, and SERCA2a (sarco/endoplasmic reticulum calcium ATPase 2a). Subsequent PCE treatment improved the lipid profile and the cardiac function. Additionally, PCE administration was associated with elevated myocardial levels of eNOS, PKG, and SERCA2a, while no significant changes in the vascular status were observed. Western blot analysis further revealed hypercholesterolemia-induced increase and PCE-associated reduction in heme oxygenase-1 expression. The observed effects of anthocyanins indicate their potential as a valuable addition to the treatment regimen for atherosclerosis-associated cardiac dysfunction.
Collapse
Affiliation(s)
- Reka Szekeres
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Rita Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Dana Diana Romanescu
- Department of Diabetology, Pelican Clinical Hospital, 410087 Oradea, Romania;
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Balazs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Anna Szilagyi
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Barbara Takacs
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Vera Tarjanyi
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Beata Pelles-Tasko
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Ildiko Forgacs
- Center for Complex Systems and Microbiome Innovations, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, H-4032 Debrecen, Hungary; (I.F.); (J.R.)
| | - Judit Remenyik
- Center for Complex Systems and Microbiome Innovations, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, H-4032 Debrecen, Hungary; (I.F.); (J.R.)
| | - Zoltan Szilvassy
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| | - Bela Juhasz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (R.S.); (D.P.); (R.K.); (M.B.); (B.V.); (R.G.); (A.S.); (B.T.); (V.T.); (B.P.-T.); (Z.S.)
| |
Collapse
|
207
|
Gao L, Wang T, Zhuoma D, Yuan R, Huang S, Li B. Farrerol attenuates glutamate-induced apoptosis in HT22 cells via the Nrf2/heme oxygenase-1 pathway. Biosci Biotechnol Biochem 2023; 87:1009-1016. [PMID: 37348480 DOI: 10.1093/bbb/zbad084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
Farrerol is a flavonoid found in plants with a wide range of pharmacological effects, including protection and enhancement of nerve cell function, as well as antioxidant and antibacterial properties, among others. Neurodegenerative diseases are irreversible neurological disorders resulting from the loss of neuronal cells in the brain and spinal cord. In this experiment, we investigated the neuroprotective and antioxidant effects of farrerol on glutamate-induced HT22 cells. Our results showed that farrerol inhibited reactive oxygen species expression, apoptosis, mitochondrial damage, and the activation of caspases 3 and 9 in HT22 cells induced by glutamate. Additionally, farrerol potentially regulated the Nrf2/heme oxygenase-1 (HO-1) signaling pathway, as it attenuated the nuclear translocation of Nrf2 and promoted the expression of HO-1. These findings suggest that farrerol has potential as a new therapeutic option.
Collapse
Affiliation(s)
- Liying Gao
- Department of Pharmacy, Key Laboratory of Pharmaceutical Research for Metabolic Diseases, Qingdao University of Science & Technology, Qingdao, China
| | - Tong Wang
- Department of Pharmacy, Key Laboratory of Pharmaceutical Research for Metabolic Diseases, Qingdao University of Science & Technology, Qingdao, China
| | - Dongzhi Zhuoma
- Department of Medicament, College of Medicine, Tibet University, Lhasa, China
| | - Ruiying Yuan
- Department of Medicament, College of Medicine, Tibet University, Lhasa, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Shan Huang
- Department of Pharmacy, Key Laboratory of Pharmaceutical Research for Metabolic Diseases, Qingdao University of Science & Technology, Qingdao, China
| | - Bin Li
- Department of Pharmacy, Key Laboratory of Pharmaceutical Research for Metabolic Diseases, Qingdao University of Science & Technology, Qingdao, China
| |
Collapse
|
208
|
Almukainzi M, El-Masry TA, Selim H, Saleh A, El-Sheekh M, Makhlof MEM, El-Bouseary MM. New Insight on the Cytoprotective/Antioxidant Pathway Keap1/Nrf2/HO-1 Modulation by Ulva intestinalis Extract and Its Selenium Nanoparticles in Rats with Carrageenan-Induced Paw Edema. Mar Drugs 2023; 21:459. [PMID: 37755072 PMCID: PMC10533125 DOI: 10.3390/md21090459] [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: 06/22/2023] [Revised: 08/09/2023] [Accepted: 08/19/2023] [Indexed: 09/28/2023] Open
Abstract
Currently, there is growing interest in exploring natural bioactive compounds with anti-inflammatory potential to overcome the side effects associated with the well-known synthetic chemicals. Algae are a rich source of bioactive molecules with numerous applications in medicine. Herein, the anti-inflammatory effect of Ulva intestinalis alone or selenium nanoparticles loaded with U. intestinalis (UISeNPs), after being fully characterized analytically, was investigated by a carrageenan-induced inflammation model. The pretreated groups with free U. intestinalis extract (III and IV) and the rats pretreated with UISeNPs (groups V and VI) showed significant increases in the gene expression of Keap1, with fold increases of 1.9, 2.27, 2.4, and 3.32, respectively. Similarly, a remarkable increase in the Nrf2 gene expression, with 2.09-, 2.36-, 2.59-, and 3.7-fold increases, was shown in the same groups, respectively. Additionally, the groups III, IV, V, and VI revealed a significantly increased HO-1 gene expression with a fold increase of 1.48, 1.61, 1.87, and 2.84, respectively. Thus, both U. intestinalis extract and the UISeNPs boost the expression of the cytoprotective/antioxidant pathway Keap1/Nrf2/HO-1, with the UISeNPs having the upper hand over the free extract. In conclusion, U. intestinalis and UISeNPs have proven promising anti-inflammatory activity through mediating different underlying mechanisms.
Collapse
Affiliation(s)
- May Almukainzi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia; (M.A.); (A.S.)
| | - Thanaa A. El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Hend Selim
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia; (M.A.); (A.S.)
| | - Mostafa El-Sheekh
- Botany Department, Faculty of Science, Tanta University, Tanta 31527, Egypt;
| | - Mofida E. M. Makhlof
- Botany and Microbiology Department, Faculty of Science, Damanhour University, Damanhour 22511, Egypt;
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
209
|
Yan Q, Liu S, Sun Y, Chen C, Yang S, Lin M, Long J, Yao J, Lin Y, Yi F, Meng L, Tan Y, Ai Q, Chen N, Yang Y. Targeting oxidative stress as a preventive and therapeutic approach for cardiovascular disease. J Transl Med 2023; 21:519. [PMID: 37533007 PMCID: PMC10394930 DOI: 10.1186/s12967-023-04361-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023] Open
Abstract
Cardiovascular diseases (CVDs) continue to exert a significant impact on global mortality rates, encompassing conditions like pulmonary arterial hypertension (PAH), atherosclerosis (AS), and myocardial infarction (MI). Oxidative stress (OS) plays a crucial role in the pathogenesis and advancement of CVDs, highlighting its significance as a contributing factor. Maintaining an equilibrium between reactive oxygen species (ROS) and antioxidant systems not only aids in mitigating oxidative stress but also confers protective benefits on cardiac health. Herbal monomers can inhibit OS in CVDs by activating multiple signaling pathways, such as increasing the activity of endogenous antioxidant systems and decreasing the level of ROS expression. Given the actions of herbal monomers to significantly protect the normal function of the heart and reduce the damage caused by OS to the organism. Hence, it is imperative to recognize the significance of herbal monomers as prospective therapeutic interventions for mitigating oxidative damage in CVDs. This paper aims to comprehensively review the origins and mechanisms underlying OS, elucidate the intricate association between CVDs and OS, and explore the therapeutic potential of antioxidant treatment utilizing herbal monomers. Furthermore, particular emphasis will be placed on examining the cardioprotective effects of herbal monomers by evaluating their impact on cardiac signaling pathways subsequent to treatment.
Collapse
Affiliation(s)
- Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal&Child Health Care, Changsha, People's Republic of China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiao Yao
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Fan Yi
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, 100048, China
| | - Lei Meng
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yong Tan
- Department of Nephrology, Xiangtan Central Hospital, Xiangtan, 411100, China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
210
|
Shi YQ, Zhu XT, Zhang SN, Ma YF, Han YH, Jiang Y, Zhang YH. Premature ovarian insufficiency: a review on the role of oxidative stress and the application of antioxidants. Front Endocrinol (Lausanne) 2023; 14:1172481. [PMID: 37600717 PMCID: PMC10436748 DOI: 10.3389/fendo.2023.1172481] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023] Open
Abstract
Normal levels of reactive oxygen species (ROS) play an important role in regulating follicular growth, angiogenesis and sex hormone synthesis in ovarian tissue. When the balance between ROS and antioxidants is disrupted, however, it can cause serious consequences of oxidative stress (OS), and the quantity and quality of oocytes will decline. Therefore, this review discusses the interrelationship between OS and premature ovarian insufficiency (POI), the potential mechanisms and the methods by which antioxidants can improve POI through controlling the level of OS. We found that OS can mediate changes in genetic materials, signal pathways, transcription factors and ovarian microenvironment, resulting in abnormal apoptosis of ovarian granulosa cells (GCs) and abnormal meiosis as well as decreased mitochondrial Deoxyribonucleic Acid(mtDNA) and other changes, thus accelerating the process of ovarian aging. However, antioxidants, mesenchymal stem cells (MSCs), biological enzymes and other antioxidants can delay the disease process of POI by reducing the ROS level in vivo.
Collapse
Affiliation(s)
- Yu-Qian Shi
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xi-Ting Zhu
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Su-Na Zhang
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yi-Fu Ma
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan-Hua Han
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yue Jiang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yue-Hui Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
211
|
Ma L, Zhao Z, Zhao Y, Gao Y, Zhao L, Li S. Weizmannia coagulans JA845 improves atherosclerosis induced by vitamin D3 and high-fat diet in rats through modulating lipid metabolism, oxidative stress, and endothelial vascular injury. J Appl Microbiol 2023; 134:lxad165. [PMID: 37516440 DOI: 10.1093/jambio/lxad165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/17/2023] [Accepted: 07/28/2023] [Indexed: 07/31/2023]
Abstract
AIMS Probiotics have been proved to be strongly linked to the occurrence and progression of atherosclerosis. This study aimed to investigate the improved effects and mechanisms underlying a potential probiotic, Weizmannia coagulans JA845, on atherosclerosis. METHODS AND RESULTS Male Sprague-Dawley rats supported on a high-fat diet with vitamin D3 supplementation were subjected to W. coagulans JA845 treatment. W. coagulans JA845 obviously alleviated histological abnormalities of the abdominal aorta. After 6 weeks of W. coagulans JA845 administration, levels of TG, TC, LDL, ox-LDL, ROS, and MDA in the JA845 group decreased significantly, and those of HDL, GSH-Px, and SOD were markedly elevated. Treatment with W. coagulans JA845 also inhibited the secretion of ICAM-1 and VCAM-1 and regulated the plasma NO and eNOS content. In brief, administration of W. coagulans JA845 promoted the expression of the SIRT3/SOD2/FOXO3A pathway, inhibited the lipid metabolism pathway, SREBP-1c/FAS/DGAT2, and suppressed the JNK2/P38 MAPK/VEGF pathway implicated in endothelial injury. CONCLUSIONS These results indicated W. coagulans JA845 improved atherosclerosis by regulating lipid metabolism, antioxidative stress, and protecting against endothelial injury.
Collapse
Affiliation(s)
- Liying Ma
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, P.R. China
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences /National R&D Center for Milk Processing, Changchun 130033, P.R. China
| | - Zijian Zhao
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences /National R&D Center for Milk Processing, Changchun 130033, P.R. China
| | - Yujuan Zhao
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences /National R&D Center for Milk Processing, Changchun 130033, P.R. China
| | - Yansong Gao
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences /National R&D Center for Milk Processing, Changchun 130033, P.R. China
| | - Lei Zhao
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, P.R. China
| | - Shengyu Li
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences /National R&D Center for Milk Processing, Changchun 130033, P.R. China
| |
Collapse
|
212
|
Liu Z, Zhong L, Wang L, Li M, Chen C. Integrating network pharmacology and experimental studies for uncovering the molecular mechanisms of Dioscorea bulbifera L. in the treatment of thyroid cancer. Heliyon 2023; 9:e18886. [PMID: 37600377 PMCID: PMC10432970 DOI: 10.1016/j.heliyon.2023.e18886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Thyroid cancer (TC), a common endocrine malignant tumor with a higher incidence in females than in males, is a serious threat to human life and health. Although current clinical treatments can alleviate this disease, the recurrence rate remains high. Tuber Dioscoreae Bulbiferae, also called Huang-Yao-Zi (HYZ), has remarkable curative properties, few side effects and is used for the treatment of sore throat, goiter, hemoptysis, and other diseases in traditional Chinese medicine (TCM). Existing clinical studies have found that HYZ can improve the clinical symptoms of TC patients and reduce tumor volume, while in vitro studies have found that HYZ can induce the death of thyroid cancer cells. However, the mechanism of HYZ in the treatment of TC is still unclear. Methods In this study, based on network pharmacology and bioinformatics, the target and molecular mechanism of HYZ in the treatment of TC were preliminarily explored. The results suggest that the antithyroid cancer effect of HYZ may be related to the PI3K-Akt and focal adhesion pathways. Then, a TC cell model was established to further explore the detail molecular mechanisms. Results Fortunately, HYZ induced apoptosis in KMH-2 cells and regulated the expression of apoptosis-related proteins and genes. At the same time, HYZ can also significantly inhibit the migration and invasion of TC cells. Further studies showed that the pharmacological activities of HYZ were related to the regulation of the PI3K-Akt and focal adhesion pathways. Conclusion Our study provides a reference for further animal or clinical studies investigating the effectiveness and molecular mechanisms of HYZ against thyroid cancer.
Collapse
Affiliation(s)
- Ziqi Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Lian Zhong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Lingyu Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Meiyan Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Chao Chen
- Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Scienceand Technology of China, Chengdu 610072, P.R. China
| |
Collapse
|
213
|
Li Z, Li B, Chen Z, Xu J, El Sabbagh A, Zhao Y, Du R, Rong L, Tian J, Cui Q. Licochalcone A plays dual antiviral roles by inhibiting RSV and protecting against host damage. J Med Virol 2023; 95:e29059. [PMID: 37635463 DOI: 10.1002/jmv.29059] [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: 07/20/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023]
Abstract
Respiratory syncytial virus (RSV) causes lower respiratory tract diseases and bronchiolitis in children and elderly individuals. There are no effective drugs currently available to treat RSV infection. In this study, we report that Licochalcone A (LCA) can inhibit RSV replication and mitigate RSV-induced cell damage in vitro, and that LCA exerts a protective effect by reducing the viral titer and inflammation in the lungs of infected mice in vivo. We suggest that the mechanism of action occurs through pathways of antioxidant stress and inflammation. Further mechanistic results demonstrate that LCA can induce nuclear factor erythroid 2-related factor 2 (Nrf2) translocation into the nucleus, activate heme oxygenase 1 (HO-1), and inhibit reactive oxygen species-induced oxidative stress. LCA also works to reverse the decrease in I-kappa-B-alpha (IкBα) levels caused by RSV, which in turn inhibits inflammation through the associated nuclear factor kappa B and tumor necrosis factor-α signaling pathways. The combined action of the two cross-talking pathways protects hosts from RSV-induced damage. To conclude, our study is the first of its kind to establish evidence of LCA as a viable treatment for RSV infection.
Collapse
Affiliation(s)
- Zhongyuan Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Baohong Li
- Innovative Institute of Chinse Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zinuo Chen
- Innovative Institute of Chinse Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinke Xu
- Shandong Center for Disease Control and Prevention, Jinan, China
| | - Asma El Sabbagh
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Yangang Zhao
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - Ruikun Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Innovative Institute of Chinse Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Jingzhen Tian
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - Qinghua Cui
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Innovative Institute of Chinse Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
214
|
Arangia A, Ragno A, Cordaro M, D’Amico R, Siracusa R, Fusco R, Marino Merlo F, Smeriglio A, Impellizzeri D, Cuzzocrea S, Mandalari G, Di Paola R. Antioxidant Activity of a Sicilian Almond Skin Extract Using In Vitro and In Vivo Models. Int J Mol Sci 2023; 24:12115. [PMID: 37569490 PMCID: PMC10418603 DOI: 10.3390/ijms241512115] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Almond skins are known for their antioxidative and anti-inflammatory properties, which are mainly due to the presence of polyphenols. The aim of the present study was to evaluate the antioxidant and anti-inflammatory effects of almond skin extract (ASE) obtained from the Sicilian cultivar "Fascionello" and to evaluate the possible mechanisms of action using an in vitro model of human monocytic U937 cells as well as an in vivo model of carrageenan (CAR)-induced paw edema. The in vitro studies demonstrated that pretreatment with ASE inhibited the formation of ROS and apoptosis. The in vivo studies showed that ASE restored the CAR-induced tissue changes; restored the activity of endogenous antioxidant enzymes, such as superoxide dismutase, catalase, and glutathione; and decreased neutrophil infiltration, lipid peroxidation, and the release of proinflammatory mediators. The anti-inflammatory and antioxidant effects of ASE could be associated with the inhibition of the pro-inflammatory nuclear NF-κB and the activation of the nuclear factor-erythroid 2-related factor 2 (Nrf2) antioxidant pathways. In conclusion, almond skin could reduce the levels of inflammation and oxidative stress and could be beneficial in the treatment of several disorders.
Collapse
Affiliation(s)
- Alessia Arangia
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Agnese Ragno
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Marika Cordaro
- Department of Biomedical, Dental, Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Francesca Marino Merlo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.A.)
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
215
|
Maneesai P, Wattanathorn J, Potue P, Khamseekaew J, Rattanakanokchai S, Thukham-Mee W, Muchimapura S, Pakdeechote P. Cardiovascular complications are resolved by tuna protein hydrolysate supplementation in rats fed with a high-fat diet. Sci Rep 2023; 13:12280. [PMID: 37507421 PMCID: PMC10382531 DOI: 10.1038/s41598-023-39538-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/26/2023] [Indexed: 07/30/2023] Open
Abstract
This study is aimed to investigate whether tuna protein hydrolysate (TPH) supplementation could alleviate cardiovascular complications induced by a high-fat diet (HFD) in rats. Rats were fed a HFD for 16 weeks and given TPH (100 mg/kg, 300 mg/kg, or 500 mg/kg) or metformin (100 mg/kg) (n = 8) for the last four weeks. TPH had the following effects: resolved their impaired glucose tolerance, hyperglycemia, dyslipidemia, obesity, and hypertension (p < 0.05); alleviated left ventricular dysfunction and hypertrophy (p < 0.05), and vascular dysfunction and hypertrophy (p < 0.05); adipocyte hypertrophy; increases in circulating leptin and tumor necrosis factor (TNF-α) were mitigated (p < 0.05); increased renin-angiotensin system (RAS), oxidative stress, and decreased nitric oxide metabolites were modulated (p < 0.05). TPH restored the expression of angiotensin II receptor type 1 (AT1R)/NADPH oxidase 2 (NOX2), endothelial nitric oxide synthase (eNOS), nuclear factor erythroid 2-related factor (Nrf2)/heme oxygenase-1 (HO-1), and peroxisome proliferator-activated receptor γ (PPARγ)/the nuclear factor kappa B (NF-κB) protein in cardiovascular tissue (p < 0.05). In metabolic syndrome (MS) rats, metformin and TPH had comparable effects. In conclusion, TPH alleviated cardiovascular complications related to MS. It suppressed RAS, oxidative stress, and inflammation that were associated with modulation of AT1R/NOX2, eNOS, Nrf2/HO-1, and PPARγ/NF-κB expression.
Collapse
Affiliation(s)
- Putcharawipa Maneesai
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Jintanaporn Wattanathorn
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Prapassorn Potue
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Juthamas Khamseekaew
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | | | - Wipawee Thukham-Mee
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Supaporn Muchimapura
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Poungrat Pakdeechote
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
216
|
Gils C, Hansen DL, Nybo M, Frederiksen H. Elevated Hemolysis Index is associated with higher risk of cardiovascular diseases. Clin Chem Lab Med 2023; 61:1497-1505. [PMID: 36814140 DOI: 10.1515/cclm-2023-0114] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVES In vivo hemolysis is associated with thromboembolism. Although an increased Hemolysis Index (HI) can be due to in vitro as well as in vivo hemolysis, both reflects a more fragile erythrocyte population. We therefore hypothesized that HI above upper reference limit would be associated with an increased risk of cardiovascular disease (CVD). METHODS We identified persons with two elevated HI (HI+) from blood samples analyzed at a university hospital laboratory from 2012 to 2017. We compared their risk of CVD with the risk in matched comparators with normal HI and from the general population. HI+ persons and comparators were followed from start date (date of the second elevated HI) until the first of the main outcome: CVD, emigration, death, or end of observation time on December 31, 2018. RESULTS In 43,102 unique HI+ persons, the risk of developing CVD was 40% higher compared with the general population and 13% higher compared with the matched blood sample cohort. HI+ was associated with a significantly increased cumulative incidence of both arterial and venous CVD compared with the matched blood sample cohort and the general population (respectively 47 and 14% for arterial CVD; 78 and 24% for venous CVD). Moreover, overall mortality risk was significantly higher in patients with HI+ than in the two comparator groups. CONCLUSIONS Elevated HI is associated with increased risk of arterial and venous CVD and with increased mortality. Our findings imply that HI may contribute as a CVD risk biomarker.
Collapse
Affiliation(s)
- Charlotte Gils
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Dennis Lund Hansen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Mads Nybo
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Henrik Frederiksen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Hematology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
217
|
Huang X, Li Z, Zhang L, Yang Y, Wang Y, Li S, Li G, Feng H, Yang X. miR-205-5p inhibits homocysteine-induced pulmonary microvascular endothelium dysfunction by targeting FOXO1. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1456-1466. [PMID: 37491880 PMCID: PMC10520487 DOI: 10.3724/abbs.2023127] [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: 01/12/2023] [Accepted: 03/31/2023] [Indexed: 07/27/2023] Open
Abstract
Homocysteine (Hcy) is a risk factor for multiple chronic diseases, and vascular endothelial cell injury has been regarded as the initiating step for this process. miRNAs are involved in Hcy-induced endothelial dysfunction, while the underlying mechanism and roles of miRNAs in pulmonary endothelial dysfunction induced by homocysteine are unknown. Here, we find that miR-205-5p alleviates pulmonary endothelial dysfunction by targeting FOXO1 in CBS +/‒ mice to protect against Hcy-induced pulmonary endothelial dysfunction. Mechanistically, we show that Hcy can lead to DNA hypermethylation of the miR-205-5p promoter due to the increased binding of DNMT1 to its promoter, which contributes to reduction of miR-205-5p expression. In summary, miR-205-5p promoter hypermethylation causes downregulation of miR-205-5p expression, resulting in a reduction in miR-205-5p binding to FOXO1 during homocysteine-induced pulmonary endothelial dysfunction. Our data indicate that miR-205-5p may be a potential therapeutic target against Hcy-induced pulmonary injury.
Collapse
Affiliation(s)
- Xiaobo Huang
- Department of Respiratory and Critical Care MedicineSecond Affiliated Hospital of Ningxia Medical University (The First People′s Hospital of Yinchuan)Yinchuan750001China
| | - Zhen Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| | - Ling Zhang
- Department of PathologyPeople’s Hospital of Ningxia Hui Autonomous RegionYinchuan750004China
| | - Yali Yang
- Department of PathologyGeneral Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Yanjia Wang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
| | - Sirui Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
| | - Guizhong Li
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
| | | | - Xiaoling Yang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases ResearchNingxia Medical UniversityYinchuan750004China
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
| |
Collapse
|
218
|
Zeng J, Liao S, Liang Z, Li C, Luo Y, Wang K, Zhang D, Lan L, Hu S, Li W, Lin R, Jie Z, Hu Y, Dai S, Zhang Z. Schisandrin A regulates the Nrf2 signaling pathway and inhibits NLRP3 inflammasome activation to interfere with pyroptosis in a mouse model of COPD. Eur J Med Res 2023; 28:217. [PMID: 37400851 PMCID: PMC10316617 DOI: 10.1186/s40001-023-01190-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/23/2023] [Indexed: 07/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a serious chronic lung disease. Schisandrin A (SchA) is one of the most important active ingredients in Schisandra chinensis and has been used to treat various lung diseases in several countries. Here, we studied the pharmacological effect of SchA on airway inflammation induced by cigarette smoke (CS) and explored the therapeutic mechanism of SchA in COPD model mice. Our results showed that SchA treatment significantly improved the lung function of CS-induced COPD model mice and reduced the recruitment of leukocytes and hypersecretion of interleukin-6 (IL-6), interleukin-1β (IL-1β) and tumor necrosis factor α (TNF-α) in bronchoalveolar lavage fluid (BALF). H&E staining showed that SchA treatment could effectively reduce emphysema, immune cell infiltration and airway wall destruction. In addition, we found that SchA treatment can stimulate the expression of heme oxygenase-1 (HO-1) through the nuclear factor-erythroid 2-related factor (Nrf2) pathway, significantly reduce oxidative stress, increase catalase (CAT) and superoxide dismutase (SOD) levels, and suppress the level of malondialdehyde (MDA) in COPD model mice. Moreover, SchA treatment suppressed the generation of the NLRP3/ASC/Caspase1 inflammasome complex to inhibit the inflammatory response caused by IL-1β and IL-18 and pyroptosis caused by GSDMD. In conclusion, our study shows that SchA treatment can inhibit the production of ROS and the activation of the NLRP3 inflammasome by upregulating Nrf-2, thereby producing anti-inflammatory effects and reducing lung injury in COPD model mice. More importantly, SchA exhibited similar anti-inflammatory effects to dexamethasone in COPD model mice, and we did not observe substantial side effects of SchA treatment. The high safety of SchA makes it a potential candidate drug for the treatment of COPD.
Collapse
Affiliation(s)
- Jiamin Zeng
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Sida Liao
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhu Liang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Caiping Li
- Guangzhou Medical University, Guangzhou, China
| | - Yuewen Luo
- Guangzhou Medical University, Guangzhou, China
| | - Kexin Wang
- Guangzhou Medical University, Guangzhou, China
| | - Dapeng Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lan Lan
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Suzhen Hu
- Huangdao District Chinese Medicine Hospital, Qingdao, China
| | - Wanyan Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ran Lin
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zichen Jie
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanlong Hu
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Shiting Dai
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Zhimin Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
219
|
Ahmad A, Orassay A, Majaz S, Saeed A, Sadvokassova D, Berdigaliyev A, Ahmad S, Wang LX, Xie Y. Computational analysis of target genes in monkeypox virus infection and potential therapeutic precursors. Expert Rev Anti Infect Ther 2023; 21:1153-1161. [PMID: 37711024 DOI: 10.1080/14787210.2023.2259614] [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: 01/09/2023] [Revised: 07/23/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Monkeypox is an orthopoxvirus that is responsible for zoonotic infections in humans. The virus has recently spread rapidly and the WHO has listed it as an international public health emergency of concern. RESEARCH DESIGN AND METHODS Here, we used network analysis and gene enrichment protocols and analyzed datasets of MPXV infection that induced host cell gene expression list and subsequently mapped them against two herbal target gene lists which highlighted considerable coherence in pharmacological attributes with COVID-19. Molecular docking and simulation were performed for the screened compounds. RESULTS Our results identified β-carotene and kaempferol possessing tremendous ability against the MPXV PLD protein. Both compounds were subjected to each of 100 ns molecular dynamics simulation and were found native to the PLD pocket. MM-PB (GB) SA analyses indicated -25.4, -40.1 kcal/mol and -17.2, -26.4kcal/mol of ΔGbind to the active pocket of PLD. Our data suggest the adaptive nature of the MPXV PLD active pocket toward hydrophobic inhibitors. CONCLUSION These results will be of high importance for the viral researchers to be tested in wet lab settings in designing potential inhibitors.
Collapse
Affiliation(s)
- Ashfaq Ahmad
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Aliya Orassay
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
| | - Sidra Majaz
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Aamir Saeed
- Department of Bioinformatics, Hazara University, Mansehra, Pakistan
| | - Darya Sadvokassova
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
| | - Alan Berdigaliyev
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
| | - Salar Ahmad
- Department of Surgery, Tehsil Head Quarter (THQ) Hospital, Dargai Malakand, Pakistan
| | - Lian-Xiang Wang
- Department of Crops Research, Heze Research Institute, Heze, China
| | - Yingqiu Xie
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
| |
Collapse
|
220
|
Li W, Xing Q, Liu Z, Liu R, Hu Y, Yan Q, Liu X, Zhang J. The signaling pathways of traditional Chinese medicine in treating diabetic retinopathy. Front Pharmacol 2023; 14:1165649. [PMID: 37405050 PMCID: PMC10315578 DOI: 10.3389/fphar.2023.1165649] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/06/2023] [Indexed: 07/06/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the common diabetic microvascular complications that occurs in the eyes and is closely associated with vision loss in working adults. However, the clinical treatment of DR is limited or accompanied by a large number of complications. Therefore, the development of new drugs for the treatment of DR is urgently needed. Traditional Chinese medicine (TCM) is widely used to treat DR in China, and its multi-pathway and multi-level characteristics can effectively address the complex pathogenesis of DR. Growing evidence suggests that inflammation, angiogenesis, and oxidative stress are the core pathological mechanisms in the development of DR. This study innovatively considers the aforementioned processes as the fundamental unit and sheds light on the molecular mechanisms and potential of TCM against DR in terms of signaling pathways. The results showed that NF-κB, MAPK/NF-κB, TLR4/NF-κB, VEGF/VEGFR2, HIF-1α/VEGF, STAT3, and Nrf2/HO-1 are the key signaling pathways for the treatment of DR by TCMs, which involved curcumolide, erianin, quercetin, blueberry anthocyanins, puerarin, arjunolic acid, ethanol extract of Scutellaria barbata D. Don, Celosia argentea L. extract, ethanol extract of Dendrobium chrysotoxum Lindl., Shengpuhuang-tang, and LuoTong formula. The purpose of this review is to update and summarize the signaling pathways of TCM in the treatment of DR and provide ideas for the development of new drugs against DR in the future.
Collapse
Affiliation(s)
- Wencan Li
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Qichang Xing
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Zheng Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Renzhu Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Yixiang Hu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Qingzi Yan
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Jiani Zhang
- Department of Pharmacy, Xiangtan Central Hospital, Xiangtan, Hunan, China
| |
Collapse
|
221
|
Markina YV, Kirichenko TV, Tolstik TV, Bogatyreva AI, Zotova US, Cherednichenko VR, Postnov AY, Markin AM. Target and Cell Therapy for Atherosclerosis and CVD. Int J Mol Sci 2023; 24:10308. [PMID: 37373454 DOI: 10.3390/ijms241210308] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Cardiovascular diseases (CVD) and, in particular, atherosclerosis, remain the main cause of death in the world today. Unfortunately, in most cases, CVD therapy begins after the onset of clinical symptoms and is aimed at eliminating them. In this regard, early pathogenetic therapy for CVD remains an urgent problem in modern science and healthcare. Cell therapy, aimed at eliminating tissue damage underlying the pathogenesis of some pathologies, including CVD, by replacing it with various cells, is of the greatest interest. Currently, cell therapy is the most actively developed and potentially the most effective treatment strategy for CVD associated with atherosclerosis. However, this type of therapy has some limitations. In this review, we have tried to summarize the main targets of cell therapy for CVD and atherosclerosis in particular based on the analysis using the PubMed and Scopus databases up to May 2023.
Collapse
Affiliation(s)
- Yuliya V Markina
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Taisiya V Tolstik
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Ulyana S Zotova
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Anton Yu Postnov
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Alexander M Markin
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
- Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow 117198, Russia
| |
Collapse
|
222
|
Yalameha B, Reza Nejabati H. Urinary Exosomal Metabolites: Overlooked Clue for Predicting Cardiovascular Risk. Clin Chim Acta 2023:117445. [PMID: 37315726 DOI: 10.1016/j.cca.2023.117445] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 06/16/2023]
Abstract
Over the last decade, increasing research has focused on urinary exosomes (UEs) in biological fluids and their relationship with physiological and pathological processes. UEs are membranous vesicles with a size of 40-100 nm, containing a number of bioactive molecules such as proteins, lipids, mRNAs, and miRNAs. These vesicles are an inexpensive non-invasive source that can be used in clinical settings to differentiate healthy patients from diseased patients, thereby serving as potential biomarkers for the early identification of disease. Recent studies have reported the isolation of small molecules called exosomal metabolites from individuals' urine with different diseases. These metabolites could utilize for a variety of purposes, such as the discovery of biomarkers, investigation of mechanisms related to disease development, and importantly prediction of cardiovascular diseases (CVDs) risk factors, including thrombosis, inflammation, oxidative stress, hyperlipidemia as well as homocysteine. It has been indicated that alteration in urinary metabolites of N1-methylnicotinamide, 4-aminohippuric acid, and citric acid can be valuable in predicting cardiovascular risk factors, providing a novel approach to evaluating the pathological status of CVDs. Since the UEs metabolome has been clearly and precisely so far unexplored in CVDs, the present study has specifically addressed the role of the mentioned metabolites in the prediction of CVDs risk factors.
Collapse
Affiliation(s)
- Banafsheh Yalameha
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
223
|
Liang Q, Guo R, Tsao JR, He Y, Wang C, Jiang J, Zhang D, Chen T, Yue T, Hu K. Salidroside alleviates oxidative stress in dry eye disease by activating autophagy through AMPK-Sirt1 pathway. Int Immunopharmacol 2023; 121:110397. [PMID: 37302369 DOI: 10.1016/j.intimp.2023.110397] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/14/2023] [Accepted: 05/24/2023] [Indexed: 06/13/2023]
Abstract
Dry eye disease (DED) is a multifactorial disease, and oxidative stress plays a crucial role in its pathogenesis. Recently, multiple studies have shown that upregulation of autophagy can protect the cornea from oxidative stress damage. The present study investigated the therapeutic effects of salidroside, the main component of Rhodiola crenulata, in both in vivo and in vitro dry eye models. The results showed that topical eye drop treatment with salidroside restored corneal epithelium damage, increased tear secretion, and reduced cornea inflammation in the DED mice. Salidroside activated autophagy through AMP-activated protein kinase (AMPK)-sirtuin-1 (Sirt1) signaling pathway, which promoted the nuclear translocation of nuclear factor erythroid-2-related factor 2 (Nrf2) and increased the expression of downstream antioxidant factors heme oxygenase-1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1). This process restored antioxidant enzyme activity, reduced reactive oxygen species (ROS) accumulation, and alleviated oxidative stress. The application of autophagy inhibitor chloroquine and AMPK inhibitor Compound C reversed the therapeutic efficacy of salidroside, validating the above findings. In conclusion, our data suggest that salidroside is a promising candidate for DED treatment.
Collapse
Affiliation(s)
- Qi Liang
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Rongjie Guo
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Jia-Ruei Tsao
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Yun He
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Chenchen Wang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 618 Fengqi East Rd, Hangzhou, Zhejiang, China
| | - Jiaxuan Jiang
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Di Zhang
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Taige Chen
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China; Department of Rheumatology and Immunology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Tingting Yue
- Department of Neurosurgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China.
| | - Kai Hu
- Department of Ophthalmology, Affiliated Drum Tower Hospital, Medical School, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China.
| |
Collapse
|
224
|
Qiu L, Chen K, Wang X, Zhao Y. RETRACTED: Tangzhiqing-mediated NRF2 reduces autophagy-dependent ferroptosis to mitigate diabetes-related cognitive impairment neuronal damage. Rejuvenation Res 2023. [PMID: 37279293 DOI: 10.1089/rej.2023.0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
The publisher of Rejuvenation Research officially retracts the article entitled, "Tangzhiqing-mediated NRF2 reduces autophagy-dependent ferroptosis to mitigate diabetes-related cognitive impairment neuronal damage," by Lingyan Qiu, Mr. Kai Chen, Prof. Xu Wang, and Ms. Yun Zhao. (Rejuvenation Res 2023; epub 6 Jun; doi: 10.1089/rej.2023.0013). After the acceptance and Instant Online publication of the paper, the authors were contacted repeatedly regarding their page proofs, and for further clarification of unresolved issues within the paper. All attempts to reach the authors were unsuccessful. Concurrently, the publisher identified a problematic overlap with a paper published in 2023 in Endocrine, Metabolic & Immune Disorders - Drug Targets.1 This paper was subsequently withdrawn. These troubling details have led the editorial leadership of Rejuvenation Research to lose confidence in the validity of the submission and to retract the paper. All authors were notified of the decision to retract the paper via email. The lead author, Lingyan Qiu, and the corresponding author, Xu Wang, quickly responded and appealed the decision to retract. The appeal was denied. Reference 1. https://www.eurekaselect.com/article/132631. Withdrawn: Experimental study on NRF2 mediated by Chinese medicine tangzhiqing to reduce autophagy-dependent ferroptosis and alleviate neuron damage in HT22 mice with diabetes-related cognitive disorder. 22 June, 2023; DOI: 10.2174/1871530323666230622151649 Diabetes is a chronic condition defined by the body's inability to process glucose. The most common form, diabetes mellitus, reflects the body's insulin resistance, which leads to long-term raised glucose blood levels. These levels can cause oxidative damage, cell stress, and excessive autophagy throughout the body, including the nervous system. Diabetes-related cognitive impairment (DCI) results from chronic elevation of blood glucose, and as diabetes cases continue to rise, so too do comorbidities such as DCI. Although there are medications to address high blood glucose, there are few that can inhibit excessive autophagy and cell death. Therefore, we investigated if the Traditional Chinese Medicine, Tangzhiqing (TZQ), can reduce the impact of DCI in a high-glucose cell model. We used commercially available kits to evaluate cell viability, mitochondrial activity, and oxidative stress. We found that TZQ treatment increased cell viability, ensured continued mitochondrial activity, and reduced reactive oxygen species. We also found that TZQ functions by increasing NRF2 activity, which decreases the ferroptotic-associated pathways that involve p62, HO-1, and GPX4. Therefore, TZQ should be further investigated for its role in reducing DCI.
Collapse
Affiliation(s)
- Lingyan Qiu
- Nanjing University of Chinese Medicine, 66478, 138 Xianlin Avenue, Qixia District, Nanjing, Jiangsu, China, Nanjing, China, 210046;
| | - Kai Chen
- Nanjing University of Traditional Chinese Medicine, 66478, Nanjing, Jiangsu, China;
| | - Xu Wang
- Nanjing University of Traditional Chinese Medicine, 66478, Nanjing, Jiangsu, China;
| | - Yun Zhao
- Nanjing University of Traditional Chinese Medicine, 66478, Nanjing, Jiangsu, China;
| |
Collapse
|
225
|
Gul S, Attaullah S, Alsugoor MH, Bawazeer S, Shah SA, Khan S, Salahuddin HS, Ullah M. Folicitin abrogates scopolamine induced oxidative stress, hyperlipidemia mediated neuronal synapse and memory dysfunction in mice. Heliyon 2023; 9:e16930. [PMID: 37416682 PMCID: PMC10320035 DOI: 10.1016/j.heliyon.2023.e16930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 07/08/2023] Open
Abstract
No effective drug treatment is available for Alzheimer disease, thus the need arise to develop efficient drugs for its treatment. Natural products have pronounced capability in treating Alzheimer disease therefore current study aimed to evaluate the neuro-protective capability of folicitin against scopolamine-induced Alzheimer disease neuropathology in mice. Experimental mice were divided into four groups i.e. control (single dose of 250 μL saline), scopolamine-administered group (1 mg/kg administered for three weeks), scopolamine plus folicitin-administered group (scopolamine 1 mg/kg administration for three weeks followed by folicitin administration for last two weeks) and folicitin-administered group (20 mg/kg administered for 5 alternate days). Results of behavioral tests and Western blot indicated that folicitin has the capability of recovering the memory against scopolamine-induced memory impairment by reducing the oxidative stress through up-regulating the endogenous antioxidant system like nuclear factor erythroid 2-related factor and Heme oxygenase-1 while prohibiting phosphorylated c-Jun N-terminal kinase. Similarly, folicitin also improved the synaptic dysfunction by up-regulating SYP and PSD95. Scopolamine-induced hyperglycemia and hyperlipidemia were abolished by folicitin as evidenced through random blood glucose test, glucose tolerance test and lipid profile test. All these results revealed that folicitin being a potent anti-oxidant is capable of improving synaptic dysfunction and reducing oxidative stress through Nrf-2/HO-1 pathway, thus plays a key role in treating Alzheimer disease as well as possess hyperglycemic and hyperlipidemic effect. Furthermore, a detailed study is suggested.
Collapse
Affiliation(s)
- Seema Gul
- Department of Zoology, Islamia College Peshawar, 25120, Khyber Pakhtunkhwa, Pakistan
| | - Sobia Attaullah
- Department of Zoology, Islamia College Peshawar, 25120, Khyber Pakhtunkhwa, Pakistan
| | - Mahdi H. Alsugoor
- Umme Al-Qura University, Faculty of Pharmacy, Department of Pharmacognosy, Makkah, Saudi Arabia
| | - Sami Bawazeer
- Umme Al-Qura University, Faculty of Pharmacy, Department of Pharmacognosy, Makkah, Saudi Arabia
| | - Shahid Ali Shah
- Neuro Molecular Medicine Research Centre (NMMRC), Ring Road, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Sanaullah Khan
- Department of Zoology, University of Peshawar, Peshawar, 25120, Khyber Pakhtunkhwa, Pakistan
| | | | - Mujeeb Ullah
- Department of Zoology, Islamia College Peshawar, 25120, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
226
|
Gou T, Hu M, Xu M, Chen Y, Chen R, Zhou T, Liu J, Guo L, Ao H, Ye Q. Novel wine in an old bottle: Preventive and therapeutic potentials of andrographolide in atherosclerotic cardiovascular diseases. J Pharm Anal 2023; 13:563-589. [PMID: 37440909 PMCID: PMC10334359 DOI: 10.1016/j.jpha.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 07/15/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) frequently results in sudden death and poses a serious threat to public health worldwide. The drugs approved for the prevention and treatment of ASCVD are usually used in combination but are inefficient owing to their side effects and single therapeutic targets. Therefore, the use of natural products in developing drugs for the prevention and treatment of ASCVD has received great scholarly attention. Andrographolide (AG) is a diterpenoid lactone compound extracted from Andrographis paniculata. In addition to its use in conditions such as sore throat, AG can be used to prevent and treat ASCVD. It is different from drugs that are commonly used in the prevention and treatment of ASCVD and can not only treat obesity, diabetes, hyperlipidaemia and ASCVD but also inhibit the pathological process of atherosclerosis (AS) including lipid accumulation, inflammation, oxidative stress and cellular abnormalities by regulating various targets and pathways. However, the pharmacological mechanisms of AG underlying the prevention and treatment of ASCVD have not been corroborated, which may hinder its clinical development and application. Therefore, this review summarizes the physiological and pathological mechanisms underlying the development of ASCVD and the in vivo and in vitro pharmacological effects of AG on the relative risk factors of AS and ASCVD. The findings support the use of the old pharmacological compound ('old bottle') as a novel drug ('novel wine') for the prevention and treatment of ASCVD. Additionally, this review summarizes studies on the availability as well as pharmaceutical and pharmacokinetic properties of AG, aiming to provide more information regarding the clinical application and further research and development of AG.
Collapse
Affiliation(s)
- Tingting Gou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Minghao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Min Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuchen Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Rong Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Junjing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiang Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
227
|
Zhang Y, Wang P, Jin MX, Zhou YQ, Ye L, Zhu XJ, Li HF, Zhou M, Li Y, Li S, Liang KY, Wang Y, Gao Y, Pan MX, Zhou SQ, Peng Q. Schisandrin B Improves the Hypothermic Preservation of Celsior Solution in Human Umbilical Cord Mesenchymal Stem Cells. Tissue Eng Regen Med 2023; 20:447-459. [PMID: 36947320 PMCID: PMC10219924 DOI: 10.1007/s13770-023-00531-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Human umbilical cord mesenchymal stem cells (hUCMSCs) have emerged as promising therapy for immune and inflammatory diseases. However, how to maintain the activity and unique properties during cold storage and transportation is one of the key factors affecting the therapeutic efficiency of hUCMSCs. Schisandrin B (SchB) has many functions in cell protection as a natural medicine. In this study, we investigated the protective effects of SchB on the hypothermic preservation of hUCMSCs. METHODS hUCMSCs were isolated from Wharton's jelly. Subsequently, hUCMSCs were exposed to cold storage (4 °C) and 24-h re-warming. After that, cells viability, surface markers, immunomodulatory effects, reactive oxygen species (ROS), mitochondrial integrity, apoptosis-related and antioxidant proteins expression level were evaluated. RESULTS SchB significantly alleviated the cells injury and maintained unique properties such as differentiation potential, level of surface markers and immunomodulatory effects of hUCMSCs. The protective effects of SchB on hUCMSCs after hypothermic storage seemed associated with its inhibition of apoptosis and the anti-oxidative stress effect mediated by nuclear factor erythroid 2-related factor 2 signaling. CONCLUSION These results demonstrate SchB could be used as an agent for hypothermic preservation of hUCMSCs.
Collapse
Affiliation(s)
- Ying Zhang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Peng Wang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Mei-Xian Jin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Ying-Qi Zhou
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Liang Ye
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Xiao-Juan Zhu
- Department of Anesthesiology, First People's Hospital of Kashi, Kashi, 844000, China
| | - Hui-Fang Li
- Department of Anesthesiology, First People's Hospital of Kashi, Kashi, 844000, China
| | - Ming Zhou
- Department of Anesthesiology, First People's Hospital of Kashi, Kashi, 844000, China
| | - Yang Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Shao Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Kang-Yan Liang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Yi Wang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Ming-Xin Pan
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Shu-Qin Zhou
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China.
| | - Qing Peng
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
228
|
Li C, Liu M, Deng L, Luo D, Ma R, Lu Q. Oxyberberine ameliorates TNBS-induced colitis in rats through suppressing inflammation and oxidative stress via Keap1/Nrf2/NF-κB signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154899. [PMID: 37247589 DOI: 10.1016/j.phymed.2023.154899] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic, unspecific inflammatory bowel disorder lacking effective therapeutic targets and radical drugs. Oxyberberine (OBB), a novel intestinal flora-elicited oxidative metabolite of berberine (BBR), has been revealed to exhibit diverse pharmacological properties. PURPOSE In this follow-up study, we attempted to shed light on the possible therapeutic effect and latent mechanism of OBB on 2, 4, 6-trinitrobenzenesulfonic acid (TNBS)-evoked UC in rats. METHODS UC rats were established via a gentle enema of TNBS. Rats were sacrificed after intragastric administration of drugs for seven days. The weight reduction, disease activity index, macroscopic and histological colonic alterations were assessed. Further investigation on molecular mechanisms was conducted by ELISA, qRT-PCR, immunohistochemistry, or Western blot. RESULTS OBB treatment remarkably decreased the weight loss, macroscopic scores, and colonal weight/length ratio, as well as mitigated the colonic pathological deterioration and MPO vitality in colitis rats, achieving a superior protective effect to BBR. Additionally, OBB modulated the disequilibrium between pro- and anti-inflammatory factors by promoting the production of IL-13 and IL-4, and lowering the contents of TNF-α, IL-2, IL-8, and IL-22. Furthermore, OBB pretreatment dramatically ameliorated oxidative stress via enhancing antioxidant defense genes expressions (including HO-1, GCLM, GCLC, and NQO-1), thereby increasing SOD and GSH, and decreasing MDA and ROS activities. Furthermore, OBB strikingly restrained the translocation of NF-κB p65 and phosphorylation of IκBα, promoted HO-1 expression, Keap1 degradation and Nrf2 nuclear translocation. CONCLUSION The study firstly indicated that OBB had a superior therapeutic effect than BBR against TNBS-elicited colitis in rats. The protective effect of OBB might be closely related to the modulation of Keap1/Nrf2/NF-κB-mediated inflammatory response and oxidant stress. The evidences highlight the potentiality of OBB as a prospective candidate for the amelioration of colitis.
Collapse
Affiliation(s)
- Cailan Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China; Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563000, China.
| | - Meigui Liu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, PR China
| | - Li Deng
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, China
| | - Dandan Luo
- Department of Pharmacy, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Runfang Ma
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, PR China
| | - Qiang Lu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, PR China.
| |
Collapse
|
229
|
Liu Y, Zhou F, Shu HZ, Lin LG, Zhang QW. Germacrane-type sesquiterpenoids from the flowers of Chrysanthemum indicum with hepatoprotective activity. Food Chem Toxicol 2023; 177:113850. [PMID: 37225032 DOI: 10.1016/j.fct.2023.113850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/06/2023] [Accepted: 05/20/2023] [Indexed: 05/26/2023]
Abstract
Two new germacrane-type sesquiterpenoids, chrysanthemolides A (1) and B (2), and four known germacrane-type sesquiterpenoids, hanphyllin (3), 3β-hydroxy-11α,13-dihydro-costunolide (4), costunolide (5), and 6,7-dimethylmethylene-4-aldehyde-1β-hydroxy-10(15)-ene-(4Z)-dicyclodecylene (6), were isolated and identified from the flowers of Chrysanthemum indicum. The structures of the new compounds were elucidated via high resolution electrospray ionization mass spectrometry (HR-ESI-MS), 1D and 2D nuclear magnetic resonance (NMR) spectra and electronic circular dichroism (ECD). Meanwhile, all the isolates were tested for their hepatoprotective activity in tert-butyl hydroperoxide (t-BHP) injured AML12 cells. Compounds 1, 2, and 4 showed significant protective effects at 40 μM, comparable with the positive control resveratrol at 10 μM. As the most potent one, compound 1 was chosen for further studies. Compound 1 dose-dependently increased the viability of t-BHP-injured AML12 cells. Furthermore, compound 1 decreased reactive oxygen species accumulation, while increased glutathione level, heme oxygenase-1 level and superoxide dismutase activity, through anchoring in the binding site of Kelch domain of the Kelch-like ECH-associated protein 1 (Keap1) to promote the dissociation of nuclear factor erythroid 2-related factor 2 from Keap1 and translocation to nuclei. In summary, germacrane-type sesquiterpenoids from C. indicum might be further developed to protect liver against oxidative damage.
Collapse
Affiliation(s)
- Yu Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, 999078, China
| | - Fei Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, 999078, China
| | - Hong-Zhen Shu
- Department of Pharmacy, Sichuan Vocational College of Health and Rehabilitation, Zigong, 643000, China
| | - Li-Gen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, 999078, China; Department of Pharmaceutical Sciences and Technology, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao, 999078, China.
| | - Qing-Wen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, 999078, China; Department of Pharmaceutical Sciences and Technology, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao, 999078, China.
| |
Collapse
|
230
|
Tang BL, Liu Y, Zhang JL, Lu ML, Wang HX. Ginsenoside Rg1 ameliorates hypoxia-induced pulmonary arterial hypertension by inhibiting endothelial-to-mesenchymal transition and inflammation by regulating CCN1. Biomed Pharmacother 2023; 164:114920. [PMID: 37216706 DOI: 10.1016/j.biopha.2023.114920] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 05/13/2023] [Indexed: 05/24/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic obstructive disease characterized by vascular remodeling. Studies have confirmed that ginsenoside Rg1 can improve pulmonary hypertension to a certain extent, but the potential mechanism by which it improves hypoxia-induced PAH remains unclear. The aim of this study was to investigate the therapeutic effect of ginsenoside Rg1 on hypoxia-induced PAH. The results showed that hypoxia promoted inflammation, EndMT, and vascular remodeling, which were accompanied by decreased CCN1 levels and increased p-NFκB p65, TGF-β1, and p-Smad 2/3 levels. Treatment with ginsenoside Rg1, recombinant CCN1, BAY-11-7082, and SB-431542 could prevent hypoxia-induced vascular remodeling, reduce the expression of the hypoxia-induced inflammatory cytokines TNF-α and IL-1β, inhibit the expression of the mesenchymal markers α-SMA and Vimentin and restore the expression of the endothelial markers CD31 and VE-cadherin to improve hypoxia-induced EndMT, which may be associated with the upregulation of CCN1 protein expression and downregulation of p-NFκB p65, TGF-β1, and p-Smad 2/3 in rats and cells. siRNA CCN1 transfection increased the expression of p-NFκB p65, TGF-β1, and p-Smad 2/3 and accelerated the occurrence and development of inflammation and EndMT after hypoxia. In summary, our study indicated that hypoxia-induced EndMT and inflammation play a role in hypoxic pulmonary hypertension (HPH). Ginsenoside Rg1 treatment could reverse hypoxia-induced EndMT and inflammation by regulating CCN1 and has potential value in the prevention and treatment of HPH.
Collapse
Affiliation(s)
- Bai-Lin Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, China
| | - Yu Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, China
| | - Jing-Liang Zhang
- Internal Medicine-Cardiovascular Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Mei-Li Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, China
| | - Hong-Xin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
231
|
Zhou K, Liu D, Jin Y, Xia W, Zhang P, Zhou Z. Oxymatrine ameliorates osteoarthritis via the Nrf2/NF-κB axis in vitro and in vivo. Chem Biol Interact 2023; 380:110539. [PMID: 37196756 DOI: 10.1016/j.cbi.2023.110539] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/25/2023] [Accepted: 05/09/2023] [Indexed: 05/19/2023]
Abstract
PURPOSE Osteoarthritis (OA) is a common degenerative joint disorder. Currently, the underlying etiology of OA is still far from fully elucidated and there is no cure for OA progression. Previous studies have demonstrated that oxymatrine (OMT) could inhibit inflammation and oxidative stress in several animal models. However, the potential effects of OMT on OA remain largely elusive. The aim of the study is to investigate the anti-inflammatory and chondrocyte protective effect of OMT, and delineate the potential mechanism in vitro and in vivo. METHODS Western blotting, RT-qPCR, ELISA and tissue staining were employed to explore the mechanisms by which OMT exerted a protective effect on IL-1β-induced production of pro-inflammation cytokines and extracellular matrix (ECM) degradation in primary murine chondrocytes and DMM mouse models. RESULTS The results showed that OMT reduced the IL-1β-induced over-production of pro-inflammation cytokines and ECM degradation. Mechanistically, OMT inhibited the NF-κB pathway via activating Nrf2. In vivo studies also demonstrated that OMT ameliorated OA progression. CONCLUSIONS OMT reduced pro-inflammation cytokines, ECM degradation and OA progression via activating Nrf2 and inhibiting NF-κB pathway.
Collapse
Affiliation(s)
- Kailong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dong Liu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yesheng Jin
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Wei Xia
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Peng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Zhiqiang Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
232
|
Luo M, Zheng Y, Tang S, Gu L, Zhu Y, Ying R, Liu Y, Ma J, Guo R, Gao P, Zhang C. Radical oxygen species: an important breakthrough point for botanical drugs to regulate oxidative stress and treat the disorder of glycolipid metabolism. Front Pharmacol 2023; 14:1166178. [PMID: 37251336 PMCID: PMC10213330 DOI: 10.3389/fphar.2023.1166178] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
Background: The incidence of glycolipid metabolic diseases is extremely high worldwide, which greatly hinders people's life expectancy and patients' quality of life. Oxidative stress (OS) aggravates the development of diseases in glycolipid metabolism. Radical oxygen species (ROS) is a key factor in the signal transduction of OS, which can regulate cell apoptosis and contribute to inflammation. Currently, chemotherapies are the main method to treat disorders of glycolipid metabolism, but this can lead to drug resistance and damage to normal organs. Botanical drugs are an important source of new drugs. They are widely found in nature with availability, high practicality, and low cost. There is increasing evidence that herbal medicine has definite therapeutic effects on glycolipid metabolic diseases. Objective: This study aims to provide a valuable method for the treatment of glycolipid metabolic diseases with botanical drugs from the perspective of ROS regulation by botanical drugs and to further promote the development of effective drugs for the clinical treatment of glycolipid metabolic diseases. Methods: Using herb*, plant medicine, Chinese herbal medicine, phytochemicals, natural medicine, phytomedicine, plant extract, botanical drug, ROS, oxygen free radicals, oxygen radical, oxidizing agent, glucose and lipid metabolism, saccharometabolism, glycometabolism, lipid metabolism, blood glucose, lipoprotein, triglyceride, fatty liver, atherosclerosis, obesity, diabetes, dysglycemia, NAFLD, and DM as keywords or subject terms, relevant literature was retrieved from Web of Science and PubMed databases from 2013 to 2022 and was summarized. Results: Botanical drugs can regulate ROS by regulating mitochondrial function, endoplasmic reticulum, phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT), erythroid 2-related factor 2 (Nrf-2), nuclear factor κB (NF-κB), and other signaling pathways to improve OS and treat glucolipid metabolic diseases. Conclusion: The regulation of ROS by botanical drugs is multi-mechanism and multifaceted. Both cell studies and animal experiments have demonstrated the effectiveness of botanical drugs in the treatment of glycolipid metabolic diseases by regulating ROS. However, studies on safety need to be further improved, and more studies are needed to support the clinical application of botanical drugs.
Collapse
Affiliation(s)
- Maocai Luo
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuhong Zheng
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyun Tang
- GCP Center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linsen Gu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Zhu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rongtao Ying
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yufei Liu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianli Ma
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruixin Guo
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peiyang Gao
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
233
|
Wang G, Luo Y, Gao X, Liang Y, Yang F, Wu J, Fang D, Luo M. MicroRNA regulation of phenotypic transformations in vascular smooth muscle: relevance to vascular remodeling. Cell Mol Life Sci 2023; 80:144. [PMID: 37165163 PMCID: PMC11071847 DOI: 10.1007/s00018-023-04793-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/10/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023]
Abstract
Alterations in the vascular smooth muscle cells (VSMC) phenotype play a critical role in the pathogenesis of several cardiovascular diseases, including hypertension, atherosclerosis, and restenosis after angioplasty. MicroRNAs (miRNAs) are a class of endogenous noncoding RNAs (approximately 19-25 nucleotides in length) that function as regulators in various physiological and pathophysiological events. Recent studies have suggested that aberrant miRNAs' expression might underlie VSMC phenotypic transformation, appearing to regulate the phenotypic transformations of VSMCs by targeting specific genes that either participate in the maintenance of the contractile phenotype or contribute to the transformation to alternate phenotypes, and affecting atherosclerosis, hypertension, and coronary artery disease by altering VSMC proliferation, migration, differentiation, inflammation, calcification, oxidative stress, and apoptosis, suggesting an important regulatory role in vascular remodeling for maintaining vascular homeostasis. This review outlines recent progress in the discovery of miRNAs and elucidation of their mechanisms of action and functions in VSMC phenotypic regulation. Importantly, as the literature supports roles for miRNAs in modulating vascular remodeling and for maintaining vascular homeostasis, this area of research will likely provide new insights into clinical diagnosis and prognosis and ultimately facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Gang Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yulin Luo
- GCP Center, Affiliated Hospital (Traditional Chinese Medicine) of Southwest Medical University, Luzhou, China
| | - Xiaojun Gao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Liang
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Feifei Yang
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Dan Fang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
234
|
Fan P, Meng H, Hao W, Zheng Y, Li H, Zhang Z, Du L, Guo X, Wang D, Wang Y, Wu H. Cardamonin targets KEAP1/NRF2 signaling for protection against atherosclerosis. Food Funct 2023; 14:4905-4920. [PMID: 37157847 DOI: 10.1039/d3fo00967j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Atherosclerosis (AS)-induced cardiovascular disease is a leading cause of death worldwide. To date, there is still a lack of effective approaches for AS intervention. Cardamonin (CAD) is a bioactive food component, but its effect on AS is unknown. In this work, CAD was investigated for its effect on AS using low-density lipoprotein receptor knockout mice and tumor necrosis factor-alpha (TNF-α)-stimulated endothelial cells (ECs). After a 12-week intervention, CAD was found to significantly prevent AS formation in the aortic root and aortic tree, reduce the necrotic core area, and inhibit aortic inflammation and oxidative stress. Moreover, CAD quenched TNF-α-provoked inflammation and oxidative stress in ECs. RNA-sequencing identified nuclear factor erythroid-2 related factor 2 (NFE2L2, NRF2)/heme oxidase 1 (HO1) signaling to be drastically activated by CAD. CAD is a known activator of the aryl hydrocarbon receptor (AHR) which is a transcription factor of the NFE2L2 gene. Surprisingly, AHR was not required for CAD's action on the activation of NRF2/HO1 signaling since AHR gene silencing did not reverse this effect. Furthermore, a molecular docking assay showed a strong binding potential of CAD to the Kelch domain of the Kelch-like ECH-associated protein 1 (KEAP1) which sequesters NRF2 in the cytoplasm. Both CAD and the Kelch domain inhibitor Ki696 promoted NRF2 nuclear translocation, whereas the combination of CAD and Ki696 did not yield a greater effect compared with either CAD or Ki696, confirming the interaction of CAD with the Kelch domain. This work provides an experimental basis for CAD as a novel and effective bioactive food component in future AS interventions.
Collapse
Affiliation(s)
- Pengfei Fan
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Huali Meng
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Wenhao Hao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Yan Zheng
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Xin Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| | - Dongliang Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), 74 Zhongshan Road II, Guangzhou 510080, China
| | - Yunyan Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Rd., Jinan, Shandong 250012, China.
| | - Hao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China.
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China
| |
Collapse
|
235
|
Wang G, Sun Y, Yang Q, Dai D, Zhang L, Fan H, Zhang W, Dong J, Zhao P. Liensinine, a alkaloid from lotus plumule, mitigates lipopolysaccharide-induced sepsis-associated encephalopathy through modulation of nuclear factor erythroid 2-related factor-mediated inflammatory biomarkers and mitochondria apoptosis. Food Chem Toxicol 2023; 177:113813. [PMID: 37150347 DOI: 10.1016/j.fct.2023.113813] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023]
Abstract
The present study aims to investigate the role of liensinine in life-threatened sepsis-associated encephalopathy (SAE) mice and the underlying mechanism. Here, seventy-two mice were divided into six groups, including the control group, SAE group, liensinine-treated group, and three doses of liensinine-treated SAE groups. Lipopolysaccharide triggered cerebrum necrosis and disrupted the integrity and permeability of blood-brain barrier (BBB). While liensinine restored cerebrum structure and improved BBB integrity with upregulated tight junction proteins, decreased evans blue leakage and fibrinogen expression with decreased matrix metalloproteinases 2/9 in serum, thereby reducing BBB permeability. Moreover, lipopolysaccharide triggered cerebrum oxidative stress and inflammation, whereas liensinine enhanced antioxidant enzymes activities and weakened malondialdehyde through nuclear factor erythroid 2-related factor. Meanwhile, liensinine inhibited inflammation by activating inducible nitric oxide synthase. Tunel staining combined with transmission electron microscope indicated that lipopolysaccharide induced cerebrum apoptosis, whereas liensinine blocked apoptosis through decreasing B-cell lymphoma-2 associated X (Bax) expression and cytochrome C (Cyto-c) release, increasing B-cell lymphoma-2 (Bcl-2) expression, blocking apoptosome assembly, inhibiting caspase-3 activation, thereby suppressing intrinsic mitochondria apoptosis. Recovering of inflammatory homeostasis and inhibition of mitochondria apoptosis by liensinine ultimately restored cognitive function in SAE mice. Altogether, liensinine attenuated lipopolysaccharide-induced SAE via modulation of Nrf2-mediated inflammatory biomarkers and mitochondria apoptosis.
Collapse
Affiliation(s)
- Guanglu Wang
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, 222000, China; Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yong Sun
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Qiankun Yang
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, 222000, China; Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Dapeng Dai
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Le Zhang
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Hui Fan
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, 222000, China; Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wei Zhang
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, 222000, China; Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Panpan Zhao
- Institute of Neuroscience, The First People's Hospital of Lianyungang, Lianyungang, 222000, China.
| |
Collapse
|
236
|
Zhai Y, Wang T, Fu Y, Yu T, Ding Y, Nie H. Ferulic Acid: A Review of Pharmacology, Toxicology, and Therapeutic Effects on Pulmonary Diseases. Int J Mol Sci 2023; 24:ijms24098011. [PMID: 37175715 PMCID: PMC10178416 DOI: 10.3390/ijms24098011] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Ferulic acid (FA), a prevalent dietary phytochemical, has many pharmacological effects, including anti-oxidation and anti-inflammation effects, and has been widely used in the pharmaceutical, food, and cosmetics industries. Many studies have shown that FA can significantly downregulate the expression of reactive oxygen species and activate nuclear factor erythroid-2-related factor-2/heme oxygenase-1 signaling, exerting anti-oxidative effects. The anti-inflammatory effect of FA is mainly related to the p38 mitogen-activated protein kinase and nuclear factor-kappaB signaling pathways. FA has demonstrated potential clinical applications in the treatment of pulmonary diseases. The transforming growth factor-β1/small mothers against decapentaplegic 3 signaling pathway can be blocked by FA, thereby alleviating pulmonary fibrosis. Moreover, in the context of asthma, the T helper cell 1/2 imbalance is restored by FA. Furthermore, FA ameliorates acute lung injury by inhibiting nuclear factor-kappaB and mitogen-activated protein kinase pathways via toll-like receptor 4, consequently decreasing the expression of downstream inflammatory mediators. Additionally, there is a moderate neuraminidase inhibitory activity showing a tendency to reduce the interleukin-8 level in response to influenza virus infections. Although the application of FA has broad prospects, more preclinical mechanism-based research should be carried out to test these applications in clinical settings. This review not only covers the literature on the pharmacological effects and mechanisms of FA, but also discusses the therapeutic role and toxicology of FA in several pulmonary diseases.
Collapse
Affiliation(s)
- Yiman Zhai
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Tingyu Wang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yunmei Fu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| |
Collapse
|
237
|
Feng M, Gui Y, An J, Cao X, Lu W, Yang G, Jian S, Hu B, Wen C. The thioredoxin expression of Cristaria plicata is regulated by Nrf2/ARE pathway under microcystin stimulation. Int J Biol Macromol 2023; 242:124509. [PMID: 37085063 DOI: 10.1016/j.ijbiomac.2023.124509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
Thioredoxin plays an important role in inhibiting apoptosis and protecting cells from oxidative stress. This study was aimed to clarify how the expression of Trx from Cristaria plicata is regulated by Nrf2/ARE pathway. The expression of CpTrx mRNA was significantly up-regulated in gill and kidney tissues under microcystin stress. The Nrf2 gene of Cristaria plicata was identified to possess an auto active domain bit. While CpNrf2 was knocked down by specific small RNA, CpTrx mRNA expression was significantly down-regulated. The promoter of CpTrx gene had high transcriptional activity, and this basic transcriptional activity persisted after ARE element mutation. The region of promoter -206 to +217 bp was a core promoter region and had forward regulatory elements. Gel shift Assay exhibited that the CpTrx promoter could bind to the purified proteins CpNrf2 and CpMafK in vitro. The binding phenomenon disappeared after the ARE element mutation in promoter region. Subcellular localization experiments displayed that fluorescence overlap between CpNrf2 and Trx promoter increased under microcystin toxin stress. These results suggested that Trx expression was regulated by Nrf2/ARE pathway under oxidative stress.
Collapse
Affiliation(s)
- Maolin Feng
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China
| | - Yingping Gui
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China
| | - Jinhua An
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China
| | - XinYing Cao
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China
| | - Wuting Lu
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China
| | - Gang Yang
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China
| | - Shaoqing Jian
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China
| | - Baoqing Hu
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China
| | - Chungen Wen
- College of Life Science, Education Ministry Key Laboratory of Poyang Lake Environment and Resource Utilization, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
238
|
Yang L, Lu Y, Zhang Z, Chen Y, Chen N, Chen F, Qi Y, Han C, Xu Y, Chen M, Shen M, Wang S, Zeng H, Su Y, Hu M, Wang J. Oxymatrine boosts hematopoietic regeneration by modulating MAPK/ERK phosphorylation after irradiation-induced hematopoietic injury. Exp Cell Res 2023; 427:113603. [PMID: 37075826 DOI: 10.1016/j.yexcr.2023.113603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/04/2023] [Accepted: 04/16/2023] [Indexed: 04/21/2023]
Abstract
Hematopoietic toxicity due to ionizing radiation (IR) is a leading cause of death in nuclear incidents, occupational hazards, and cancer therapy. Oxymatrine (OM), an extract originating from the root of Sophora flavescens (Kushen), possesses extensive pharmacological properties. In this study, we demonstrate that OM treatment accelerates hematological recovery and increases the survival rate of mice subjected to irradiation. This outcome is accompanied by an increase in functional hematopoietic stem cells (HSCs), resulting in an enhanced hematopoietic reconstitution ability. Mechanistically, we observed significant activation of the MAPK signaling pathway, accelerated cellular proliferation, and decreased cell apoptosis. Notably, we identified marked increases in the cell cycle transcriptional regulator Cyclin D1 (Ccnd1) and the anti-apoptotic protein BCL2 in HSC after OM treatment. Further investigation revealed that the expression of Ccnd1 transcript and BCL2 levels were reversed upon specific inhibition of ERK1/2 phosphorylation, effectively negating the rescuing effect of OM. Moreover, we determined that targeted inhibition of ERK1/2 activation significantly counteracted the regenerative effect of OM on human HSCs. Taken together, our results suggest a crucial role for OM in hematopoietic reconstitution following IR via MAPK signaling pathway-mediated mechanisms, providing theoretical support for innovative therapeutic applications of OM in addressing IR-induced injuries in humans.
Collapse
Affiliation(s)
- Lijing Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Yukai Lu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Zihao Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Yin Chen
- Department of Gynaecology and Obstetrics, 958 Hospital of PLA Army, Chongqing, 400038, China.
| | - Naicheng Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Fang Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Yan Qi
- Department of Hematology, Daping Hospital, Third Military Medical University, Chongqing, 400038, China.
| | - Changhao Han
- Department of Hematology, Daping Hospital, Third Military Medical University, Chongqing, 400038, China.
| | - Yang Xu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Mo Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Mingqiang Shen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Song Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Hao Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Yongping Su
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Mengjia Hu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China; Chinese PLA Center for Disease Control and Prevention, No. 20 Dongda Street, Fengtai District, Beijing, 100071, China.
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
239
|
Liu R, Zhang X, Nie L, Sun S, Liu J, Chen H. Heme oxygenase 1 in erythropoiesis: an important regulator beyond catalyzing heme catabolism. Ann Hematol 2023; 102:1323-1332. [PMID: 37046065 DOI: 10.1007/s00277-023-05193-7] [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: 01/11/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023]
Abstract
Heme oxygenase 1 (HO-1), encoded by the HMOX-1 gene, is the main heme oxygenase that catalyzes the degradation of heme into iron, carbon monoxide, and biliverdin. HMOX-1 gene expression is stimulated by oxidative stress and regulated at transcriptional and post-transcriptional levels. After translation, subcellular location and protein stability of HO-1 are also altered by different extracellular and intracellular stimuli. HO-1 plays a key role in regulating iron homeostasis and cell protection and has become a new target for disease treatment. Erythropoiesis is a tightly controlled, iron-dependent process that begins with hematopoietic stem cells and maturates to red blood cells. HO-1 is expressed in hematopoietic stem/progenitor cells, hematopoietic niche cells, erythroblasts, and especially erythroblastic island and phagocytic macrophages. HO-1 functions importantly in the entire erythroid development process by influencing hematopoietic stem cell proliferation, erythroid lineage engagement, terminal erythroid differentiation, and even senescent RBC erythrophagocytosis. HO-1 is also related to stress erythropoiesis and certain red blood cell diseases. Elucidation of HO-1 regulation and function in erythropoiesis will be of great significance for the treatment of related diseases.
Collapse
Affiliation(s)
- Rui Liu
- Molecular Biology Research Center, School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan Province, 410078, People's Republic of China
| | - Xuzhi Zhang
- Molecular Biology Research Center, School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan Province, 410078, People's Republic of China
- Xiangya School of Medicine, Central South University, Changsha, Hunan Province, 410013, People's Republic of China
| | - Ling Nie
- Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, People's Republic of China
| | - Shuming Sun
- Molecular Biology Research Center, School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan Province, 410078, People's Republic of China
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan Province, 410078, People's Republic of China
| | - Huiyong Chen
- Molecular Biology Research Center, School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan Province, 410078, People's Republic of China.
| |
Collapse
|
240
|
Xie D, Song L, Xiang D, Gao X, Zhao W. Salvianolic acid A alleviates atherosclerosis by inhibiting inflammation through Trc8-mediated 3-hydroxy-3-methylglutaryl-coenzyme A reductase degradation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154694. [PMID: 36804757 DOI: 10.1016/j.phymed.2023.154694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 01/12/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Atherosclerosis is the most prevalent cardiovascular disease and remains the major contributor to death and mortality globally. Salvianolic acid A (SalA) is a water-soluble phenolic acid that benefits atherosclerosis. However, the mechanisms of SalA protecting against atherosclerosis remain unclear. PURPOSE We aimed to determine whether SalA prevents atherosclerosis by modulating 3-Hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) degradation via the ubiquitin-proteasomal pathway. METHODS The animal and cellular models of atherosclerosis were established by subjecting apolipoprotein E (ApoE) knockout mice to a high-fat diet (HFD) and exposing human umbilical vein endothelial cells (HUVECs) to oxidized low-density lipoprotein (ox-LDL), respectively. RESULTS Our results showed that similar to atorvastatin, SalA suppressed atherosclerotic plaque formation, improved serum lipid accumulation, and reduced cholesterol levels in HFD-fed ApoE-/- mice. Moreover, SalA protected HUVECs from ox-LDL-caused cell viability reduction and lipid accumulation. The mechanism study revealed that SalA reduced the production of proinflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6, and augmented the generation of the anti-inflammatory cytokine IL-10 in ApoE-/- mice and HUVECs, accompanied by increased HMGCR ubiquitination and degradation via translocation in renal carcinoma on chromosome 8 (Trc8), insulin-induced gene (Insig)1 and Insig2. Furthermore, the knockdown of Trc8 abolished the SalA-induced HMGCR degradation and anti-atherosclerosis activity. CONCLUSION SalA rescues atherosclerosis by inhibiting inflammation through the Trc8-regulated degradation of HMGCR. These findings underscore Trc8 as a potential target of atherosclerosis.
Collapse
Affiliation(s)
- Dan Xie
- Emergency Department, Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, China
| | - Lijun Song
- School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Dongyang Xiang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550000, China
| | - Xiangyu Gao
- Emergency Department, Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu 215300, China.
| | - Wenchang Zhao
- School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
241
|
Zheng X, Qiu J, Zhang H, Gao N, Jiang T, Gong Y, Zhang W, Li Z, Feng X, Hong Z. PD184352 exerts anti-inflammatory and antioxidant effects by promoting activation of the Nrf2/HO-1 axis. Biochem Pharmacol 2023; 211:115542. [PMID: 37028460 DOI: 10.1016/j.bcp.2023.115542] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/15/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023]
Abstract
Osteoarthritis (OA) is a disabling joint disease characterized by cartilage degeneration. Reactive oxygen species (ROS)-induced oxidative stress is an important cause of early chondrocyte death. For this reason, we investigated PD184352, a small molecule inhibitor with potential anti-inflammatory and antioxidant activity. We evaluated the protective effect of PD184352 against destabilized medial meniscus (DMM)-induced OA in mice. The knee joints of the PD184352-treated group had higher Nrf2 expression and milder cartilage damage. Moreover, in in vitro experiments, PD184352 suppressed IL-1β-induced NO, iNOS, PGE2 production, and attenuated pyroptosis. PD184352 treatment promoted antioxidant protein expression and reduced the accumulation of ROS by activating the Nrf2/HO-1 axis. Finally, the anti-inflammatory and antioxidant effects of PD184352 were shown to be partially dependent on Nrf2 activation. Our study reveals the potential role of PD184352 as an antioxidant and provides a new strategy for OA treatment.
Collapse
|
242
|
Yang C, Liu H, Peng X, Li X, Rao G, Xie Z, Yang Q, Du L, Xie C. Key circRNAs, lncRNAs, and mRNAs of ShenQi Compound in Protecting Vascular Endothelial Cells From High Glucose Identified by Whole Transcriptome Sequencing. J Cardiovasc Pharmacol 2023; 81:300-316. [PMID: 36701487 PMCID: PMC10079301 DOI: 10.1097/fjc.0000000000001403] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023]
Abstract
ABSTRACT Vascular endothelial cells, which make up the inner wall of blood arteries, are susceptible to damage from oxidative stress and apoptosis caused by hyperglycemia. According to certain reports, noncoding RNAs are involved in controlling oxidative stress and apoptosis. ShenQi Compound (SQC), a traditional herbal remedy, has been successfully treating diabetic vascular disease in China for more than 20 years. Although it is well established that SQC protects the vascular endothelium, the molecular mechanism remains unknown. Goto-Kakizaki rats, spontaneous type II diabetes rats, that consistently consume a high-fat diet were chosen as model animals. Six groups (control group, model group, metformin group, and 7.2 g/kg/d SQC group, 14.4 g/kg/d SQC group, and 28.8 g/kg/d SQC group) were included in this work, 15 rats each group. The approach of administration was gavage, and the same volume (5.0 mL/kg/d) was given in each group, once a day, 12 weeks. The thoracic aortas were removed after the rats were sacrificed. Oxidative reduction profile in thoracic aorta, histopathological observation of thoracic aorta, endothelial cell apoptosis in thoracic aorta, whole transcriptome sequencing, bioinformatic analyses, and qRT-PCR were conducted. As a result, SQC prevented the oxidative stress and apoptosis induced by a high glucose concentration. Under hyperglycemia condition, noncoding RNAs, including 1 downregulated novel circRNA (circRNA.3121), 3 downregulated lncRNAs (Skil.cSep08, Shawso.aSep08-unspliced, and MSTRG.164.2), and 1 upregulated mRNA (Pcdh17), were clearly reverse regulated by SQC. SQC plays a role in protecting vascular endothelial cells from high glucose mainly by mediating ncRNA to inhibit cell apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Chan Yang
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Hanyu Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Sichuan, Chengdu, China
| | - Xi Peng
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Xinqiong Li
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Guocheng Rao
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Ziyan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Sichuan, Chengdu, China
| | - Qiangfei Yang
- Jianyang City People's Hospital, Sichuan, China; and
| | - Lian Du
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Sichuan, Chengdu, China
| |
Collapse
|
243
|
Su S, Ma Z, Wu H, Xu Z, Yi H. Oxidative stress as a culprit in diabetic kidney disease. Life Sci 2023; 322:121661. [PMID: 37028547 DOI: 10.1016/j.lfs.2023.121661] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/26/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023]
Abstract
Diabetic kidney disease (DKD) has become the leading cause of end-stage renal disease (ESRD), and the prevalence of DKD has increased worldwide during recent years. DKD is associated with poor therapeutic outcomes in most patients, but there is limited understanding of its pathogenesis. This review suggests that oxidative stress interacts with many other factors in causing DKD. Highly active mitochondria and NAD(P)H oxidase are major sources of oxidants, and they significantly affect the risk for DKD. Oxidative stress and inflammation may be considered reciprocal causes of DKD, in that each is a cause and an effect of DKD. Reactive oxygen species (ROS) can act as second messengers in various signaling pathways and as regulators of metabolism, activation, proliferation, differentiation, and apoptosis of immune cells. Epigenetic modifications, such as DNA methylation, histone modifications, and non-coding RNAs can modulate oxidative stress. The development of new technologies and identification of new epigenetic mechanisms may provide novel opportunities for the diagnosis and treatment of DKD. Clinical trials demonstrated that novel therapies which reduce oxidative stress can slow the progression of DKD. These therapies include the NRF2 activator bardoxolone methyl, new blood glucose-lowering drugs such as sodium-glucose cotransporter 2 inhibitors, and glucagon-like peptide-1 receptor agonists. Future studies should focus on improving early diagnosis and the development of more effective combination treatments for this multifactorial disease.
Collapse
|
244
|
Liu S, Bi H, Jiang M, Chen Y, Jiang M. An update on the role of TRIM/NLRP3 signaling pathway in atherosclerosis. Biomed Pharmacother 2023; 160:114321. [PMID: 36736278 DOI: 10.1016/j.biopha.2023.114321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/14/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of large and medium arteries that includes lipid metabolism disorder and recruitment of immune cells to the artery wall. An increasing number of studies have confirmed that inflammasome over-activation is associated with the onset and progression of atherosclerosis. The NLRP3 inflammasome, in particular, has been proven to increase the incidence rate of cardiovascular diseases (CVD) by promoting pro-inflammatory cytokine release and reducing plaque stability. The strict control of inflammasome and prevention of excessive inflammatory reactions have been the research focus of inflammatory diseases. Tripartite motif (TRIM) is a protein family with a conservative structure and rapid evolution. Several studies have demonstrated the TRIM family's regulatory role in mediating inflammation. This review aims to clarify the relationship between TRIMs and NLRP3 inflammasome and provide insights for future research and treatment discovery.
Collapse
Affiliation(s)
- Sibo Liu
- The QUEEN MARY school, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China
| | - Hongfeng Bi
- Medical Equipment Department, Dongying Shengli Oilfield Central Hospital, Dongying, Shandong 257034, China
| | - Meiling Jiang
- Department of obstetrics, Dongying Shengli Oilfield Central Hospital, Dongying, Shandong 257034, China
| | - Yuanli Chen
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Meixiu Jiang
- The Institute of Translational Medicine, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
245
|
Tabolacci E, Tringali G, Nobile V, Duca S, Pizzoferrato M, Bottoni P, Maria Elisabetta C. Rutin Protects Fibroblasts from UVA Radiation through Stimulation of Nrf2 Pathway. Antioxidants (Basel) 2023; 12:antiox12040820. [PMID: 37107196 PMCID: PMC10135198 DOI: 10.3390/antiox12040820] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
This study explores the photoprotective effects of rutin, a bioflavonoid found in some vegetables and fruits, against UVA-induced damage in human skin fibroblasts. Our results show that rutin increases cell viability and reduces the high levels of ROS generated by photo-oxidative stress (1 and 2 h of UVA exposure). These effects are related to rutin’s ability to modulate the Nrf2 transcriptional pathway. Interestingly, activation of the Nrf2 signaling pathway results in an increase in reduced glutathione and Bcl2/Bax ratio, and the subsequent protection of mitochondrial respiratory capacity. These results demonstrate how rutin may play a potentially cytoprotective role against UVA-induced skin damage through a purely antiapoptotic mechanism.
Collapse
|
246
|
Dong C, Song C, He Z, Song Q, Song T, Liu J, Xiong Y, Su X, Zhou J, Yang S, Liao W. Protective efficacy of Schizandrin B on ameliorating nephrolithiasis via regulating GSK3β/Nrf2 signaling-mediated ferroptosis in vivo and in vitro. Int Immunopharmacol 2023; 117:110042. [PMID: 36940552 DOI: 10.1016/j.intimp.2023.110042] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/25/2023] [Accepted: 03/12/2023] [Indexed: 03/23/2023]
Abstract
Schizandrin B (SchB) protects against oxidative, inflammatory, and ferroptotic injury. Oxidative stress and inflammation are indispensably involved in nephrolithiasis and ferroptosis also plays an important role in stone formation. It is unclear whether SchB can ameliorate nephrolithiasis; its underlying mechanism is also unknown. First, we employed bioinformatics to investigate the mechanisms of nephrolithiasis. To evaluate the efficacy of SchB, HK-2 cell models of oxalate-induced damage, Erastin-induced ferroptosis, and the Sprague Dawley rat model of Ethylene Glycol-induced nephrolithiasis were established. Then, Nrf2 siRNA and GSK3β overexpression plasmids were transfected into HK-2 cells to elucidate the role of SchB in regulating oxidative stress-mediated ferroptosis. In our study, oxidative stress and inflammation were strongly associated with nephrolithiasis. Administration of SchB attenuated the cell viability, dysfunctional mitochondria, oxidative stress and inflammatory response in vitro and alleviated renal injury and crystal deposition in vivo. SchB treatment also reduced the levels of cellular Fe2+ accumulation, lipid peroxidation and MDA, and regulated ferroptosis-related proteins, including XCT, GPX4, FTH1 and CD71, in Erastin-induced or oxalate-induced HK-2 cells. Mechanistically, SchB facilitated Nrf2 nuclear translocation, and silencing Nrf2 or overexpressing GSK3β worsened oxalate-induced oxidative injury and abolished the beneficial effect of SchB against ferroptosis in vitro. To summarize, SchB could alleviate nephrolithiasis by positively regulating GSK3β/Nrf2 signaling-mediated ferroptosis.
Collapse
Affiliation(s)
- Caitao Dong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Chao Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Ziqi He
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Qianlin Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Tianbao Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Junwei Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Yunhe Xiong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Xiaozhe Su
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Jiawei Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Sixing Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China.
| | - Wenbiao Liao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
247
|
Zhang X, Liang F, Li T, Jiang Y, Ren F. Metformin ameliorates calcium oxalate crystallization and stone formation by activating the Nrf2/HO-1 signaling pathway: Two birds with one stone. Arch Biochem Biophys 2023; 739:109568. [PMID: 36914110 DOI: 10.1016/j.abb.2023.109568] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/12/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023]
Abstract
Deposition of calcium oxalate (CaOx) crystals and oxidative stress-induced injury of renal tubular epithelial cell are the primary pathogenic factors of nephrolithiasis. In this study we investigated the beneficial effects of metformin hydrochloride (MH) against nephrolithiasis and explored the underlying molecular mechanism. Our results demonstrated that MH inhibited the formation of CaOx crystals and promoted the transformation of thermodynamically stable CaOx monohydrate (COM) to more unstable CaOx dihydrate (COD). MH treatment effectively ameliorated oxalate-induced oxidative injury and mitochondrial damage in renal tubular cells and reduced CaOx crystal deposition in rat kidneys. MH also attenuated oxidative stress by lowering MDA level and enhancing SOD activity in HK-2 and NRK-52E cells and in a rat model of nephrolithiasis. In both HK-2 and NRK-52E cells, COM exposure significantlylowered the expressions of HO-1 and Nrf2, which was rescued by MH treatment even in the presence of Nrf2 and HO-1 inhibitors. In rats with nephrolithiasis, MH treatment significantly rescued the down-regulation of the mRNA and protein expression of Nrf2 and HO-1 in the kidneys. These results demonstrate that MH can alleviate CaOx crystal deposition and kidney tissue injury in rats with nephrolithiasis by suppressing oxidative stress and activating the Nrf2/HO-1 signaling pathway, suggesting the potential value of MH in the treatment of nephrolithiasis.
Collapse
Affiliation(s)
- Xiaofang Zhang
- Department of Pharmacy, Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Futu Liang
- Department of Pharmacy, Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Tianyang Li
- Department of Pharmacy, Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Yaodong Jiang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China.
| | - Fei Ren
- Department of Pharmacy, Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|
248
|
Li X, Li Z, Dong X, Wu Y, Li B, Kuang B, Chen G, Zhang L. Astragaloside IV attenuates myocardial dysfunction in diabetic cardiomyopathy rats through downregulation of CD36-mediated ferroptosis. Phytother Res 2023. [PMID: 36882189 DOI: 10.1002/ptr.7798] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 03/09/2023]
Abstract
Diabetic cardiomyopathy (DCM), one of the major complications of type 2 diabetes, is a leading cause of heart failure and death in advanced diabetes. Although there is an association between DCM and ferroptosis in cardiomyocytes, the internal mechanism of ferroptosis leading to DCM development remains unknown. CD36 is a key molecule in lipid metabolism that mediates ferroptosis. Astragaloside IV (AS-IV) confers various pharmacological effects such as antioxidant, anti-inflammatory, and immunomodulatory. In this study, we demonstrated that AS-IV was able to recover the dysfunction of DCM. In vivo experiments showed that AS-IV ameliorated myocardial injury and improved contractile function, attenuated lipid deposition, and decreased the expression level of CD36 and ferroptosis-related factors in DCM rats. In vitro experiments showed that AS-IV decreased CD36 expression and inhibited lipid accumulation and ferroptosis in PA-induced cardiomyocytes. The results demonstrated that AS-IV decreased cardiomyocyte injury and myocardial dysfunction by inhibiting ferroptosis mediated by CD36 in DCM rats. Therefore, AS-IV regulated the lipid metabolism of cardiomyocytes and inhibited cellular ferroptosis, which may have potential clinical value in DCM treatment.
Collapse
Affiliation(s)
- Xin Li
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Ziwei Li
- Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Dong
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Yu Wu
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Baohua Li
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Bin Kuang
- Dongguan Hospital of Traditional Chinese Medicine, Dongguan, China
| | - Gangyi Chen
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Liangyou Zhang
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
249
|
Sharma A, Jaiswal V, Park M, Lee HJ. Biogenic silver NPs alleviate LPS-induced neuroinflammation in a human fetal brain-derived cell line: Molecular switch to the M2 phenotype, modulation of TLR4/MyD88 and Nrf2/HO-1 signaling pathways, and molecular docking analysis. BIOMATERIALS ADVANCES 2023; 148:213363. [PMID: 36881963 DOI: 10.1016/j.bioadv.2023.213363] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
Silver nanoparticles (AgNPs) have inconsistent findings against inflammation. Although a wealth of literature on the beneficial effects of green-synthesized AgNPs has been published, a detailed mechanistic study of green AgNPs on the protective effects against lipopolysaccharide (LPS)-induced neuroinflammation using human microglial cells (HMC3) has not yet been reported. For the first time, we studied the inhibitory effect of biogenic AgNPs on inflammation and oxidative stress induced by LPS in HMC3 cells. X-ray photoelectron spectroscopy, Fourier-transform infrared spectroscopy, and transmission electron microscopy were used to characterize AgNPs produced from honeyberry. Co-treatment with AgNPs significantly reduced mRNA expressions of inflammatory molecules such as interleukin (IL)-6 and tumor necrosis factor-α, while increasing the expressions of anti-inflammatory markers such as IL-10 and transforming growth factor (TGF)-β. HMC3 cells were also switched from M1 to M2, as shown by lower expression of M1 markers such as cluster of differentiation (CD)80, CD86, and CD68 and higher expression of M2 markers such as CD206, CD163, and triggering receptors expressed on myeloid cells (TREM2). Furthermore, AgNPs inhibited LPS-induced toll-like receptor (TLR)4 signaling, as evidenced by decreased expression of myeloid differentiation factor 88 (MyD88) and TLR4. In addition, AgNPs reduced the production of reactive oxygen species (ROS) and enhanced the expression of nuclear factor-E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), while decreasing the expression of inducible nitric oxide synthase. The docking score of the honeyberry phytoconstituents ranged from -14.93 to - 4.28 KJ/mol. In conclusion, biogenic AgNPs protect against neuroinflammation and oxidative stress by targeting TLR4/MyD88 and Nrf2/HO-1 signaling pathways in a LPS-induced in vitro model. Biogenic AgNPs could be utilized as potential nanomedicine against LPS-induced inflammatory disorders.
Collapse
Affiliation(s)
- Anshul Sharma
- College of BioNano Technology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea
| | - Varun Jaiswal
- College of BioNano Technology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea
| | - Miey Park
- College of BioNano Technology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea; Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Republic of Korea
| | - Hae-Jeung Lee
- College of BioNano Technology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea; Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea.
| |
Collapse
|
250
|
Zhang S, Chen Y, Yang Z, Xiang H, Kang P, Li J. Active substances and molecular mechanisms of the anti-myocardial ischemia effects of Carthami flos by network pharmacology and in vitro experiments. Heliyon 2023; 9:e13877. [PMID: 36895345 PMCID: PMC9988582 DOI: 10.1016/j.heliyon.2023.e13877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Myocardial ischemia is a predominant cardiovascular disorder that can result in a series of life-threatening cardiovascular diseases. Carthami flos (CF), the flower of Carthamus tinctorius L., is a commonly used herbal medicine in Chinese medicine for treating coronary atherosclerotic heart diseases based on its anti-myocardial ischemia (MI) effects. This paper aimed to investigate the active substances and mechanisms of the anti-MI effects of CF by network pharmacology and in vitro experiments. The results indicated that 9 constituents showed high degree of association with multiple targets of MI, including quercetin, kaempferol, β-sitosterol, luteolin, baicalein, safflomin A, safflomin C, safflower-yellow-B and hydroxysafflor yellow A. In addition, AKT1, EGFR, CASP3, MYC, JUN, ALB, CTNNB1, VEGFA, ESR1, and IL1B were screened as the leading targets with a degree number ≥50. Bioinformatic annotation of GO-MF and KEGG showed that the anti-MI effects of CF are related to apoptosis and response to antioxidative stress pathways. The in vitro results showed that CF reduced LDH and CK levels, alleviated cell cycle arrest, and decreased ROS levels in H2O2-treated H9c2 cells. In addition, CF also promoted the nuclear shift of Nrf2 and the mRNA expressions of Akt, Nrf2 and Bcl-2 but decreased the expression of caspase-3 in H2O2-treated H9c2 cells. Collectively, the anti-MI effects of CF involve inhibiting apoptosis and antioxidative stress in cardiomyoblasts by regulating Akt/Nrf2/Caspase-3/Bcl-2, and the possible active substances of CF are quercetin, kaempferol, β-sitosterol, luteolin, baicalein, safflomin C, safflower-yellow-B, and hydroxysafflor yellow A. The results of this study will be helpful for further drug development of CF and its active monomers.
Collapse
Affiliation(s)
- Sirong Zhang
- Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan 628000, PR China
| | - Yu Chen
- Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan 628000, PR China
- Corresponding author.
| | - Zhilin Yang
- Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan 628000, PR China
| | - Hai Xiang
- Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan 628000, PR China
| | - Pan Kang
- Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan 628000, PR China
| | - Jiang Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
- Corresponding author.
| |
Collapse
|