1
|
Li Y, Xue X, Peng M, Zheng X, Tao H, Guo S. UPLC-MS/MS analysis of Salvia miltiorrhiza Bge. Extract and its ameliorative effect of endothelial cell metabolism in isoproterenol-induced myocardial injury. Fitoterapia 2025; 183:106554. [PMID: 40254169 DOI: 10.1016/j.fitote.2025.106554] [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: 01/25/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Salvia miltiorrhiza Bge. (S. miltiorrhiza) is an important herbal medicine, and the researches mainly focused on the roots of S. miltiorrhiza rather than the above-ground parts. Therefore, the detailed research on the above-ground parts of S. miltiorrhiza is still unclear. This study aimed to elucidate the constituents of the above-ground parts of S. miltiorrhiza extract (SME) and evaluate its effect on endothelial cell injury. For this purpose, the constituents of SME were identified by ultra performance liquid chromatography tandem mass spectrometry, and 41 compounds were determined, including terpenes, phenylpropanoids, flavonoids, sterols and phenolic acids. The effect of SME on endothelial cell was evaluated using isoproterenol (ISO)-treated C57BL/6 mice and hypoxia-treated HUVECs, and the results revealed that SME could improve endothelial cell injury in ISO-induced myocardial infarction model and hypoxia-induced HUVECs injury. In this process, SME could inhibit reactive oxygen species production, decrease the levels of malondialdehyde, lactic dehydrogenase, pyruvic acid, intracellular iron ions and sugar, and increase the levels of glutathione. Meanwhile, SME could improve the abnormal expressions of apoptosis-, glycolysis- and ferroptosis-related proteins. Taken together, our data demonstrated that SME treatment could alleviate endothelial cell injury by regulating HIF-1α mediated glycolysis and ferroptosis signals.
Collapse
Affiliation(s)
- Yan Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaofei Xue
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Meng Peng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaolin Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hailong Tao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Sen Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
2
|
Zhou M, Qin Z, Zhu X, Ruan Y, Ling H, Li C, Gan X. Pyruvate dehydrogenase kinases: key regulators of cellular metabolism and therapeutic targets for metabolic diseases. J Physiol Biochem 2025; 81:21-34. [PMID: 40117090 DOI: 10.1007/s13105-025-01068-9] [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: 09/06/2024] [Accepted: 01/27/2025] [Indexed: 03/23/2025]
Abstract
Pyruvate dehydrogenase kinases (PDKs) can regulate the conversion of pyruvate to acetyl coenzyme A through the mitochondrial pyruvate dehydrogenase complex (PDHC). As the rate-limiting enzymes of PDHC, PDKs link glycolysis to the tricarboxylic acid cycle. Pathological changes in many diseases involve alterations in cellular metabolism, which are partly reflected in changes in mitochondrial function. The intermediate role of PDKs in metabolic processes allows for the influence of both glycolysis and oxidative phosphorylation. Recent studies have shown that PDKs play a crucial role in regulating metabolic reprogramming, mitochondrial function and cellular activities in both oncological studies and various non-oncological diseases. This paper aims to clarify the molecular regulatory mechanisms of PDKs; review the relationship of PDKs with cellular metabolic reprogramming, regulation of ROS, and apoptosis; and the present status of research on PDKs in osteoporosis, diabetes mellitus, and vascular diseases. With this review, we have increased our understanding and insight at the molecular level, providing new insights into targeting PDKs to reverse metabolism-related diseases.
Collapse
Affiliation(s)
- Min Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ziqi Qin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiting Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yifeng Ruan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huiling Ling
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chen Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
3
|
Luan H, Wang Z, Zhang Z, Hou B, Liu Z, Yang L, Yang M, Ma Y, Zhang B. Brassica oleracea L. extract ameliorates isoproterenol-induced myocardial injury by regulating HIF-1α-mediated glycolysis. Fitoterapia 2024; 172:105715. [PMID: 37907131 DOI: 10.1016/j.fitote.2023.105715] [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: 07/07/2023] [Revised: 09/07/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023]
Abstract
Brassica oleracea L. (BO) is an important vegetable with proven health benefits. This study aimed to elucidate the constituents of BO leaf extract (BOE) and evaluate its effect on myocardial injury. For this purpose, the constituents of BOE were identified using ultra-high performance liquid chromatography with quadrupole time-of- flight mass spectrometry, and 26 compounds were determined, including glucosinolates, sulfur compounds, alkaloids, phenolic acids, flavones, and two other kinds of compounds. The effects of BOE on myocardial cells were evaluated using isoproterenol (ISO)-treated H9C2 cells and Wistar rats, and the results revealed that BOE could inhibit cardiomyocyte hypertrophy and reduce the levels of B-type natriuretic peptide, nitric oxide, reactive oxygen species, lactic acid, and pyruvic acid. Meanwhile, BOE could increase the levels of mitochondrial membrane potential. Moreover, BOE could reduce the levels of apoptosis- and glycolysis-related proteins. Taken together, our data demonstrated that BOE treatment could alleviate ISO-induced myocardial cell injury by downregulating apoptosis and glycolysis signals.
Collapse
Affiliation(s)
- Huiling Luan
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Zhenhui Wang
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Zhenzhen Zhang
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Baohua Hou
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Zhenzhen Liu
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Lanping Yang
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Mengmeng Yang
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Yile Ma
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China
| | - Baobao Zhang
- Department of Pharmacy, School of Medicine, Henan Polytechnic University, Jiaozuo 454000, People's Republic of China.
| |
Collapse
|
4
|
Quiroga J, Alarcón P, Ramírez MF, Manosalva C, Teuber S, Carretta MD, Burgos RA. d-lactate-induced ETosis in cattle polymorphonuclear leucocytes is dependent on the release of mitochondrial reactive oxygen species and the PI3K/Akt/HIF-1 and GSK-3β pathways. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 145:104728. [PMID: 37164278 DOI: 10.1016/j.dci.2023.104728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/12/2023]
Abstract
d-lactate is a metabolite originating from bacterial metabolism that accumulates as a result of dietary disturbances in cattle, leading to ruminal acidosis. d-lactate exerts functions as a metabolic signal inducing metabolic reprogramming and extracellular trap (ET) release in polymorphonuclear leucocytes (PMNs). We previously demonstrated that d-lactate induces metabolic reprogramming via hypoxia-induced factor 1 alpha (HIF-1α) stabilization in bovine fibroblast-like synoviocytes (FLSs). In the present study, the role of HIF-1 in ET formation induced by d-lactate was assessed. HIF-1α stabilization in PMNs was controlled by mitochondrial reactive oxygen species (mtROS) release. Furthermore, inhibition of mitochondrial complex I and scavenging of mtROS decreased d-lactate-triggered ETosis. d-lactate-enhanced HIF-1α accumulation was dependent on the PI3K/Akt pathway but independent of GSK-3β activity. Pharmacological blockade of the PI3K/Akt/HIF-1 and GSK-3β axes inhibited d-lactate-triggered ETosis and downregulated PDK1 and LDHA expression. However, only GSK-3β inhibition decreased the expression of glycogen metabolism enzymes and prevented the decline in glycogen stores induced by d-lactate exposure. The results of this study suggest that mtROS, PI3K/Akt/HIF-1 and GSK-3β axes regulate carbohydrate metabolism adaptations that support d-lactate-induced ET formation in cattle.
Collapse
Affiliation(s)
- John Quiroga
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Alarcón
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - María Fernanda Ramírez
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Manosalva
- Institute of Pharmacy, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Stefanie Teuber
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - María Daniella Carretta
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael Agustín Burgos
- Laboratory of Inflammation Pharmacology and Immunometabolism, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
5
|
Che K, Yang Y, Zhang J, Feng L, Xie Y, Li Q, Qiu J. Oral pyruvate prevents high-intensity interval exercise-induced metabolic acidosis in rats by promoting lactate dehydrogenase reaction. Front Nutr 2023; 10:1096986. [PMID: 37090767 PMCID: PMC10117856 DOI: 10.3389/fnut.2023.1096986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction There is no denying the clinical benefits of exogenous pyruvate in the treatment of pathological metabolic acidosis. However, whether it can prevent exercise physiological metabolic acidosis, delay the occurrence of exercise fatigue, and improve the beneficial effects of exercise and its internal mechanism remain unclear. Methods We randomly divided 24 male SD rats into 3 groups: one group was a control without exercise (CC, n = 8), and the other two groups were supplemented with 616 mg/kg/day pyruvate (EP, n = 8) or distilled water of equal volume (EC, n = 8). These groups completed acute high-intensity interval exercise (HIIE) after 7 days of supplementation. The acid metabolism variables were measured immediately after exercise including blood pH (pHe), base excess (BE), HCO3 -, blood lactic acid and skeletal muscle pH (pHi). The redox state was determined by measuring the oxidized coenzyme I/reduced coenzyme I (nicotinamide adenine dinucleotide [NAD+]/reduced NAD+ [NADH]) ratio and lactate/pyruvate (L/P) ratio. In addition, the activities of lactate dehydrogenase A (LDHA), hexokinase (HK), phosphofructokinase (PFK) and pyruvate kinase (PK) were determined by ELISA. Results Pyruvate supplementation significantly reversed the decrease of pHe, BE, HCO3 - and pHi values after HIIE (p < 0.001), while significantly increased the activities of LDHA (p = 0.048), HK (p = 0.006), and PFK (p = 0.047). Compared with the CC, the NAD+/NADH (p = 0.008) ratio and the activities of LDHA (p = 0.002), HK (p < 0.001), PFK (p < 0.001), and PK (p = 0.006) were significantly improved in EP group. Discussion This study provides compelling evidence that oral pyruvate attenuates HIIE-induced intracellular and extracellular acidification, possibly due to increased activity of LDHA, which promotes the absorption of H+ in the LDH reaction. The beneficial effects of improving the redox state and glycolysis rate were also shown. Our results suggest that pyruvate can be used as an oral nutritional supplement to buffer HIIE induced metabolic acidosis.
Collapse
Affiliation(s)
- Kaixuan Che
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
| | - Yanping Yang
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
| | - Jun Zhang
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
| | - Lin Feng
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
| | - Yan Xie
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
| | - Qinlong Li
- Department of Exercise Physiology, Exercise Science School, Beijing Sport University, Beijing, China
| | - Junqiang Qiu
- Department of Exercise Biochemistry, Exercise Science School, Beijing Sport University, Beijing, China
- Beijing Sports Nutrition Engineering Research Center, Beijing, China
- *Correspondence: Junqiang Qiu,
| |
Collapse
|
6
|
Zhang L, Zhang Y, Zhou J, Yao Y, Li R, Zhou M, Chen S, Qiao Z, Yang K. Combined transcriptome and proteome analysis of yak PASMCs under hypoxic and normoxic conditions. PeerJ 2022; 10:e14369. [PMID: 36452079 PMCID: PMC9703989 DOI: 10.7717/peerj.14369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/19/2022] [Indexed: 11/26/2022] Open
Abstract
Background Yaks are animals that have lived in plateau environments for generations. Yaks can adapt to the hypoxic plateau environment and also pass this adaptability on to the next generation. The lungs are the most important respiratory organs for mammals to adapt to their environment. Pulmonary artery smooth muscle cells play an important role in vascular remodeling under hypoxia, but the genetic mechanism underpinning the yak's ability to adapt to challenging plateau conditions is still unknown. Methods A tandem mass tag (TMT) proteomics study together with an RNA-seq transcriptome analysis were carried out on pulmonary artery smooth muscle cells (PASMCs) that had been grown for 72 hours in both normoxic (20% O2) and hypoxic (1% O2) environments. RNA and TP (total protein) were collected from the hypoxic and normoxic groups for RNA-seq transcriptome sequencing and TMT marker protein quantification, and RT-qPCR validation was performed. Results A total of 17,711 genes and 6,859 proteins were identified. Further, 5,969 differentially expressed genes (DEGs) and 531 differentially expressed proteins (DEPs) were identified in the comparison group, including 2,924 and 186 upregulated genes and proteins and 3,045 and 345 down-regulated genes and proteins, respectively. The transcriptomic and proteomic analyses revealed that 109 DEGs and DEPs were highly positively correlated, with 77 genes showing the same expression trend. Nine overlapping genes were identified in the HIF-1 signaling pathway, glycolysis / gluconeogenesis, central carbon metabolism in cancer, PPAR signaling pathway, AMPK signaling pathway, and cholesterol metabolism (PGAM1, PGK1, TPI1, HMOX1, IGF1R, OLR1, SCD, FABP4 and LDLR), suggesting that these differentially expressed genes and protein functional classifications are related to the hypoxia-adaptive pathways. Overall, our study offers abundant data for further analysis of the molecular mechanisms in yak PASMCs and their adaptability to different oxygen concentrations.
Collapse
Affiliation(s)
- Lan Zhang
- Life Science and Engineering College, Northwest Minzu University, Lan, China
| | - Yiyang Zhang
- Life Science and Engineering College, Northwest Minzu University, Lan, China,Biomedical Research Center, Northwest Minzu University, Lan Zhou, China,Gansu Tech Innovation Center of Animal Cell, Lan Zhou, China
| | - Juan Zhou
- Life Science and Engineering College, Northwest Minzu University, Lan, China
| | - Yifan Yao
- Life Science and Engineering College, Northwest Minzu University, Lan, China,Biomedical Research Center, Northwest Minzu University, Lan Zhou, China,Gansu Tech Innovation Center of Animal Cell, Lan Zhou, China
| | - Rui Li
- Life Science and Engineering College, Northwest Minzu University, Lan, China,Biomedical Research Center, Northwest Minzu University, Lan Zhou, China,Gansu Tech Innovation Center of Animal Cell, Lan Zhou, China
| | - Manlin Zhou
- Life Science and Engineering College, Northwest Minzu University, Lan, China
| | - Shuwu Chen
- Life Science and Engineering College, Northwest Minzu University, Lan, China,Biomedical Research Center, Northwest Minzu University, Lan Zhou, China,Gansu Tech Innovation Center of Animal Cell, Lan Zhou, China
| | - Zilin Qiao
- Life Science and Engineering College, Northwest Minzu University, Lan, China,Biomedical Research Center, Northwest Minzu University, Lan Zhou, China,Gansu Tech Innovation Center of Animal Cell, Lan Zhou, China
| | - Kun Yang
- Life Science and Engineering College, Northwest Minzu University, Lan, China,Biomedical Research Center, Northwest Minzu University, Lan Zhou, China,Gansu Tech Innovation Center of Animal Cell, Lan Zhou, China
| |
Collapse
|
7
|
Ultraviolet Radiation Promoted Hypoxia-Induced Apoptosis in HL-60 Human Promyelocytic Leukemia Cell Line. JOURNAL OF ONCOLOGY 2022; 2022:7702481. [PMID: 36353706 PMCID: PMC9640238 DOI: 10.1155/2022/7702481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/25/2022] [Accepted: 10/12/2022] [Indexed: 01/24/2023]
Abstract
Minimal residual disease (MRD) is an important reason for the failure of autologous hematopoietic stem cell transplantation (auto-HSCT). Reducing MRD in grafts is particularly important to improve the efficacy of auto-HSCT. Previously, we reported that ultraviolet light-emitting diode (UV LED) suppressed the expression of Bcl-2 to induce apoptosis in HL-60 cells. Leukemia can lead to severe hypoxia of the bone marrow. Therefore, this study aimed to investigate the effect of UV LED on leukemia cells under hypoxia. HL-60 cells were irradiated with a UV LED (30 J/m2) and simulated under hypoxia with cobalt chloride. We found that UV LED irradiation or CoCl2 inhibited proliferation, induced apoptosis, decreased the Bcl-2/Bax ratio, and increased the levels of caspase 3, cleaved-caspase 3, and caspase 9 in HL-60 cells. In particular, the combined application of UV and CoCl2 significantly enhanced the apoptosis of HL-60 cells. In conclusion, UV LED in hypoxia exacerbated the inhibition of proliferation and induction of apoptosis and necrosis in HL-60 cells via the regulation of caspase 3/9 and the Bcl-2/Bax ratio-dependent pathway. The application of UV LEDs in hypoxia conditions may be a promising approach to kill residual drug-resistant leukemia cells in autologous grafts.
Collapse
|
8
|
Shan Y, Xie K, Zhou Q, He R, Chen Z, Feng W. Sevoflurane alleviates myocardial ischemia/reperfusion injury via actitation of heat shock protein-70 in patients undergoing double valve replacement surgery. Am J Transl Res 2022; 14:5529-5540. [PMID: 36105042 PMCID: PMC9452318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 03/03/2022] [Indexed: 06/15/2023]
Abstract
This study investigated the cardioprotective effect(s) of sevoflurane in rheumatic heart disease patients undergoing double valve replacement surgery (DVRS) under cardiopulmonary bypass (CPB) and its potential mechanisms (ChiCTR2100051220 on http://www.ChiCTR.org.cn). Forty-six patients were randomly assigned to undergo propofol or sevoflurane anesthesia during surgery. The levels of myocardial injury markers, inflammatory cytokines, heat shock protein-70 (HSP70), and superoxide dismutase (SOD) activity were measured from blood samples. Mean arterial pressure, cardiac index, and stroke volume index were significantly higher in the sevoflurane group than in the propofol group at the end of CPB. However, there were no significant differences in operative duration, length of CPB or aortic cross-clamp time, auto-resuscitation heart rate, drainage within 48 h after surgery, time to extubation, and recovery time after DVRS. The dose of inotropic agents (dopamine and noradrenaline) was significantly lower in the sevoflurane group than in the propofol group. Sevoflurane was associated with smaller increases in the levels of myocardial injury-associated markers (CK-MB and cardiac troponin I [cTnI]) and inflammatory cytokines (interleukin [IL]-6, IL-8, and tumor-necrosis factor-alpha [TNF-α]); however, there was a greater increase in HSP70 levels compared with propofol after surgery. Moreover, SOD activity after surgery was significantly higher in the sevoflurane group than in the propofol group. Increased HSP70 levels in the sevoflurane group were positively correlated with cTnI, IL-6, IL-8, and TNF-α levels, and negatively correlated with SOD activity. These results suggest a cardioprotective effect of sevoflurane during DVRS. Sevoflurane may reduce biomarkers of cardiac injury through its anti-inflammatory effects via upregulation of HSP70.
Collapse
Affiliation(s)
- Yue Shan
- Department of Anesthesiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang, China
| | - Kai Xie
- Department of Anesthesiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang, China
| | - Qifu Zhou
- Department of Anesthesiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang, China
| | - Rui He
- Department of Anesthesiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang, China
| | - Zhonghua Chen
- Department of Anesthesiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang, China
| | - Weizhong Feng
- Department of Cardiac Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine)Shaoxing 312000, Zhejiang, China
| |
Collapse
|
9
|
Yi X, Qi M, Huang M, Zhou S, Xiong J. Honokiol Inhibits HIF-1α-Mediated Glycolysis to Halt Breast Cancer Growth. Front Pharmacol 2022; 13:796763. [PMID: 35350760 PMCID: PMC8957822 DOI: 10.3389/fphar.2022.796763] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 02/02/2022] [Indexed: 12/28/2022] Open
Abstract
Background: Hypoxia-inducible factor-1α (HIF-1α) induces the expression of glycolysis-related genes, which plays a direct and key role in Warburg effect. In a recent study, honokiol (HNK) was identified as one of the potential agents that inhibited the HIF-1α signaling pathway. Because the HIF- 1α pathway is closely associated with glycolysis, we investigated whether HNK inhibited HIF-1α-mediated glycolysis. Methods: The effects of HNK on HIF-1α-mediated glycolysis and other glycolysis-related genes’ expressions, cancer cells apoptosis and tumor growth were studied in various human breast cancer models in vitro and in vivo. We performed the following tests: extracellular acidification and oxygen consumption rate assays, glucose uptake, lactate, and ATP assays for testing glycolysis; WST-1 assay for investigating cell viability; colony formation assay for determining clonogenicity; flow cytometry for assessing cell apoptosis; qPCR and Western blot for determining the expression of HIF-1α, GLUT1, HK2 and PDK1. The mechanisms of which HNK functions as a direct inhibitor of HIF-1α were verified through the ubiquitination assay, the Co-IP assay, and the cycloheximide (CHX) pulse-chase assay. Results: HNK increased the oxygen consumption rate while decreased the extracellular acidification rate in breast cancer cells; it further reduced glucose uptake, lactic acid production and ATP production in cancer cells. The inhibitory effect of HNK on glycolysis is HIF-1α-dependent. HNK also downregulated the expression of HIF-1α and its downstream regulators, including GLUT1, HK2 and PDK1. A mechanistic study demonstrated that HNK enhanced the self-ubiquitination of HIF-1α by recruiting two E3 ubiquitin ligases (UFL1 and BRE1B). In vitro, HNK inhibited cell proliferation and clonogenicity, as well as induced apoptosis of cancer cells. These effects were also HIF1α-dependent. In vivo, HNK inhibited tumor growth and HIF-1α-mediated glycolysis. Conclusion: HNK has an inhibitory effect on HIF-1α-mediated glycolysis in human breast cancer. Our research revealed a new mechanism of HNK as an anti-cancer drug, thus representing a novel strategy to improve the prognosis of cancer.
Collapse
Affiliation(s)
- Xianglan Yi
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengxin Qi
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingxiang Huang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Zhou
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Xiong
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Cardioprotective Mechanisms of Interrupted Anesthetic Preconditioning with Sevoflurane in the Setting of Ischemia/Reperfusion Injury in Rats. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Anesthetic preconditioning (AP) is known to mimic ischemic preconditioning. The purpose of this study was to investigate the effects of an interrupted sevoflurane administration protocol on myocardial ischemia/reperfusion (I/R) injury. Methods: Male Wistar rats (n = 60) were ventilated for 30 min with room air (control group, CG) or with a mixture of air and sevoflurane (1 minimum alveolar concentration—MAC) in 5-min cycles, alternating with 5-min wash-out periods (preconditioned groups). Cytokines implicated in the AP response were measured. An (I/R) lesion was produced immediately after the sham intervention (CG) and preconditioning protocol (early AP group, EAPG) or 24 h after the intervention (late AP group, LAPG). The area of fibrosis, the degree of apoptosis and the number of c-kit+ cells was estimated for each group. Results: Cytokine levels were increased post AP. The area of fibrosis decreased in both EAPG and LAPG compared to the CG (p < 0.0001). When compared to the CG, the degree of apoptosis was reduced in both LAPG (p = 0.006) and EAPG (p = 0.007) and the number of c-kit+ cells was the greatest for the LAPG (p < 0.0001). Conclusions: Sevoflurane preconditioning, using an interrupted anesthesia protocol, is efficient in myocardial protection and could be beneficial to reduce perioperative or periprocedural ischemia in patients with increased cardiovascular risk.
Collapse
|
11
|
Li Q, Guo S, Yang C, Liu X, Chen X, He J, Tong C, Ding Y, Peng C, Geng Y, Mu X, Liu T, Li F, Wang Y, Gao R. High-fat diet-induced obesity primes fatty acid β-oxidation impairment and consequent ovarian dysfunction during early pregnancy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:887. [PMID: 34164521 PMCID: PMC8184480 DOI: 10.21037/atm-21-2027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Obesity is associated with many adverse effects on female fertility. Obese women have a higher likelihood of developing ovulatory dysfunction due to dysregulation of the hypothalamic-pituitary-ovarian axis. However, the effect of obesity on ovarian function during early pregnancy needs to be further assessed. Methods C57BL6/J mice were given a high-fat diet (HFD) for 12 weeks to induce obesity. An in vitro high-fat model was established by treating the human ovarian granulosa cell line KGN with oleic acid and palmitic acid. Ovarian morphology of obese mice in early pregnancy was assessed by hematoxylin and eosin staining and ovarian function was assessed by enzyme-linked immunosorbent assay, western blotting, and immunohistochemistry. Oil Red O staining and transmission electron microscopy were used to detect fatty acid accumulation. Specific markers relating to the ovarian functional mechanism were assessed by real-time PCR, western blotting, lactate detection, adenosine triphosphate (ATP) detection, biochemical analyses, and enzyme-linked immunosorbent assay. Results The results of this study showed that during early pregnancy, the number of corpus lutea, serum estradiol and progesterone levels, and the expression of the steroid biosynthesis-related protein CYP19A1 (aromatase), CYP11A1 (cholesterol side chain cleavage enzyme), and StAR (steroidogenic acute regulatory protein), were significantly increased in HFD mice. Mice fed an HFD also showed a significant increase in ovarian lipid accumulation on day 7 of pregnancy. Genes involved in fatty acid synthesis (Acsl4 and Elovl5), and fatty acid uptake and transport (Slc27a4), together with the β-oxidation rate-limiting enzyme Cpt1a, were significantly upregulated in HFD mice. Specifically, there was abnormal elevation of ATP and aberrant expression of tricarboxylic acid cycle (TCA)- and electron transport chain (ETC)-related genes in the ovaries of pregnant HFD mice. KGN cells treated with etomoxir targeting β-oxidation of fatty acid showed decreased TCA cycle and ETC related gene expression. The elevation of ATP and estradiol and progesterone levels was reversed. Conclusions During early pregnancy, HFD-induced obesity increases fatty acid β-oxidation, which in turn increases TCA cycle and ETC related gene expression, leading to increased ATP production and ovarian dysfunction.
Collapse
Affiliation(s)
- Qingying Li
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Sujuan Guo
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Chengshun Yang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Xueqing Liu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Chao Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yubin Ding
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Chuan Peng
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanqing Geng
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Xinyi Mu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Taihang Liu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Fangfang Li
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Yingxiong Wang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| |
Collapse
|