1
|
He X, Han Y, Chen L, Yun Y, Huang Y, Borjigin G, Nashun B. Mechanisms of Adipose Tissue Metabolism in Naturally Grazing Sheep at Different Growth Stages: Insights from mRNA and miRNA Profiles. Int J Mol Sci 2025; 26:3324. [PMID: 40244192 PMCID: PMC11989906 DOI: 10.3390/ijms26073324] [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: 02/19/2025] [Revised: 03/30/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
Adipose tissue metabolism plays a crucial role in sheep meat quality and the optimization of adipose tissue utilization. To reveal the molecular mechanisms of adipose tissue metabolism during growth in naturally grazing sheep, we investigated the mRNA and miRNA profiles in subcutaneous adipose tissue (SAT) from naturally grazing Sunit sheep at 6, 18, and 30 months of age (Mth-6, Mth-18, and Mth-30). We identified 927 differentially expressed (DE) genes and 134 DE miRNAs in the SAT of sheep at different growth stages. Specifically, the expressions of ACACA, FASN, DGAT2, GPAM, SCD, ELOVL6, HSD17B12, TECR, PKM, TKT, PCK1, CD44, and THBS2S genes were significantly upregulated in Mth-18 and Mth-30 compared to that in Mth-6. These genes promoted fatty acid synthesis, triglyceride synthesis, gluconeogenesis, and extracellular matrix-receptor interaction and decreased glycolysis, leading to increased adipocyte proliferation and fat deposition. Notably, our findings suggested that the reduced activity of the AMPK signaling pathway may be regulated by CAMKK2 and PP2A during sheep growth. Furthermore, our results revealed several DE miRNAs, mml-miR-320b, chi-miR-1388-3p, bta-miR-6715, oar-miR-143, and miR-424, that potentially influence fat metabolism. Overall, this study provides a theoretical basis and new insights into the molecular mechanisms of adipose tissue metabolism during growth in naturally grazing sheep.
Collapse
Affiliation(s)
- Xige He
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| | - Yunfei Han
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.H.); (L.C.); (Y.H.); (G.B.)
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot 010031, China
| | - Lu Chen
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.H.); (L.C.); (Y.H.); (G.B.)
| | - Yueying Yun
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China;
| | - Yajuan Huang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.H.); (L.C.); (Y.H.); (G.B.)
| | - Gerelt Borjigin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.H.); (L.C.); (Y.H.); (G.B.)
| | - Buhe Nashun
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot 010070, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010040, China
| |
Collapse
|
2
|
Mi J, Zhao L, Shen Y, Mo S, Kuang Y. PFKP Lactylation Promotes the Ovarian Cancer Progression Through Targeting PTEN. Biochem Genet 2024:10.1007/s10528-024-10990-4. [PMID: 39638933 DOI: 10.1007/s10528-024-10990-4] [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: 09/03/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Ovarian cancer (OC) ranks among the most prevalent malignancies affecting females globally and is a leading cause of cancer-related mortality in women. This study sought to elucidate the influence of phosphofructokinase P (PFKP) on the progression of OC. A cohort of sixty OC patients was enrolled. OC cells were exposed to both normoxic and hypoxic conditions. Expression levels of PFKP and phosphatase and tensin homolog (PTEN) were quantified using real time quantitative polymerase chain reaction (RT-qPCR) and Western blot analyses. Immunofluorescence confirmed these protein expression patterns. Glycolysis-related parameters, encompassing glucose uptake, extracellular lactate levels, extracellular acidification rates, and oxygen consumption rates, were assessed using commercially available kits. Lactylation status of PFKP was evaluated via immunoprecipitation followed by Western blot analysis. An OC xenograft mouse model was also established. Findings indicated elevated PFKP expression in OC tissues and cells. Additionally, PFKP knockdown attenuated glycolysis and counteracted the hypoxia-induced enhancement of glycolytic activity in OC cells. Mutation of the lysine (K) residue at position 392 diminished PFKP lactylation. Further investigations revealed that PFKP depletion upregulated PTEN expression in hypoxia-treated OC cells. Besides, PTEN suppression increased the glycolysis in hypoxia-treated OC cells. Animal study results demonstrated that PFKP inhibition curtailed OC tumor growth by modulating PTEN expression. Collectively, these results suggested that lactylation of PFKP at the K392 residue promoted glycolysis in OC cells by regulating PTEN, thereby facilitating the disease's progression.
Collapse
Affiliation(s)
- Jianfeng Mi
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, No.6 Shaungyong Road, Nanning, 532200, China
| | - Ling Zhao
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, No.6 Shaungyong Road, Nanning, 532200, China
| | - Yonglong Shen
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, No.6 Shaungyong Road, Nanning, 532200, China
| | - Shien Mo
- Department of Gynecology, The Sec People's Hospital of Qinzhou, Qinzhou, China
| | - Yan Kuang
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, No.6 Shaungyong Road, Nanning, 532200, China.
| |
Collapse
|
3
|
Hua T, Zhang G, Yao Y, Jia H, Liu W. Research progress of megakaryocytes and platelets in lung injury. Ann Med 2024; 56:2362871. [PMID: 38902986 PMCID: PMC11195464 DOI: 10.1080/07853890.2024.2362871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 05/17/2024] [Indexed: 06/22/2024] Open
Abstract
The lung is an important site of extramedullary platelet formation, and megakaryocytes in the lung participate in immune responses in addition to platelet production. In acute lung injury and chronic lung injury, megakaryocytes and platelets play a promoting or protective role through different mechanisms. The authors reviewed the role of megakaryocytes and platelets in common clinical lung injuries with different course of disease and different pathogenic factors in order to provide new thinking for the diagnosis and treatment of lung injuries.
Collapse
Affiliation(s)
- Tianzhen Hua
- Department of Burns and Plastic Surgery, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Guangliang Zhang
- Department of Burns and Plastic Surgery, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Yi Yao
- Department of Burns and Plastic Surgery, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Haoran Jia
- Department of Burns and Plastic Surgery, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Wei Liu
- Department of Burns and Plastic Surgery, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
4
|
Yan F, Yuan WQ, Wu SM, Yang YH, Cui DJ. Novel mechanisms of intestinal flora regulation in high-altitude hypoxia. Heliyon 2024; 10:e38220. [PMID: 39498080 PMCID: PMC11534185 DOI: 10.1016/j.heliyon.2024.e38220] [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: 03/05/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 11/07/2024] Open
Abstract
Background This study investigates the molecular mechanisms behind firmicutes-mediated macrophage (Mψ) polarization and glycolytic metabolic reprogramming through HIF-1α in response to intrinsic mucosal barrier injury induced by high-altitude hypoxia. Methods Establishing a hypoxia mouse model of high altitude, we utilized single-cell transcriptome sequencing to identify key cell types involved in regulating intestinal mucosal barrier damage caused by high-altitude hypoxia. Through proteomic analysis of colonic tissue Mψ and metabolomic analysis of Mψ metabolites, we determined crucial proteins and metabolic pathways influencing intestinal mucosal barrier damage induced by high-altitude hypoxia. Mechanistic validation was conducted using RAW264.7 Mψ in vitro by assessing cell viability with CCK-8 assay following treatment with different metabolites. The hypoxia mouse model was further validated in vivo by transplanting gut microbiota of Firmicutes. Histological examinations through H&E staining assessed colonic cell morphology and structure, while the FITC-dextran assay evaluated intestinal tissue permeability. Hypoxia probe signal intensity in mouse colonic tissue was assessed via metronidazole staining. Various experimental techniques, including flow cytometry, immunofluorescence, ELISA, Western blot, and RT-qPCR, were employed to study the impact of HIF-1α/glycolysis pathway and different gut microbiota metabolites on Mψ polarization. Results Bioinformatics analysis revealed that single-cell transcriptomics identified Mψ as a key cell type, with their polarization pattern playing a crucial role in the intestinal mucosal barrier damage induced by high-altitude hypoxia. Proteomics combined with metabolomics analysis indicated that HIF-1α and the glycolytic pathway are pivotal proteins and signaling pathways in the intestinal mucosal barrier damage caused by high-altitude hypoxia. In vitro cell experiments demonstrated that activation of the glycolytic pathway by HIF-1α led to a significant upregulation of mRNA levels of IL-1β, IL-6, and TNFα while downregulating mRNA levels of IL-10 and TGFβ, thereby promoting M1 Mψ activation and inhibiting M2 Mψ polarization. Further mechanistic validation experiments revealed that the metabolite butyric acid from Firmicutes bacteria significantly downregulated the protein expression of HIF-1α, GCK, PFK, PKM, and LDH, thus inhibiting the HIF-1α/glycolytic pathway that suppresses M1 Mψ and activates M2 Mψ, consequently alleviating the hypoxic symptoms in RAW264.7 cells. Subsequent animal experiments confirmed that Firmicutes bacteria inhibited the HIF-1α/glycolytic pathway to modulate Mψ polarization, thereby mitigating intestinal mucosal barrier damage in high-altitude hypoxic mice. Conclusion The study reveals that firmicutes, through the inhibition of the HIF-1α/glycolysis pathway, mitigate Mψ polarization, thereby alleviating intrinsic mucosal barrier injury in high-altitude hypoxia.
Collapse
Affiliation(s)
- Fang Yan
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Medical College of Guizhou University, Guiyang, Guizhou, China
| | - Wen-qiang Yuan
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Medical College of Guizhou University, Guiyang, Guizhou, China
| | - Shi-min Wu
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Zunyi Medical University, Zunyi, 563006, China
| | - Yun-han Yang
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Medical College of Guizhou University, Guiyang, Guizhou, China
| | - De-jun Cui
- Department of Gastroenterology, National Institution of Drug Clinical Trial, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
- Medical College of Guizhou University, Guiyang, Guizhou, China
| |
Collapse
|
5
|
Yang X, Li J, Ma Y, Dong X, Qu J, Liang F, Liu J. Curcumin-mediated enhancement of lung barrier function in rats with high-altitude-associated acute lung injury via inhibition of inflammatory response. Respir Res 2024; 25:354. [PMID: 39342264 PMCID: PMC11439224 DOI: 10.1186/s12931-024-02975-z] [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: 06/20/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Exposure to a hypobaric hypoxic environment at high altitudes can lead to lung injury. In this study, we aimed to determine whether curcumin (Cur) could improve lung barrier function and protect against high-altitude-associated acute lung injury. METHODS Two hundred healthy rats were randomly divided into standard control, high-altitude control (HC), salidroside (40 mg/kg, positive control), and Cur (200 mg/kg) groups. Each group was further divided into five subgroups. Basic vital signs, lung injury histopathology, routine blood parameters, plasma lactate level, and arterial blood gas indicators were evaluated. Protein and inflammatory factor (tumor necrosis factor α (TNF-α), interleukin [IL]-1β, IL-6, and IL-10) concentrations in bronchoalveolar lavage fluid (BALF) were determined using the bicinchoninic acid method and enzyme-linked immunosorbent assay, respectively. Inflammation-related and lung barrier function-related proteins were analyzed using immunoblotting. RESULTS Cur improved blood routine indicators such as hemoglobin and hematocrit and reduced the BALF protein content and TNF-α, IL-1β, and IL-6 levels compared with those in the HC group. It increased IL-10 levels and reduced pulmonary capillary congestion, alveolar hemorrhage, and the degree of pulmonary interstitial edema. It increased oxygen partial pressure, oxygen saturation, carbonic acid hydrogen radical, and base excess levels, and the expression of zonula occludens 1, occludin, claudin-4, and reduced carbon dioxide partial pressure, plasma lactic acid, and the expression of phospho-nuclear factor kappa. CONCLUSIONS Exposure to a high-altitude environment for 48 h resulted in severe lung injury in rats. Cur improved lung barrier function and alleviated acute lung injury in rats at high altitudes.
Collapse
Affiliation(s)
- Xinyue Yang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
- Graduate School , Xinjiang Medical University, Urumqi, 830000, China
| | - Jiajia Li
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830000, China
| | - Yan Ma
- Department of Anesthesiology, Xinjiang Medical University Affiliated First Hospital, Urumqi, 830054, China
| | - Xiang Dong
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
| | - Jinquan Qu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
| | - Feixing Liang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China.
| |
Collapse
|
6
|
Yang W, Lei X, Liu F, Sui X, Yang Y, Xiao Z, Cui Z, Sun Y, Yang J, Yang X, Lin X, Bao Z, Li W, Ma Y, Wang Y, Luo Y. Meldonium, as a potential neuroprotective agent, promotes neuronal survival by protecting mitochondria in cerebral ischemia-reperfusion injury. J Transl Med 2024; 22:771. [PMID: 39148053 PMCID: PMC11325598 DOI: 10.1186/s12967-024-05222-7] [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: 01/08/2024] [Accepted: 04/19/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Stroke is a globally dangerous disease capable of causing irreversible neuronal damage with limited therapeutic options. Meldonium, an inhibitor of carnitine-dependent metabolism, is considered an anti-ischemic drug. However, the mechanisms through which meldonium improves ischemic injury and its potential to protect neurons remain largely unknown. METHODS A rat model with middle cerebral artery occlusion (MCAO) was used to investigate meldonium's neuroprotective efficacy in vivo. Infarct volume, neurological deficit score, histopathology, neuronal apoptosis, motor function, morphological alteration and antioxidant capacity were explored via 2,3,5-Triphenyltetrazolium chloride staining, Longa scoring method, hematoxylin and eosin staining, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay, rotarod test, transmission electron microscopy and Oxidative stress index related kit. A primary rat hippocampal neuron model subjected to oxygen-glucose deprivation reperfusion was used to study meldonium's protective ability in vitro. Neuronal viability, mitochondrial membrane potential, mitochondrial morphology, respiratory function, ATP production, and its potential mechanism were assayed by MTT cell proliferation and cytotoxicity assay kit, cell-permeant MitoTracker® probes, mitochondrial stress, real-time ATP rate and western blotting. RESULTS Meldonium markedly reduced the infarct size, improved neurological function and motor ability, and inhibited neuronal apoptosis in vivo. Meldonium enhanced the morphology, antioxidant capacity, and ATP production of mitochondria and inhibited the opening of the mitochondrial permeability transition pore in the cerebral cortex and hippocampus during cerebral ischemia-reperfusion injury (CIRI) in rats. Additionally, meldonium improved the damaged fusion process and respiratory function of neuronal mitochondria in vitro. Further investigation revealed that meldonium activated the Akt/GSK-3β signaling pathway to inhibit mitochondria-dependent neuronal apoptosis. CONCLUSION Our study demonstrated that meldonium shows a neuroprotective function during CIRI by preserving the mitochondrial function, thus prevented neurons from apoptosis.
Collapse
Affiliation(s)
- Weijie Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiuxing Lei
- Lu'An Hospital of Traditional Chinese Medicine, Anhui, China
| | - Fengying Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xin Sui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yi Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhenyu Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ziqi Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yangyang Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jun Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xinyi Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xueyang Lin
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhenghao Bao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Weidong Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yingkai Ma
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| |
Collapse
|
7
|
Liu F, He H, Yang W, Wang D, Sui X, Sun Y, Wang S, Yang Y, Xiao Z, Yang J, Wang Y, Luo Y. Novel energy optimizer, meldonium, rapidly restores acute hypobaric hypoxia-induced brain injury by targeting phosphoglycerate kinase 1. Cell Commun Signal 2024; 22:383. [PMID: 39075489 PMCID: PMC11285322 DOI: 10.1186/s12964-024-01757-w] [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: 02/28/2024] [Accepted: 07/19/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Acute hypobaric hypoxia-induced brain injury has been a challenge in the health management of mountaineers; therefore, new neuroprotective agents are urgently required. Meldonium, a well-known cardioprotective drug, has been reported to have neuroprotective effects. However, the relevant mechanisms have not been elucidated. We hypothesized that meldonium may play a potentially novel role in hypobaric hypoxia cerebral injury. METHODS We initially evaluated the neuroprotection efficacy of meldonium against acute hypoxia in mice and primary hippocampal neurons. The potential molecular targets of meldonium were screened using drug-target binding Huprot™ microarray chip and mass spectrometry analyses after which they were validated with surface plasmon resonance (SPR), molecular docking, and pull-down assay. The functional effects of such binding were explored through gene knockdown and overexpression. RESULTS The study clearly shows that pretreatment with meldonium rapidly attenuates neuronal pathological damage, cerebral blood flow changes, and mitochondrial damage and its cascade response to oxidative stress injury, thereby improving survival rates in mice brain and primary hippocampal neurons, revealing the remarkable pharmacological efficacy of meldonium in acute high-altitude brain injury. On the one hand, we confirmed that meldonium directly interacts with phosphoglycerate kinase 1 (PGK1) to promote its activity, which improved glycolysis and pyruvate metabolism to promote ATP production. On the other hand, meldonium also ameliorates mitochondrial damage by PGK1 translocating to mitochondria under acute hypoxia to regulate the activity of TNF receptor-associated protein 1 (TRAP1) molecular chaperones. CONCLUSION These results further explain the mechanism of meldonium as an energy optimizer and provide a strategy for preventing acute hypobaric hypoxia brain injury at high altitudes.
Collapse
Affiliation(s)
- Fengying Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Huanhuan He
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Weijie Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Daohui Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xin Sui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yangyang Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Shuai Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yi Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Zhenyu Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jun Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| |
Collapse
|
8
|
Mao Z, Wang C, Liu J, Li X, Duan H, Ye Y, Liu H, Lv L, Xue G, He Z, Wuren T, Wang H. Superoxide dismutase 1-modified dental pulp stem cells alleviate high-altitude pulmonary edema by inhibiting oxidative stress through the Nrf2/HO-1 pathway. Gene Ther 2024; 31:422-433. [PMID: 38834681 DOI: 10.1038/s41434-024-00457-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
High-altitude pulmonary edema (HAPE) is a deadly form of altitude sickness, and there is no effective treatment for HAPE. Dental pulp stem cells (DPSCs) are a type of mesenchymal stem cell isolated from dental pulp tissues and possess various functions, such as anti-inflammatory and anti-oxidative stress. DPSCs have been used to treat a variety of diseases, but there are no studies on treating HAPE. In this study, Sprague-Dawley rats were exposed to acute low-pressure hypoxia to establish the HAPE model, and SOD1-modified DPSCs (DPSCsHiSOD1) were administered through the tail vein. Pulmonary arterial pressure, lung water content (LWC), total lung protein content of bronchoalveolar lavage fluid (BALF) and lung homogenates, oxidative stress, and inflammatory indicators were detected to evaluate the effects of DPSCsHiSOD1 on HAPE. Rat type II alveolar epithelial cells (RLE-6TN) were used to investigate the effects and mechanism of DPSCsHiSOD1 on hypoxia injury. We found that DPSCs could treat HAPE, and the effect was better than that of dexamethasone treatment. SOD1 modification could enhance the function of DPSCs in improving the structure of lung tissue, decreasing pulmonary arterial pressure and LWC, and reducing the total lung protein content of BALF and lung homogenates, through anti-oxidative stress and anti-inflammatory effects. Furthermore, we found that DPSCsHiSOD1 could protect RLE-6TN from hypoxic injury by reducing the accumulation of reactive oxygen species (ROS) and activating the Nrf2/HO-1 pathway. Our findings confirm that SOD1 modification could enhance the anti-oxidative stress ability of DPSCs through the Nrf2/HO-1 signalling pathway. DPSCs, especially DPSCsHiSOD1, could be a potential treatment for HAPE. Schematic diagram of the antioxidant stress mechanism of DPSCs in the treatment of high-altitude pulmonary edema. DPSCs can alleviate oxidative stress by releasing superoxide dismutase 1, thereby reducing ROS production and activating the Nrf2/HO-1 signalling pathway to ameliorate lung cell injury in HAPE.
Collapse
Affiliation(s)
- Zhuang Mao
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Changyao Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
- School of Life Sciences, Hebei University, Baoding, 071002, China
| | - Juanli Liu
- Key Laboratory for Application of High-Altitude Medicine, Qinghai University, Xi'ning, 810008, China
- Research Center for High Altitude Medicine, Qinghai University, Xi'ning, 810008, China
- Department of Critical Care Medicine, Qinghai Provincial People's Hospital, Xi'ning, 810007, China
| | - Xue Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
- College of Life Science, Anhui Medical University, Hefei, 230032, China
| | - Han Duan
- School of Life Sciences, Hebei University, Baoding, 071002, China
| | - Yi Ye
- Key Laboratory for Application of High-Altitude Medicine, Qinghai University, Xi'ning, 810008, China
- Research Center for High Altitude Medicine, Qinghai University, Xi'ning, 810008, China
| | - Huifang Liu
- Key Laboratory for Application of High-Altitude Medicine, Qinghai University, Xi'ning, 810008, China
- Research Center for High Altitude Medicine, Qinghai University, Xi'ning, 810008, China
| | - Lin Lv
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Guanzhen Xue
- Key Laboratory for Application of High-Altitude Medicine, Qinghai University, Xi'ning, 810008, China
- Research Center for High Altitude Medicine, Qinghai University, Xi'ning, 810008, China
| | - Zhichao He
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
- College of Life Science, Anhui Medical University, Hefei, 230032, China
| | - Tana Wuren
- Key Laboratory for Application of High-Altitude Medicine, Qinghai University, Xi'ning, 810008, China.
- Research Center for High Altitude Medicine, Qinghai University, Xi'ning, 810008, China.
| | - Hua Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- School of Life Sciences, Hebei University, Baoding, 071002, China.
- College of Life Science, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
9
|
Pușcaș A, Buț MG, Vari CE, Ősz BE, Ștefănescu R, Filip C, Jîtcă G, Istrate TI, Tero-Vescan A. Meldonium Supplementation in Professional Athletes: Career Destroyer or Lifesaver? Cureus 2024; 16:e63634. [PMID: 39092347 PMCID: PMC11292090 DOI: 10.7759/cureus.63634] [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] [Accepted: 06/29/2024] [Indexed: 08/04/2024] Open
Abstract
Meldonium is a substance with known anti-anginal effects demonstrated by numerous studies and human clinical trials; however, it does not possess marketing authorization within the European Union, only in ex-Soviet republics. Since 2016, meldonium has been included by the World Anti-doping Agency (WADA) on the S4 list of metabolic modulators. In performance athletes, meldonium is now considered a doping agent due to its capacity to decrease lactate production during and after exercise, its capability to enhance the storage and utilization of glycogen, and its protective action against oxidative stress. Together, these attributes can significantly improve aerobic endurance, cardiac function, and capacity as well as shorten recovery times (allowing higher intensity training), thereby enhancing performance. The purpose of this review is to highlight the most important mechanisms underlying the protective effect of meldonium against mitochondrial dysfunction (MD), which is responsible for oxidative stress, inflammation, and the cardiac changes known as "athletic heart syndrome." Meldonium acts as an inhibitor of γ-butyrobetaine hydroxylase (BBOX), preventing the de novo synthesis of carnitine and its absorption at the intestinal level via the organic cation/carnitine transporter 2 (OCTN2) and directing the oxidation of fatty acids to the peroxisomes. The decrease in mitochondrial β-oxidation of fatty acids leads to a reduction in lipid peroxidation products that cause oxidative stress and prevent the formation of acyl/acetyl-carnitines involved in numerous pathological disorders. Given the recent findings of the potentially detrimental effects of prolonged high-intensity exercise on cardiovascular health and coronary atherosclerosis, there may be legitimate arguments for the justification of the use of substances like meldonium as protective supplements for athletes.
Collapse
Affiliation(s)
- Amalia Pușcaș
- Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| | - Mădălina-Georgiana Buț
- Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| | - Camil-Eugen Vari
- Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| | - Bianca-Eugenia Ősz
- Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| | - Ruxandra Ștefănescu
- Pharmacognosy and Phytotherapy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| | - Cristina Filip
- Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| | - George Jîtcă
- Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| | - Tudor-Ionuț Istrate
- Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| | - Amelia Tero-Vescan
- Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
| |
Collapse
|
10
|
Kamińska D, Skrzycki M. Lipid droplets, autophagy, and ER stress as key (survival) pathways during ischemia-reperfusion of transplanted grafts. Cell Biol Int 2024; 48:253-279. [PMID: 38178581 DOI: 10.1002/cbin.12114] [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: 07/13/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 01/06/2024]
Abstract
Ischemia-reperfusion injury is an event concerning any organ under a procedure of transplantation. The early result of ischemia is hypoxia, which causes malfunction of mitochondria and decrease in cellular ATP. This leads to disruption of cellular metabolism. Reperfusion also results in cell damage due to reoxygenation and increased production of reactive oxygen species, and later by induced inflammation. In damaged and hypoxic cells, the endoplasmic reticulum (ER) stress pathway is activated by increased amount of damaged or misfolded proteins, accumulation of free fatty acids and other lipids due to inability of their oxidation (lipotoxicity). ER stress is an adaptive response and a survival pathway, however, its prolonged activity eventually lead to induction of apoptosis. Sustaining cell functionality in stress conditions is a great challenge for transplant surgeons as it is crucial for maintaining a desired level of graft vitality. Pathways counteracting negative consequences of ischemia-reperfusion are autophagy and lipid droplets (LD) metabolism. Autophagy remove damaged organelles and molecules driving them to lysosomes, digested simpler compounds are energy source for the cell. Mitophagy and ER-phagy results in improvement of cell energetic balance and alleviation of ER stress. This is important in sustaining metabolic homeostasis and thus cell survival. LD metabolism is connected with autophagy as LD are degraded by lipophagy, a source of free fatty acids and glycerol-thus autophagy and LD can readily remove lipotoxic compounds in the cell. In conclusion, monitoring and pharmaceutic regulation of those pathways during transplantation procedure might result in increased/improved vitality of transplanted organ.
Collapse
Affiliation(s)
- Daria Kamińska
- Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, Poland
| | - Michał Skrzycki
- Chair and Department of Biochemistry, Medical University of Warsaw, Warszawa, Poland
| |
Collapse
|
11
|
Tian L, Jia Z, Yan Y, Jia Q, Shi W, Cui S, Chen H, Han Y, Zhao X, He K. Low-dose of caffeine alleviates high altitude pulmonary edema via regulating mitochondrial quality control process in AT1 cells. Front Pharmacol 2023; 14:1155414. [PMID: 37081967 PMCID: PMC10110878 DOI: 10.3389/fphar.2023.1155414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Backgrounds: High-altitude pulmonary edema (HAPE) is a life-threatening disease without effective drugs. Caffeine is a small molecule compound with antioxidant biological activity used to treat respiratory distress syndrome. However, it is unclear whether caffeine plays a role in alleviating HAPE.Methods: We combined a series of biological experiments and label-free quantitative proteomics analysis to detect the effect of caffeine on treating HAPE and explore its mechanism in vivo and in vitro.Results: Dry and wet weight ratio and HE staining of pulmonary tissues showed that the HAPE model was constructed successfully, and caffeine relieved pulmonary edema. The proteomic results of mice lungs indicated that regulating mitochondria might be the mechanism by which caffeine reduced HAPE. We found that caffeine blocked the reduction of ATP production and oxygen consumption rate, decreased ROS accumulation, and stabilized mitochondrial membrane potential to protect AT1 cells from oxidative stress damage under hypoxia. Caffeine promoted the PINK1/parkin-dependent mitophagy and enhanced mitochondrial fission to maintain the mitochondria quality control process.Conclusion: Low-dose of caffeine alleviated HAPE by promoting PINK1/parkin-dependent mitophagy and mitochondrial fission to control the mitochondria quality. Therefore, caffeine could be a potential treatment for HAPE.
Collapse
Affiliation(s)
- Liuyang Tian
- School of Medicine, Nankai University, Tianjin, China
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
| | - Zhilong Jia
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Center for Artificial Intelligence in Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| | - Yan Yan
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Qian Jia
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Wenjie Shi
- Technical Research Centre for Prevention and Control of Birth Defects, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Saijia Cui
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Huining Chen
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Yang Han
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Xiaojing Zhao
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- Research Center for Translational Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| | - Kunlun He
- School of Medicine, Nankai University, Tianjin, China
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- National Engineering Research Center for Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhilong Jia, ; Xiaojing Zhao, ; Kunlun He,
| |
Collapse
|
12
|
Liu F, Sui X, Wang Q, Li J, Yang W, Yang Y, Xiao Z, Sun Y, Guo X, Yang X, Yang J, Wang Y, Luo Y. Insights into the pharmacodynamics and pharmacokinetics of meldonium after exposure to acute high altitude. Front Pharmacol 2023; 14:1119046. [PMID: 36909160 PMCID: PMC9992410 DOI: 10.3389/fphar.2023.1119046] [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: 12/08/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Objective: Meldonium, a well-known cardioprotective drug, has been reported to be protective against pulmonary injury at high altitudes; however, the pharmacodynamics of meldonium in other vital organs under acute high-altitude injury are less investigated and the related pharmacokinetics have not been fully elucidated. Methods and Results: The present study examined the basic pharmacodynamics and pharmacokinetics (PK) in rat exposure to acute high-altitude hypoxia after intragastrical and intravenous pre-administration of meldonium. The results indicate that meldonium can improve acute hypoxia-induced pathological damage in brain and lung tissues, and restore blood biochemistry and routine blood index of heart, liver and kidney tissues under a simulated acute high-altitude environment. Furthermore, compared to the normoxia group, rats exposed to simulated high-altitude hypoxia and premedicated with intragastrical meldonium showed linear kinetics in the dose range of 25-100 mg/kg, with a significantly increase in the area under curve (AUC) and reduced clearance rate. No significant differences in these meldonium of PK parameters were observed with intravenous administration. Additionally, meldonium was involved in the regulation of succinic acid and 3-hydroxypropionic acid. Conclusion: These results will contribute to our understanding of the preclinical PK properties of meldonium and its acute high-altitude protective effects.
Collapse
Affiliation(s)
- Fengying Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xin Sui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Qian Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jinglai Li
- Guollence Pharmaceutical Technology Co., Ltd., Beijing, China
| | - Weijie Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yi Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhenyu Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yangyang Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaoxuan Guo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,School of Pharmaceutical Science, Liaoning University, Shenyang, China
| | - Xinyi Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jun Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
13
|
Zhuravleva MV, Granovskaya MV, Zaslavskaya KY, Kazaishvili YG, Scherbakova VS, Andreev-Andrievskiy AA, Pozdnyakov DI, Vyssokikh MY. SYNERGIC EFFECT OF PREPARATION WITH COORDINATION COMPLEX “TRIMETHYDRAZINIUM PROPIONATE+ETHYMTH METHYLHYDROXYPIRIDINE SUCCINATE” ON ENERGY METABOLISM AND CELL RESPIRATION. PHARMACY & PHARMACOLOGY 2022. [DOI: 10.19163/2307-9266-2022-10-4-387-399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The article presents the results of an in vitro study of the synergetic effect evaluation of the combined preparation based on coordination complex ethylmethylhydroxypyridine succinate and trimethylhydrazinium propionate on energy metabolism and cell respiration.The aim of the study was to evaluate the mitochondria-directed action of the metabolic and antioxidant preparation based on succinic acid coordination complex with trimethylhydrazinium in relation to optimizing the energy metabolism in the cells under the oxidative stress conditions, as well as against the background of ischemic processes.Materials and methods. The study of the hydroxysuccinate complex effect of the drug Brainmax® components was carried out on isolated mouse liver mitochondria. In the course of the study, the potential of mitochondria, the generation rate of hydrogen peroxide during the respiration, the respiration rate were evaluated in the following positions: a) unstimulated by malate and pyruvate, b) stimulated by malate and pyruvate (complex I substrates), by succinate (complex II substrates), c) against the background of the initial section of the electron transport chain blockade by rotenone, d) in phosphorylation blockade by oligomycin, e) against the background of the FCCP-induced uncoupling, and f) in cyanide-blocked complex IV (cytochrome C oxidase).Results. It has been shown that the succinic acid coordination complex with trimethylhydrazinium, which is the active principle of the Brainmax® drug, significantly reduced the transmembrane potential of mitochondria (IC50=197±5 µM), compared with the widely used preparations of ethylmethylhydroxypyridine succinate and trimethylhydrazinium propionate, which facilitates the transfer of the produced ATP into the cell and preserves a vital activity of mitochondria even under stress. In the study of the mitochondrial respiration stimulated by the substrates of complex I (NADP-coenzyme Q-oxidoreductase), pyruvate and malate, the studied drug led to a more pronounced increase in the oxygen consumption with IC50=75±6 µМ. When evaluating the effect of the complex on the production of ATP by mitochondria, the most pronounced effect was observed with the addition of studied complex, which indicated to the uncoupling of respiration and oxidative phosphorylation at the given concentrations of the studied compounds. When assessing the effect of the complex on the production of hydrogen peroxide by isolated mitochondria, a significant decrease in the peroxide production was shown in the samples containing the complex of trimethylhydrazinium propionate and EMHPS.Conclusion. Based on totality of the results obtained, it can be assumed that a favorable conformation of the pharmacophore groups of ethylmethylhydroxypyridine succinate and trimethylhydrozinium propionate coordination complex included in the composition of Brainmax® leads to a synergetic interaction and more pronounced pharmacological effects on target cells. This complex provides stabilization of a mitochondrial function, intensification of the adenosine triphosphate energy production and the optimization of energy processes in the cell, reduces the severity of the oxidative stress and eliminates undesirable effects of an ischemic-hypoxic tissue damage.
Collapse
Affiliation(s)
- M. V. Zhuravleva
- Scientific Center for Expertise of Medicinal Products
Sechenov First Moscow State Medical University (Sechenov University)
| | | | | | | | | | | | - D. I. Pozdnyakov
- Pyatigorsk Medical and Pharmaceutical Institute – branch of Volgograd State Medical University
| | - M. Yu. Vyssokikh
- A.N. Belozersky Institute of Physico-Chemical Biology of Moscow State University named after M.V. Lomonosov
| |
Collapse
|