1
|
Tian J, Ke X, Zhang Y, Qu J, Fu S, Xia Y, Yang W, Zeng Y, Fan J, Li Y, Fan B. Safety evaluation of alpha-glycerylphosphorylcholine as a novel food. Food Chem Toxicol 2025; 195:115123. [PMID: 39577616 DOI: 10.1016/j.fct.2024.115123] [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: 08/16/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
To evaluate the safety of alpha-glycerylphosphorylcholine (α-GPC) as a novel food, the study of acute oral toxicity, subchronic toxicity, teratogenic toxicity and genotoxicity were conducted. In acute oral toxicity, no toxic effects were observed in rats of both genders administrated 10.0 g/kg BW α-GPC. In 90-day oral toxicity, female high-dose group (2,000 mg/kg) had lower body weight, body weight gain, empty stomach body weight, total protein (TP), albumin (ALB), and higher alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) in contrast to control group. In teratogenic toxicity, the body weights of pregnant rats on the 9th day (d9), the 12th day (d12), the 15th day (d15), and the 20th day (d20), body weight gains, and net body weight gains in high-dose group (2,000 mg/kg) decreased, the other parameters had no difference compared to control group. In genotoxicity tests (Mammalian erythrocyte micronucleus, Chromosome aberration and Ames test), all dose groups didn't display significant change compared with negative control group. Based on above results, α-GPC is actually low hazard novel food, has a NOAEL of 1,000 mg/kg BW for female rats and 2,000 mg/kg BW for male rats following 13-week oral exposure, has a NOAEL of 1,000 mg/kg BW for pregnant rats and 2,000 mg/kg BW for fetal rats in teratogenic toxicity, has no genotoxicity in vitro or in vivo.
Collapse
Affiliation(s)
- Jie Tian
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Xianghong Ke
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Yinjing Zhang
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Jingjing Qu
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Shaohua Fu
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Ying Xia
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Wenxiang Yang
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Yanhua Zeng
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Jun Fan
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China
| | - Yanmei Li
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China.
| | - Bolin Fan
- Hubei Provincial Center for Disease Control and Prevention & NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Wuhan, 430079, China.
| |
Collapse
|
2
|
Lee HJ, Cho HR, Bang M, Lee YS, Kim YJ, Chong K. Potential Risk of Choline Alfoscerate on Isoflurane-Induced Toxicity in Primary Human Astrocytes. J Korean Neurosurg Soc 2024; 67:418-430. [PMID: 37859347 PMCID: PMC11220420 DOI: 10.3340/jkns.2023.0208] [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: 10/05/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVE Isoflurane, a widely used common inhalational anesthetic agent, can induce brain toxicity. The challenge lies in protecting neurologically compromised patients from neurotoxic anesthetics. Choline alfoscerate (L-α-Glycerophosphorylcholine, α-GPC) is recognized for its neuroprotective properties against oxidative stress and inflammation, but its optimal therapeutic window and indications are still under investigation. This study explores the impact of α-GPC on human astrocytes, the most abundant cells in the brain that protect against oxidative stress, under isoflurane exposure. METHODS This study was designed to examine changes in factors related to isoflurane-induced toxicity following α-GPC administration. Primary human astrocytes were pretreated with varying doses of α-GPC (ranging from 0.1 to 10.0 μM) for 24 hours prior to 2.5% isoflurane exposure. In vitro analysis of cell morphology, water-soluble tetrazolium salt-1 assay, quantitative real-time polymerase chain reaction, proteome profiler array, and transcriptome sequencing were conducted. RESULTS A significant morphological damage to human astrocytes was observed in the group that had been pretreated with 10.0 mM of α-GPC and exposed to 2.5% isoflurane. A decrease in cell viability was identified in the group pretreated with 10.0 μM of α-GPC and exposed to 2.5% isoflurane compared to the group exposed only to 2.5% isoflurane. Quantitative real-time polymerase chain reaction revealed that mRNA expression of heme-oxygenase 1 and hypoxia-inducible factor-1α, which were reduced by isoflurane, was further suppressed by 10.0 μM α-GPC pretreatment. The proteome profiler array demonstrated that α-GPC pretreatment influenced a variety of factors associated with apoptosis induced by oxidative stress. Additionally, transcriptome sequencing identified pathways significantly related to changes in isoflurane-induced toxicity caused by α-GPC pretreatment. CONCLUSION The findings suggest that α-GPC pretreatment could potentially enhance the vulnerability of primary human astrocytes to isoflurane-induced toxicity by diminishing the expression of antioxidant factors, potentially leading to amplified cell damage.
Collapse
Affiliation(s)
- Hyun Jung Lee
- Department of Anesthesiology and Pain Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Hye Rim Cho
- Department of Neurosurgery, Korea University Medicine, Korea University College of Medicine, Seoul, Korea
| | - Minji Bang
- Photo-Theranosis and Bioinformatics for Tumor Laboratory, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeo Song Lee
- Department of Neurosurgery, Korea University Medicine, Korea University College of Medicine, Seoul, Korea
| | - Youn Jin Kim
- Department of Anesthesiology and Pain Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Kyuha Chong
- Photo-Theranosis and Bioinformatics for Tumor Laboratory, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Yang Z, Chen S, Sun W, Yang Y, Xu Y, Tang Y, Jiang W, Li J, Zhang Y. Study on the mechanisms by which pumpkin polysaccharides regulate abnormal glucose and lipid metabolism in diabetic mice under oxidative stress. Int J Biol Macromol 2024; 270:132249. [PMID: 38729500 DOI: 10.1016/j.ijbiomac.2024.132249] [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: 11/04/2023] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Pumpkin polysaccharide (PPe-H) can perform physiological functions through its antioxidative and hypoglycemic effects; however, the mechanisms through which PPe-H regulates abnormal glucose and lipid metabolism caused by oxidative stress injury remain unclear. In the present study, streptozotocin was used to generate an acute diabetic mouse model, and the effects of PPe-H on glucose and lipid metabolism impaired by oxidative stress in diabetic mice were studied. PPe-H significantly reduced blood glucose levels and enhanced the oral glucose tolerance of diabetic mice under stress injury (p < 0.05). The analysis of liver antioxidant enzymes showed that PPe-H significantly enhanced the activities of SOD and CAT (p < 0.05), increased the GSH level, and decreased the level of MDA (p < 0.05). Transcriptomic and metabolomic analyses of the liver tissues of mice revealed characteristic differences in the genetic and metabolic levels of the samples, which showed that PPe-H treatment may play a positive role in regulating the metabolism of methionine, cysteine, glycerol phospholipid, and linoleic acid. These results indicated that PPe-H alleviated the symptoms of hyperglycemia by regulating metabolites related to oxidative stress and glycolipid metabolism in diabetic mice.
Collapse
Affiliation(s)
- Zeen Yang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Shengdong Chen
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Wenxuan Sun
- Liang Xin College, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Yechen Yang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Yuxuan Xu
- Liang Xin College, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Yuxuan Tang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Wen Jiang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Jia Li
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Yongjun Zhang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China.
| |
Collapse
|
4
|
Hao H, Jia X, Ren T, Du Y, Wang J. Novel insight into the mechanism underlying synergistic cytotoxicity from two components in 5-Fluorouracil-phenylalanine co-crystal based on cell metabolomics. Eur J Pharm Biopharm 2022; 180:181-189. [DOI: 10.1016/j.ejpb.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/04/2022]
|
5
|
The Effects of Alpha-Glycerylphosphorylcholine on Heart Rate Variability and Hemodynamic Variables Following Sprint Interval Exercise in Overweight and Obese Women. Nutrients 2022; 14:nu14193970. [PMID: 36235623 PMCID: PMC9572742 DOI: 10.3390/nu14193970] [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: 08/19/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
The current study examined the effects of Alpha-Glycerylphosphorylcholine (A-GPC) on heart rate variability (HRV) and hemodynamic responses following a sprint interval exercise (SIE) in women who were overweight or obese. Participants (n = 12, 31.0 ± 4.6 years; 29.4 ± 2.1 kg/m2) consumed 1000 mg of A-GPC or a placebo after eating breakfast in a randomized, double-blind cross-over design. After 60 min, participants performed two bouts of the SIE (30 s Wingate) interspersed with 4 min of active recovery (40 rpm). Hemodynamic variables and HRV domains were measured before and 60 min after the A-GPC consumption, immediately after SIE, and every 15 min up to 120 min during recovery. A-GPC consumption increased resting levels of both the time domain (Standard Deviation of RR wave intervals [SDNN] and percentage of interval differences of adjacent RR intervals greater than 50 ms [pNN50%]) and frequency domain (high frequency [HF] and low frequency [LF]) variables of HRV (p < 0.05). Moreover, HRV variables (except for LF/HF) decreased (p < 0.05) immediately after SIE in the A-GPC and placebo sessions. Systolic and diastolic blood pressure increased (p < 0.05) immediately after SIE in both trials. Both HRV and hemodynamic variables recovered (p < 0.05) faster in the A-GPC compared to the placebo session. We concluded that A-GPC consumption recovers HRV and blood pressure faster following strenuous exercise in overweight and obese women, and that it might favorably modify cardiac autonomic function.
Collapse
|
6
|
NMR-Based Metabolomic Analysis of Cardiac Tissues Clarifies Molecular Mechanisms of CVB3-Induced Viral Myocarditis and Dilated Cardiomyopathy. Molecules 2022; 27:molecules27186115. [PMID: 36144851 PMCID: PMC9500976 DOI: 10.3390/molecules27186115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Viral myocarditis (VMC), which is defined as inflammation of the myocardium with consequent myocardial injury, may develop chronic disease eventually leading to dilated cardiomyopathy (DCM). Molecular mechanisms underlying the progression from acute VMC (aVMC), to chronic VMC (cVMC) and finally to DCM, are still unclear. Here, we established mouse models of VMC and DCM with Coxsackievirus B3 infection and conducted NMR-based metabolomic analysis of aqueous metabolites extracted from cardiac tissues of three histologically classified groups including aVMC, cVMC and DCM. We showed that these three pathological groups were metabolically distinct from their normal counterparts and identified three impaired metabolic pathways shared by these pathological groups relative to normal controls, including nicotinate and nicotinamide metabolism; alanine, aspartate and glutamate metabolism; and D-glutamine and D-glutamate metabolism. We also identified two extra impaired metabolic pathways in the aVMC group, including glycine, serine and threonine metabolism; and taurine and hypotaurine metabolism Furthermore, we identified potential cardiac biomarkers for metabolically distinguishing these three pathological stages from normal controls. Our results indicate that the metabolomic analysis of cardiac tissues can provide valuable insights into the molecular mechanisms underlying the progression from acute VMC to DCM.
Collapse
|
7
|
In-depth investigation of the mechanisms of Echinacea purpurea polysaccharide mitigating alcoholic liver injury in mice via gut microbiota informatics and liver metabolomics. Int J Biol Macromol 2022; 209:1327-1338. [PMID: 35461865 DOI: 10.1016/j.ijbiomac.2022.04.131] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/12/2022] [Accepted: 04/17/2022] [Indexed: 12/12/2022]
Abstract
Accumulating evidence suggests that the pathogenesis of alcoholic liver disease (ALD) is strongly correlated with abnormalities of the gut-liver axis. Echinacea purpurea polysaccharide (EPP) is a homogeneous polysaccharide, which has been shown to mitigate ALD. However, the effects of EPP on gut microbiome and consequently on hepatic metabolism have yet to be explored. In this study, the microbiome and metabolomics were combined to explore the effects of EPP on gut microbiota and hepatic metabolism, and the relationship between both was further revealed by Spearman correlation analysis. Results exhibited EPP reversed alcohol-induced disturbances in gut microbiota, evidenced by increased abundance of Muribaculaceae, Lactobacillus, and Bacteroides and decreased abundance of Escherichia_Shigella and Enterococcus. Besides, EPP promoted the production of n-butyric acid, a short-chain fatty acid that maintains the integrity of the intestinal barrier. Moreover, EPP improved alterations in hepatic metabolites, and characteristic metabolites such as Berberine and Ponasterone as well as key metabolic pathways, particularly Nitrogen metabolism, were identified. Furthermore, correlation analysis suggested significant associations between gut microbes and hepatic metabolites, which in turn confirmed EPP alleviated ALD via the gut-liver axis. Therefore, these findings elucidated in-depth mechanisms of EPP against ALD and provided a new target for intervention in alcohol-related diseases.
Collapse
|
8
|
Kong Q, Gu J, Lu R, Huang C, Hu X, Wu W, Lin D. NMR-Based Metabolomic Analysis of Sera in Mouse Models of CVB3-Induced Viral Myocarditis and Dilated Cardiomyopathy. Biomolecules 2022; 12:biom12010112. [PMID: 35053260 PMCID: PMC8773787 DOI: 10.3390/biom12010112] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 02/04/2023] Open
Abstract
Viral myocarditis (VMC) is an inflammatory heart condition which can induce dilated cardiomyopathy (DCM). However, molecular mechanisms underlying the progression of VMC into DCM remain exclusive. Here, we established mouse models of VMC and DCM by infecting male BALB/c mice with Coxsackievirus B3 (CVB3), and performed NMR-based metabonomic analyses of mouse sera. The mouse models covered three pathological stages including: acute VMC (aVMC), chronic VMC (cVMC) and DCM. We recorded 1D 1H-NMR spectra on serum samples and conducted multivariate statistical analysis on the NMR data. We found that metabolic profiles of these three pathological stages were distinct from their normal controls (CON), and identified significant metabolites primarily responsible for the metabolic distinctions. We identified significantly disturbed metabolic pathways in the aVMC, cVMC and DCM stages relative to CON, including: taurine and hypotaurine metabolism; pyruvate metabolism; glycine, serine and threonine metabolism; glycerolipid metabolism. Additionally, we identified potential biomarkers for discriminating a VMC, cVMC and DCM from CON including: taurine, valine and acetate for aVMC; glycerol, valine and leucine for cVMC; citrate, glycine and isoleucine for DCM. This work lays the basis for mechanistically understanding the progression from acute VMC to DCM, and is beneficial to exploitation of potential biomarkers for prognosis and diagnosis of heart diseases.
Collapse
Affiliation(s)
- Qing Kong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China;
| | - Jinping Gu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (J.G.); (R.L.); (X.H.)
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Ruohan Lu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (J.G.); (R.L.); (X.H.)
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361024, China;
| | - Xiaomin Hu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (J.G.); (R.L.); (X.H.)
| | - Weifeng Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China;
- Correspondence: (W.W.); (D.L.); Tel.: +86-771-5358955 (W.W.); +86-592-2186078 (D.L.)
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (J.G.); (R.L.); (X.H.)
- Correspondence: (W.W.); (D.L.); Tel.: +86-771-5358955 (W.W.); +86-592-2186078 (D.L.)
| |
Collapse
|
9
|
Burgaletto C, Di Benedetto G, Munafò A, Bernardini R, Cantarella G. Beneficial Effects of Choline Alphoscerate on Amyloid-β Neurotoxicity in an In vitro Model of Alzheimer's Disease. Curr Alzheimer Res 2021; 18:298-309. [PMID: 34102970 DOI: 10.2174/1567205018666210608093658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/17/2021] [Accepted: 04/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of neurodegenerative disorder characterized by cognitive impairment, which represents an urgent public health concern. Given the worldwide impact of AD, there is a compelling need for effective therapies to slow down or halt this disorder. OBJECTIVE Choline alphoscerate (α-GPC) represents a potentially effective cholinergic neurotrans- mission enhancing agent with an interesting clinical profile in cognitive dysfunctions improve- ment, although only scanty data are available about the mechanisms underlying such beneficial ef- fects. METHOD The SH-SY5Y neuronal cell line, differentiated for 1 week with 10 μm of all-trans-reti- noic acid (RA), to achieve a switch towards a cholinergic phenotype, was used as an in vitro model of AD. SH-SY5Y cells were pre-treated for 1h with α-GPC (100nM) and treated for 72 h with Aβ25-35 (10μM). RESULTS α-GPC was able to antagonize Aβ25-35 mediated neurotoxicity and attenuate the Aβ-in- duced phosphorylation of the Tau protein. Moreover, α-GPC exerted its beneficial effects by em- ploying the NGF/TrkA system, knocked down in AD and, consequently, by sustaining the expres- sion level of synaptic vesicle proteins, such as synaptophysin. CONCLUSION Taken together, our data suggest that α-GPC can have a role in neuroprotection in the course of toxic challenges with Aβ. Thus, a deeper understanding of the mechanism underlying its beneficial effect, could provide new insights into potential future pharmacological applications of its functional cholinergic enhancement, with the aim to mitigate AD and could represent the basis for innovative therapy.Recent Advances in Anti-Infective Drug Discovery.
Collapse
Affiliation(s)
- Chiara Burgaletto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Giulia Di Benedetto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Antonio Munafò
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| |
Collapse
|
10
|
Warmbrunn MV, Koopen AM, de Clercq NC, de Groot PF, Kootte RS, Bouter KEC, ter Horst KW, Hartstra AV, Serlie MJ, Ackermans MT, Soeters MR, van Raalte DH, Davids M, Nieuwdorp M, Groen AK. Metabolite Profile of Treatment-Naive Metabolic Syndrome Subjects in Relation to Cardiovascular Disease Risk. Metabolites 2021; 11:metabo11040236. [PMID: 33924347 PMCID: PMC8069178 DOI: 10.3390/metabo11040236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/02/2021] [Accepted: 04/10/2021] [Indexed: 12/26/2022] Open
Abstract
Metabolic syndrome (MetSyn) is an important risk factor for type 2 diabetes and cardiovascular diseases (CVD). This study aimed to find distinct plasma metabolite profiles between insulin-resistant and non-insulin resistant subjects with MetSyn and evaluate if MetSyn metabolite profiles are related to CVD risk and lipid fluxes. In a cross-sectional study, untargeted metabolomics of treatment-naive males with MetSyn (n = 132) were analyzed together with clinical parameters. In a subset of MetSyn participants, CVD risk was calculated using the Framingham score (n = 111), and lipolysis (n = 39) was measured by a two-step hyperinsulinemic euglycemic clamp using [1,1,2,3,3-2H5] glycerol to calculate lipolysis suppression rates. Peripheral insulin resistance was related to fatty acid metabolism and glycerolphosphorylcholine. Interestingly, although insulin resistance is considered to be a risk factor for CVD, we observed that there was little correspondence between metabolites associated with insulin resistance and metabolites associated with CVD risk. The latter mainly belonged to the androgenic steroid, fatty acid, phosphatidylethanolamine, and phophatidylcholine pathways. These data provide new insights into metabolic changes in mild MetSyn pathophysiology and MetSyn CVD risk related to lipid metabolism. Prospective studies may focus on the pathophysiological role of the here-identified biomarkers.
Collapse
Affiliation(s)
- Moritz V. Warmbrunn
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
- Correspondence: (M.V.W.); (A.K.G.)
| | - Annefleur M. Koopen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
| | - Nicolien C. de Clercq
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
| | - Pieter F. de Groot
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
| | - Ruud S. Kootte
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
| | - Kristien E. C. Bouter
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
| | - Kasper W. ter Horst
- Department of Endocrinology and Metabolism, Amsterdam UMC, Location AMC at University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (K.W.t.H.); (M.J.S.); (M.T.A.); (M.R.S.)
| | - Annick V. Hartstra
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
| | - Mireille J. Serlie
- Department of Endocrinology and Metabolism, Amsterdam UMC, Location AMC at University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (K.W.t.H.); (M.J.S.); (M.T.A.); (M.R.S.)
| | - Mariette T. Ackermans
- Department of Endocrinology and Metabolism, Amsterdam UMC, Location AMC at University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (K.W.t.H.); (M.J.S.); (M.T.A.); (M.R.S.)
| | - Maarten R. Soeters
- Department of Endocrinology and Metabolism, Amsterdam UMC, Location AMC at University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (K.W.t.H.); (M.J.S.); (M.T.A.); (M.R.S.)
| | - Daniel H. van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam UMC, Location VUMC at VU University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
- Amsterdam UMC, Amsterdam Cardiovascular Sciences, VU University, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Mark Davids
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
- Wallenberg Laboratory, University of Gothenburg, Bruna Stråket 16, SE-413 45 Göteborg, Sweden
| | - Albert K. Groen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.M.K.); (N.C.d.C.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (A.V.H.); (M.D.); (M.N.)
- Correspondence: (M.V.W.); (A.K.G.)
| |
Collapse
|
11
|
Gáspár R, Gömöri K, Kiss B, Szántai Á, Pálóczi J, Varga ZV, Pipis J, Váradi B, Ágg B, Csont T, Ferdinandy P, Barteková M, Görbe A. Decorin Protects Cardiac Myocytes against Simulated Ischemia/Reperfusion Injury. Molecules 2020; 25:molecules25153426. [PMID: 32731559 PMCID: PMC7436189 DOI: 10.3390/molecules25153426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 01/13/2023] Open
Abstract
Search for new cardioprotective therapies is of great importance since no cardioprotective drugs are available on the market. In line with this need, several natural biomolecules have been extensively tested for their potential cardioprotective effects. Previously, we have shown that biglycan, a member of a diverse group of small leucine-rich proteoglycans, enhanced the expression of cardioprotective genes and decreased ischemia/reperfusion-induced cardiomyocyte death via a TLR-4 dependent mechanism. Therefore, in the present study we aimed to test whether decorin, a small leucine-rich proteoglycan closely related to biglycan, could exert cardiocytoprotection and to reveal possible downstream signaling pathways. Methods: Primary cardiomyocytes isolated from neonatal and adult rat hearts were treated with 0 (Vehicle), 1, 3, 10, 30 and 100 nM decorin as 20 h pretreatment and maintained throughout simulated ischemia and reperfusion (SI/R). In separate experiments, to test the mechanism of decorin-induced cardio protection, 3 nM decorin was applied in combination with inhibitors of known survival pathways, that is, the NOS inhibitor L-NAME, the PKG inhibitor KT-5823 and the TLR-4 inhibitor TAK-242, respectively. mRNA expression changes were measured after SI/R injury. Results: Cell viability of both neonatal and adult cardiomyocytes was significantly decreased due to SI/R injury. Decorin at 1, 3 and 10 nM concentrations significantly increased the survival of both neonatal and adult myocytes after SI/R. At 3nM (the most pronounced protective concentration), it had no effect on apoptotic rate of neonatal cardiac myocytes. No one of the inhibitors of survival pathways (L-NAME, KT-5823, TAK-242) influenced the cardiocytoprotective effect of decorin. MYND-type containing 19 (Zmynd19) and eukaryotic translation initiation factor 4E nuclear import factor 1 (Eif4enif1) were significantly upregulated due to the decorin treatment. In conclusion, this is the first demonstration that decorin exerts a direct cardiocytoprotective effect possibly independent of NO-cGMP-PKG and TLR-4 dependent survival signaling.
Collapse
Affiliation(s)
- Renáta Gáspár
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom ter 9, H-6720 Szeged, Hungary; (R.G.); (T.C.)
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Szeged, Dom ter 12, H-6720 Szeged, Hungary; (K.G.); (Á.S.); (J.P.)
| | - Bernadett Kiss
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Ágnes Szántai
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Szeged, Dom ter 12, H-6720 Szeged, Hungary; (K.G.); (Á.S.); (J.P.)
| | - János Pálóczi
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Szeged, Dom ter 12, H-6720 Szeged, Hungary; (K.G.); (Á.S.); (J.P.)
| | - Zoltán V. Varga
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
| | - Judit Pipis
- Pharmahungary Group, Hajnoczy utca 6, H-6722 Szeged, Hungary;
| | - Barnabás Váradi
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
| | - Bence Ágg
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, Hajnoczy utca 6, H-6722 Szeged, Hungary;
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dom ter 9, H-6720 Szeged, Hungary; (R.G.); (T.C.)
| | - Péter Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, Hajnoczy utca 6, H-6722 Szeged, Hungary;
| | - Monika Barteková
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovak
- Institute of Physiology, Comenius University in Bratislava, Sasinkova 2, 813 72 Bratislava, Slovak
- Correspondence: (M.B.); (A.G.)
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Szeged, Dom ter 12, H-6720 Szeged, Hungary; (K.G.); (Á.S.); (J.P.)
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, H-1089 Budapest, Hungary; (B.K.); (Z.V.V.); (B.V.); (B.Á.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, Hajnoczy utca 6, H-6722 Szeged, Hungary;
- Correspondence: (M.B.); (A.G.)
| |
Collapse
|