1
|
Li H, Liang J, Han M, Gao Z. Polyphenols synergistic drugs to ameliorate non-alcoholic fatty liver disease via signal pathway and gut microbiota: A review. J Adv Res 2025; 68:43-62. [PMID: 38471648 PMCID: PMC11785558 DOI: 10.1016/j.jare.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver disease with an increasing incidence worldwide. Single drug therapy may have toxic side effects and disrupt gut microbiota balance. Polyphenols are widely used in disease intervention due to their distinctive nutritional properties and medicinal value, which a potential gut microbiota modulator. However, there is a lack of comprehensive review to explore the efficacy and mechanism of combined therapy with drugs and polyphenols for NAFLD. AIM OF REVIEW Based on this, this review firstly discusses the link between NAFLD and gut microbiota, and outlines the effects of polyphenols and drugs on gut microbiota. Secondly, it examined recent advances in the treatment and intervention of NAFLD with drugs and polyphenols and the therapeutic effect of the combination of the two. Finally, we highlight the underlying mechanisms of polyphenol combined drug therapy in NAFLD. This is mainly in terms of signaling pathways (NF-κB, AMPK, Nrf2, JAK/STAT, PPAR, SREBP-1c, PI3K/Akt and TLR) and gut microbiota. Furthermore, some emerging mechanisms such as microRNA potential biomarker therapies may provide therapeutic avenues for NAFLD. KEY SCIENTIFIC CONCEPTS OF REVIEW Drawing inspiration from combination drug strategies, the use of active substances in combination with drugs for NAFLD intervention holds transformative and prospective potential, both improve NAFLD and restore gut microbiota balance while reducing the required drug dosage. This review systematically discusses the bidirectional interactions between gut microbiota and NAFLD, and summarizes the potential mechanisms of polyphenol synergistic drugs in the treatment of NAFLD by modulating signaling pathways and gut microbiota. Future researches should develop multi-omics technology to identify patients who benefit from polyphenols combination drugs and devising individualized treatment plans to enhance its therapeutic effect.
Collapse
Affiliation(s)
- Hongcai Li
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Jingjing Liang
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Mengzhen Han
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Zhenpeng Gao
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China.
| |
Collapse
|
2
|
Civelek E, Karaman EF, Özden S, Büyükpınarbaşılı N, Uydeş Doğan BS, Kaleli Durman D. Evaluation of the effects of pioglitazone on perivascular adipose tissue function, properties, and structure in a rat model of type-2 diabetes. Can J Physiol Pharmacol 2025; 103:12-28. [PMID: 39361973 DOI: 10.1139/cjpp-2024-0084] [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] [Indexed: 10/05/2024]
Abstract
Perivascular adipose tissue (PVAT) plays an important role in many physiological and pathological processes, such as regulation of vascular tone. The aim of this study is to evaluate the effects of pioglitazone on functional, structural, and biochemical properties of PVAT in an experimental model of type-2 diabetes (T2DM). T2DM was induced by high-fat-diet/low-dose-streptozotocin in rats, and pioglitazone (20 mg/kg/p.o.) was administered for 6 weeks. Changes in biochemical parameters, PVAT-mass, vascular-reactivity in thoracic-aorta, as well as PVAT adipocytokine and PPARG-expression levels, and histopathology were evaluated. Pioglitazone administration improved blood glucose and lipid profiles in T2DM. Pioglitazone did not change the anticontractile effect of PVAT on aortic contractile reactivity and besides, had no influence on endothelium-dependent and -independent relaxation responses. Pioglitazone administration increased PVAT-mass and tumor necrotizing factor-α levels, while adiponectin, leptin, and interleukin-6 levels were unchanged. Also, a prominent increase was observed in PPARG-expression in T2DM-Pio group. Moreover, pioglitazone decreased liver steatosis, aortic wall thickening, and myocardial damage, whereas increased adipocyte size and adiposity in PVAT. Overall, pioglitazone treatment changed the mass and in part the inflammatory profile of PVAT but did not modify vasoreactivity in T2DM. This study provides novel findings in relationship with the adipogenic effect of pioglitazone and PVAT function.
Collapse
MESH Headings
- Animals
- Pioglitazone/pharmacology
- Pioglitazone/therapeutic use
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Adipose Tissue/pathology
- Male
- Rats
- Hypoglycemic Agents/pharmacology
- PPAR gamma/metabolism
- Rats, Wistar
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/pathology
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/physiopathology
- Adipokines/metabolism
- Disease Models, Animal
Collapse
Affiliation(s)
- Erkan Civelek
- Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
- Istanbul University, Graduate School of Health Sciences, Istanbul, Turkey
| | - Ecem Fatma Karaman
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul, Turkey
- Biruni University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul, Turkey
| | - Sibel Özden
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul, Turkey
| | - Nur Büyükpınarbaşılı
- Bezmialem Vakif University, Faculty of Medicine, Department of Medical Pathology, Istanbul, Turkey
| | - B Sönmez Uydeş Doğan
- Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
| | - Deniz Kaleli Durman
- Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
| |
Collapse
|
3
|
Cinakova A, Krenek P, Klimas J, Kralova E. Adding SGLT2 Cotransporter Inhibitor to PPARγ Activator Does Not Provide an Additive Effect in the Management of Diabetes-Induced Vascular Dysfunction. Pharmacology 2023; 108:565-575. [PMID: 37844554 DOI: 10.1159/000533592] [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: 05/23/2023] [Accepted: 08/10/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION Endothelial dysfunction (ED) plays a key role in the pathogenesis of diabetic vascular complications. In monotherapy, dapagliflozin (Dapa) as well as pioglitazone (Pio) prevent the progression of target organ damage in both type 1 (T1DM) and type 2 diabetes. We investigated whether the simultaneous PPAR-γ activation and SGLT2 cotransporter inhibition significantly alleviate ED-related pathological processes and thus normalize vascular response in experimental T1DM. METHODS Experimental diabetes was induced by streptozotocin (STZ; 55 mg/kg, i.p.) in Wistar rats. Dapa (10 mg/kg), Pio (12 mg/kg), or their combination were administrated to the STZ rats orally. Six weeks after STZ administration, the aorta was excised for functional studies and real-time qPCR analysis. RESULTS In the aorta of diabetic rats, impaired endothelium-dependent and independent relaxation were accompanied by the imbalance between vasoactive factors (eNos, Et1) and overexpression of inflammation (Tnfα, Il1b, Il6, Icam, Vcam) and oxidative stress (Cybb) markers. Pio monotherapy normalized response to vasoactive substances and restored balance between Et1-eNos expression, while Dapa treatment was ineffective. Nevertheless, Dapa and Pio monotherapy significantly reverted inflammation and oxidative stress markers to normal values. The combination treatment exhibited an additive effect in modulating Il6 expression, reaching the effect of Pio monotherapy in other measured parameters. CONCLUSION Particularly, Pio exerts a vasoprotective character when used in monotherapy. When combined with Dapa, it does not exhibit an expected additive effect within modulating vasoreactivity or oxidative stress, though having a significant influence on IL6 downregulation.
Collapse
Affiliation(s)
- Aneta Cinakova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Peter Krenek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Eva Kralova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
4
|
Rai H, Fitzgerald S, Coughlan JJ, Spence M, Colleran R, Joner M, Byrne RA. Glu298Asp variant of the endothelial nitric oxide synthase gene and acute coronary syndrome or premature coronary artery disease: A systematic review and meta-analysis. Nitric Oxide 2023; 138-139:85-95. [PMID: 37451608 DOI: 10.1016/j.niox.2023.07.001] [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: 04/14/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Several published studies have reported an association between the Glu298Asp polymorphism (rs1799983), residing in the endothelial nitric oxide synthase (NOS3) gene, and lower levels of circulating nitric oxide, as well as an increased risk of coronary artery disease (CAD). However, association status of this genetic variant with acute coronary syndrome (ACS) or premature CAD (PCAD) is still unclear. Against this background, we conducted a systematic review and study level meta-analysis to assess the association of the NOS3 Glu298Asp polymorphism with ACS or PCAD. MATERIALS AND METHODS A comprehensive online search to identify relevant studies was performed on several databases including PubMed, EMBASE, MEDLINE, Scopus, Cochrane library and Web of Science. The identified studies were stratified into two ancestral subgroups: 'European ancestry' and 'All other ancestries combined'. Study level odds ratios (ORs) and their 95% confidence intervals (CI) were pooled using random/fixed effects employing a Z test. RESULTS Out of a total of 195 distinct records identified through online search, 37 articles with 39 different studies, with a total sample size of 27,441 (11,516 cases/15,925 controls) were included for quantitative synthesis. Pooled results suggested significant associations of the NOS3 Glu298Asp polymorphism with ACS or PCAD through dominant as well as allelic genetic models (p ≤ 0.002), primarily driven by the 'All other ancestries combined' subgroup. The 'All other ancestries combined' subgroup demonstrated an additional risk of 36% for ACS or PCAD, through both dominant and allelic genetic models (OR = 1.36, 95%CI = 1.13, 1.63, p = 0.001 and OR = 1.36, 95%CI = 1.14, 1.61, p = 0.0005 respectively). On the other hand, the 'European ancestry' subgroup did not show any significant associations. Sensitivity analysis and a sub-analysis for the myocardial infarction endpoint further supported these observed associations. CONCLUSIONS This meta-analysis indicates towards an association between the NOS3 Glu298Asp polymorphism and ACS or PCAD, predominantly driven by 'All other ancestries combined' subgroup. In contrast, the 'European ancestry' subgroup did not demonstrate any significant association. Further large-scale investigations are required to confirm our derived results.
Collapse
Affiliation(s)
- Himanshu Rai
- Cardiovascular Research Institute (CVRI) Dublin, Mater Private Network, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - Sean Fitzgerald
- Cardiovascular Research Institute (CVRI) Dublin, Mater Private Network, Dublin, Ireland; School of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - J J Coughlan
- Cardiovascular Research Institute (CVRI) Dublin, Mater Private Network, Dublin, Ireland
| | - Mark Spence
- Cardiovascular Research Institute (CVRI) Dublin, Mater Private Network, Dublin, Ireland; School of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Roisin Colleran
- Cardiovascular Research Institute (CVRI) Dublin, Mater Private Network, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Michael Joner
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Robert A Byrne
- Cardiovascular Research Institute (CVRI) Dublin, Mater Private Network, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
5
|
Alshabi AM, Alkahtani SA, Shaikh IA, Habeeb MS. Caffeine modulates pharmacokinetic and pharmacodynamic profiles of pioglitazone in diabetic rats: Impact on therapeutics. Saudi Med J 2021; 42:151-160. [PMID: 33563733 PMCID: PMC7989285 DOI: 10.15537/smj.2021.2.25695] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/06/2021] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES To determine the influence of caffeine on pharmacokinetics and pharmacodynamics of pioglitazone (PIO) in diabetic rats. METHODS This was a preclinical study conducted in the College of Pharmacy, Najran University, Saudi Arabia, using 5 groups of Wistar rats: normal rats, untreated diabetic rats, diabetic rats + caffeine (20 mg/kg), diabetic rats + PIO (10 mg/kg), and diabetic rats + PIO (10 mg/kg) + caffeine (20 mg/kg). The drugs were administered for 14 days, and fasting plasma glucose concentrations were determined on the first day, and thereafter at weekly intervals. On day 14, rat sacrifice was followed with assay of levels of biomarkers. To estimate the pharmacokinetic parameters, the diabetic animals were assigned to 2 groups: one group received PIO (10 mg/kg), while the other received PIO + caffeine (20 mg/kg). Blood samples were drawn from the retro-orbital plexus at different time intervals, and pharmacokinetic parameters were measured using high performance liquid chromatography. RESULTS Caffeine caused statistically marked increases in area under the curve, Cmax, Tmax, and half-life of PIO, and decreased clearance. Combination of PIO and caffeine produced a synergistic effect on percentage reduction in blood glucose, with 67.1% reductions observed on day 7 and 68.9% reductions observed on day 14. Liver and cardiac biomarkers were significantly decreased, suggesting cardioprotective and hepatoprotective effects. CONCLUSION Co-administration of PIO with caffeine enhances its antidiabetic effect, probably due to enhanced bioavailability of PIO, leading to clinical benefits in diabetic patients.
Collapse
Affiliation(s)
- Ali M. Alshabi
- From the Department of Clinical Pharmacy (Alshabi, Alkahtani), and from the Department of Pharmacology (Shaikh, Habeeb), College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia.
| | - Saad A. Alkahtani
- From the Department of Clinical Pharmacy (Alshabi, Alkahtani), and from the Department of Pharmacology (Shaikh, Habeeb), College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia.
| | - Ibrahim A. Shaikh
- From the Department of Clinical Pharmacy (Alshabi, Alkahtani), and from the Department of Pharmacology (Shaikh, Habeeb), College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia.
| | - Mohammed S. Habeeb
- From the Department of Clinical Pharmacy (Alshabi, Alkahtani), and from the Department of Pharmacology (Shaikh, Habeeb), College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia.
| |
Collapse
|
6
|
Nalawade V, Dixit VA, Vora A, Zade H. Development and Validation of an LC-MS/MS Method for Simultaneous Determination of Canagliflozin and Metformin HCl in Rat Plasma and its Application. CURR PHARM ANAL 2020. [DOI: 10.2174/1573412915666190312161823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background:
Food and herbal extracts rich in Quercetin (QRT) are often self-medicated by
diabetics and can potentially alter the pharmacokinetics (PK) of Metformin HCl (MET) and Canagliflozin
(CNG) leading to food or herb-drug interactions and reduced therapeutic efficacy. However, the
impact of these flavonoids on the pharmacokinetic behaviour of MET and CNG is mostly unknown.
Methods:
A simple one-step protein precipitation method was developed for the determination of MET
and CNG from rat plasma. The mobile phase chosen was MeOH 65% and 35% water containing 0.1%
formic acid at a flow rate of 1mL/min.
Results:
The retention time of MET, internal standard (Valsartan) and CNG was 1.83, 6.2 and 8.2 min,
respectively. The method was found to be linear in the range of 200 - 8000 ng/mL for CNG and 100 =
4000 ng/ml for MET. Precision and accuracy of the method were below 20% at LLOQ and below 15%
for LQC, MQC, and HQC.
Conclusion:
The method was successfully applied for the determination of PK of MET and CNG by
using 100 μL of rat plasma. QRT co-administration affects the PK parameters of MET and CNG. This
alteration in PK parameters might be of significant use for clinicians and patients.
Collapse
Affiliation(s)
- Vivek Nalawade
- SVKM’s, NMIMS Deemed to be University, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, Mumbai, Maharashtra, India
| | - Vaibhav A. Dixit
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani (BITSPilani), VidyaVihar Campus, street number 41, Pilani, 333031, Rajasthan, India
| | - Amisha Vora
- SVKM’s, NMIMS Deemed to be University, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, Mumbai, Maharashtra, India
| | - Himashu Zade
- SVKM’s, NMIMS Deemed to be University, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, Mumbai, Maharashtra, India
| |
Collapse
|
7
|
Tan L, Song A, Ren L, Wang C, Song G. Effect of pioglitazone on skeletal muscle lipid deposition in the insulin resistance rat model induced by high fructose diet under AMPK signaling pathway. Saudi J Biol Sci 2020; 27:1317-1323. [PMID: 32346341 PMCID: PMC7182985 DOI: 10.1016/j.sjbs.2020.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/28/2020] [Accepted: 03/08/2020] [Indexed: 01/19/2023] Open
Abstract
To study the changes of lipid deposition in skeletal muscle of insulin resistance rat and the effect of pioglitazone intervention on the expression of AMPK pathway related genes in rat, a rat model of insulin resistance was induced and constructed by high fructose diet as an test group, and normal rats were used as a control group. First, the effect of pioglitazone intervention on serum lipids-related indicators and mRNA expression levels of fat-related genes in skeletal muscle in rats was investigated. Then skeletal muscle sections were made and stained with oil red O to investigate the effect of pioglitazone intervention on lipid deposition in skeletal muscle of rats. Finally, the effects of pioglitazone intervention therapy on the mRNA and protein expression of related genes in the AMPK signaling pathway in skeletal muscle tissue of rat were explored by real-time quantitative PCR (qRT-PCR) and Western-blotting technology. The results showed that the blood glucose (BG), insulin (INS), adiponectin (ADPN), free fatty acid (FFA), triglyceride (TG), and cholesterol (TC) levels in serum of the test group were higher than the control group (P < 0.05); the visceral fat weight and abdominal fat index of the test group were significantly higher than the control group (P < 0.01); after the pioglitazone intervention, all blood lipid-related indexes in the rat model were significantly lower than before the intervention (P < 0.05); skeletal muscle section staining results showed that the number of lipid droplets in skeletal muscle of rat model was significantly reduced after pioglitazone intervention; and pioglitazone intervention can significantly increase the mRNA and protein expression levels of p-ACC, GLUT7, PGC-1α, and CPT1 genes in the skeletal muscles of experimental rats (P < 0.05). Accordingly, it can be concluded that pioglitazone can play a role in treating insulin resistance by regulating the expression of related genes of AMPK, ACC, etc. in the AMPK signaling pathway.
Collapse
Affiliation(s)
- Lixin Tan
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - An Song
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100191, PR China
| | - Luping Ren
- Hebei Key Laboratory of Metabolic Disease, Shijiazhuang, Hebei 050051, PR China
| | - Chao Wang
- Hebei Key Laboratory of Metabolic Disease, Shijiazhuang, Hebei 050051, PR China
| | - Guangyao Song
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei 050051, PR China
| |
Collapse
|
8
|
Quercetin Exerts Age-Dependent Beneficial Effects on Blood Pressure and Vascular Function, But Is Inefficient in Preventing Myocardial Ischemia-Reperfusion Injury in Zucker Diabetic Fatty Rats. Molecules 2020; 25:molecules25010187. [PMID: 31906454 PMCID: PMC6983107 DOI: 10.3390/molecules25010187] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 12/28/2022] Open
Abstract
Background: Quercetin (QCT) was shown to exert beneficial cardiovascular effects in young healthy animals. The aim of the present study was to determine cardiovascular benefits of QCT in older, 6-month and 1-year-old Zucker diabetic fatty (ZDF) rats (model of type 2 diabetes). Methods: Lean (fa/+) and obese (fa/fa) ZDF rats of both ages were treated with QCT for 6 weeks (20 mg/kg/day). Isolated hearts were exposed to ischemia-reperfusion (I/R) injury (30 min/2 h). Endothelium-dependent vascular relaxation was measured in isolated aortas. Expression of selected proteins in heart tissue was detected by Western blotting. Results: QCT reduced systolic blood pressure in both lean and obese 6-month-old rats but had no effect in 1-year-old rats. Diabetes worsened vascular relaxation in both ages. QCT improved vascular relaxation in 6-month-old but worsened in 1-year-old obese rats and had no impact in lean controls of both ages. QCT did not exert cardioprotective effects against I/R injury and even worsened post-ischemic recovery in 1-year-old hearts. QCT up-regulated expression of eNOS in younger and PKCε expression in older rats but did not activate whole PI3K/Akt pathway. Conclusions: QCT might be beneficial for vascular function in diabetes type 2; however, increasing age and/or progression of diabetes may confound its vasculoprotective effects. QCT seems to be inefficient in preventing myocardial I/R injury in type 2 diabetes and/or higher age. Impaired activation of PI3K/Akt kinase pathway might be, at least in part, responsible for failing cardioprotection in these subjects.
Collapse
|
9
|
Luo Z, Jia A, Lu Z, Muhammad I, Adenrele A, Song Y. Associations of the NOS3 rs1799983 polymorphism with circulating nitric oxide and lipid levels: a systematic review and meta-analysis. Postgrad Med J 2019; 95:361-371. [PMID: 31138610 DOI: 10.1136/postgradmedj-2019-136396] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/15/2019] [Accepted: 05/04/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Circulating nitric oxide (NO) and lipid levels are closely associated with coronary artery disease (CAD). It is unclear whether the rs1799983 polymorphism in endothelial nitric oxide synthase (NOS3) gene is associated with plasma levels of NO and lipids. This systematic review and meta-analysis (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) aimed to clarify the relationships between the rs1799983 polymorphism and plasma levels of NO and lipids. METHODS Sixteen studies (2702 subjects) and 59 studies (14 148 subjects) were identified for the association analyses for NO and lipids, respectively. Mean difference (MD) and 95% CI were used to estimate the effects of the rs1799983 polymorphism on plasma NO and lipid levels. The primary outcome variable was NO, and the secondary outcomes included triglycerides, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C). RESULTS Carriers of the T allele had lower levels of NO (MD -0.27 μmol/L, 95% CI -0.42 to -0.12 μmol/L, p<0.001) and HDL-C (MD -0.07 mmol/L, 95% CI -0.14 to -0.00 mmol/L, p=0.04), and higher levels of TC (MD 0.13 mmol/L, 95% CI 0.06 to 0.20 mmol/L, p<0.001) and LDL-C (MD 0.14 mmol/L, 95% CI 0.05 to 0.22 mmol/L, p=0.002) than the non-carriers. Triglyceride levels were comparable between the genotypes. CONCLUSION The association between the NOS3 rs1799983 polymorphism and CAD may be partly mediated by abnormal NO and lipid levels caused by the T allele.
Collapse
Affiliation(s)
- Zhi Luo
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong city, Sichuan province, China
| | - Aimei Jia
- School of Preclinical Medicine, North Sichuan Medical College, Nanchong city, Sichuan province, China
| | - Zhan Lu
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong city, Sichuan province, China
| | - Irfan Muhammad
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong city, Sichuan province, China
| | - Adebayo Adenrele
- Department of Anatomy, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - Yongyan Song
- School of Preclinical Medicine, North Sichuan Medical College, Nanchong city, Sichuan province, China
| |
Collapse
|
10
|
Maaliki D, Shaito AA, Pintus G, El-Yazbi A, Eid AH. Flavonoids in hypertension: a brief review of the underlying mechanisms. Curr Opin Pharmacol 2019; 45:57-65. [DOI: 10.1016/j.coph.2019.04.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/12/2019] [Accepted: 04/16/2019] [Indexed: 12/21/2022]
|
11
|
Ghosh A, Gao L, Thakur A, Siu PM, Lai CWK. Role of free fatty acids in endothelial dysfunction. J Biomed Sci 2017; 24:50. [PMID: 28750629 PMCID: PMC5530532 DOI: 10.1186/s12929-017-0357-5] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
Plasma free fatty acids levels are increased in subjects with obesity and type 2 diabetes, playing detrimental roles in the pathogenesis of atherosclerosis and cardiovascular diseases. Increasing evidence showing that dysfunction of the vascular endothelium, the inner lining of the blood vessels, is the key player in the pathogenesis of atherosclerosis. In this review, we aimed to summarize the roles and the underlying mechanisms using the evidence collected from clinical and experimental studies about free fatty acid-mediated endothelial dysfunction. Because of the multifaceted roles of plasma free fatty acids in mediating endothelial dysfunction, elevated free fatty acid level is now considered as an important link in the onset of endothelial dysfunction due to metabolic syndromes such as diabetes and obesity. Free fatty acid-mediated endothelial dysfunction involves several mechanisms including impaired insulin signaling and nitric oxide production, oxidative stress, inflammation and the activation of the renin-angiotensin system and apoptosis in the endothelial cells. Therefore, targeting the signaling pathways involved in free fatty acid-induced endothelial dysfunction could serve as a preventive approach to protect against the occurrence of endothelial dysfunction and the subsequent complications such as atherosclerosis.
Collapse
Affiliation(s)
- Arijit Ghosh
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, HKSAR, China
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, China
| | - Lei Gao
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, HKSAR, China
| | - Abhimanyu Thakur
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, China
| | - Parco M. Siu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, HKSAR, China
| | - Christopher W. K. Lai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, HKSAR, China
| |
Collapse
|