1
|
Dias CC, Caetano CFF, Costa GAJ, Coelho AA, Lemos JVM, de Paula DS, Lima JPM, de Barros Silva PG. Treatment with cyclosporine attenuates the inflammatory process and severity of bisphosphonate-induced osteonecrosis of the jaws in rats. Inflammopharmacology 2025; 33:2007-2022. [PMID: 39992590 DOI: 10.1007/s10787-025-01673-6] [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: 10/17/2024] [Accepted: 01/31/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION Osteonecrosis usually occurs with necrotic bone exposure in the mandible asymptomatically for long periods but can evolve to present pain, fistula, odor, bleeding, and suppuration. OBJECTIVE To evaluate the influence of cyclosporine treatment and its influence on osteonecrosis in a rat model. METHODS The animals were randomly divided into 05 groups (n = 8/group). The negative control group (SAL), positive control group treated with zoledronic acid (ZA + SAL), and test groups were treated with cyclosporine A (CsA) at 5, 2.5, and 1.25 mg/kg and treated with ZA. The left lower second molars were extracted. The animals were euthanized 1 month after tooth extraction. Digital radiographs, histological slides, and immunoexpression of IL-2, IL-6, TNF-α, PPAR-γ, c-Fos, c-Jun, FoxP3, and INF-γ were analyzed. Western blot assays were performed to investigate the expression of RORyT. In addition, hematological analysis, body mass variation, and femur mechanical tests were performed. RESULTS Radiographs showed that in the groups treated with ZA, there was an increase in the radiolucent area suggestive of osteonecrosis, and treatment with cyclosporine did not reduce this parameter (p < 0.001). In the western blot analysis, animals treated with ZA showed increased expression of RORyT (1.887 ± 0.114) compared to the saline group (0.799 ± 0.107), and treatment with the highest dose of cyclosporine (0.652 ± 0.070) reduced this expression (p < 0.001). DISCUSSION Studies have observed bone health in animals treated with CsA. Treatment with this immunosuppressant showed a bone-protective effect of CsA, which corroborates our findings. CONCLUSION Treatment with CsA reduced the immunoexpression of pro-inflammatory cytokines such as IL-2 and TNF-α and decreased the expression of RORyT.
Collapse
Affiliation(s)
- Camila Costa Dias
- Post-Graduate Program in Dental Sciences, Unichristus, Fortaleza, Brazil
| | | | | | - Antônio Alexandre Coelho
- Department of Dentistry, Unichristus, Vereador Paulo Mamede, 130 - Cocó, Fortaleza, CE, 60192-350, Brazil
| | - José Vitor Mota Lemos
- Department of Dentistry, Unichristus, Vereador Paulo Mamede, 130 - Cocó, Fortaleza, CE, 60192-350, Brazil
| | - Dayrine Silveira de Paula
- Department of Dentistry, Unichristus, Vereador Paulo Mamede, 130 - Cocó, Fortaleza, CE, 60192-350, Brazil
- Post-Graduate Program in Dental Sciences, Unichristus, Fortaleza, Brazil
| | - Juliana Paiva Marques Lima
- Department of Dentistry, Unichristus, Vereador Paulo Mamede, 130 - Cocó, Fortaleza, CE, 60192-350, Brazil
- Post-Graduate Program in Dental Sciences, Unichristus, Fortaleza, Brazil
| | - Paulo Goberlânio de Barros Silva
- Department of Dentistry, Unichristus, Vereador Paulo Mamede, 130 - Cocó, Fortaleza, CE, 60192-350, Brazil.
- Post-Graduate Program in Dental Sciences, Unichristus, Fortaleza, Brazil.
| |
Collapse
|
2
|
Jiang Y, Tong W, Li Y, Ma Q, Chen Y. Melatonin inhibits the formation of intraplaque neovessels in ApoE-/- mice via PPARγ- RhoA-ROCK pathway. Biochem Biophys Res Commun 2024; 696:149391. [PMID: 38184922 DOI: 10.1016/j.bbrc.2023.149391] [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: 09/28/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND According to former research, the atherosclerotic plaque is thought to be aggravated by intraplaque neovessels (IPN) and intraplaque hemorrhage (IPH). Intriguingly, a lower incidence of IPH was found in plaque treated with melatonin. In this study, we attempted to investigate the impact and underlying mechanism regarding the influences of melatonin upon IPN. METHODS A mouse model was established by subjecting the high fat diet (HFD)-fed ApoE-/- mice to tandem stenosis (TS) surgery with melatonin and GW9662, a PPARγ antagonist, being given by gavage. In vitro experiment was conducted with HUVECs exposing to according treatments of VEGF, melatonin, GW9662, or Y27632. RESULTS Plaque and IPN were attenuated by treatment with melatonin, which was then reversed by blocking PPARγ. Western blotting results showed that melatonin increased PPARγ and decreased RhoA/ROCK signaling in carotid artery. Elevated RhoA/ROCK signaling was observed in melatonin-treated mice when PPARγ was blocked. In accordance with it, experiments using protein and mRNA from HUVECs revealed that melatonin inhibited the RhoA/ROCK signaling by enhancing PPARγ. According to in vitro study, melatonin was able to inhibit cell migration and angiogenesis, which was aborted by GW9662. Blockage of ROCK using Y27632 was able to cease the effect of GW9662 and restored the suppression on cell migration and angiogenesis by melatonin. CONCLUSIONS Our study demonstrates that melatonin is able to curb development of plaque and IPN formation by inhibiting the migration of endothelial cells via PPARγ- RhoA-ROCK pathway. That provides a therapeutic potential for both melatonin and PPARγ agonist targeting IPN, IPH, and atherosclerotic plaque.
Collapse
Affiliation(s)
- YuFan Jiang
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Tong
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yueyang Li
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qiang Ma
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - YunDai Chen
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
3
|
Nourmohammadi K, Bayrami A, Naderi R, Shirpoor A, Soraya H. Moderate exercise mitigates cardiac dysfunction and injury induced by cyclosporine A through activation of the PGI 2 / PPAR-γ signaling pathway. Res Pharm Sci 2023; 18:696-707. [PMID: 39005570 PMCID: PMC11246107 DOI: 10.4103/1735-5362.389958] [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: 03/04/2023] [Revised: 06/20/2023] [Accepted: 09/11/2023] [Indexed: 07/16/2024] Open
Abstract
Background and purpose The present study investigated the role of the prostaglandin I2/peroxisome proliferator activator receptor (PGI2/PPAR) signaling pathway in cardiac cell proliferation, apoptosis, and systemic hemodynamic variables under cyclosporine A (CsA) exposure alone or combined with moderate exercises. Experimental approach Twenty-four male Wistar rats were classified into three groups, namely, control, CsA, and CsA + exercise. Findings/Results After 42 days of treatment, the findings showed a significant enhancement in the expression of the β-MHC gene, enhancement in protein expression of Bax and caspase-3, and a significant decline in the protein expression of Bcl-2 expression, as well as increased proliferation intensity in the heart tissue of the CsA group compared to the control group. Systolic pressure, pulse pressure, mean arterial pressure (MAP), QT and QRS duration, and T wave amplitude, as well as QTc amount in the CsA group, showed a significant increase compared to the control group. PPAR-γ and PGI2 showed no significant changes compared to the control group. Moderate exercise along with CsA significantly enhanced the protein expression of PPAR-γ and PGI2 and declined protein expression of Bax, and caspase-3 compared to those in the CsA group. In the CsA + exercise group, systolic pressure, MAP, and Twave showed a significant decrease compared to the CsA group. Moderate exercises along CsA improved heart cell proliferation intensity and significantly reduced β- MHC gene expression compared to the CsA group. Conclusions and implications The results showed moderate exercise alleviated CsA-induced heart tissue apoptosis and proliferation with the corresponding activation of the PGI2/PPAR-γ pathway.
Collapse
Affiliation(s)
- Khatereh Nourmohammadi
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Abolfazl Bayrami
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Roya Naderi
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Alireza Shirpoor
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
4
|
Di D, Zhang R, Zhou H, Wei M, Cui Y, Zhang J, Yuan T, Liu Q, Zhou T, Liu J, Wang Q. Exposure to phenols, chlorophenol pesticides, phthalate and PAHs and mortality risk: A prospective study based on 6 rounds of NHANES. CHEMOSPHERE 2023; 329:138650. [PMID: 37037349 DOI: 10.1016/j.chemosphere.2023.138650] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 05/03/2023]
Abstract
OBJECTIVES Human exposure to various endocrine disrupting chemicals (EDCs) is widespread and long-lasting. The primary objective of this study was to prospectively evaluate the association of combined exposure of phenols, chlorophenol pesticides, phthalate and polycyclic aromatic hydrocarbons (PAHs) and mortality risk in a representative US population. METHODS The data on urinary levels of phenols, chlorophenol pesticides, phthalates, and PAH metabolites, were collected from participants aged ≥20 years in six rounds of the National Health and Nutrition Examination Survey (NHANES) (2003-2014). NHANES-linked death records up to December 31, 2015 were used to ascertain mortality status and cause of death. Cox proportional hazards and competing risk models were mainly used for chemical and mortality risk association analysis. The weighted quantile sum (WQS) regression and the least absolute shrinkage and selection operator regression were employed to estimate the association between EDC co-exposure and mortality risk. RESULTS High levels of mono-n-butyl phthalate, monobenzyl phthalate, and 1-napthol were significantly associated with increased risk of all cause, cardiovascular disease (CVD) and cancer mortality among all participants. WQS index was associated with the risks of all-cause (hazard ratio [HR] = 1.389, 95%CI: 1.155-1.669) and CVD mortality (HR = 1.925, 95%CI: 1.152-3.216). High co-exposure scores were associated with elevated all-cause (HR = 2.842, 95% CI: 1.2.094-3.858), CVD (HR = 1.855, 95% CI: 1.525-2.255), and cancer mortality risks (HR = 2.961, 95% CI: 1.468-5.972). The results of subgroup analysis, competing risk model, and sensitivity analysis were generally consistent with the findings from the main analyses, indicating the robustness of our findings. CONCLUSIONS This study provided the first epidemiological evidence that co-exposure to EDC at fairly low levels contributed to elevated mortality risk among US adults. The underlying mechanisms for the effects of EDC co-exposure on human health are worthy of future exploration.
Collapse
Affiliation(s)
- Dongsheng Di
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ruyi Zhang
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haolong Zhou
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Muhong Wei
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan Cui
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jianli Zhang
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tingting Yuan
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Liu
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tingting Zhou
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Junan Liu
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qi Wang
- MOE Key Lab of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
5
|
Sun JY, Du LJ, Shi XR, Zhang YY, Liu Y, Wang YL, Chen BY, Liu T, Zhu H, Liu Y, Ruan CC, Gan Z, Ying H, Yin Z, Gao PJ, Yan X, Li RG, Duan SZ. An IL-6/STAT3/MR/FGF21 axis mediates heart-liver cross-talk after myocardial infarction. SCIENCE ADVANCES 2023; 9:eade4110. [PMID: 37018396 PMCID: PMC10075967 DOI: 10.1126/sciadv.ade4110] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 03/07/2023] [Indexed: 06/19/2023]
Abstract
The liver plays a protective role in myocardial infarction (MI). However, very little is known about the mechanisms. Here, we identify mineralocorticoid receptor (MR) as a pivotal nexus that conveys communications between the liver and the heart during MI. Hepatocyte MR deficiency and MR antagonist spironolactone both improve cardiac repair after MI through regulation on hepatic fibroblast growth factor 21 (FGF21), illustrating an MR/FGF21 axis that underlies the liver-to-heart protection against MI. In addition, an upstreaming acute interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) pathway transmits the heart-to-liver signal to suppress MR expression after MI. Hepatocyte Il6 receptor deficiency and Stat3 deficiency both aggravate cardiac injury through their regulation on the MR/FGF21 axis. Therefore, we have unveiled an IL-6/STAT3/MR/FGF21 signaling axis that mediates heart-liver cross-talk during MI. Targeting the signaling axis and the cross-talk could provide new strategies to treat MI and heart failure.
Collapse
Affiliation(s)
- Jian-Yong Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Xue-Rui Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yu-Yao Zhang
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yong-Li Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Bo-Yan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Hong Zhu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing 210061, China
| | - Hao Ying
- CAS Key Laboratory of Nutrition, Metabolism, and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai 519000, Guangdong, China
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, Guangdong, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China
| |
Collapse
|
6
|
Guixé‐Muntet S, Biquard L, Szabo G, Dufour J, Tacke F, Francque S, Rautou P, Gracia‐Sancho J. Review article: vascular effects of PPARs in the context of NASH. Aliment Pharmacol Ther 2022; 56:209-223. [PMID: 35661191 PMCID: PMC9328268 DOI: 10.1111/apt.17046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/04/2021] [Accepted: 05/08/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors known to regulate glucose and fatty acid metabolism, inflammation, endothelial function and fibrosis. PPAR isoforms have been extensively studied in metabolic diseases, including type 2 diabetes and cardiovascular diseases. Recent data extend the key role of PPARs to liver diseases coursing with vascular dysfunction, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). AIM This review summarises and discusses the pathobiological role of PPARs in cardiovascular diseases with a special focus on their impact and therapeutic potential in NAFLD and NASH. RESULTS AND CONCLUSIONS PPARs may be attractive for the treatment of NASH due to their liver-specific effects but also because of their efficacy in improving cardiovascular outcomes, which may later impact liver disease. Assessment of cardiovascular disease in the context of NASH trials is, therefore, of the utmost importance, both from a safety and efficacy perspective.
Collapse
Affiliation(s)
- Sergi Guixé‐Muntet
- Liver Vascular Biology Research GroupIDIBAPS Biomedical Research Institute & CIBEREHDBarcelonaSpain
| | - Louise Biquard
- Université de Paris, Inserm, CNRSCentre de recherche sur l'InflammationUMR1149ParisFrance
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Jean‐François Dufour
- Department of Visceral Surgery and Medicine & Department for Biomedical ResearchInselspital, University of BernBernSwitzerland
| | - Frank Tacke
- Department of Hepatology & GastroenterologyCharité Universitätsmedizin Berlin, Campus Virchow‐Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Sven Francque
- Department of Gastroenterology and HepatologyAntwerp University HospitalAntwerpBelgium,Translational Sciences in Inflammation and ImmunologyInflaMed Centre of Excellence, Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Pierre‐Emmanuel Rautou
- Université de Paris, AP‐HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGESTCentre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE‐LIVER, Centre de recherche sur l'inflammationParisFrance
| | - Jordi Gracia‐Sancho
- Liver Vascular Biology Research GroupIDIBAPS Biomedical Research Institute & CIBEREHDBarcelonaSpain,Department of Visceral Surgery and Medicine & Department for Biomedical ResearchInselspital, University of BernBernSwitzerland
| |
Collapse
|
7
|
Liu T, Yan T, Jia X, Liu J, Ma R, Wang Y, Wang X, Liang Y, Xiao Y, Dong Y. Systematic exploration of the potential material basis and molecular mechanism of the Mongolian medicine Nutmeg-5 in improving cardiac remodeling after myocardial infarction. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114847. [PMID: 34800647 DOI: 10.1016/j.jep.2021.114847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nutmeg-5, which consists of Myristica fragrans Houtt., Aucklandia lappa Decne., Inula helenium L., Fructus Choerospondiatis and Piper longum L., is an ancient and classic formula in traditional Mongolian medicine that is widely used in the treatment of ischemic heart disease. However, its material basis and pharmacological mechanisms remain to be fully elucidated. AIM OF THE STUDY The aim of this study was to explore the potential material basis and molecular mechanism of Nutmeg-5 in improving cardiac remodeling after myocardial infarction (MI). MATERIALS AND METHODS The constituents of Nutmeg-5 absorbed into the blood were identified by high-performance liquid chromatography-mass spectrometry (HPLC-MS/MS). A mouse MI model was induced in male Kunming mice by permanent ligation of the left anterior descending coronary artery (LDA) ligation. Echocardiography was performed to assess cardiac function. The protective effect of Nutmeg-5 and compound Danshen dripping pills as positive control medicine on post-MI cardiac remodeling was evaluated by tissue histology and determination of the serum protein levels of biomarkers of myocardial injury. RNA sequencing analysis of mouse left ventricle tissue was performed to explore the molecular mechanism of Nutmeg-5 in cardiac remodeling after MI. RESULTS A total of 27 constituents absorbed into blood were identified in rat plasma following gavage administration of Nutmeg-5 (0.54 g/kg) for 1 h. We found that ventricular remodeling after MI was significantly improved after Nutmeg-5 treatment in mice, which was demonstrated by decreased mortality, better cardiac function, decreased heart weight to body weight and heart weight to tibia length ratios, and attenuated cardiac fibrosis and myocardial injury. RNA sequencing revealed that the protective effect of Nutmeg-5 on cardiac remodeling after MI was associated with improved heart metabolism. Further study found that Nutmeg-5 treatment could preserve the ultrastructure of mitochondria and upregulate gene expression related to mitochondrial function and structure. HIF-1α (hypoxia inducible factor 1, alpha subunit) expression was significantly upregulated in the hearts of MI mice and significantly suppressed in the hearts of Nutmeg-5-treated mice. In addition, Nutmeg-5 treatment significantly activated the peroxisome proliferator-activated receptor alpha signaling pathway, which was inhibited in the hearts of MI mice. CONCLUSIONS Nutmeg-5 attenuates cardiac remodeling after MI by improving heart metabolism and preserving mitochondrial dysfunction by inhibiting HIF-1α expression in the mouse heart after MI.
Collapse
Affiliation(s)
- Tianlong Liu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, PR China
| | - Tingting Yan
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Hohhot, 010110, PR China; Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Inner Mongolia Medical University, Hohhot, 010110, PR China
| | - Xin Jia
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, PR China; Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Hohhot, 010110, PR China; Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Inner Mongolia Medical University, Hohhot, 010110, PR China
| | - Jing Liu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, PR China
| | - Ruilian Ma
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, PR China
| | - Yi Wang
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010059, PR China
| | - Xianjue Wang
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, 010050, Inner Mongolia, PR China
| | - Yabin Liang
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, 010050, Inner Mongolia, PR China
| | - Yunfeng Xiao
- Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Inner Mongolia Medical University, Hohhot, 010110, PR China; Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, China
| | - Yu Dong
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Hohhot, 010110, PR China; Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| |
Collapse
|
8
|
Tardelli M, Stulnig TM. Aquaporin regulation in metabolic organs. VITAMINS AND HORMONES 2021; 112:71-93. [PMID: 32061350 DOI: 10.1016/bs.vh.2019.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aquaporins (AQPs) are a family of 13 small trans-membrane proteins, which facilitate shuttling of glycerol, water and urea. The peculiar role of AQPs in glycerol transport makes them attractive targets in metabolic organs since glycerol represents the backbone of triglyceride synthesis. Importantly, AQPs are known to be regulated by various nuclear receptors which in turn govern lipid and glucose metabolism as well as inflammatory cascades. Here, we review the role of AQPs regulation in metabolic organs exploring their physiological impact in health and disease.
Collapse
Affiliation(s)
- Matteo Tardelli
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Cornell Department of Medicine, Weill Cornell Medical College, New York, NY, United States; Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas M Stulnig
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Wang C, Zhou J, Wang J, Li S, Fukunaga A, Yodoi J, Tian H. Progress in the mechanism and targeted drug therapy for COPD. Signal Transduct Target Ther 2020; 5:248. [PMID: 33110061 PMCID: PMC7588592 DOI: 10.1038/s41392-020-00345-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is emphysema and/or chronic bronchitis characterised by long-term breathing problems and poor airflow. The prevalence of COPD has increased over the last decade and the drugs most commonly used to treat it, such as glucocorticoids and bronchodilators, have significant therapeutic effects; however, they also cause side effects, including infection and immunosuppression. Here we reviewed the pathogenesis and progression of COPD and elaborated on the effects and mechanisms of newly developed molecular targeted COPD therapeutic drugs. Among these new drugs, we focussed on thioredoxin (Trx). Trx effectively prevents the progression of COPD by regulating redox status and protease/anti-protease balance, blocking the NF-κB and MAPK signalling pathways, suppressing the activation and migration of inflammatory cells and the production of cytokines, inhibiting the synthesis and the activation of adhesion factors and growth factors, and controlling the cAMP-PKA and PI3K/Akt signalling pathways. The mechanism by which Trx affects COPD is different from glucocorticoid-based mechanisms which regulate the inflammatory reaction in association with suppressing immune responses. In addition, Trx also improves the insensitivity of COPD to steroids by inhibiting the production and internalisation of macrophage migration inhibitory factor (MIF). Taken together, these findings suggest that Trx may be the ideal drug for treating COPD.
Collapse
Affiliation(s)
- Cuixue Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Jiedong Zhou
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Jinquan Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Shujing Li
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Atsushi Fukunaga
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Junji Yodoi
- Laboratory of Infection and Prevention, Department of Biological Response, Institute for Virus Research, Kyoto University, Kyoto, 606-8501, Japan
| | - Hai Tian
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China.
- Jiaozhimei Biotechnology (Shaoxing) Co, Ltd, Shaoxing, 312000, China.
| |
Collapse
|
10
|
Papazafiropoulou AK, Koros C, Melidonis A, Antonopoulos S. Diabetes and dementia - the two faces of Janus. Arch Med Sci Atheroscler Dis 2020; 5:e186-e197. [PMID: 32832719 PMCID: PMC7433787 DOI: 10.5114/amsad.2020.97433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/18/2020] [Indexed: 01/03/2023] Open
Abstract
Patients with type 2 diabetes are at high risk for cognitive decline and dementia. Despite the limited data on the possible pathogenetic mechanisms, evidence suggests that cognitive decline, and thus dementia and Alzheimer's disease, might arise from a complex interplay between type 2 diabetes and the aging brain, including decreased insulin signalling and glucose metabolism, mitochondrial dysfunction, neuroinflammation, and vascular disease. Furthermore, there is increasing interest on the effects of antidiabetic agents on cognitive decline. There are many studies showing that antidiabetic agents might have beneficial effects on the brain, mainly through inhibition of oxidative stress, inflammation, and apoptosis. In addition, experimental studies on patients with diabetes and Alzheimer's disease have shown beneficial effects on synaptic plasticity, metabolism of amyloid-β, and microtubule-associated protein tau. Therefore, in the present review, we discuss the effects of antidiabetic agents in relation to cognitive decline, and in particular dementia and Alzheimer's disease, in patients with type 2 diabetes.
Collapse
Affiliation(s)
| | - Chris Koros
- 1 Department of Neurology, National and Kapodistrian University of Athens Medical School, Eginition Hospital, Athens, Greece
| | - Andreas Melidonis
- Diabetes and Cardiometabolic Centre, Metropolitan Hospital, Piraeus, Greece
| | - Stavros Antonopoulos
- 1 Department of Internal Medicine and Diabetes Centre, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
11
|
JNK and cardiometabolic dysfunction. Biosci Rep 2019; 39:BSR20190267. [PMID: 31270248 PMCID: PMC6639461 DOI: 10.1042/bsr20190267] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiometabolic syndrome (CMS) describes the cluster of metabolic and cardiovascular diseases that are generally characterized by impaired glucose tolerance, intra-abdominal adiposity, dyslipidemia, and hypertension. CMS currently affects more than 25% of the world’s population and the rates of diseases are rapidly rising. These CMS conditions represent critical risk factors for cardiovascular diseases including atherosclerosis, heart failure, myocardial infarction, and peripheral artery disease (PAD). Therefore, it is imperative to elucidate the underlying signaling involved in disease onset and progression. The c-Jun N-terminal Kinases (JNKs) are a family of stress signaling kinases that have been recently indicated in CMS. The purpose of this review is to examine the in vivo implications of JNK as a potential therapeutic target for CMS. As the constellation of diseases associated with CMS are complex and involve multiple tissues and environmental triggers, carefully examining what is known about the JNK pathway will be important for specificity in treatment strategies.
Collapse
|
12
|
Abdel-Rahman EA, Bhattacharya S, Buabeid M, Majrashi M, Bloemer J, Tao YX, Dhanasekaran M, Escobar M, Amin R, Suppiramaniam V. PPAR-δ Activation Ameliorates Diabetes-Induced Cognitive Dysfunction by Modulating Integrin-linked Kinase and AMPA Receptor Function. J Am Coll Nutr 2019; 38:693-702. [PMID: 31008686 DOI: 10.1080/07315724.2019.1598307] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
An estimated 9% of the American population experiences type II diabetes mellitus (T2DM) due to diet or genetic predisposition. Recent reports indicate that patients with T2DM are at increased risk for cognitive dysfunctions, as observed in conditions like Alzheimer's disease (AD). In addition, AD is the leading cause of dementia, highlighting the urgency of developing novel therapeutic targets for T2DM-induced cognitive deficits. The peroxisome proliferator activated receptor-δ (PPAR-δ) is highly expressed in the brain and has been shown to play an important role in spatial memory and hippocampal neurogenesis. However, the effect of PPAR-δ agonists on T2DM-induced cognitive impairment has not been explored. In this study, the effects of GW0742 (a selective PPAR-δ agonist) on hippocampal synaptic transmission, plasticity, and spatial memory were investigated in the db/db mouse model of T2DM. Oral administration of GW0742 for 2 weeks significantly improved hippocampal long-term potentiation. In addition, GW0742 effectively prevented deficits in hippocampal dependent spatial memory in db/db mice. PPAR-δ-mediated improvements in synaptic plasticity and behavior were accompanied by a significant recovery in hippocampal α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor-mediated synaptic transmission. Our findings suggest that activation of PPAR-δ might ameliorate T2DM-induced impairments in hippocampal synaptic plasticity and memory.
Collapse
Affiliation(s)
- Engy A Abdel-Rahman
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama, USA.,Department of Pharmacology, Faculty of Medicine, Assuit University, Assuit, Egypt**
| | - Subhrajit Bhattacharya
- Department of Pharmacology, Rollins Research Center, Emory University, Atlanta, Georgia, USA**
| | - Manal Buabeid
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama, USA.,College of Pharmacy and Health Sciences, Ajman University, Ajman, UAE**
| | - Mohammed Majrashi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama, USA.,Department of Pharmacology, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama, USA
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA.,Center for Neuroscience Initiative, Auburn University, Auburn, Alabama, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama, USA.,Center for Neuroscience Initiative, Auburn University, Auburn, Alabama, USA
| | - Martha Escobar
- Department of Psychology, Auburn University, Auburn, Alabama, USA.,Department of Psychology, Oakland University, Rochester, Michigan, USA*
| | - Rajesh Amin
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama, USA.,Center for Neuroscience Initiative, Auburn University, Auburn, Alabama, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama, USA.,Center for Neuroscience Initiative, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
13
|
Rooney MR, Lutsey PL, Bhatti P, Prizment A. Urinary 2,5-dicholorophenol and 2,4-dichlorophenol concentrations and prevalent disease among adults in the National Health and Nutrition Examination Survey (NHANES). Occup Environ Med 2019; 76:181-188. [PMID: 30377258 PMCID: PMC6377840 DOI: 10.1136/oemed-2018-105278] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/08/2018] [Accepted: 10/15/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To test cross-sectional associations between urinary concentrations of 2,5-dichlorophenol (2,5-DCP) and 2,4-dichlorophenol (2,4-DCP) with the prevalence of cardiovascular disease (CVD), cancer, lung disease, thyroid problems and liver conditions. METHODS Logistic regression was used to evaluate associations of urinary concentrations of 2,5-DCP and 2,4-DCP with prevalence of various medical conditions among 3617 National Health and Nutrition Examination Survey participants from 2007-2008 and 2009-2010. ORs and 95% CIs for each disease were estimated. All regression models were adjusted for urinary creatinine. RESULTS We observed a monotonically increasing association between quartiles of 2,5-DCP and prevalence of CVD. After adjustment for sociodemographic and lifestyle characteristics, participants with the highest versus lowest quartile of urinary 2,5-DCP had an OR=1.84 (95% CI 1.26 to 2.70) (p linear trend=0.006). The association was similar with further adjustment for established clinical CVD risk factors. Higher 2,5-DCP was also associated with prevalence of all cancers combined (ORQ4 vs Q1=1.50 (95% CI 1.00 to 2.26); p trend=0.05) and, in exploratory analyses, with gynaecological cancers (ORQ4 vs Q1=4.15 (95% CI 1.51 to 11.40; p trend=0.01)). No associations were detected between 2,5-DCP and lung diseases, thyroid problems or liver conditions, nor between 2,4-DCP and prevalent disease. CONCLUSION In this nationally representative study, higher urinary 2,5-DCP concentrations were associated with greater prevalence of CVD and all cancers combined. Further examination may be warranted to assess whether chronic exposure to 2,5-DCP is associated with incidence of adverse health outcomes.
Collapse
Affiliation(s)
- Mary R Rooney
- Division of Epidemiology & Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Pamela L Lutsey
- Division of Epidemiology & Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Parveen Bhatti
- Cancer Control Research, BC Cancer, Vancouver BC, Canada
- Program in Epidemiology, Public Health Sciences Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Anna Prizment
- Division of Epidemiology & Community Health, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
14
|
Beta-caryophyllene protects against diet-induced dyslipidemia and vascular inflammation in rats: Involvement of CB2 and PPAR-γ receptors. Chem Biol Interact 2019; 297:16-24. [DOI: 10.1016/j.cbi.2018.10.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/23/2018] [Accepted: 10/17/2018] [Indexed: 02/07/2023]
|
15
|
Tardelli M, Claudel T, Bruschi FV, Trauner M. Nuclear Receptor Regulation of Aquaglyceroporins in Metabolic Organs. Int J Mol Sci 2018; 19:E1777. [PMID: 29914059 PMCID: PMC6032257 DOI: 10.3390/ijms19061777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Nuclear receptors, such as the farnesoid X receptor (FXR) and the peroxisome proliferator-activated receptors gamma and alpha (PPAR-γ, -α), are major metabolic regulators in adipose tissue and the liver, where they govern lipid, glucose, and bile acid homeostasis, as well as inflammatory cascades. Glycerol and free fatty acids are the end products of lipid droplet catabolism driven by PPARs. Aquaporins (AQPs), a family of 13 small transmembrane proteins, facilitate the shuttling of water, urea, and/or glycerol. The peculiar role of AQPs in glycerol transport makes them pivotal targets in lipid metabolism, especially considering their tissue-specific regulation by the nuclear receptors PPARγ and PPARα. Here, we review the role of nuclear receptors in the regulation of glycerol shuttling in liver and adipose tissue through the function and expression of AQPs.
Collapse
Affiliation(s)
- Matteo Tardelli
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | - Francesca Virginia Bruschi
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| |
Collapse
|
16
|
Subramanian V, Borchard S, Azimzadeh O, Sievert W, Merl-Pham J, Mancuso M, Pasquali E, Multhoff G, Popper B, Zischka H, Atkinson MJ, Tapio S. PPARα Is Necessary for Radiation-Induced Activation of Noncanonical TGFβ Signaling in the Heart. J Proteome Res 2018; 17:1677-1689. [PMID: 29560722 DOI: 10.1021/acs.jproteome.8b00001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High-dose ionizing radiation is known to induce adverse effects such as inflammation and fibrosis in the heart. Transcriptional regulators PPARα and TGFβ are known to be involved in this radiation response. PPARα, an anti-inflammatory transcription factor controlling cardiac energy metabolism, is inactivated by irradiation. The pro-inflammatory and pro-fibrotic TGFβ is activated by irradiation via SMAD-dependent and SMAD-independent pathways. The goal of this study was to investigate how altering the level of PPARα influences the radiation response of these signaling pathways. For this purpose, we used genetically modified C57Bl/6 mice with wild type (+/+), heterozygous (+/-) or homozygous (-/-) PPARα genotype. Mice were locally irradiated to the heart using doses of 8 or 16 Gy; the controls were sham-irradiated. The heart tissue was investigated using label-free proteomics 20 weeks after the irradiation and the predicted pathways were validated using immunoblotting, ELISA, and immunohistochemistry. The heterozygous PPARα mice showed most radiation-induced changes in the cardiac proteome, whereas the homozygous PPARα mice showed the least changes. Irradiation induced SMAD-dependent TGFβ signaling independently of the PPARα status, but the presence of PPARα was necessary for the activation of the SMAD-independent pathway. These data indicate a central role of PPARα in cardiac response to ionizing radiation.
Collapse
Affiliation(s)
| | | | | | - Wolfgang Sievert
- Center for Translational Cancer Research (TranslaTUM), Radiation Immuno Oncology Group , Campus Klinikum rechts der Isar, Technical University of Munich , Munich 81675 , Germany
| | | | - Mariateresa Mancuso
- Laboratory of Radiation Biology and Biomedicine , Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA) , Rome 00196 , Italy
| | - Emanuela Pasquali
- Laboratory of Radiation Biology and Biomedicine , Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA) , Rome 00196 , Italy
| | - Gabriele Multhoff
- Center for Translational Cancer Research (TranslaTUM), Radiation Immuno Oncology Group , Campus Klinikum rechts der Isar, Technical University of Munich , Munich 81675 , Germany
| | - Bastian Popper
- Department of Cell Biology and Core Facility Animal Models (CAM), Biomedical Center , Ludwig-Maximilians University Munich , Planegg 80539 , Germany
| | | | | | | |
Collapse
|
17
|
Abstract
The heart failure accounts for the highest mortality rate all over the world. The development of preventive therapeutic approaches is still in their infancy. Owing to the extremely high energy demand of the heart, the bioenergetics pathways need to respond efficiently based on substrate availability. The metabolic regulation of such heart bioenergetics is mediated by various rate limiting enzymes involved in energy metabolism. Although all the pertinent mechanisms are not clearly understood, the progressive decline in the activity of metabolic enzymes leading to diminished ATP production is known to cause progression of the heart failure. Therefore, metabolic therapy that can maintain the appropriate activities of metabolic enzymes can be a promising approach for the prevention and treatment of the heart failure. The flavonoids that constitute various human dietary ingredients also effectively offer a variety of health benefits. The flavonoids target a variety of metabolic enzymes and facilitate effective management of the equilibrium between production and utilization of energy in the heart. This review discusses the broad impact of metabolic enzymes in the heart functions and explains how the dysregulated enzyme activity causes the heart failure. In addition, the prospects of targeting dysregulated metabolic enzymes by developing flavonoid-based metabolic approaches are discussed.
Collapse
|
18
|
Abushouk AI, El-Husseny MWA, Bahbah EI, Elmaraezy A, Ali AA, Ashraf A, Abdel-Daim MM. Peroxisome proliferator-activated receptors as therapeutic targets for heart failure. Biomed Pharmacother 2017; 95:692-700. [PMID: 28886529 DOI: 10.1016/j.biopha.2017.08.083] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/05/2017] [Accepted: 08/23/2017] [Indexed: 01/06/2023] Open
Abstract
Heart failure (HF) is a common clinical syndrome that affects more than 23 million individuals worldwide. Despite the marked advances in its management, the mortality rates in HF patients have remained unacceptably high. Peroxisome proliferator-activated receptors (PPARs) are nuclear transcription regulators, involved in the regulation of fatty acid and glucose metabolism. PPAR agonists are currently used for the treatment of type II diabetes mellitus and hyperlipidemia; however, their role as therapeutic agents for HF remains under investigation. Preclinical studies have shown that pharmacological modulation of PPARs can upregulate the expression of fatty acid oxidation genes in cardiomyocytes. Moreover, PPAR agonists were proven able to improve ventricular contractility and reduce cardiac remodelling in animal models through their anti-inflammatory, anti-oxidant, anti-fibrotic, and anti-apoptotic activities. Whether these effects can be replicated in humans is yet to be proven. This article reviews the interactions of PPARs with the pathophysiological mechanisms of HF and how the pharmacological modulation of these receptors can be of benefit for HF patients.
Collapse
Affiliation(s)
| | | | - Eshak I Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Ahmed Elmaraezy
- NovaMed Medical Research Association, Cairo, Egypt; Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Aya Ashraf Ali
- Faculty of Medicine, Minia University, Minia, Egypt; Minia Medical Research Society, Minia University, Minia, Egypt
| | - Asmaa Ashraf
- Faculty of Medicine, Minia University, Minia, Egypt; Minia Medical Research Society, Minia University, Minia, Egypt
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt; Department of Ophthalmology and Micro-Technology, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
19
|
Sun Y, Han M, Shen Z, Huang H, Miao X. Anti-hypertensive and cardioprotective effects of a novel apitherapy formulation via upregulation of peroxisome proliferator-activated receptor-α and -γ in spontaneous hypertensive rats. Saudi J Biol Sci 2017; 25:213-219. [PMID: 29472767 PMCID: PMC5816011 DOI: 10.1016/j.sjbs.2017.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 02/01/2023] Open
Abstract
Ventricular remodeling is associated with many heart diseases, and ventricular remodeling induced by hypertension can be fatal independent of hypertension. In this study, we prepared a novel apitherapy formulation, designated Bao-Yuan-Ling (BYL), which contained propolis, royal jelly, and bee venom, to treat spontaneous hypertensive rats (SHRs). We then evaluated the pharmacology of BYL and the potential mechanisms through which BYL affects hypertension and ventricular remodeling. We found that BYL treatment could reduce blood pressure in SHRs. Thereafter, we found that BYL treatment reduced serum levels of angiotensin II, endothelin 1, and transforming growth factor-β and improved the myocardial structure. Moreover, the results of quantitative real-time polymerase chain reaction indicated that BYL treatment could upregulate the mRNA expression of peroxisome proliferator-activated receptor (PPAR)-α and PPAR-γ. Thus, we could conclude that BYL had hypotensive and cardioprotective effects in SHRs, potentially through improvement of myocardial energy metabolism.
Collapse
Affiliation(s)
- Yanru Sun
- College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350000, China.,College of Bee Science, Fujian Agriculture and Forestry University, Fuzhou 350000, China
| | - Mingfeng Han
- National United Engineering Laboratory of Natural Biological Toxins, Fuzhou 350000, China
| | - Zhenhuang Shen
- National United Engineering Laboratory of Natural Biological Toxins, Fuzhou 350000, China
| | - Haibo Huang
- National United Engineering Laboratory of Natural Biological Toxins, Fuzhou 350000, China
| | - Xiaoqing Miao
- College of Bee Science, Fujian Agriculture and Forestry University, Fuzhou 350000, China.,National United Engineering Laboratory of Natural Biological Toxins, Fuzhou 350000, China
| |
Collapse
|
20
|
Azimzadeh O, Tapio S. Proteomics landscape of radiation-induced cardiovascular disease: somewhere over the paradigm. Expert Rev Proteomics 2017; 14:987-996. [PMID: 28976223 DOI: 10.1080/14789450.2017.1388743] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Epidemiological studies clearly show that thoracic or whole body exposure to ionizing radiation increases the risk of cardiac morbidity and mortality. Radiation-induced cardiovascular disease (CVD) has been intensively studied during the last ten years but the underlying molecular mechanisms are still poorly understood. Areas covered: Heart proteomics is a powerful tool holding promise for the future research. The central focus of this review is to compare proteomics data on radiation-induced CVD with data arising from proteomics of healthy and diseased cardiac tissue in general. In this context we highlight common and unique features of radiation-related and other heart pathologies. Future prospects and challenges of the field are discussed. Expert commentary: Data from comprehensive cardiac proteomics have deepened the knowledge of molecular mechanisms involved in radiation-induced cardiac dysfunction. State-of-the-art proteomics has the potential to identify novel diagnostic and therapeutic markers of this disease.
Collapse
Affiliation(s)
- Omid Azimzadeh
- a Institute of Radiation Biology , Helmholtz Zentrum München, German Research Center for Environmental Health GmbH , Neuherberg , Germany
| | - Soile Tapio
- a Institute of Radiation Biology , Helmholtz Zentrum München, German Research Center for Environmental Health GmbH , Neuherberg , Germany
| |
Collapse
|
21
|
Maccallini C, Mollica A, Amoroso R. The Positive Regulation of eNOS Signaling by PPAR Agonists in Cardiovascular Diseases. Am J Cardiovasc Drugs 2017; 17:273-281. [PMID: 28315197 DOI: 10.1007/s40256-017-0220-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Increasing evidence shows that activation of peroxisome proliferator-activated receptors (PPARs) plays an essential role in the regulation of vascular endothelial function through a range of mechanisms, including non-metabolic. Among these, the PPAR-mediated activation of endothelial nitric oxide synthase (eNOS) appears to be of considerable importance. The regulated and sustained bioavailability of nitric oxide (NO) in the endothelium is essential to avoid the development of cardiovascular diseases such as hypertension or atherosclerosis. Therefore, a deeper understanding of the different effects of specific PPAR ligands on NO bioavailability could be useful in the development of novel or multi-targeted PPAR agonists. In this review, we report the most meaningful and up-to-date in vitro and in vivo studies of the regulation of NO production performed by different PPAR agonists. Insights into the molecular mechanisms of PPAR-mediated eNOS activation are also provided. Although findings from animal studies in which the activation of PPARα, PPARβ/δ, or PPARγ have provided clear vasoprotective effects have been promising, several benefits from PPAR agonists are offset by unwanted outcomes. Therefore, new insights could be useful in the development of tissue-targeted PPAR agonists with more tolerable side effects to improve treatment options for cardiovascular diseases.
Collapse
|
22
|
Tapio S. Using proteomics to explore the effects of radiation on the heart - impacts for medicine. Expert Rev Proteomics 2017; 14:277-279. [DOI: 10.1080/14789450.2017.1294067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Soile Tapio
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| |
Collapse
|
23
|
Ramipril restores PPARβ/δ and PPARγ expressions and reduces cardiac NADPH oxidase but fails to restore cardiac function and accompanied myosin heavy chain ratio shift in severe anthracycline-induced cardiomyopathy in rat. Eur J Pharmacol 2016; 791:244-253. [DOI: 10.1016/j.ejphar.2016.08.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 01/25/2023]
|
24
|
Pellegrin M, Szostak J, Bouzourène K, Aubert JF, Berthelot A, Nussberger J, Laurant P, Mazzolai L. Running Exercise and Angiotensin II Type I Receptor Blocker Telmisartan Are Equally Effective in Preventing Angiotensin II-Mediated Vulnerable Atherosclerotic Lesions. J Cardiovasc Pharmacol Ther 2016; 22:159-168. [PMID: 27246357 DOI: 10.1177/1074248416652235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The present study was conducted to directly compare the efficacy of running exercise and telmisartan treatment on angiotensin (Ang) II-mediated atherosclerosis and plaque vulnerability. MATERIALS AND METHODS Apolipoprotein E-deficient (ApoE-/-) mice with Ang II-mediated atherosclerosis (2-kidney, 1-clip [2K1C] renovascular hypertension model) were randomized into 3 groups: treadmill running exercise (RUN), telmisartan treatment (TEL), and sedentary untreated controls (SED) for 5 weeks. Atherosclerosis was assessed using histological and immunohistochemical analyses. Gene expression was determined by real-time reverse transcription polymerase chain reaction. RESULTS TEL but not RUN mice significantly decreased (50%) atherosclerotic lesion size compared to SED. RUN and TEL promoted plaque stabilization to a similar degree in ApoE-/- 2K1C mice. However, plaque composition and vascular inflammatory markers were differently affected: RUN decreased plaque macrophage infiltration (35%), whereas TEL reduced lipid core size (88%); RUN significantly increased aortic peroxisome proliferator-activated receptor (PPAR)-α, -δ, and -γ expression, whereas TEL significantly modulated T-helper 1/T-helper 2 (Th1/Th2) aortic response toward an anti-inflammatory state (decreased aortic interleukin [IL] 2 to IL-10 and IL-2 to IL-13 expression ratios). Plaque smooth muscle cell content was similarly increased (128% and 141%, respectively). Aortic AT1 and AT2 receptor expression as well as aortic CD11c/CD206 and IL-1β/IL-1ra expression ratios were not significantly modulated by either RUN or TEL. CONCLUSION Running exercise and telmisartan treatment are equally effective in preventing Ang II-mediated plaque vulnerability but through distinct cellular and molecular mechanisms. Our findings further support the use of exercise training and selective AT1 receptor blocker therapies for atherosclerotic cardiovascular disease prevention.
Collapse
Affiliation(s)
- Maxime Pellegrin
- 1 Division of Angiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Justyna Szostak
- 1 Division of Angiology, University Hospital of Lausanne, Lausanne, Switzerland.,2 Sciences Séparatives Biologiques et Pharmaceutiques, UFR STAPS/SMP, Université de Franche-Comté, Besançon, France
| | - Karima Bouzourène
- 1 Division of Angiology, University Hospital of Lausanne, Lausanne, Switzerland
| | | | - Alain Berthelot
- 2 Sciences Séparatives Biologiques et Pharmaceutiques, UFR STAPS/SMP, Université de Franche-Comté, Besançon, France
| | - Jürg Nussberger
- 1 Division of Angiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Pascal Laurant
- 3 Laboratoire Pharm-Ecologie Cardiovasculaire, UFRip Sciences Technologie et Santé, Université Avignon et des Pays de Vaucluse, Avignon, France
| | - Lucia Mazzolai
- 1 Division of Angiology, University Hospital of Lausanne, Lausanne, Switzerland
| |
Collapse
|
25
|
Czarnowska E, Bierła JB, Toczek M, Tyrankiewicz U, Pająk B, Domal-Kwiatkowska D, Ratajska A, Smoleński RT, Mende U, Chłopicki S. Narrow time window of metabolic changes associated with transition to overt heart failure in Tgaq*44 mice. Pharmacol Rep 2016; 68:707-14. [PMID: 27126697 DOI: 10.1016/j.pharep.2016.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND The timing and consequences of alternations in substrate utilization in heart failure (HF) and their relationship with structural changes remain unclear. This study aimed to analyze metabolic changes associated with transition to overt heart failure in transgenic mouse model of HF resulting from cardiac-specific overexpression of constitutively active Gαq*. METHODS Structural changes quantified by morphometry, relative cardiac mRNA and protein expression of PPARα, FAT/CD36, CPT-1, GLUT-4 and glycolytic efficiency following administration of 1-(13)C glucose were investigated in 4-14-month-old Tgαq*44 mice (TG), compared with age-matched FVB wild type mice (WT). RESULTS Initial hypertrophy in TG (4-10-month of age) was featured by an accelerated glycolytic pathway that was not accompanied by structural changes in cardiomyocytes. In 10-month-old TG, cardiomyocyte elongation and hypertrophic remodeling and increased glycolytic flux was accompanied by relatively low expression of FAT/CD36, CPT-1 and PPARα. During the transition phase (12-month-old TG), a pronounced increase in PPARα with an increase in relative fatty acid (FA) flux was associated with anomalies of cardiomyocytes with accumulation of lipid droplets and glycogen as well as cell death. At the stage of overt heart failure (14-month-old TG), an accelerated glycolytic pathway with a decline in FA oxidation was accompanied by further structural changes. CONCLUSION Tgαq*44 mice display three distinct phases of metabolic/structural changes during hypertrophy and progression to HF, with relatively short period of increase in FA metabolism, highlighting a narrow metabolic changes associated with transition to overt heart failure in Tgaq*44 mice that have therapeutic significance.
Collapse
Affiliation(s)
- Elżbieta Czarnowska
- Department of Pathology, The Children's Memorial Health Institute, Warszawa, Poland.
| | - Joanna B Bierła
- Department of Pathology, The Children's Memorial Health Institute, Warszawa, Poland
| | - Marta Toczek
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Urszula Tyrankiewicz
- Department of Magnetic Resonance Imaging Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland; Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
| | - Beata Pająk
- Electron Microscopy Platform, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warszawa, Poland; Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warszawa, Poland
| | - Dorota Domal-Kwiatkowska
- Department of Biochemistry, Medical University of Silesia, School of Pharmacy with the Division of Laboratory Medicine, Sosnowiec, Poland
| | - Anna Ratajska
- Department of Pathology, The Medical University of Warsaw, Warszawa, Poland
| | | | - Ulrike Mende
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Stefan Chłopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland; Department of Experimental Pharmacology, Jagiellonian University Medical College, Kraków, Poland.
| |
Collapse
|
26
|
The Correlation of PPARα Activity and Cardiomyocyte Metabolism and Structure in Idiopathic Dilated Cardiomyopathy during Heart Failure Progression. PPAR Res 2016; 2016:7508026. [PMID: 26981112 PMCID: PMC4770161 DOI: 10.1155/2016/7508026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/11/2016] [Indexed: 12/20/2022] Open
Abstract
This study aimed to define relationship between PPARα expression and metabolic-structural characteristics during HF progression in hearts with DCM phenotype. Tissue endomyocardial biopsy samples divided into three groups according to LVEF ((I) 45–50%, n = 10; (II) 30–40%, n = 15; (III) <30%, n = 15; and control (donor hearts, >60%, n = 6)) were investigated. The PPARα mRNA expression in the failing hearts was low in Group (I), high in Group (II), and comparable to that of the control in Group (III). There were analogous changes in the expression of FAT/CD36 and CPT-1 mRNA in contrast to continuous overexpression of GLUT-4 mRNA and significant increase of PDK-4 mRNA in Group (II). In addition, significant structural changes of cardiomyocytes with glycogen accumulation were accompanied by increased expression of PPARα. For the entire study population with HF levels of FAT/CD36 mRNA showed a strong tendency of negative correlation with LVEF. In conclusion, PPARα elevated levels may be a direct cause of adverse remodeling, both metabolic and structural. Thus, there is limited time window for therapy modulating cardiac metabolism and protecting cardiomyocyte structure in failing heart.
Collapse
|
27
|
Hamza MS, Kumar C, Chia SM, Anandalakshmi V, Boo N, Strapps W, Robinson M, Caguyong M, Bartz S, Tadin-Strapps M, van Gool A, Shih SJ. Alterations in the hepatic transcriptional landscape after RNAi mediated ApoB silencing in cynomolgus monkeys. Atherosclerosis 2015; 242:383-95. [DOI: 10.1016/j.atherosclerosis.2015.07.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 06/09/2015] [Accepted: 07/18/2015] [Indexed: 12/25/2022]
|
28
|
Auer PL, Nalls M, Meschia JF, Worrall BB, Longstreth WT, Seshadri S, Kooperberg C, Burger KM, Carlson CS, Carty CL, Chen WM, Cupples LA, DeStefano AL, Fornage M, Hardy J, Hsu L, Jackson RD, Jarvik GP, Kim DS, Lakshminarayan K, Lange LA, Manichaikul A, Quinlan AR, Singleton AB, Thornton TA, Nickerson DA, Peters U, Rich SS. Rare and Coding Region Genetic Variants Associated With Risk of Ischemic Stroke: The NHLBI Exome Sequence Project. JAMA Neurol 2015; 72:781-8. [PMID: 25961151 PMCID: PMC4673986 DOI: 10.1001/jamaneurol.2015.0582] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
IMPORTANCE Stroke is the second leading cause of death and the third leading cause of years of life lost. Genetic factors contribute to stroke prevalence, and candidate gene and genome-wide association studies (GWAS) have identified variants associated with ischemic stroke risk. These variants often have small effects without obvious biological significance. Exome sequencing may discover predicted protein-altering variants with a potentially large effect on ischemic stroke risk. OBJECTIVE To investigate the contribution of rare and common genetic variants to ischemic stroke risk by targeting the protein-coding regions of the human genome. DESIGN, SETTING, AND PARTICIPANTS The National Heart, Lung, and Blood Institute (NHLBI) Exome Sequencing Project (ESP) analyzed approximately 6000 participants from numerous cohorts of European and African ancestry. For discovery, 365 cases of ischemic stroke (small-vessel and large-vessel subtypes) and 809 European ancestry controls were sequenced; for replication, 47 affected sibpairs concordant for stroke subtype and an African American case-control series were sequenced, with 1672 cases and 4509 European ancestry controls genotyped. The ESP's exome sequencing and genotyping started on January 1, 2010, and continued through June 30, 2012. Analyses were conducted on the full data set between July 12, 2012, and July 13, 2013. MAIN OUTCOMES AND MEASURES Discovery of new variants or genes contributing to ischemic stroke risk and subtype (primary analysis) and determination of support for protein-coding variants contributing to risk in previously published candidate genes (secondary analysis). RESULTS We identified 2 novel genes associated with an increased risk of ischemic stroke: a protein-coding variant in PDE4DIP (rs1778155; odds ratio, 2.15; P = 2.63 × 10(-8)) with an intracellular signal transduction mechanism and in ACOT4 (rs35724886; odds ratio, 2.04; P = 1.24 × 10(-7)) with a fatty acid metabolism; confirmation of PDE4DIP was observed in affected sibpair families with large-vessel stroke subtype and in African Americans. Replication of protein-coding variants in candidate genes was observed for 2 previously reported GWAS associations: ZFHX3 (cardioembolic stroke) and ABCA1 (large-vessel stroke). CONCLUSIONS AND RELEVANCE Exome sequencing discovered 2 novel genes and mechanisms, PDE4DIP and ACOT4, associated with increased risk for ischemic stroke. In addition, ZFHX3 and ABCA1 were discovered to have protein-coding variants associated with ischemic stroke. These results suggest that genetic variation in novel pathways contributes to ischemic stroke risk and serves as a target for prediction, prevention, and therapy.
Collapse
Affiliation(s)
- Paul L Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin, Milwaukee2Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mike Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | | | | | - W T Longstreth
- Department of Neurology, University of Washington, Seattle
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kathleen M Burger
- Department of Neurology, George Washington University Hospital, Washington, DC
| | - Christopher S Carlson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Cara L Carty
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Wei-Min Chen
- Center for Public Health Genomics, University of Virginia, Charlottesville
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Myriam Fornage
- Institute for Molecular Medicine, University of Texas Health Science Center, Houston
| | - John Hardy
- Department of Neuroscience, Reta Lila Weston Institute, University College London, London, England
| | - Li Hsu
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rebecca D Jackson
- Department of Internal Medicine, The Ohio State University, Columbus
| | - Gail P Jarvik
- Department of Medicine, University of Washington, Seattle
| | - Daniel S Kim
- Department of Genome Sciences, University of Washington, Seattle
| | | | - Leslie A Lange
- Department of Genetics, University of North Carolina, Chapel Hill
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville
| | - Aaron R Quinlan
- Center for Public Health Genomics, University of Virginia, Charlottesville
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | | | | | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville
| |
Collapse
|
29
|
Thomas M, Bayha C, Klein K, Müller S, Weiss TS, Schwab M, Zanger UM. The truncated splice variant of peroxisome proliferator-activated receptor alpha, PPARα-tr, autonomously regulates proliferative and pro-inflammatory genes. BMC Cancer 2015; 15:488. [PMID: 26122096 PMCID: PMC4485637 DOI: 10.1186/s12885-015-1500-x] [Citation(s) in RCA: 23] [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: 01/22/2015] [Accepted: 06/19/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The peroxisome proliferator-activated receptor alpha (PPARα) controls lipid/energy homeostasis and inflammatory responses. The truncated splice variant PPARα-tr was suggested to exert a dominant negative function despite being unable to bind consensus PPARα DNA response elements. METHODS The distribution and variability factor of each PPARα variant were assessed in the well-characterized cohort of human liver samples (N = 150) on the mRNA and protein levels. Specific siRNA-mediated downregulation of each transcript as well as specific overexpression with subsequent qRT-PCR analysis of downstream genes was used for investigation of specific functional roles of PPARα-wt and PPARα-tr forms in primary human hepatocytes. RESULTS Bioinformatic analyses of genome-wide liver expression profiling data suggested a possible role of PPARα-tr in downregulating proliferative and pro-inflammatory genes. Specific gene silencing of both forms in primary human hepatocytes showed that induction of metabolic PPARα-target genes by agonist WY14,643 was prevented by PPARα-wt knock-down but neither prevented nor augmented by PPARα-tr knock-down. WY14,643 treatment did not induce proliferative genes including MYC, CDK1, and PCNA, and knock-down of PPARα-wt had no effect, while PPARα-tr knock-down caused up to 3-fold induction of these genes. Similarly, induction of pro-inflammatory genes IL1B, PTGS2, and CCL2 by IL-6 was augmented by knock-down of PPARα-tr but not of PPARα-wt. In contrast to human proliferative genes, orthologous mouse genes were readily inducible by WY14,643 in PPARα-tr non-expressing AML12 mouse hepatocytes. Induction was augmented by overexpression of PPARα-wt and attenuated by overexpression of PPARα-tr. Pro-inflammatory genes including IL-1β, CCL2 and TNFα were induced by WY14,643 in mouse and human cells and both PPARα forms attenuated induction. As potential mechanism of PPARα-tr inhibitory action we suggest crosstalk with WNT/β-catenin pathway. Finally, treatment with WY14,643 in the presence of PPARα-tr resulted in the significant reduction of cell viability of AML12 and human ovarian cancer cell line, SKOV3. CONCLUSIONS Our data suggest that the truncated PPARα splice variant functions as an endogenous inhibitor of proliferative and pro-inflammatory genes in human cells and that its absence in mouse may explain species-specific differences in fibrate-induced hepatocarcinogenesis.
Collapse
Affiliation(s)
- Maria Thomas
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70736, Stuttgart, and University of Tuebingen, Tuebingen, Germany.
| | - Christine Bayha
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70736, Stuttgart, and University of Tuebingen, Tuebingen, Germany.
| | - Kathrin Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70736, Stuttgart, and University of Tuebingen, Tuebingen, Germany.
| | - Simon Müller
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70736, Stuttgart, and University of Tuebingen, Tuebingen, Germany.
- Present address: MUON-STAT, Klugestraße 28, 70197, Stuttgart, Germany.
| | - Thomas S Weiss
- University Children Hospital (KUNO), Regensburg University Hospital, Regensburg, Germany.
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70736, Stuttgart, and University of Tuebingen, Tuebingen, Germany.
- Department of Clinical Pharmacology, University of Tuebingen, Tuebingen, Germany.
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70736, Stuttgart, and University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
30
|
Li QY, Chen L, Yan MM, Shi XJ, Zhong MK. Tectorigenin regulates adipogenic differentiation and adipocytokines secretion via PPARγ and IKK/NF-κB signaling. PHARMACEUTICAL BIOLOGY 2015; 53:1567-1575. [PMID: 25856699 DOI: 10.3109/13880209.2014.993038] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Obesity is associated with a number of diseases with metabolic abnormalities such as type 2 diabetes (T2D). OBJECTIVE We investigate the effects of tectorigenin on 3T3-L1 preadipocyte differentiation and adipocytokines secretion. MATERIALS AND METHODS The effects of tectorigenin on adipocyte differentiation were studied using Oil Red O staining. Effects of tectorigenin on adipogenesis-related genes expression and adipocytokines secretion were measured by the real-time quantitative RT-PCR and ELISA method, respectively. Reporter gene assays were performed to determine the PPARγ and NF-κB transactivation. We also used [(3)H]-2-deoxy-d-glucose to study the glucose uptake, and the IKK/NF-κB signaling pathway was assessed by western blot analysis. HFD/STZ rats were used to evaluate the therapeutic efficacies of tectorigenin. RESULTS Tectorigenin 10, 25, 50, and 75 μM inhibited 3T3-L1 adipogenesis and related genes transcription. TNF-α-induced changes of IL-6, MCP-1, as well as adiponectin in 3T3-L1 were markedly reversed by tectorigenin at 75 μM. Further investigation using reporter gene revealed that tectorigenin was a partial PPARγ agonist with an IC50 value of 13.3 μM. Tectorigenin improved basal and insulin-stimulated glucose uptake in mature 3T3-L1 adipocytes. Moreover, tectorigenin antagonized TNF-α-induced NF-κB transactivation and p65 nuclear translocation. Although tectorigenin (50 and 100 mg/kg) displayed the ability to promote insulin sensitivity and improve glucose metabolism in HFD/STZ rats, it did not cause significant side effects such as body weight gain, fluid retention, or cardiac hypertrophy. DISCUSSION AND CONCLUSION These results suggest that tectorigenin may ameliorate hyperglycemia by blocking preadipocyte differentiation and adipocytokines secretion in which PPARγ and NF-κB signaling pathways were involved.
Collapse
Affiliation(s)
- Qun-Yi Li
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University , Shanghai , China
| | | | | | | | | |
Collapse
|
31
|
Lee SJ, Jia Y. The effect of bioactive compounds in tea on lipid metabolism and obesity through regulation of peroxisome proliferator-activated receptors. Curr Opin Lipidol 2015; 26:3-9. [PMID: 25551797 DOI: 10.1097/mol.0000000000000145] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE OF REVIEW The hypolipidemic and antiobesogenic effects of tea intake have been associated with bioactive compounds that regulate peroxisome proliferator-activated receptors (PPARs). This review describes the recent research on two of these compounds, (-)-epigallocatechin gallate (EGCG) and linalool. RECENT FINDINGS Catechins (specifically EGCG) are key bioactive compounds found in tea, and a recent study has shown that linalool may also be an active tea compound. These compounds act on lipid metabolism by regulating PPAR subtypes. EGCG inhibits the key adipogenic transcription factor PPARγ while activating PPARα, whereas linalool is a PPARα agonist activating hepatic fatty acid uptake and subsequent oxidation to reduce plasma triglyceride levels. SUMMARY The collective activities of EGCG and linalool in tea may exert hypolipidemic and antiobesogenic effects by regulating PPARs. The research summarized in this review expands our understanding of the biological and physiological mechanisms of the bioactive compounds found in tea.
Collapse
Affiliation(s)
- Sung-Joon Lee
- aDepartment of Biotechnology, Graduate School of Life Sciences & Biotechnology and Brain Korea 21-PLUS Program bDepartment of Food Biosciences and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | | |
Collapse
|
32
|
Peroxisome Proliferator-Activated Receptor-Gamma Agonists for Alzheimer’s Disease and Amnestic Mild Cognitive Impairment: A Systematic Review and Meta-Analysis. Drugs Aging 2014; 32:57-65. [DOI: 10.1007/s40266-014-0228-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
33
|
Effect of peroxisome proliferator-activated receptor gamma agonist on heart of rabbits with acute myocardial ischemia/reperfusion injury. ASIAN PAC J TROP MED 2014; 7:271-5. [PMID: 24507674 DOI: 10.1016/s1995-7645(14)60036-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/15/2014] [Accepted: 02/15/2014] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To explore protective effect of rosiglitazone on myocardial ischemia reperfusion injury. METHODS A total of 48 male SD rats were randomly divided into control group (A), I/R group(B), high dose of rosiglitazone (C), low dose of rosiglitazone (D). Plasm concentration of creatine kinase (CK), CK-MB, hsCRP, Superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), nitric oxide (NO) and endothelin (ET) were measured 1 h later after I/R. 24 h after I/R hearts were harvested to observe pathological and ultrastructural changes. Immunohistochemistry and western blotting was used to test CD40 expression in myocardial tissue. Area of myocardial infarction were tested, arrhythmia rate during I/R was recorded. RESULTS Plasm concentration of creatine kinase (CK), CK-MB, hsCRP, NO, MDA and ET were decreased in group C, D compared with group B. Plasm concentration of T-SOD and GSH-Px was increased significantly in group C, D compared with group B. Compared with group B, pathological and ultrastructural changes in group C, D were slightly. Myocardial infarction area and arrhythmia rate were lower in group C, D compare with group B. CONCLUSIONS Rosiglitazone can protect myocardium from I/R injury by enhancing T-SOD and GSH-Px concentration, inhibit inflammatory reaction, improve endothelial function, reduce oxidative stress and calcium overload.
Collapse
|
34
|
Telmisartan inhibits the proinflammatory effects of homocysteine on human endothelial cells through activation of the peroxisome proliferator-activated receptor-δ pathway. Int J Mol Med 2014; 34:828-34. [PMID: 24994548 DOI: 10.3892/ijmm.2014.1834] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 06/11/2014] [Indexed: 11/05/2022] Open
Abstract
The aim of this study was to investigate the inhibition capacity of telmisartan to endothelial inflammation induced by homocysteine (Hcy) and discuss the proposed mechanism in vitro. Human umbilical vein endothelial cells (HUVECs) were prepared by collagenase digestion and cultured in vitro. An increase in monocyte chemoattractant protein-1 (MCP-1) and vascular cell adhesion molecule-1 (VCAM-1) was analyzed by reverse transcription-polymerase chain reaction (RT-PCR) as markers of Hcy-induced endothelial inflammation. HL-60 cell adhesion to HUVECs was measured by rose bengal staining. Nuclear, cytosolic and total nuclear transcription factor-κB (NF-κB) p65 levels were analyzed by western blotting. Peroxisome proliferator-activated receptor-δ (PPARδ) expression by HUVECs exposed to Hcy with or without telmisartan pretreatment was analyzed by RT-PCR and western blotting. Hcy significantly increased the levels of MCP-1 mRNA, VCAM-1 mRNA and monocyte binding to HUVECs. These effects were significantly attenuated by pretreatment with telmisartan and PPARδ agonists. The effect of telmisartan was inhibited by PPARδ antagonists. The Hcy-mediated downregulation of PPARδ mRNA and protein of HUVECs was inhibited by telmisartan. Hcy-mediated upregulation of NF-κB p65 protein levels in nuclear extracts was inhibited by telmisartan and PPARδ agonists. In conclusion, telmisartan exerts potent anti-inflammatory effects in endothelial cells, probably via a binary mechanism involving PPARδ activation and inhibition of the nuclear translocation of NF-κB.
Collapse
|
35
|
van Bilsen M, Planavila A. Fatty acids and cardiac disease: fuel carrying a message. Acta Physiol (Oxf) 2014; 211:476-90. [PMID: 24773697 DOI: 10.1111/apha.12308] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/02/2014] [Accepted: 04/25/2014] [Indexed: 12/20/2022]
Abstract
From the viewpoint of the prevention of cardiovascular disease (CVD) burden, there has been a continuous interest in the detrimental effects of the Western-type high-fat diet for more than half a century. More recently, this general view has been subject to change as epidemiological studies showed that replacing fat by carbohydrate may even be worse and that various polyunsaturated fatty acids (FA) have beneficial rather than detrimental effects on CVD outcome. At the same time, advances in lipid biology have provided insight into the mechanisms by which the different lipid components of the Western diet affect the cardiovascular system. In fact, this still is a rapidly growing field of research and in recent years novel FA derivatives and FA receptors have been discovered. This includes fish-oil derived FA-derivatives with anti-inflammatory properties, the so-called resolvins, and various G-protein-coupled receptors that recognize FA as ligands. In the present review, we will extensively discuss the role of FA and their metabolites on cardiac disease, with special emphasis on the role of the different saturated and polyunsaturated FA and their respective metabolites in cellular signal transduction and the possible implications for the development of cardiac hypertrophy and cardiac failure.
Collapse
Affiliation(s)
- M. van Bilsen
- Department of Physiology; Cardiovascular Research Institute Maastricht; Maastricht University; Maastricht the Netherlands
| | - A. Planavila
- Departament de Bioquímica i Biologia Molecular; Institut de Biomedicina de la Universitat de Barcelona (IBUB); Universitat de Barcelona and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Barcelona Spain
| |
Collapse
|
36
|
Regulation of the human thioredoxin gene promoter and its key substrates: a study of functional and putative regulatory elements. Biochim Biophys Acta Gen Subj 2013; 1840:303-14. [PMID: 24041992 DOI: 10.1016/j.bbagen.2013.09.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Revised: 07/26/2013] [Accepted: 09/06/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND The thioredoxin system maintains redox balance through the action of thioredoxin and thioredoxin reductase. Thioredoxin regulates the activity of various substrates, including those that function to counteract cellular oxidative stress. These include the peroxiredoxins, methionine sulfoxide reductase A and specific transcription factors. Of particular relevance is Redox Factor-1, which in turn activates other redox-regulated transcription factors. SCOPE OF REVIEW Experimentally defined transcription factor binding sites in the human thioredoxin and thioredoxin reductase gene promoters together with promoters of the major thioredoxin system substrates involved in regulating cellular redox status are discussed. An in silico approach was used to identify potential putative binding sites for these transcription factors in all of these promoters. MAJOR CONCLUSIONS Our analysis reveals that many redox gene promoters contain the same transcription factor binding sites. Several of these transcription factors are in turn redox regulated. The ARE is present in several of these promoters and is bound by Nrf2 during various oxidative stress stimuli to upregulate gene expression. Other transcription factors also bind to these promoters during the same oxidative stress stimuli, with this redundancy supporting the importance of the antioxidant response. Putative transcription factor sites were identified in silico, which in combination with specific regulatory knowledge for that gene promoter may inform future experiments. GENERAL SIGNIFICANCE Redox proteins are involved in many cellular signalling pathways and aberrant expression can lead to disease or other pathological conditions. Therefore understanding how their expression is regulated is relevant for developing therapeutic agents that target these pathways.
Collapse
|
37
|
Kianbakht S, Dabaghian FH. Improved glycemic control and lipid profile in hyperlipidemic type 2 diabetic patients consuming Salvia officinalis L. leaf extract: a randomized placebo. Controlled clinical trial. Complement Ther Med 2013; 21:441-6. [PMID: 24050577 DOI: 10.1016/j.ctim.2013.07.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 05/26/2013] [Accepted: 07/10/2013] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES Evaluation of the efficacy and safety of Salvia officinalis L. (S. officinalis) leaf extract in the treatment of hyperlipidemic type 2 diabetic patients. DESIGN Randomized placebo-controlled parallel group study. SETTING Diabetes Clinic (Karaj City, Alborz Province of Iran). INTERVENTIONS The efficacy and safety of taking S. officinalis leaf extract (one 500 mg capsule t.i.d. for 3 months) in treatment of 40 hyperlipidemic (hypercholesterolemic and/or hypertriglyceridemic) type 2 diabetic patients were evaluated and compared with the placebo group (n=40). MAIN OUTCOME MEASURES Fasting blood levels of glucose, glycosylated hemoglobin (HbA1c), total cholesterol, triglyceride, LDL-C (low density lipoprotein cholesterol), HDL-C (high density lipoprotein cholesterol), SGOT (serum glutamic-oxaloacetic transaminase), SGPT (serum glutamic-pyruvic transaminase) and creatinine. RESULTS The extract lowered fasting glucose, HbA1c, total cholesterol, triglyceride and LDL-C but increased HDL-C compared to baseline at endpoint. Percent difference mean (95% confidence interval) between the extract and placebo groups in terms of effects on fasting glucose, HbA1c, total cholesterol, triglyceride, LDL-C and HDL-C at endpoint were 32.2 (26.5, 37.9), 22.7 (16.8, 28.6), 16.9 (9.7, 24.1), 56.4 (36.1, 76.7), 35.6 (29.9, 41.3) and 27.6 (15.8, 39.4) (P=0.001, P=0.01, P=0.01, P=0.009, P<0.001 and P=0.008), respectively. Moreover, the extract did not have any significant effects on the other parameters compared to the placebo group at endpoint (P>0.05). No adverse effects were reported. CONCLUSIONS S. officinalis leaves may be safe and have anti-hyperglycemic and lipid profile improving effects in hyperlipidemic type 2 diabetic patients.
Collapse
Affiliation(s)
- S Kianbakht
- Department of Pharmacology and Applied Medicine, Research Institute of Medicinal Plants, ACECR, Karaj, Iran.
| | | |
Collapse
|
38
|
Wang W, Shi X, Yuan Y, Zhu H, Zhou W, Huang H, Feng M. Inhibitory effect of apolipoprotein A-I on matrix metalloproteinase-2 expression in vivo and in vitro. Acta Biochim Biophys Sin (Shanghai) 2013; 45:194-202. [PMID: 23299078 DOI: 10.1093/abbs/gms121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In the present study, we investigated the effects of apolipoprotein A-I (apoA-I) on matrix metalloproteinase-2 (MMP-2) expression in vivo and in vitro. First, we detected the effects of apoA-I on aorta MMP-2, peroxisome proliferator-activated receptor α/γ (PPAR α/γ), cyclooxygenase-2 (COX-2), and nuclear factor kappa B (NF-κB) expressions in atherosclerotic rabbit models using immunohistochemical methods. The results showed that the expressions of MMP-2, COX-2, and NF-κB were decreased in aortas of atherosclerotic rabbits treated with apoA-I, while PPAR α/γ expression was increased. Then, we chose the important inflammation cells, macrophages to testify those effects in vitro. Macrophages were divided into six groups and treated with different concentrations of apoA-I, the mRNA expressions of MMP-2, PPAR α/γ, and COX-2 were then determined using reverse-transcription polymerase chain reaction, and protein expression of PPAR γ, NF-κB were detected by western blot analysis. The levels of MMP-2 and PPAR α in cultured supernatants were determined using enzyme-linked immunosorbent assays. Interestingly, the in vitro results were similar to the results of the in vivo study. After incubation with apoA-I for 24 h, the expressions of MMP-2, COX-2, and NF-κB were decreased, while PPAR α/γ expression was increased. In consideration of their particular roles in the process of making plaque stable in vivo and in vitro, we speculate that the inhibitory effect of apoA-I on MMP-2 expression may have a close relationship with the effects of apoA-I on PPAR α/γ, COX-2, and NF-κB expressions. Although further research is needed to clarify the underlying mechanisms of these effects, our findings provide a novel insight into the anti-atherosclerotic plaque rupture effects of apoA-I.
Collapse
Affiliation(s)
- Weina Wang
- Department of Pharmacology, Fudan University, Shanghai 201203, China
| | | | | | | | | | | | | |
Collapse
|
39
|
Shiue YL, Chen LR, Tsai CJ, Yeh CY, Huang CT. Emerging roles of peroxisome proliferator-activated receptors in the pituitary gland in female reproduction. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.gmbhs.2013.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
40
|
Reversible inhibition of vasoconstriction by thiazolidinediones related to PI3K/Akt inhibition in vascular smooth muscle cells. Biochem Pharmacol 2013. [DOI: 10.1016/j.bcp.2012.11.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
41
|
Wy14643 improves vascular function in the aorta of the spontaneously hypertensive rat mainly by activating peroxisome proliferator-activated receptors alpha. Eur J Pharmacol 2012; 696:101-10. [DOI: 10.1016/j.ejphar.2012.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 09/05/2012] [Accepted: 09/17/2012] [Indexed: 12/15/2022]
|
42
|
Andriankaja OM, Galicia J, Dong G, Xiao W, Alawi F, Graves DT. Gene expression dynamics during diabetic periodontitis. J Dent Res 2012; 91:1160-5. [PMID: 23103632 DOI: 10.1177/0022034512465292] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Diabetes impairs the resolution of periodontal inflammation. We explored pathways altered by inflammation in the diabetic periodontium by using ligatures to induce periodontitis in type-2 diabetic Goto-Kakizaki rats. Ligatures were removed after 7 days, and rats were then treated with TNF inhibitor (pegsunercept) or vehicle alone and euthanized 4 days later. RNA was extracted from periodontal tissue, examined by mRNA profiling, and further analyzed by functional criteria. We found that 1,754 genes were significantly up-regulated and 1,243 were down-regulated by pegsunercept (p < 0.05). Functional analysis revealed up-regulation of neuron-associated and retina-associated gene clusters as well as those related to cell activity and signaling. Others were down-regulated by TNF inhibition and included genes associated with host defense, apoptosis, cell signaling and activity, and coagulation/hemostasis/complement. For selected genes, findings with microarray and rt-PCR agreed. PPAR-α was investigated further by immunohistochemistry due to its anti-inflammatory function and was found to be up-regulated in the gingiva during the resolution of periodontal inflammation and suppressed by diabetes. The results indicate that diabetes-enhanced inflammation both up- and down-regulates genes involved in cellular activity and cell signaling, while it predominantly up-regulates genes involved in the host response, apoptosis, and coagulation/homeostasis/complement and down-regulates mRNA levels of neuron, retina, and energy/metabolism-associated genes.
Collapse
Affiliation(s)
- O M Andriankaja
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | |
Collapse
|
43
|
Altieri P, Spallarossa P, Barisione C, Garibaldi S, Garuti A, Fabbi P, Ghigliotti G, Brunelli C. Inhibition of doxorubicin-induced senescence by PPARδ activation agonists in cardiac muscle cells: cooperation between PPARδ and Bcl6. PLoS One 2012; 7:e46126. [PMID: 23049957 PMCID: PMC3458009 DOI: 10.1371/journal.pone.0046126] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 08/28/2012] [Indexed: 01/29/2023] Open
Abstract
Senescence and apoptosis are two distinct cellular programs that are activated in response to a variety of stresses. Low or high doses of the same stressor, i.e., the anticancer drug doxorubicin, may either induce apoptosis or senescence, respectively, in cardiac muscle cells. We have demonstrated that PPARδ, a ligand-activated transcriptional factor that controls lipid metabolism, insulin sensitivity and inflammation, is also involved in the doxorubicin-induced senescence program. This occurs through its interference with the transcriptional repressor protein B cell lymphoma-6 (Bcl6). Low doses of doxorubicin increase the expression of PPARδ that sequesters Bcl6, thus preventing it from exerting its anti-senescent effects. We also found that L-165041, a specific PPARδ activator, is highly effective in protecting cardiomyocytes from doxorubicin-induced senescence through a Bcl6 related mechanism. In fact, L-165041 increases Bcl6 expression via p38, JNK and Akt activation, and at the same time it induces the release of Bcl6 from PPARδ, thereby enabling Bcl6 to bind to its target genes. L-165041 also prevented apoptosis induced by higher doses of doxorubicin. However, while experiments performed with siRNA analysis techniques very clearly showed the weight of Bcl6 in the cellular senescence program, no role was found for Bcl6 in the anti-apoptotic effects of L-165041, thus confirming that senescence and apoptosis are two very distinct stress response cellular programs. This study increases our understanding of the molecular mechanism of anthracycline cardiotoxicity and suggests a potential role for PPARδ agonists as cardioprotective agents.
Collapse
Affiliation(s)
- Paola Altieri
- Research Centre of Cardiovascular Biology, University of Genova, Genova, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Reel B, Guzeloglu M, Bagriyanik A, Atmaca S, Aykut K, Albayrak G, Hazan E. The effects of PPAR-γ agonist pioglitazone on renal ischemia/reperfusion injury in rats. J Surg Res 2012; 182:176-84. [PMID: 22981741 DOI: 10.1016/j.jss.2012.08.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 07/20/2012] [Accepted: 08/09/2012] [Indexed: 01/19/2023]
Abstract
BACKGROUND Acute renal failure due to renal ischemia/reperfusion (IR) injury is a significant clinical problem in cardiovascular surgery. Reactive oxygen species and inflammation play essential roles in the pathophysiology of IR injury. Matrix metalloproteinases (MMPs) are enzymes that play important roles in inflammation and mediate extracellular matrix degradation. It is known that peroxisome proliferator-activated receptor-γ agonists have antiinflammatory and antioxidant effects. In the present study, we aimed to investigate the effects of pioglitazone, a synthetic peroxisome proliferator-activated receptor-γ agonist, on MMPs and oxidative stress in a renal IR injury model in rats. MATERIALS AND METHODS Male Wistar albino rats were divided into three groups: control (n = 7), placebo (n = 7; saline/p.o.), and pioglitazone (n = 7; 5 mg/kg/day/p.o.). In the control group, a right nephrectomy was conducted without left renal IR injury. In the placebo and pioglitazone groups, pretreatments were started 3 d before operation. In both groups, left renal pedicles were clamped for 60 min and then reperfused for 60 min. Paraffinized renal sections were evaluated histopathologically. Furthermore, expressions of MMP-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-2, superoxide dismutase 1 (SOD1), and p47-phox/p67-phox subunits of NADPH oxidase were determined by immunostaining and scoring. RESULTS In the placebo group, renal IR injury induced diffuse tubular necrosis and intense acute inflammation, but pioglitazone inhibited these effects. MMP-2, MMP-9, and TIMP-2 expression increased in the placebo group. However, while MMP-2 and -9 expression decreased, TIMP-2 expression did not change in the pioglitazone group. p47-phox/p67-phox expression increased in the placebo group, but SOD1 expression did not change. Pioglitazone diminished p47-phox/p67-phox expression, whereas it enhanced SOD1 expression. CONCLUSION Our results suggest that pioglitazone might be helpful to reduce renal IR injury because of its antiinflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Buket Reel
- Faculty of Pharmacy, Department of Pharmacology, Ege University, Izmir, Turkey
| | | | | | | | | | | | | |
Collapse
|
45
|
Guzeloglu M, Reel B, Atmaca S, Bagrıyanık A, Hazan E. The effects of PPARγ agonist rosiglitazone on neointimal hyperplasia in rabbit carotid anastomosis model. J Cardiothorac Surg 2012; 7:57. [PMID: 22716287 PMCID: PMC3487767 DOI: 10.1186/1749-8090-7-57] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 06/08/2012] [Indexed: 11/13/2022] Open
Abstract
Background Neointimal hyperplasia involving smooth muscle cell (SMC) proliferation, migration and extracellular matrix (ECM) degradation is an important component of atherosclerosis. It develops as a response to vascular injury after balloon angioplasty and vascular graft placement. Matrix metalloproteinases (MMPs) induce SMC proliferation, migration and contribute to intimal hyperplasia by degrading ECM. PPARγ agonists inhibit SMC proliferation, migration and lesion formation. In this study, we aimed to investigate the effects of PPARγ agonist rosiglitazone on neointimal hyperplasia and gelatinase (MMP-2 and MMP-9) expressions in rabbit carotid anastomosis model. Methods New Zealand white rabbits (n = 13, 2.7–3.2 kg) were divided into placebo and treatment groups. Right carotid artery (CA) was transected and both ends were anastomosed. Treatment group (n = 6) received rosiglitazone (3 mg/kg/day/p.o.) and placebo group (n = 7) received PBS (phosphate buffered saline, 2.5 ml/kg/day/p.o.) for 4 weeks postoperatively. After the sacrification, right and left CAs were isolated. Morphometric analyses and immunohistochemical examinations for gelatinases were performed. Results Intimal area (0.055 ± 0.005 control vs 0.291 ± 0.020 μm2 anastomosed, p < 0,05) and index (0.117 ± 0.002 control vs 0.574 ± 0.013 anastomosed, p < 0,01) significantly increased in anastomosed arteries compared to control arteries from placebo group. However, in rosiglitazone-treated group, intimal area (0.291 ± 0.020 PBS vs 0.143 ± 0.027 rosiglitazone, p < 0,05) and index (0.574 ± 0.013 PBS vs 0.263 ± 0.0078 rosiglitazone, p < 0,01) significantly decreased. Furthermore, gelatinase immunopositivity was found to have significantly increased in anastomosed arteries from placebo group and decreased with rosiglitazone treatment. Conclusions These results suggest that rosiglitazone may prevent neointimal hyperplasia, which is the most important factor involved in late graft failure, by inhibiting gelatinase enzyme expression.
Collapse
Affiliation(s)
- Mehmet Guzeloglu
- Department of Cardiovascular Surgery, Faculty of Medicine, İzmir University, Izmir, Turkey.
| | | | | | | | | |
Collapse
|
46
|
Jia Y, Kim JY, Jun HJ, Kim SJ, Lee JH, Hoang MH, Hwang KY, Um SJ, Chang HI, Lee SJ. The natural carotenoid astaxanthin, a PPAR-α agonist and PPAR-γ antagonist, reduces hepatic lipid accumulation by rewiring the transcriptome in lipid-loaded hepatocytes. Mol Nutr Food Res 2012; 56:878-88. [DOI: 10.1002/mnfr.201100798] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | | | | | - Sun-Joong Kim
- Department of Biotechnology; Graduate School of Biotechnology; Korea University; Seoul; Republic of Korea
| | | | | | - Kwang-Yeon Hwang
- Department of Biotechnology; Graduate School of Biotechnology; Korea University; Seoul; Republic of Korea
| | - Soo-Jong Um
- Department of Bioscience and Biotechnology; Sejong University; Seoul; Republic of Korea
| | - Hyo Ihl Chang
- Department of Biotechnology; Graduate School of Biotechnology; Korea University; Seoul; Republic of Korea
| | | |
Collapse
|
47
|
Protection from Metabolic Dysregulation, Obesity, and Atherosclerosis by Citrus Flavonoids: Activation of Hepatic PGC1α-Mediated Fatty Acid Oxidation. PPAR Res 2012; 2012:857142. [PMID: 22701469 PMCID: PMC3369495 DOI: 10.1155/2012/857142] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 04/02/2012] [Indexed: 12/25/2022] Open
Abstract
Studies in a multitude of models including cell culture, animal and clinical studies demonstrate that citrus-derived flavonoids have therapeutic potential to attenuate dyslipidemia, correct hyperinsulinemia and hyperglycemia, and reduce atherosclerosis. Emerging evidence suggests the metabolic regulators, PPARα and PGC1α, are targets of the citrus flavonoids, and their activation may be at least partially responsible for mediating their metabolic effects. Molecular studies will add significantly to the concept of these flavonoids as viable and promising therapeutic agents to treat the dysregulation of lipid homeostasis, metabolic disease, and its cardiovascular complications.
Collapse
|
48
|
2-Hydroxy-4ʼ-Methoxychalcone Inhibits Proliferation and Inflammation of Human Aortic Smooth Muscle Cells by Increasing the Expression of Peroxisome Proliferator–Activated Receptor Gamma. J Cardiovasc Pharmacol 2012; 59:339-51. [DOI: 10.1097/fjc.0b013e3182440486] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Bouchard-Mercier A, Paradis AM, Pérusse L, Vohl MC. Associations between polymorphisms in genes involved in fatty acid metabolism and dietary fat intakes. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2012; 5:1-12. [PMID: 22414759 DOI: 10.1159/000336511] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 01/16/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND Obesity prevalence is growing in our population. Twin studies have estimated the heritability of dietary intakes to about 30%. The objective of this study was to verify whether polymorphisms in genes involved in fatty acid metabolism are associated with dietary fat intakes. METHODS Seven hundred participants were recruited. A validated food frequency questionnaire was used to assess dietary intakes. PCR-RFLP and TAQMAN methodology were used to genotype PPARα Leu162Val, PPARγ Pro12Ala, PPARδ -87T>C, PPARGC1α Gly482Ser, FASN Val1483Ile and SREBF1 c.*619C>G. Statistical analyses were executed with SAS statistical package. RESULTS Carriers of the Ala12 allele of PPARγ Pro12Ala polymorphism had higher intakes of total fat (p = 0.04). For FASN Val1483Ile polymorphism, significant gene-sex interaction effects were found for total fat and saturated fat intakes (p = 0.02 and p = 0.002, respectively). No significant difference in fat intakes was observed for PPARα Leu162Val, PPARδ -87T>C, PPARGC1α Gly482Ser and SREBF1 c.*619C>G polymorphisms. CONCLUSIONS Polymorphisms in PPARγ and FASN seem to be associated with dietary fat intakes. Genetic variants are important to take into account when studying dietary intakes.
Collapse
|
50
|
Matera MG, Calzetta L, Segreti A, Cazzola M. Emerging drugs for chronic obstructive pulmonary disease. Expert Opin Emerg Drugs 2012; 17:61-82. [DOI: 10.1517/14728214.2012.660917] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|