1
|
Singh A, Chaudhary R. Potentials of peroxisome proliferator-activated receptor (PPAR) α, β/δ, and γ: An in-depth and comprehensive review of their molecular mechanisms, cellular Signalling, immune responses and therapeutic implications in multiple diseases. Int Immunopharmacol 2025; 155:114616. [PMID: 40222274 DOI: 10.1016/j.intimp.2025.114616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Peroxisome proliferator-activated receptors (PPARs), ligand-activated transcription factors, have emerged as a key regulator of various biological processes, underscoring their relevance in the pathophysiology and treatment of numerous diseases. PPARs are primarily recognized for their critical role in lipid and glucose metabolism, which underpins their therapeutic applications in managing type 2 diabetes mellitus. Beyond metabolic disorders, they have gained attention for their involvement in immune modulation, making them potential targets for autoimmune-related inflammatory diseases. Furthermore, PPAR's ability to regulate proliferation, differentiation, and apoptosis has positioned them as promising candidates in oncology. Their anti-inflammatory and anti-fibrotic properties further highlight their potential in dermatological and cardiovascular conditions, where dysregulated inflammatory responses contribute to disease progression. Recent advancements have elucidated the molecular mechanisms of different PPAR isoforms, including their regulation of key signalling pathways such as NF-κB and MAPK, which are crucial in inflammation and cellular stress responses. Additionally, their interactions with co-factors and post-translational modifications further diversify their functional roles. The therapeutic potential of various PPAR agonists has been extensively explored, although challenges related to side effects and target specificity remain. This growing body of evidence underscores the significance of PPARs in understanding the molecular basis of diseases and advancing therapeutic interventions, paving way for targeted treatment approach across a wide spectrum of medical conditions. Here, we provide a comprehensive and detailed perspective of PPARs and their potential across different health conditions to advance our understanding, elucidate underlying mechanisms, and facilitate the development of potential treatment strategies.
Collapse
Affiliation(s)
- Alpana Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
2
|
Hong H, Wu Y, Li Y, Han Y, Cao X, Wu VWY, Chan TTH, Zhou J, Cao Q, Lui KO, Wong CK, Dai Z, Tian XY. Endothelial PPARδ Ablation Exacerbates Vascular Hyperpermeability via STAT1/CXCL10 Signaling in Acute Lung Injury. Circ Res 2025; 136:735-751. [PMID: 39996324 DOI: 10.1161/circresaha.124.325855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/16/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Vascular hyperpermeability is one of the hallmarks of acute lung injury, contributing to excessive inflammation and respiratory failure. The PPARδ (peroxisome proliferator-activated receptor delta) is an anti-inflammatory transcription factor, although its role in endothelial barrier function remains unclear. Here, we studied the essential role of PPARδ in maintaining vascular endothelial barrier integrity during lung inflammation and investigated the underlying mechanisms. METHODS Endothelial cell (EC)-selective PPARδ knockout mice (PpardEC-KO) and littermate control mice (PpardEC-WT) received lipopolysaccharide injection to induce acute lung injury. Lung inflammation, pulmonary vascular leakage, and mouse mortality were monitored. Single-cell RNA sequencing was performed on sorted mouse lung ECs. RESULTS PpardEC-KO mice exhibited aggravated lung inflammation, characterized by increased leukocyte infiltration, elevated production of proinflammatory cytokines, and higher mortality rates. The enhanced inflammatory responses were associated with increased protein leakage, interstitial edema, and impaired endothelial barrier structure, leading to vascular hyperpermeability in PpardEC-KO mice. Mechanistically, with single-cell RNA sequencing, we identified the emergence of an interferon-activated capillary EC population marked by CXCL10 (C-X-C motif chemokine 10) expression following lipopolysaccharide challenge. PPARδ silencing significantly increased CXCL10 expression in ECs through activating STAT1 (Signal transducer and activator of transcription 1). Notably, CXCL10 treatment induced degradation of tight junction proteins ZO-1 (zonula occludens protein 1) and claudin-5 through the ubiquitin-proteasome system, disrupting membrane junction continuity in ECs. Administration of anti-CXCL10 antibody or CXCL10 receptor antagonist AMG487 suppressed both lipopolysaccharide-induced lung inflammation and vascular leakage in PpardEC-KO mice. CONCLUSIONS These results highlighted a novel anti-inflammatory role of PPARδ in ECs by suppressing CXCL10-mediating vascular hyperpermeability. Targeting the CXCL10 signaling shows therapeutic potential against vascular injury in acute lung injury.
Collapse
Affiliation(s)
- Huiling Hong
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Yalan Wu
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, China (Y.W.)
| | - Yangxian Li
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Yumeng Han
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Xiaoyun Cao
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Vivian Wei Yan Wu
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| | - Thomas Ting Hei Chan
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Jingying Zhou
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Qin Cao
- School of Biomedical Sciences (T.T.H.C., J.Z., Q.C.), The Chinese University of Hong Kong
| | - Kathy O Lui
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Chun-Kwok Wong
- Department of Chemical Pathology (X.C., K.O.L., C.-K.W.), The Chinese University of Hong Kong
| | - Zhiyu Dai
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona (Z.D.)
| | - Xiao Yu Tian
- School of Biomedical Sciences, CUHK Shenzhen Research Institute, Heart and Vascular Institute, CUHK-GIBH CAS Joint Research Laboratory on Stem Cell and Regenerative Medicine (H.H., Y.W., Y.L., Y.H., X.C., V.W.Y.W., X.Y.T.), The Chinese University of Hong Kong
| |
Collapse
|
3
|
Zheng Y, Shao M, Zheng Y, Sun W, Qin S, Sun Z, Zhu L, Guan Y, Wang Q, Wang Y, Li L. PPARs in atherosclerosis: The spatial and temporal features from mechanism to druggable targets. J Adv Res 2025; 69:225-244. [PMID: 38555000 PMCID: PMC11954843 DOI: 10.1016/j.jare.2024.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Atherosclerosis is a chronic and complex disease caused by lipid disorder, inflammation, and other factors. It is closely related to cardiovascular diseases, the chief cause of death globally. Peroxisome proliferator-activated receptors (PPARs) are valuable anti-atherosclerosis targets that showcase multiple roles at different pathological stages of atherosclerosis and for cell types at different tissue sites. AIM OF REVIEW Considering the spatial and temporal characteristics of the pathological evolution of atherosclerosis, the roles and pharmacological and clinical studies of PPARs were summarized systematically and updated under different pathological stages and in different vascular cells of atherosclerosis. Moreover, selective PPAR modulators and PPAR-pan agonists can exert their synergistic effects meanwhile reducing the side effects, thereby providing novel insight into future drug development for precise spatial-temporal therapeutic strategy of anti-atherosclerosis targeting PPARs. KEY SCIENTIFIC Concepts of Review: Based on the spatial and temporal characteristics of atherosclerosis, we have proposed the importance of stage- and cell type-dependent precision therapy. Initially, PPARs improve endothelial cells' dysfunction by inhibiting inflammation and oxidative stress and then regulate macrophages' lipid metabolism and polarization to improve fatty streak. Finally, PPARs reduce fibrous cap formation by suppressing the proliferation and migration of vascular smooth muscle cells (VSMCs). Therefore, research on the cell type-specific mechanisms of PPARs can provide the foundation for space-time drug treatment. Moreover, pharmacological studies have demonstrated that several drugs or compounds can exert their effects by the activation of PPARs. Selective PPAR modulators (that specifically activate gene subsets of PPARs) can exert tissue and cell-specific effects. Furthermore, the dual- or pan-PPAR agonist could perform a better role in balancing efficacy and side effects. Therefore, research on cells/tissue-specific activation of PPARs and PPAR-pan agonists can provide the basis for precision therapy and drug development of PPARs.
Collapse
Affiliation(s)
- Yi Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingyan Shao
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanfei Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Si Qin
- Lab of Food Function and Nutrigenomics, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Ziwei Sun
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Linghui Zhu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuanyuan Guan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qi Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Lingru Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
4
|
Franco Acevedo A, Mack JJ, Valenzuela NM. The transcriptional repressor B cell lymphoma 6 regulates CXCR3 chemokine and human leukocyte antigen II expression in endothelial cells. Am J Transplant 2024; 24:2157-2173. [PMID: 39074669 DOI: 10.1016/j.ajt.2024.07.026] [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: 03/12/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 07/31/2024]
Abstract
Interferon gamma (IFN-γ) induces an endothelial proimmunogenic phenotype through the JAK/STAT1 pathway, which can shape the activation of alloreactive leukocytes in transplant rejection. In immune cells, the DNA-binding protein B cell lymphoma 6 (BCL6) controls the transcription of inflammatory genes. This study tested if BCL6 modulates IFN-γ-induced gene expression in endothelial cells. In vitro, BCL6 was IFN-γ-inducible in primary human endothelium, along with CXCR3 chemokines and human leukocyte antigen (HLA). BCL6, HLA II, and CXCL9 were also increased in human cardiac transplants during acute rejection. Knockdown of BCL6 augmented, whereas overexpression and BTB domain inhibitors (BCL6-BTBi) suppressed, HLA II and CXCR3 chemokine expression but not HLA I. Further, BCL6 had a greater effect on HLA-DR and DP but was less involved in regulating HLA-DQ expression. The effect correlated with BCL6 binding motifs in or near affected genes. The BCL6 DNA recognition sequence was highly similar to that of STAT1, and BTBi reduced STAT1's transcriptional activity in vitro. Our results show for the first time that BCL6 selectively controls IFN-γ-induced endothelial gene expression, advancing our understanding of the endogenous mechanisms regulating donor immunogenicity.
Collapse
Affiliation(s)
- Adriana Franco Acevedo
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, USA
| | - Julia J Mack
- Department of Cardiology, University of California, Los Angeles, USA
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, USA.
| |
Collapse
|
5
|
Lim HJ, Kwak HJ. Selective PPARδ Agonist GW501516 Protects Against LPS-Induced Macrophage Inflammation and Acute Liver Failure in Mice via Suppressing Inflammatory Mediators. Molecules 2024; 29:5189. [PMID: 39519830 PMCID: PMC11547330 DOI: 10.3390/molecules29215189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammation is critical in the development of acute liver failure (ALF). Peroxisome proliferator-activated receptor delta (PPARδ) regulates anti-inflammatory responses and is protective in several diseases such as obesity and cancer. However, the beneficial effects and underlying mechanisms of PPARδ agonist GW501516 in ALF remain unclear. This study investigated the molecular mechanisms underlying the anti-inflammatory effects of GW501516 in macrophages and assessed its protective potential against lipopolysaccharide (LPS)/galactosamine (GalN)-induced ALF. In vivo administration of GW501516 significantly reduced LPS/GalN-induced hepatotoxicity, as evidenced by lower mortality, decreased liver damage, and attenuated secretion of IL-1β, IL-6, and TNF-α. GW501516 treatment also decreased LPS-induced nitric oxide synthase 2 (NOS2) expression and nitric oxide (NO) production in RAW264.7 cells, an effect reversed by PPARδ siRNA. Additionally, GW501516 inhibited LPS-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK), suggesting that inactivation of these MAPKs contributes to its effects. The secretion of IL-6, TNF-α, and NF-κB DNA-binding activity were also suppressed by GW501516, while the nuclear translocation of the NF-κB p65 subunit was unaffected. In conclusion, our findings suggest that GW501516 exerts protective effects in ALF by inhibiting the production of inflammatory mediators. Therefore, GW501516 may act as a potential agent for developing anti-inflammatory therapies for ALF.
Collapse
Affiliation(s)
- Hyun-Joung Lim
- Division of Cardiovascular Diseases Research, Department of Chronic Diseases Convergence, National Institute of Health, Cheongju 28159, Republic of Korea;
| | - Hyun Jeong Kwak
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
6
|
Wang Y, Wang E, Anany M, Füllsack S, Huo YH, Dutta S, Ji B, Hoeppner LH, Kilari S, Misra S, Caulfield T, Vander Kooi CW, Wajant H, Mukhopadhyay D. The crosstalk between neuropilin-1 and tumor necrosis factor-α in endothelial cells. Front Cell Dev Biol 2024; 12:1210944. [PMID: 38994453 PMCID: PMC11236538 DOI: 10.3389/fcell.2024.1210944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Tumor necrosis factor-α (TNFα) is a master cytokine which induces expression of chemokines and adhesion molecules, such as intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), in endothelial cells to initiate the vascular inflammatory response. In this study, we identified neuropilin-1 (NRP1), a co-receptor of several structurally diverse ligands, as a modulator of TNFα-induced inflammatory response of endothelial cells. NRP1 shRNA expression suppressed TNFα-stimulated leukocyte adhesion and expression of ICAM-1 and VCAM-1 in human umbilical vein endothelial cells (HUVECs). Likewise, it reduced TNFα-induced phosphorylation of MAPK p38 but did not significantly affect other TNF-induced signaling pathways, such as the classical NFκB and the AKT pathway. Immunofluorescent staining demonstrated co-localization of NRP1 with the two receptors of TNF, TNFR1 and TNFR2. Co-immunoprecipitation further confirmed that NRP1 was in the same protein complex or membrane compartment as TNFR1 and TNFR2, respectively. Modulation of NRP1 expression, however, neither affected TNFR levels in the cell membrane nor the receptor binding affinities of TNFα. Although a direct interface between NRP1 and TNFα/TNFR1 appeared possible from a protein docking model, a direct interaction was not supported by binding assays in cell-free microplates and cultured cells. Furthermore, TNFα was shown to downregulate NRP1 in a time-dependent manner through TNFR1-NFκB pathway in HUVECs. Taken together, our study reveals a novel reciprocal crosstalk between NRP1 and TNFα in vascular endothelial cells.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cardiovascular Medicine, Rochester, MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Mohamed Anany
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Department of Microbial Biotechnology, Institute of Biotechnology, National Research Centre, Giza, Egypt
| | - Simone Füllsack
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Yu Henry Huo
- Department of Cardiovascular Medicine, Rochester, MN, United States
| | - Shamit Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Baoan Ji
- Department of Cancer Biology, Jacksonville, FL, United States
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | | | - Sanjay Misra
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Thomas Caulfield
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Craig W Vander Kooi
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, United States
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
7
|
Cooreman MP, Vonghia L, Francque SM. MASLD/MASH and type 2 diabetes: Two sides of the same coin? From single PPAR to pan-PPAR agonists. Diabetes Res Clin Pract 2024; 212:111688. [PMID: 38697298 DOI: 10.1016/j.diabres.2024.111688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 05/04/2024]
Abstract
Type 2 diabetes (T2D) and metabolic dysfunction-associated steatotic liver disease (MASLD), mainly related to nutrition and lack of physical activity, are both very common conditions, share several disease pathways and clinical manifestations, and increasingly co-occur with disease progression. Insulin resistance is an upstream node in the biology of both conditions and triggers liver parenchymal injury, inflammation and fibrosis. Peroxisome proliferator-activated receptor (PPAR) nuclear transcription factors are master regulators of energy homeostasis - insulin signaling in liver, adipose and skeletal muscle tissue - and affect immune and fibrogenesis pathways. Among distinct yet overlapping effects, PPARα regulates lipid metabolism and energy expenditure, PPARβ/δ has anti-inflammatory effects and increases glucose uptake by skeletal muscle, while PPARγ improves insulin sensitivity and exerts direct antifibrotic effects on hepatic stellate cells. Together PPARs thus represent pharmacological targets across the entire biology of MASH. Single PPAR agonists are approved for hypertriglyceridemia (PPARα) and T2D (PPARγ), but these, as well as dual PPAR agonists, have shown mixed results as anti-MASH treatments in clinical trials. Agonists of all three PPAR isoforms have the potential to improve the full disease spectrum from insulin resistance to fibrosis, and correspondingly to improve cardiometabolic and hepatic health, as has been shown (phase II data) with the pan-PPAR agonist lanifibranor.
Collapse
Affiliation(s)
- Michael P Cooreman
- Research and Development, Inventiva, Daix, France; Research and Development, Inventiva, New York, NY, USA.
| | - Luisa Vonghia
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium; InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Sven M Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium; InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
8
|
Wolpe AG, Luse MA, Baryiames C, Schug WJ, Wolpe JB, Johnstone SR, Dunaway LS, Juśkiewicz ZJ, Loeb SA, Askew Page HR, Chen YL, Sabapathy V, Pavelec CM, Wakefield B, Cifuentes-Pagano E, Artamonov MV, Somlyo AV, Straub AC, Sharma R, Beier F, Barrett EJ, Leitinger N, Pagano PJ, Sonkusare SK, Redemann S, Columbus L, Penuela S, Isakson BE. Pannexin-3 stabilizes the transcription factor Bcl6 in a channel-independent manner to protect against vascular oxidative stress. Sci Signal 2024; 17:eadg2622. [PMID: 38289985 PMCID: PMC11960805 DOI: 10.1126/scisignal.adg2622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
Targeted degradation regulates the activity of the transcriptional repressor Bcl6 and its ability to suppress oxidative stress and inflammation. Here, we report that abundance of endothelial Bcl6 is determined by its interaction with Golgi-localized pannexin 3 (Panx3) and that Bcl6 transcriptional activity protects against vascular oxidative stress. Consistent with data from obese, hypertensive humans, mice with an endothelial cell-specific deficiency in Panx3 had spontaneous systemic hypertension without obvious changes in channel function, as assessed by Ca2+ handling, ATP amounts, or Golgi luminal pH. Panx3 bound to Bcl6, and its absence reduced Bcl6 protein abundance, suggesting that the interaction with Panx3 stabilized Bcl6 by preventing its degradation. Panx3 deficiency was associated with increased expression of the gene encoding the H2O2-producing enzyme Nox4, which is normally repressed by Bcl6, resulting in H2O2-induced oxidative damage in the vasculature. Catalase rescued impaired vasodilation in mice lacking endothelial Panx3. Administration of a newly developed peptide to inhibit the Panx3-Bcl6 interaction recapitulated the increase in Nox4 expression and in blood pressure seen in mice with endothelial Panx3 deficiency. Panx3-Bcl6-Nox4 dysregulation occurred in obesity-related hypertension, but not when hypertension was induced in the absence of obesity. Our findings provide insight into a channel-independent role of Panx3 wherein its interaction with Bcl6 determines vascular oxidative state, particularly under the adverse conditions of obesity.
Collapse
Affiliation(s)
- Abigail G. Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Melissa A. Luse
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | | | - Wyatt J. Schug
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Jacob B. Wolpe
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Scott R. Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Center for Vascular and Heart Research, Roanoke, VA 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060, USA
| | - Luke S. Dunaway
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Zuzanna J. Juśkiewicz
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Skylar A. Loeb
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Henry R. Askew Page
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Vikram Sabapathy
- Center for Immunity, Inflammation, and Regenerative Medicine (CIIR), University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Caitlin M. Pavelec
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Brent Wakefield
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Eugenia Cifuentes-Pagano
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mykhaylo V. Artamonov
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Avril V. Somlyo
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Adam C. Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rahul Sharma
- Center for Immunity, Inflammation, and Regenerative Medicine (CIIR), University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Frank Beier
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Eugene J. Barrett
- Department of Medicine, Division of Endocrinology and Metabolism, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Norbert Leitinger
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Patrick J. Pagano
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Stefanie Redemann
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Center for Membrane and Cell Physiology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville, VA 22904, USA
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Oncology (Division of Experimental Oncology), Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5W9, Canada
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| |
Collapse
|
9
|
Imig JD. Peroxisome proliferator-activated receptors, farnesoid X receptor, and dual modulating drugs in hypertension. Front Physiol 2023; 14:1186477. [PMID: 37427406 PMCID: PMC10326315 DOI: 10.3389/fphys.2023.1186477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/15/2023] [Indexed: 07/11/2023] Open
Abstract
Hypertension characterized by an elevated blood pressure is a cardiovascular disease that afflicts greater than one in every three adults worldwide. Nuclear receptors are large superfamily of DNA-binding transcription factors that target genes to regulate metabolic and cardiovascular function. Drugs have been developed for nuclear receptors such as peroxisome proliferator-activated receptors (PPARα and PPARγ) and farnesoid X receptor (FXR). PPARα, PPARγ, and FXR agonists are used clinically to treat lipid disorders and metabolic diseases. Evidence from clinical studies and animal hypertension models have demonstrated that PPARα, PPARγ, and FXR agonism can lower blood pressure and decrease end organ damage which could be useful for the treatment of hypertension in patients with metabolic diseases. Unfortunately, PPAR and FXR agonists have unwanted clinical side effects. There have been recent developments to limit side effects for PPAR and FXR agonists. Combining PPAR and FXR agonism with soluble epoxide hydrolase (sEH) inhibition or Takeda G protein receptor 5 (TGR5) agonism has been demonstrated in preclinical studies to have actions that would decrease clinical side effects. In addition, these dual modulating drugs have been demonstrated in preclinical studies to have blood pressure lowering, anti-fibrotic, and anti-inflammatory actions. There is now an opportunity to thoroughly test these novel dual modulators in animal models of hypertension associated with metabolic diseases. In particular, these newly developed dual modulating PPAR and FXR drugs could be beneficial for the treatment of metabolic diseases, organ fibrosis, and hypertension.
Collapse
|
10
|
Zhao L, Zhang M, Liu YW, Tan Y, Yin J, Chen Y, Chen D, Ni B. Sinomenine alleviates lipopolysaccharide-induced acute lung injury via a PPARβ/δ-dependent mechanism. Eur J Pharmacol 2023:175838. [PMID: 37307937 DOI: 10.1016/j.ejphar.2023.175838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023]
Abstract
Evidence is mounting that sinomenine and peroxisome proliferator-activated receptor β/δ (PPARβ/δ) are effective against lipopolysaccharide (LPS)-induced acute lung injury (ALI) via anti-inflammatory properties. However, it is unknown whether PPARβ/δ plays a role in the protective effect of sinomenine on ALI. Here, we initially observed that preemptive administration of sinomenine markedly alleviated lung pathological changes, pulmonary edema and neutrophil infiltration, accompanied by inhibition of the expression of the pro-inflammatory cytokines Tumor necrosis factor-α (TNF-α) and Interleukin-6 (IL-6), which were largely reversed following the addition of a PPARβ/δ antagonist. Subsequently, we also noticed that sinomenine upregulated adenosine A2A receptor expression in a PPARβ/δ-dependent manner in LPS-stimulated bone marrow-derived macrophages (BMDMs). Further investigation indicated that PPARβ/δ directly bound to the functional peroxisome proliferator responsive element (PPRE) in the adenosine A2A receptor gene promoter region to enhance the expression of the adenosine A2A receptor. Sinomenine was identified as a PPARβ/δ agonist. It could bind with PPARβ/δ, and promote the nuclear translocation and transcriptional activity of PPARβ/δ. In addition, combined treatment with sinomenine and an adenosine A2A receptor agonist exhibited synergistic effects and better protective roles than their single use against ALI. Taken together, our results reveal that sinomenine exerts advantageous effects on ALI by activating of PPARβ/δ, with the subsequent upregulation of adenosine A2A receptor expression, and provide a novel and potential therapeutic application for ALI.
Collapse
Affiliation(s)
- Li Zhao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Mengjie Zhang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Yang-Wuyue Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Tan
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Jun Yin
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Yuanyuan Chen
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Dewei Chen
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China; Department of High Altitude Physiology & Biology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China.
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China; Key Laboratory of High Altitude Medicine, PLA, Chongqing, China.
| |
Collapse
|
11
|
Wagner N, Wagner KD. Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease. Int J Mol Sci 2023; 24:ijms24032345. [PMID: 36768666 PMCID: PMC9916802 DOI: 10.3390/ijms24032345] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Peroxisome proliferator activated receptors, including PPARα, PPARβ/δ, and PPARγ, are ligand-activated transcription factors belonging to the nuclear receptor superfamily. They play important roles in glucose and lipid metabolism and are also supposed to reduce inflammation and atherosclerosis. All PPARs are involved in angiogenesis, a process critically involved in cardiovascular pathology. Synthetic specific agonists exist for all PPARs. PPARα agonists (fibrates) are used to treat dyslipidemia by decreasing triglyceride and increasing high-density lipoprotein (HDL) levels. PPARγ agonists (thiazolidinediones) are used to treat Type 2 diabetes mellitus by improving insulin sensitivity. PPARα/γ (dual) agonists are supposed to treat both pathological conditions at once. In contrast, PPARβ/δ agonists are not in clinical use. Although activators of PPARs were initially considered to have favorable effects on the risk factors for cardiovascular disease, their cardiovascular safety is controversial. Here, we discuss the implications of PPARs in vascular biology regarding cardiac pathology and focus on the outcomes of clinical studies evaluating their benefits in cardiovascular diseases.
Collapse
|
12
|
Marques P, Villarroel-Vicente C, Collado A, García A, Vila L, Duplan I, Hennuyer N, Garibotto F, Enriz RD, Dacquet C, Staels B, Piqueras L, Cortes D, Sanz MJ, Cabedo N. Anti-inflammatory effects and improved metabolic derangements in ob/ob mice by a newly synthesized prenylated benzopyran with pan-PPAR activity. Pharmacol Res 2023; 187:106638. [PMID: 36586645 DOI: 10.1016/j.phrs.2022.106638] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND PURPOSE Selective peroxisome proliferator-activated receptors (PPARs) are widely used to treat metabolic complications; however, the limited effect of PPARα agonists on glucose metabolism and the adverse effects associated with selective PPARγ activators have stimulated the development of novel pan-PPAR agonists to treat metabolic disorders. Here, we synthesized a new prenylated benzopyran (BP-2) and evaluated its PPAR-activating properties, anti-inflammatory effects and impact on metabolic derangements. EXPERIMENTAL APPROACH BP-2 was used in transactivation assays to evaluate its agonism to PPARα, PPARβ/δ and PPARγ. A parallel-plate flow chamber was employed to investigate its effect on TNFα-induced leukocyte-endothelium interactions. Flow cytometry and immunofluorescence were used to determine its effects on the expression of endothelial cell adhesion molecules (CAMs) and chemokines and p38-MAPK/NF-κB activation. PPARs/RXRα interactions were determined using a gene silencing approach. Analysis of its impact on metabolic abnormalities and inflammation was performed in ob/ob mice. KEY RESULTS BP-2 displayed strong PPARα activity, with moderate and weak activity against PPARβ/δ and PPARγ, respectively. In vitro, BP-2 reduced TNFα-induced endothelial ICAM-1, VCAM-1 and fractalkine/CX3CL1 expression, suppressed mononuclear cell arrest via PPARβ/δ-RXRα interactions and decreased p38-MAPK/NF-κB activation. In vivo, BP-2 improved the circulating levels of glucose and triglycerides in ob/ob mice, suppressed T-lymphocyte/macrophage infiltration and proinflammatory markers in the liver and white adipose tissue, but increased the expression of the M2-like macrophage marker CD206. CONCLUSION AND IMPLICATIONS BP-2 emerges as a novel pan-PPAR lead candidate to normalize glycemia/triglyceridemia and minimize inflammation in metabolic disorders, likely preventing the development of further cardiovascular complications.
Collapse
Affiliation(s)
- Patrice Marques
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain
| | - Carlos Villarroel-Vicente
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain
| | - Aida Collado
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain
| | - Ainhoa García
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain
| | - Laura Vila
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain
| | - Isabelle Duplan
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U-1011-EGID, F-59000 Lille, France
| | - Nathalie Hennuyer
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U-1011-EGID, F-59000 Lille, France
| | - Francisco Garibotto
- Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis-IMIBIO-SL-CONICET, Chacabuco 917-5700, San Luis, Argentina
| | - Ricardo D Enriz
- Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis-IMIBIO-SL-CONICET, Chacabuco 917-5700, San Luis, Argentina
| | | | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U-1011-EGID, F-59000 Lille, France
| | - Laura Piqueras
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Madrid, Spain
| | - Diego Cortes
- Department of Pharmacology, University of Valencia, Valencia, Spain.
| | - María-Jesús Sanz
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Madrid, Spain.
| | - Nuria Cabedo
- Department of Pharmacology, University of Valencia, Valencia, Spain; Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain.
| |
Collapse
|
13
|
Ahmed YM, Orfali R, Abdelwahab NS, Hassan HM, Rateb ME, AboulMagd AM. Partial Synthetic PPARƳ Derivative Ameliorates Aorta Injury in Experimental Diabetic Rats Mediated by Activation of miR-126-5p Pi3k/AKT/PDK 1/mTOR Expression. Pharmaceuticals (Basel) 2022; 15:1175. [PMID: 36297290 PMCID: PMC9607084 DOI: 10.3390/ph15101175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2D) is a world wild health care issue marked by insulin resistance, a risk factor for the metabolic disorder that exaggerates endothelial dysfunction, increasing the risk of cardiovascular complications. Peroxisome proliferator-activated receptor PPAR) agonists have therapeutically mitigated hyperlipidemia and hyperglycemia in T2D patients. Therefore, we aimed to experimentally investigate the efficacy of newly designed synthetic PPARα/Ƴ partial agonists on a High-Fat Diet (HFD)/streptozotocin (STZ)-induced T2D. Female Wistar rats (200 ± 25 g body weight) were divided into four groups. The experimental groups were fed the HFD for three consecutive weeks before STZ injection (45 mg/kg/i.p) to induce T2D. Standard reference PPARƳ agonist pioglitazone and the partial synthetic PPARƳ (PIO; 20 mg/kg/BW, orally) were administered orally for 2 weeks after 72 h of STZ injection. The aorta tissue was isolated for biological ELISA, qRT-PCR, and Western blotting investigations for vascular inflammatory endothelial mediators endothelin-1 (ET-1), intracellular adhesion molecule 1 (ICAM-1), E-selectin, and anti-inflammatory vasoactive intestinal polypeptide (VIP), as well as microRNA126-5p and p-AKT/p-Pi3k/p-PDK-1/p-mTOR, endothelial Nitric Oxide Synthase (eNOS) immunohistochemical staining all are coupled with and histopathological examination. Our results revealed that HFD/STZ-induced T2D increased fasting blood glucose, ET-1, ICAM-1, E-selectin, and VIP levels, while decreasing the expression of both microRNA126-5p and p-AKT/p-Pi3k/p-PDK-1/p-mTOR phosphorylation. In contrast, the partial synthetic PPARƳ derivative evidenced a vascular alteration significantly more than reference PIO via decreasing (ET-1), ICAM-1, E-selectin, and VIP, along with increased expression of microRNA126-5p and p-AKT/p-Pi3k/p-PDK-1/p-mTOR. In conclusion, the partial synthetic PPARƳ derivative significantly affected HFD/STZ-induced T2D with vascular complications in the rat aorta.
Collapse
Affiliation(s)
- Yasmin M. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 62521, Egypt
| | - Raha Orfali
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Nada S. Abdelwahab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Nahda University, Beni-Suef 62521, Egypt
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Hossam M. Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Mostafa E. Rateb
- School of Computing, Engineering & Physical Sciences, University of the West of Scotland, Paisley PA1 2BE, UK
| | - Asmaa M. AboulMagd
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Nahda University, Beni-Suef 62521, Egypt
| |
Collapse
|
14
|
Haas MJ, Feng V, Gonzales K, Bikkina P, Angelica Landicho M, Mooradian AD. Transcription factor EB protects against endoplasmic reticulum stress in human coronary artery endothelial cells. Eur J Pharmacol 2022; 933:175274. [PMID: 36108736 DOI: 10.1016/j.ejphar.2022.175274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/24/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022]
Abstract
Oxidative stress and endoplasmic reticulum (ER) stress promote atherogenesis while transcription factor EB (TFEB) inhibits atherosclerosis. Since reducing oxidative stress with antioxidants have failed to reduce atherosclerosis possibly because of aggravation of ER stress, we studied the effect of TFEB on ER stress in human coronary artery endothelial cells. ER stress was measured using the secreted alkaline phosphatase assay. Expression and phosphorylation of key mediators of unfolded protein response (UPR). TFEB, inositol-requiring enzyme 1α (IRE1α), phospho-IRE1α, protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), phospho-PERK, and activating transcription factor 6 (ATF6) expression were measured by Western blot. The effect of TFEB gain- and loss-of-function on ER stress were assessed with a plasmid expressing a constitutively active form of TFEB and via siRNA-mediated silencing, respectively. Treatment with tunicamycin (TM) and thapsigargin (TG) increased TFEB expression by 42.8% and 42.3%, respectively. In HCAEC transfected with the TFEB siRNA, treatment with either TM, TG or high-dextrose increased IRE1α and PERK phosphorylation and ATF6 levels significantly more compared to cells transfected with the control siRNA and treated similarly. Furthermore, transient transfection with a plasmid expressing a constitutively active form of TFEB reduced ER stress. Increased expression of TFEB inhibited ER stress in HCAEC treated with pharmacologic (TM and TG) and physiologic (high-dextrose) ER stress inducers, while TFEB knockout aggravated ER stress caused by these ER stress inducers. TFEB-mediated ER stress reduction may contribute to its anti-atherogenic effects in HCAEC and may be a novel target for drug development.
Collapse
Affiliation(s)
- Michael J Haas
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL, 32209, USA.
| | - Victoria Feng
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL, 32209, USA
| | - Krista Gonzales
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL, 32209, USA
| | - Priyanka Bikkina
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL, 32209, USA
| | - Marie Angelica Landicho
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL, 32209, USA
| | - Arshag D Mooradian
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, Jacksonville, FL, 32209, USA
| |
Collapse
|
15
|
Wagner N, Wagner KD. Peroxisome Proliferator-Activated Receptors and the Hallmarks of Cancer. Cells 2022; 11:cells11152432. [PMID: 35954274 PMCID: PMC9368267 DOI: 10.3390/cells11152432] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) function as nuclear transcription factors upon the binding of physiological or pharmacological ligands and heterodimerization with retinoic X receptors. Physiological ligands include fatty acids and fatty-acid-derived compounds with low specificity for the different PPAR subtypes (alpha, beta/delta, and gamma). For each of the PPAR subtypes, specific pharmacological agonists and antagonists, as well as pan-agonists, are available. In agreement with their natural ligands, PPARs are mainly focused on as targets for the treatment of metabolic syndrome and its associated complications. Nevertheless, many publications are available that implicate PPARs in malignancies. In several instances, they are controversial for very similar models. Thus, to better predict the potential use of PPAR modulators for personalized medicine in therapies against malignancies, it seems necessary and timely to review the three PPARs in relation to the didactic concept of cancer hallmark capabilities. We previously described the functions of PPAR beta/delta with respect to the cancer hallmarks and reviewed the implications of all PPARs in angiogenesis. Thus, the current review updates our knowledge on PPAR beta and the hallmarks of cancer and extends the concept to PPAR alpha and PPAR gamma.
Collapse
Affiliation(s)
- Nicole Wagner
- Correspondence: (N.W.); (K.-D.W.); Tel.: +33-489-153-713 (K.-D.W.)
| | | |
Collapse
|
16
|
Wu Y, Lin X, Hong H, Fung YL, Cao X, Tse JKY, Li TH, Chan TF, Tian XY. Endothelium-targeted delivery of PPARδ by adeno-associated virus serotype 1 ameliorates vascular injury induced by hindlimb ischemia in obese mice. Biomed Pharmacother 2022; 151:113172. [PMID: 35644115 DOI: 10.1016/j.biopha.2022.113172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/13/2022] [Accepted: 05/22/2022] [Indexed: 11/28/2022] Open
Abstract
Diabetic vasculopathy is a major health problem worldwide. Peripheral arterial disease (PAD), and in its severe form, critical limb ischemia is a major form of diabetic vasculopathy with limited treatment options. Existing literature suggested an important role of PPARδ in vascular homeostasis. It remains elusive for using PPARδ as a potential therapeutic target due to mostly the side effects of PPARδ agonists. To explore the roles of PPARδ in endothelial homeostasis, endothelial cell (EC) selective Ppard knockout and controlled mice were subjected to hindlimb ischemia (HLI) injury. The muscle ECs were sorted for single-cell RNA sequencing (scRNA-seq) analysis. HLI was also performed in high fat diet (HFD)-induced obese mice to examine the function of PPARδ in obese mice with delayed vascular repair. Adeno-associated virus type 1 (AAV1) carrying ICAM2 promoter to target endothelium for overexpressing PPARδ was injected into the injured muscles of normal chow- and HFD-fed obese mice before HLI surgery was performed. scRNA-seq analysis of ECs in ischemic muscles revealed a pivotal role of PPARδ in endothelial homeostasis. PPARδ expression was diminished both after HLI injury, and also in obese mice, which showed further delayed vascular repair. Endothelium-targeted delivery of PPARδ by AAV1 improved perfusion recovery, increased capillary density, restored endothelial integrity, suppressed vascular inflammation, and promoted muscle regeneration in ischemic hindlimbs of both lean and obese mice. Our study indicated the effectiveness of endothelium-targeted PPARδ overexpression for restoring functional vasculature after ischemic injury, which might be a promising option of gene therapy to treat PAD and CLI.
Collapse
Affiliation(s)
- Yalan Wu
- Chinese University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, Faculty of Medicine, Hong Kong, China; Shenzhen Research Institute, Chinese University of Hong Kong, Shenzhen, China
| | - Xiao Lin
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Huiling Hong
- Chinese University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, Faculty of Medicine, Hong Kong, China; Shenzhen Research Institute, Chinese University of Hong Kong, Shenzhen, China
| | - Yee Lok Fung
- Chinese University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, Faculty of Medicine, Hong Kong, China; Shenzhen Research Institute, Chinese University of Hong Kong, Shenzhen, China
| | - Xiaoyun Cao
- Chinese University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, Faculty of Medicine, Hong Kong, China; Shenzhen Research Institute, Chinese University of Hong Kong, Shenzhen, China
| | - Joyce Ka Yu Tse
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Tsz Ho Li
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Ting Fung Chan
- Shenzhen Research Institute, Chinese University of Hong Kong, Shenzhen, China; School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Xiao Yu Tian
- Chinese University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, Faculty of Medicine, Hong Kong, China; Shenzhen Research Institute, Chinese University of Hong Kong, Shenzhen, China.
| |
Collapse
|
17
|
Xiao L, Wang N. PPAR-δ: A key nuclear receptor in vascular function and remodeling. J Mol Cell Cardiol 2022; 169:1-9. [DOI: 10.1016/j.yjmcc.2022.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/29/2022] [Accepted: 04/25/2022] [Indexed: 12/08/2022]
|
18
|
Lai B, Xie X, Li F, Cui Q, Dang E, Luo W, Wang N, Zheng Y, Wang G, Xiao L, Wang N. Xenobiotic Receptor CAR Is Highly Induced in Psoriasis and Promotes Keratinocyte Proliferation. J Invest Dermatol 2021; 141:2895-2907.e7. [PMID: 34097921 DOI: 10.1016/j.jid.2021.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 11/29/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease with abnormal epidermal proliferation. Xenobiotics contribute to the pathogenesis of psoriasis. The mechanism linking xenobiotic stimuli with epidermal proliferation remains largely unknown. In this study, we investigated the role of CAR, a nuclear receptor (NR1I3) responsible for xenobiotics detoxification. We showed that CAR and its target genes were induced in the lesions from patients with psoriasis and imiquimod-treated mice. Proinflammatory cytokines (IL-17A, IL-22, oncostatin M, IL-1α, and TNF-α) synergistically increased the expressions of CAR and its target genes in both human and mouse keratinocytes. Overexpression of CAR promoted the G1/S transition by regulating cyclin E and c-Myc expressions, whereas the silencing of CAR attenuated it. Importantly, a selective CAR agonist 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime or the proinflammatory cytokines induced cyclin E and c-Myc, which were largely blocked by clotrimazole, a selective CAR antagonist, or CAR small interfering RNA. In addition, we showed that topical application of 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene, a selective agonist for mouse CAR, exacerbated the IMQ-induced psoriasis lesions with increased expressions of proliferative and inflammatory markers. In contrast, Car-knockout mice developed significantly milder lesions. In conclusion, these results showed that CAR plays a pathogenic role and, potentially, may be a target for the treatment of psoriasis.
Collapse
Affiliation(s)
- Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Xinya Xie
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Fan Li
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Qi Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wenhuan Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Ning Wang
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zheng
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Lei Xiao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China.
| | - Nanping Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
19
|
da Cruz BO, Cardozo LFMD, Coutinho-Wolino KS, Mesquita CT, Leal VO, Mafra D, Stockler-Pinto MB. Brazil Nut Supplementation Does Not Regulate PPARβ/δ Signaling Pathway in Peripheral Blood Mononuclear Cells from Coronary Artery Disease Patients. J Am Coll Nutr 2021; 41:780-787. [PMID: 34516363 DOI: 10.1080/07315724.2021.1963882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor (PPAR)β/δ activation is a potential target for modulation of inflammation in cardiovascular disease. PPARβ/δ activation depends on the presence of a ligand, which may be pharmacological or natural, such as bioactive compounds and nutrients. Due to its composition, rich in selenium and unsaturated fatty acids, Brazil nuts have been related to reduced oxidative stress and inflammation in chronic non-communicable diseases and could regulate PPARβ/δ. This study aimed to evaluate the effects of Brazil nut supplementation on PPARβ/δ mRNA expression in patients with Coronary Artery Disease (CAD). METHODS A secondary analysis of a randomized controlled clinical trial was performed with 36 CAD patients. Patients were randomly assigned to either the Supplementation group or the control group and followed up for three months. The Supplementation group consumed 1 Brazil nut/day; the control group did not receive any intervention. At the baseline and after three months, analysis of gene expression and biochemical parameters linked to inflammatory biomarkers and oxidative stress was carried out. RESULTS In the supplementation group, no significant change was observed in PPARβ/δ (0.9 ± 0.5 vs 1.2 ± 0.6; p = 0.178) and NF-κB (1.6 ± 1.5 vs 0.8 ± 0.30, p = 0.554) mRNA expression. There were no significant changes in both groups concerning all the other biochemical parameters. CONCLUSION One Brazil nut per day for three months was not able to increase the PPARβ/δ expression in CAD patients.
Collapse
Affiliation(s)
- Beatriz Oliveira da Cruz
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil
| | - Ludmila Ferreira Medeiros de Cardozo
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil
| | - Karen Salve Coutinho-Wolino
- Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil.,Graduate Program in Nutrition Sciences, Universidade Federal Fluminense, Niterói, Brazil
| | - Claudio Tinoco Mesquita
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Hospital Universitário Antonio Pedro - EBSERH-UFF, Niterói, Brazil
| | - Viviane Oliveira Leal
- Hospital Universitário Pedro Ernesto, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise Mafra
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil.,Graduate Program in Nutrition Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Graduate Program in Medical Sciences, Universidade Federal Fluminense, Niterói, Brazil
| | - Milena Barcza Stockler-Pinto
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil.,Graduate Program in Nutrition Sciences, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
20
|
Ahn J, Son HJ, Seo HD, Ha TY, Ahn J, Lee H, Shin SH, Jung CH, Jang YJ. γ-Oryzanol Improves Exercise Endurance and Muscle Strength by Upregulating PPARδ and ERRγ Activity in Aged Mice. Mol Nutr Food Res 2021; 65:e2000652. [PMID: 33932312 DOI: 10.1002/mnfr.202000652] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/05/2021] [Indexed: 12/24/2022]
Abstract
SCOPE γ-Oryzanol, a well-known antioxidant, has been used by body builders and athletes to boost strength and increase muscle gain, without major side effects. However, the effect of γ-Oryzanol on sarcopenia and the underlying molecular mechanism is poorly understood. RESULTS Aged mice fed with the γ-Oryzanol diet do not show significant changes in muscle weight, but show increased running endurance as well as improved grip strength. The expression and activity of PPARδ and ERRγ are increased in skeletal muscle of γ-Oryzanol supplemented mice. γ-Oryzanol upregulates oxidative muscle fibers by MEF2 transcription factor, and PGC-1α and ERRα expressions. Fatty acid oxidation related genes and mitochondria biogenesis are upregulated by γ-Oryzanol. In addition, γ-Oryzanol inhibits TGF-β-Smad-NADPH oxidase 4 pathway and inflammatory cytokines such as TNF-α, IL-1β, IL-6, and p65 NF-κB subunit, which cause skeletal muscle weakness. Collectively, γ-Oryzanol attenuates muscle weakness pathway and increases oxidative capacity by increasing PPARδ and ERRγ activity, which contributes to enhance strength and improve oxidative capacity in muscles, consequently enhancing exercise capacity in aged mice. Particularly, γ-Oryzanol directly binds to PPARδ. CONCLUSIONS These are the first findings showing that γ-Oryzanol enhances skeletal muscle function in aged mice by regulating PPARδ and ERRγ activity without muscle gain.
Collapse
Affiliation(s)
- Jisong Ahn
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Department of Food Science and Technology, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - Hyo Jeong Son
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
| | - Hyo Deok Seo
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
| | - Tae Youl Ha
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jiyun Ahn
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Hyunjung Lee
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
| | - Seung Ho Shin
- Department of Food and Nutrition, Gyeongsang National University, Jinju, 52828, Republic of Korea
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Chang Hwa Jung
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Young Jin Jang
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju, 55365, Republic of Korea
- Major of Food Science & Technology, Seoul Women's University, Seoul, 01797, Republic of Korea
| |
Collapse
|
21
|
Thibaut R, Gage MC, Pineda-Torra I, Chabrier G, Venteclef N, Alzaid F. Liver macrophages and inflammation in physiology and physiopathology of non-alcoholic fatty liver disease. FEBS J 2021; 289:3024-3057. [PMID: 33860630 PMCID: PMC9290065 DOI: 10.1111/febs.15877] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/05/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022]
Abstract
Non‐alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome, being a common comorbidity of type 2 diabetes and with important links to inflammation and insulin resistance. NAFLD represents a spectrum of liver conditions ranging from steatosis in the form of ectopic lipid storage, to inflammation and fibrosis in nonalcoholic steatohepatitis (NASH). Macrophages that populate the liver play important roles in maintaining liver homeostasis under normal physiology and in promoting inflammation and mediating fibrosis in the progression of NAFLD toward to NASH. Liver macrophages are a heterogenous group of innate immune cells, originating from the yolk sac or from circulating monocytes, that are required to maintain immune tolerance while being exposed portal and pancreatic blood flow rich in nutrients and hormones. Yet, liver macrophages retain a limited capacity to raise the alarm in response to danger signals. We now know that macrophages in the liver play both inflammatory and noninflammatory roles throughout the progression of NAFLD. Macrophage responses are mediated first at the level of cell surface receptors that integrate environmental stimuli, signals are transduced through multiple levels of regulation in the cell, and specific transcriptional programmes dictate effector functions. These effector functions play paramount roles in determining the course of disease in NAFLD and even more so in the progression towards NASH. The current review covers recent reports in the physiological and pathophysiological roles of liver macrophages in NAFLD. We emphasise the responses of liver macrophages to insulin resistance and the transcriptional machinery that dictates liver macrophage function.
Collapse
Affiliation(s)
- Ronan Thibaut
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université, Université de Paris, France
| | - Matthew C Gage
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Inès Pineda-Torra
- Department of Medicine, Centre for Cardiometabolic and Vascular Science, University College London, UK
| | - Gwladys Chabrier
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Nicolas Venteclef
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université, Université de Paris, France
| | - Fawaz Alzaid
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université, Université de Paris, France
| |
Collapse
|
22
|
Perez Diaz N, Lione LA, Hutter V, Mackenzie LS. Co-Incubation with PPARβ/δ Agonists and Antagonists Modeled Using Computational Chemistry: Effect on LPS Induced Inflammatory Markers in Pulmonary Artery. Int J Mol Sci 2021; 22:ijms22063158. [PMID: 33808880 PMCID: PMC8003823 DOI: 10.3390/ijms22063158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 01/01/2023] Open
Abstract
Peroxisome proliferator activated receptor beta/delta (PPARβ/δ) is a nuclear receptor ubiquitously expressed in cells, whose signaling controls inflammation. There are large discrepancies in understanding the complex role of PPARβ/δ in disease, having both anti- and pro-effects on inflammation. After ligand activation, PPARβ/δ regulates genes by two different mechanisms; induction and transrepression, the effects of which are difficult to differentiate directly. We studied the PPARβ/δ-regulation of lipopolysaccharide (LPS) induced inflammation (indicated by release of nitrite and IL-6) of rat pulmonary artery, using different combinations of agonists (GW0742 or L-165402) and antagonists (GSK3787 or GSK0660). LPS induced release of NO and IL-6 is not significantly reduced by incubation with PPARβ/δ ligands (either agonist or antagonist), however, co-incubation with an agonist and antagonist significantly reduces LPS-induced nitrite production and Nos2 mRNA expression. In contrast, incubation with LPS and PPARβ/δ agonists leads to a significant increase in Pdk-4 and Angptl-4 mRNA expression, which is significantly decreased in the presence of PPARβ/δ antagonists. Docking using computational chemistry methods indicates that PPARβ/δ agonists form polar bonds with His287, His413 and Tyr437, while antagonists are more promiscuous about which amino acids they bind to, although they are very prone to bind Thr252 and Asn307. Dual binding in the PPARβ/δ binding pocket indicates the ligands retain similar binding energies, which suggests that co-incubation with both agonist and antagonist does not prevent the specific binding of each other to the large PPARβ/δ binding pocket. To our knowledge, this is the first time that the possibility of binding two ligands simultaneously into the PPARβ/δ binding pocket has been explored. Agonist binding followed by antagonist simultaneously switches the PPARβ/δ mode of action from induction to transrepression, which is linked with an increase in Nos2 mRNA expression and nitrite production.
Collapse
Affiliation(s)
- Noelia Perez Diaz
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK; (N.P.D.); (L.A.L.); (V.H.)
| | - Lisa A. Lione
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK; (N.P.D.); (L.A.L.); (V.H.)
| | - Victoria Hutter
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK; (N.P.D.); (L.A.L.); (V.H.)
| | - Louise S. Mackenzie
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK; (N.P.D.); (L.A.L.); (V.H.)
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK
- Correspondence:
| |
Collapse
|
23
|
Investigation of PPAR β/ δ within Human Dental Pulp Cells: A Preliminary In Vitro Study. PPAR Res 2021; 2021:8854921. [PMID: 33790957 PMCID: PMC7997762 DOI: 10.1155/2021/8854921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/15/2021] [Accepted: 02/17/2021] [Indexed: 11/17/2022] Open
Abstract
Controlling the inflammatory response to restore tissue homeostasis is a crucial step to maintain tooth vitality after pathogen removal from caries-affected dental tissues. The nuclear peroxisome proliferator-activated receptor beta/delta (PPARβ/δ) is a ligand-activated transcription factor with emerging anti-inflammatory roles in many cells and tissues. However, its expression and functions are poorly understood in human dental pulp cells (hDPCs). Thus, this study evaluated PPARβ/δ expression and assessed the anti-inflammatory effects evoked by activation of PPARβ/δ in lipopolysaccharide- (LPS-) induced hDPCs. Our results showed that hDPCs constitutively expressed PPARβ/δ mRNA/protein, and treatment with LPS increased PPARβ/δ mRNA expression. The selective PPARβ/δ agonist GW0742 significantly decreased inflammation-related mRNA expression in hDPCs (IL6, IL1β, TNFα, MMP1, and MMP2) and RAW264.7 cells (Il6 and Tnfα). Further, PPARβ/δ agonist attenuated MMP2/9 gelatinolytic activity in hDPCs. Previously LPS-conditioned hDPCs increased the migration of RAW264.7 cells through the membrane of a Transwell coculture system. Conversely, pretreatment with GW0742 markedly decreased macrophage recruitment. These findings provide among the first evidence that hDPCs express PPARβ/δ. In addition, they suggest that activation of PPARβ/δ by GW0742 can attenuate some cellular and molecular in vitro aspects related to the inflammatory process, pointing out to investigate its potential target role in dental pulp inflammation.
Collapse
|
24
|
Jung TW, Pyun DH, Kim TJ, Lee HJ, Park ES, Abd El-Aty A, Hwang EJ, Shin YK, Jeong JH. Meteorin-like protein (METRNL)/IL-41 improves LPS-induced inflammatory responses via AMPK or PPARδ-mediated signaling pathways. Adv Med Sci 2021; 66:155-161. [PMID: 33592358 DOI: 10.1016/j.advms.2021.01.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/12/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Meteorin-like protein (METRNL) (also known as IL-41), recently identified as a myokine, is released in response to muscle contraction. It improves the skeletal muscle insulin sensitivity through exerting a beneficial anti-inflammatory effect. However, no independent studies have been published to verify the effects of METRNL on human umbilical vein endothelial cells (HUVECs) and THP-1 human monocytes. MATERIALS AND METHODS The levels of NFκB and IκB phosphorylation as well as the expression of adhesion molecules were assessed by Western blotting analysis. Cell adhesion assay demonstrated the interactions between HUVEC and THP-1 cells. We used enzyme-linked immunosorbent assay (ELISA) to measure the levels of TNFα and MCP-1 in culture medium. RESULTS Treatment with METRNL suppressed the secretion of TNFα and MCP-1 as well as NFκB and IκB phosphorylation and inflammatory markers in lipopolysaccharide (LPS)-treated HUVECs and THP-1 cells. Furthermore, treatment with METRNL ameliorated LPS-induced attachment of THP-1 monocytes to HUVECs via inhibition of adhesion molecule expression and apoptosis. Treatment of HUVEC and THP-1 cells with METRNL enhanced AMPK phosphorylation and PPARδ expression in a dose-dependent manner. Small interference (si) RNA-mediated suppression of AMPK or PPARδ restored all these changes. CONCLUSIONS It has therefore been shown that METRNL ameliorates inflammatory responses through AMPK and PPARδ-dependent pathways in LPS-treated HUVEC. In sum, the current study may suggest the suppressive potential of METRNL against endothelial inflammation.
Collapse
|
25
|
Christofides A, Konstantinidou E, Jani C, Boussiotis VA. The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism 2021; 114:154338. [PMID: 32791172 PMCID: PMC7736084 DOI: 10.1016/j.metabol.2020.154338] [Citation(s) in RCA: 363] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/06/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are fatty acid-activated transcription factors of nuclear hormone receptor superfamily that regulate energy metabolism. Currently, three PPAR subtypes have been identified: PPARα, PPARγ, and PPARβ/δ. PPARα and PPARδ are highly expressed in oxidative tissues and regulate genes involved in substrate delivery and oxidative phosphorylation (OXPHOS) and regulation of energy homeostasis. In contrast, PPARγ is more important in lipogenesis and lipid synthesis, with highest expression levels in white adipose tissue (WAT). In addition to tissues regulating whole body energy homeostasis, PPARs are expressed in immune cells and have an emerging critical role in immune cell differentiation and fate commitment. In this review, we discuss the actions of PPARs in the function of the innate and the adaptive immune system and their implications in immune-mediated inflammatory conditions.
Collapse
Affiliation(s)
- Anthos Christofides
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Harvard Medical School, Boston, MA 02215, United States of America; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Eirini Konstantinidou
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Harvard Medical School, Boston, MA 02215, United States of America; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Chinmay Jani
- Department of Medicine, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Mt. Auburn Hospital, Cambridge, MA 02138, United States of America
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Harvard Medical School, Boston, MA 02215, United States of America; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America.
| |
Collapse
|
26
|
Francque S, Szabo G, Abdelmalek MF, Byrne CD, Cusi K, Dufour JF, Roden M, Sacks F, Tacke F. Nonalcoholic steatohepatitis: the role of peroxisome proliferator-activated receptors. Nat Rev Gastroenterol Hepatol 2021; 18:24-39. [PMID: 33093663 DOI: 10.1038/s41575-020-00366-5] [Citation(s) in RCA: 214] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
The increasing epidemic of obesity worldwide is linked to serious health effects, including increased prevalence of type 2 diabetes mellitus, cardiovascular disease and nonalcoholic fatty liver disease (NAFLD). NAFLD is the liver manifestation of the metabolic syndrome and includes the spectrum of liver steatosis (known as nonalcoholic fatty liver) and steatohepatitis (known as nonalcoholic steatohepatitis), which can evolve into progressive liver fibrosis and eventually cause cirrhosis. Although NAFLD is becoming the number one cause of chronic liver diseases, it is part of a systemic disease that affects many other parts of the body, including adipose tissue, pancreatic β-cells and the cardiovascular system. The pathomechanism of NAFLD is multifactorial across a spectrum of metabolic derangements and changes in the host microbiome that trigger low-grade inflammation in the liver and other organs. Peroxisome proliferator-activated receptors (PPARs) are a group of nuclear regulatory factors that provide fine tuning for key elements of glucose and fat metabolism and regulate inflammatory cell activation and fibrotic processes. This Review summarizes and discusses the current literature on NAFLD as the liver manifestation of the systemic metabolic syndrome and focuses on the role of PPARs in the pathomechanisms as well as in the potential targeting of disease.
Collapse
Affiliation(s)
- Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium. .,Translational Research in Inflammation and Immunology (TWI2N), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Christopher D Byrne
- Nutrition & Metabolism, Human Development & Health, Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA
| | - Jean-François Dufour
- Hepatology, Department of Clinical Research, University Hospital of Bern, Bern, Switzerland.,University Clinic for Visceral Surgery and Medicine, Inselspital, Bern, Switzerland
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, University Clinics Düsseldorf, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Frank Sacks
- Departments of Nutrition and Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division, Department of Medicine Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|
27
|
Nie X, Tang W, Zhang Z, Yang C, Qian L, Xie X, Qiang E, Zhao J, Zhao W, Xiao L, Wang N. Procyanidin B2 mitigates endothelial endoplasmic reticulum stress through a PPARδ-Dependent mechanism. Redox Biol 2020; 37:101728. [PMID: 32961442 PMCID: PMC7509074 DOI: 10.1016/j.redox.2020.101728] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/02/2020] [Accepted: 09/12/2020] [Indexed: 12/11/2022] Open
Abstract
Hyperglycemia-induced endothelial endoplasmic reticulum (ER) stress is implicated in the pathophysiology of diabetes and its vascular complications. Procyanidins are enriched in many plant foods and have been demonstrated to exert several beneficial effects on diabetes, cardiovascular and other metabolic diseases. In the present study, we investigated the effect of procyanidin B2 (PCB2), the most widely distributed natural procyanidin, on ER stress evoked by high glucose in endothelial cells (ECs) and the underlying mechanisms. We showed that PCB2 mitigated the high glucose-activated ER stress pathways (PERK, IRE1α and ATF6) in human vascular ECs. In addition, we found that PCB2 attenuated endothelial ER stress via the activation of peroxisome proliferator-activated receptor δ (PPARδ). We demonstrated that PCB2 directly bound to and activated PPARδ. Conversely, GSK0660, a selective PPARδ antagonist, attenuated the suppressive effect of PCB2 on the ER stress signal pathway. Functionally, PCB2 ameliorated the high glucose-impaired endothelium-dependent relaxation in mouse aortas. The protective effect of PCB2 on vasodilation was abolished in the aortas pretreated with GSK0660 or those from the EC-specific PPARδ knockout mice. Moreover, the protective effects of PCB2 on ER stress and endothelial dysfunction required the inter-dependent actions of PPARδ and AMPK. Collectively, we demonstrated that PCB2 mitigated ER stress and ameliorated vasodilation via a PPARδ-mediated mechanism beyond its classic action as a scavenger of free radicals. These findings further highlighted the novel roles of procyanidins in intervening the ER stress and metabolic disorders related to endothelial dysfunction.
Collapse
Affiliation(s)
- Xin Nie
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Weiqi Tang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China; Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zihui Zhang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Chunmiao Yang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lei Qian
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Xinya Xie
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Erjiao Qiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Jingyang Zhao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China; Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wenfei Zhao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lei Xiao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Nanping Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
28
|
Tacconi C, He Y, Ducoli L, Detmar M. Epigenetic regulation of the lineage specificity of primary human dermal lymphatic and blood vascular endothelial cells. Angiogenesis 2020; 24:67-82. [PMID: 32918672 PMCID: PMC7921079 DOI: 10.1007/s10456-020-09743-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/01/2020] [Indexed: 02/08/2023]
Abstract
Lymphatic and blood vascular endothelial cells (ECs) share several molecular and developmental features. However, these two cell types possess distinct phenotypic signatures, reflecting their different biological functions. Despite significant advances in elucidating how the specification of lymphatic and blood vascular ECs is regulated at the transcriptional level during development, the key molecular mechanisms governing their lineage identity under physiological or pathological conditions remain poorly understood. To explore the epigenomic signatures in the maintenance of EC lineage specificity, we compared the transcriptomic landscapes, histone composition (H3K4me3 and H3K27me3) and DNA methylomes of cultured matched human primary dermal lymphatic and blood vascular ECs. Our findings reveal that blood vascular lineage genes manifest a more ‘repressed’ histone composition in lymphatic ECs, whereas DNA methylation at promoters is less linked to the differential transcriptomes of lymphatic versus blood vascular ECs. Meta-analyses identified two transcriptional regulators, BCL6 and MEF2C, which potentially govern endothelial lineage specificity. Notably, the blood vascular endothelial lineage markers CD34, ESAM and FLT1 and the lymphatic endothelial lineage markers PROX1, PDPN and FLT4 exhibited highly differential epigenetic profiles and responded in distinct manners to epigenetic drug treatments. The perturbation of histone and DNA methylation selectively promoted the expression of blood vascular endothelial markers in lymphatic endothelial cells, but not vice versa. Overall, our study reveals that the fine regulation of lymphatic and blood vascular endothelial transcriptomes is maintained via several epigenetic mechanisms, which are crucial to the maintenance of endothelial cell identity.
Collapse
Affiliation(s)
- Carlotta Tacconi
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H303, 8093, Zurich, Switzerland
| | - Yuliang He
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H303, 8093, Zurich, Switzerland
| | - Luca Ducoli
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H303, 8093, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, HCI H303, 8093, Zurich, Switzerland.
| |
Collapse
|
29
|
Faulkner A, Lynam E, Purcell R, Jones C, Lopez C, Board M, Wagner KD, Wagner N, Carr C, Wheeler-Jones C. Context-dependent regulation of endothelial cell metabolism: differential effects of the PPARβ/δ agonist GW0742 and VEGF-A. Sci Rep 2020; 10:7849. [PMID: 32398728 PMCID: PMC7217938 DOI: 10.1038/s41598-020-63900-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 04/07/2020] [Indexed: 12/30/2022] Open
Abstract
Peroxisome proliferator activated receptor β/δ (PPARβ/δ) has pro-angiogenic functions, but whether PPARβ/δ modulates endothelial cell metabolism to support the dynamic phenotype remains to be established. This study characterised the metabolic response of HUVEC to the PPARβ/δ agonist, GW0742, and compared these effects with those induced by VEGF-A. In HUVEC monolayers, flux analysis revealed that VEGF-A promoted glycolysis at the expense of fatty acid oxidation (FAO), whereas GW0742 reduced both glycolysis and FAO. Only VEGF-A stimulated HUVEC migration and proliferation whereas both GW0742 and VEGF-A promoted tubulogenesis. Studies using inhibitors of PPARβ/δ or sirtuin-1 showed that the tubulogenic effect of GW0742, but not VEGF-A, was PPARβ/δ- and sirtuin-1-dependent. HUVEC were reliant on glycolysis and FAO, and inhibition of either pathway disrupted cell growth and proliferation. VEGF-A was a potent inducer of glycolysis in tubulogenic HUVEC, while FAO was maintained. In contrast, GW0742-induced tubulogenesis was associated with enhanced FAO and a modest increase in glycolysis. These novel data reveal a context-dependent regulation of endothelial metabolism by GW0742, where metabolic activity is reduced in monolayers but enhanced during tubulogenesis. These findings expand our understanding of PPARβ/δ in the endothelium and support the targeting of PPARβ/δ in regulating EC behaviour and boosting tissue maintenance and repair.
Collapse
Affiliation(s)
- Ashton Faulkner
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.,Experimental Cardiovascular Medicine, Bristol Medical School, University of Bristol, Bristol, UK
| | - Eleanor Lynam
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Robert Purcell
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Coleen Jones
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Colleen Lopez
- Department of Physiology Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Mary Board
- Department of Physiology Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Kay-Dietrich Wagner
- Université Côte d'Azur, Institute of Biology Valrose, Nice (iBV), CNRS UMR7277, INSERM U1091, Nice, France
| | - Nicole Wagner
- Université Côte d'Azur, Institute of Biology Valrose, Nice (iBV), CNRS UMR7277, INSERM U1091, Nice, France
| | - Carolyn Carr
- Department of Physiology Anatomy & Genetics, University of Oxford, Oxford, UK
| | | |
Collapse
|
30
|
Wagner N, Wagner KD. PPAR Beta/Delta and the Hallmarks of Cancer. Cells 2020; 9:cells9051133. [PMID: 32375405 PMCID: PMC7291220 DOI: 10.3390/cells9051133] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone receptor family. Three different isoforms, PPAR alpha, PPAR beta/delta and PPAR gamma have been identified. They all form heterodimers with retinoic X receptors to activate or repress downstream target genes dependent on the presence/absence of ligands and coactivators or corepressors. PPARs differ in their tissue expression profile, ligands and specific agonists and antagonists. PPARs attract attention as potential therapeutic targets for a variety of diseases. PPAR alpha and gamma agonists are in clinical use for the treatment of dyslipidemias and diabetes. For both receptors, several clinical trials as potential therapeutic targets for cancer are ongoing. In contrast, PPAR beta/delta has been suggested as a therapeutic target for metabolic syndrome. However, potential risks in the settings of cancer are less clear. A variety of studies have investigated PPAR beta/delta expression or activation/inhibition in different cancer cell models in vitro, but the relevance for cancer growth in vivo is less well documented and controversial. In this review, we summarize critically the knowledge of PPAR beta/delta functions for the different hallmarks of cancer biological capabilities, which interplay to determine cancer growth.
Collapse
|
31
|
Capozzi ME, Savage SR, McCollum GW, Hammer SS, Ramos CJ, Yang R, Bretz CA, Penn JS. The peroxisome proliferator-activated receptor-β/δ antagonist GSK0660 mitigates retinal cell inflammation and leukostasis. Exp Eye Res 2020; 190:107885. [PMID: 31758977 PMCID: PMC7426872 DOI: 10.1016/j.exer.2019.107885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/19/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022]
Abstract
Diabetic retinopathy (DR) is triggered by retinal cell damage stimulated by the diabetic milieu, including increased levels of intraocular free fatty acids. Free fatty acids may serve as an initiator of inflammatory cytokine release from Müller cells, and the resulting cytokines are potent stimulators of retinal endothelial pathology, such as leukostasis, vascular permeability, and basement membrane thickening. Our previous studies have elucidated a role for peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) in promoting several steps in the pathologic cascade in DR, including angiogenesis and expression of inflammatory mediators. Furthermore, PPARβ/δ is a known target of lipid signaling, suggesting a potential role for this transcription factor in fatty acid-induced retinal inflammation. Therefore, we hypothesized that PPARβ/δ stimulates both the induction of inflammatory mediators by Müller cells as well the paracrine induction of leukostasis in endothelial cells (EC) by Müller cell inflammatory products. To test this, we used the PPARβ/δ inhibitor, GSK0660, in primary human Müller cells (HMC), human retinal microvascular endothelial cells (HRMEC) and mouse retina. We found that palmitic acid (PA) activation of PPARβ/δ in HMC leads to the production of pro-angiogenic and/or inflammatory cytokines that may constitute DR-relevant upstream paracrine inflammatory signals to EC and other retinal cells. Downstream, EC transduce these signals and increase their synthesis and release of chemokines such as CCL8 and CXCL10 that regulate leukostasis and other cellular events related to vascular inflammation in DR. Our results indicate that PPARβ/δ inhibition mitigates these upstream (MC) as well as downstream (EC) inflammatory signaling events elicited by metabolic stimuli and inflammatory cytokines. Therefore, our data suggest that PPARβ/δ inhibition is a potential therapeutic strategy against early DR pathology.
Collapse
Affiliation(s)
- Megan E Capozzi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, USA.
| | - Sara R Savage
- Department of Pharmacology, Vanderbilt University, USA
| | - Gary W McCollum
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, USA
| | - Sandra S Hammer
- Department of Cell and Developmental Biology, Vanderbilt University, USA
| | - Carla J Ramos
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, USA
| | - Rong Yang
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, USA
| | - Colin A Bretz
- Department of Cell and Developmental Biology, Vanderbilt University, USA
| | - John S Penn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, USA; Department of Pharmacology, Vanderbilt University, USA; Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, USA; Department of Cell and Developmental Biology, Vanderbilt University, USA
| |
Collapse
|
32
|
Lee T, Park HS, Jeong JH, Jung TW. Kynurenic acid attenuates pro-inflammatory reactions in lipopolysaccharide-stimulated endothelial cells through the PPARδ/HO-1-dependent pathway. Mol Cell Endocrinol 2019; 495:110510. [PMID: 31319098 DOI: 10.1016/j.mce.2019.110510] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/21/2019] [Accepted: 07/15/2019] [Indexed: 01/10/2023]
Abstract
Kynurenic acid (KA) regulates energy homeostasis and alleviates inflammation in adipose tissue but how KA affects the atherosclerotic response in HUVECs remains unclear. We evaluated the effects of KA on lipopolysaccharide (LPS)-induced inflammation and apoptosis in HUVECs. KA enhanced peroxisome proliferator-activated receptor delta (PPARδ) expression in HUVECs and THP-1 cells and suppressed LPS-induced atherosclerotic responses through PPARδ-mediated signaling. Moreover, KA treatment mitigated LPS-induced phosphorylation of nuclear factor kappa B and pro-inflammatory cytokine release in HUVECs and THP-1 cells, and down-regulated adhesion molecules in HUVECs and adhesion of THP-1 cells to HUVECs following LPS treatment. KA treatment decreased LPS-induced inflammation and apoptosis, and also promoted heme oxygenase (HO)-1 expression, which suppresses inflammation in HUVECs. Suppression of PPARδ or HO-1 expression markedly mitigated the effects of KA on atherosclerotic responses in HUVECs. Thus, KA attenuates LPS-induced atherosclerotic responses by suppressing inflammation via the PPARδ/HO-1-dependent pathway.
Collapse
Affiliation(s)
- Taeseung Lee
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyung Sub Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Sawada M, Yamamoto H, Ogasahara A, Tanaka Y, Kihara S. β-aminoisobutyric acid protects against vascular inflammation through PGC-1β-induced antioxidative properties. Biochem Biophys Res Commun 2019; 516:963-968. [PMID: 31277947 DOI: 10.1016/j.bbrc.2019.06.141] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 06/25/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUD Among various myocyte-derived bioactive molecules (myokines), β-aminoisobutyric acid (BAIBA) is a unique myokine that attenuates skeletal muscle insulin resistance and inflammation, increases browning of white adipose tissue, and enhances hepatic fatty acid oxidation, resulting in upregulated energy expenditure of the whole body. In the present study, we investigated the effects of BAIBA on the vascular endothelial cell function. METHODS The mRNA levels of proinflammatory molecules, antioxidants, and their related transcription regulators were examined by quantitative RT-PCR in BAIBA-treated human aortic or umbilical vein endothelial cells (HAEC or HUVEC, respectively), with or without tumor necrosis factor (TNF)-α stimulation. The protein expression and phosphorylation of AMP-activated protein kinase (AMPK) and endothelial nitric oxide synthase (eNOS) were determined by Western blot analysis. RESULTS BAIBA pretreatment significantly suppressed the mRNA levels of the adhesion molecules in the TNF-α-stimulated HAEC and HUVEC. BAIBA treatment significantly increased the mRNA levels of antioxidant molecules, catalase, superoxide dismutases, thioredoxin, and gamma-glutamylcysteine ligases, together with mitochondrial biogenesis-related molecules, nuclear respiratory factor 1, and mitochondrial transcription factor A. In addition, BAIBA treatment significantly increased the transcription factors that regulated these genes [i.e., peroxisome proliferator-activated receptor (PPAR)-δ, PPAR-γ, estrogen-related receptor α (ERRα), and peroxisome proliferator-activated receptor gamma coactivator (PGC)-1β]. Adenovirus-mediated PGC-1β overexpression significantly increased the mRNA levels of all antioxidant molecules. The phosphorylation levels of AMPK and eNOS were unaltered by BAIBA. CONCLUSIONS In vascular endothelial cells, BAIBA had antiatherogenic effects through the PGC-1β-ERRα/PPAR-δ and PPAR-γ pathway. This can explain the beneficial effects of exercise on vascular endothelial function.
Collapse
Affiliation(s)
- Miho Sawada
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroyasu Yamamoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Ayako Ogasahara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuya Tanaka
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinji Kihara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
34
|
Zhang Z, Xie X, Yao Q, Liu J, Tian Y, Yang C, Xiao L, Wang N. PPARδ agonist prevents endothelial dysfunction via induction of dihydrofolate reductase gene and activation of tetrahydrobiopterin salvage pathway. Br J Pharmacol 2019; 176:2945-2961. [PMID: 31144304 PMCID: PMC6637045 DOI: 10.1111/bph.14745] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/10/2019] [Accepted: 05/08/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Impaired endothelium-dependent relaxation (EDR) is a hallmark of endothelial dysfunction. A deficiency of tetrahydrobiopterin (BH4 ) causes endothelial NOS to produce ROS rather than NO. PPARδ is an emerging target for pharmacological intervention of endothelial dysfunction. Thus, the present study examined the role of PPARδ in the regulation of dihydrofolate reductase (DHFR), a key enzyme in the BH4 salvage pathway. EXPERIMENTAL APPROACH Gene expression was measured by using qRT-PCR and western blotting. Biopterins and ROS were determined by using HPLC. NO was measured with fluorescent dye and electron paramagnetic resonance spectroscopy. Vasorelaxation was measured by Multi Myograph System. KEY RESULTS The PPARδ agonist GW501516 increased DHFR and BH4 levels in endothelial cells (ECs). The effect was blocked by PPARδ antagonist GSK0660. Chromatin immunoprecipitation identified PPAR-responsive elements within the 5'-flanking region of the human DHFR gene. The promoter activity was examined with luciferase assays using deletion reporters. Importantly, DHFR expression was suppressed by palmitic acid (PA, a saturated fatty acid) but increased by docosahexaenoic acid (DHA, a polyunsaturated fatty acid). GSK0660 prevented DHA-induced increased DHFR expression. Conversely, the suppressive effect of PA was mitigated by GW501516. In mouse aortae, GW501516 ameliorated the PA-impaired EDR. However, this vasoprotective effect was attenuated by DHFR siRNA or methotrexate. In EC-specific Ppard knockout mice, GW501516 failed to improve vasorelaxation. CONCLUSION AND IMPLICATIONS PPARδ prevented endothelial dysfunction by increasing DHFR and activating the BH4 salvage pathway. These results provide a novel mechanism for the protective roles of PPARδ against vascular diseases.
Collapse
Affiliation(s)
- Zihui Zhang
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Xinya Xie
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Qinyu Yao
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Jia Liu
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Ying Tian
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Chunmiao Yang
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Lei Xiao
- Cardiovascular Research Center, School of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Nanping Wang
- The Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| |
Collapse
|
35
|
Hydrogen sulfide improves endothelial dysfunction in hypertension by activating peroxisome proliferator-activated receptor delta/endothelial nitric oxide synthase signaling. J Hypertens 2019; 36:651-665. [PMID: 29084084 DOI: 10.1097/hjh.0000000000001605] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We aimed to elucidate the ameliorative effect of hydrogen sulfide (H2S) on endothelium-dependent relaxation disturbances via peroxisome proliferator-activated receptor delta/endothelial nitric oxide synthase (PPARδ/eNOS) pathway activation in hypertensive patients and rats. METHODS Renal arteries were collected from normotensive and hypertensive patients who underwent nephron-sparing surgery. Renal arteries from 37 patients were cultured with or without sodium H2S (NaHS) 50 μmol/l. The rats were randomly divided into four groups: Sham; Sham + NaHS, two kidneys; one clipped (2K1C); and 2K1C + NaHS. Mean arterial pressure was measured by tail-cuff plethysmography. A microvessel recording technique was used to observe the effect of NaHS on endothelium-dependent relaxation. Plasma H2S concentrations were detected using the monobromobimane method. Real-time PCR and western blotting were used to assess mRNA and protein levels of AT1, cystathionine γ-lyase, PPARδ, and phosphor-eNOS. Laser confocal scanning microscopy measured intracellular NO production in human umbilical vein endothelial cells. RESULTS NaHS improved endothelial function in hypertensive humans and rats. The 20-week administration of NaHS to 2K1C rats lowered the mean arterial pressure. In human umbilical vein endothelial cells, NaHS improved the AngII-induced production of NO. NaHS upregulated PPARδ expression, increased protein kinase B (Akt) or adenosine monophosphate kinase-activated protein kinase (AMPK) phosphorylation, and enhanced eNOS phosphorylation. A PPARδ agonist could mimic the ameliorative effect of NaHS that was suppressed by PPARδ, AMPK, or Akt inhibition. CONCLUSION H2S plays a protective function in renal arterial endothelium in hypertension by activating the PPARδ/PI3K/Akt/eNOS or PPARδ/AMPK/eNOS pathway. H2S may serve as an effective strategy against hypertension.
Collapse
|
36
|
Understanding Peroxisome Proliferator-Activated Receptors: From the Structure to the Regulatory Actions on Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1127:39-57. [DOI: 10.1007/978-3-030-11488-6_3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
37
|
Cheang WS, Wong WT, Wang L, Cheng CK, Lau CW, Ma RCW, Xu A, Wang N, Huang Y, Tian XY. Resveratrol ameliorates endothelial dysfunction in diabetic and obese mice through sirtuin 1 and peroxisome proliferator-activated receptor δ. Pharmacol Res 2019; 139:384-394. [DOI: 10.1016/j.phrs.2018.11.041] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/30/2018] [Accepted: 11/28/2018] [Indexed: 12/30/2022]
|
38
|
Chen Z, Liu F, Zheng N, Guo M, Bao L, Zhan Y, Zhang M, Zhao Y, Guo W, Ding G. Wuzhi capsule (Schisandra Sphenanthera extract) attenuates liver steatosis and inflammation during non-alcoholic fatty liver disease development. Biomed Pharmacother 2018; 110:285-293. [PMID: 30522014 DOI: 10.1016/j.biopha.2018.11.069] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 11/11/2018] [Accepted: 11/19/2018] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Wuzhi (WZ) capsule contains an ethanol extract of Schisandra sphenanthera. The efficacy of WZ in treating non-alcoholic fatty liver disease (NAFLD) has not yet been elucidated. The present study assessed the effects of WZ on NAFLD. MATERIAL AND METHODS A C57BL/6 male mouse model of NAFLD was established by feeding the animals a methionine-choline-deficient (MCD) diet. Mice fed the basal diet were used as controls. Both groups were randomly administered WZ or vehicle by gavage for 5 weeks. Body weight change, liver/body weight ratio, metabolic parameters, and histological changes were assessed. Serum levels of IL-1β, IL-6, IL-10, and TNF-α were analysed by ELISA; mRNA expression of these genes in the liver was studied by real-time PCR. Western blotting was used to analyse the protein levels of PPAR-α, PPAR-γ, MCAD, LCAD, and p65 in the liver. RESULTS After 5 weeks of the MCD diet, the liver/body weight ratio of WZ mice was higher than that of control mice. Liver histology revealed significantly less steatosis, inflammation, and necrosis, which was confirmed by decreased intrahepatic triglycerides and serum ALT in WZ-treated mice. WZ also reduced the liver mRNA expression of IL-1β, IL-6, and TNF-α and the serum levels of IL-1β and IL-6. Sensitivity to steatohepatitis due to WZ administration correlated significantly with alterations in the expression of PPAR-α/γ, as well as the NF-κB signalling pathway. CONCLUSIONS WZ plays a protective role against MCD-induced steatohepatitis. The underlying mechanism likely involves the upregulation of PPAR-α/γ and downregulation of the NF-κB signalling pathway. Based on its beneficial effects on the liver, WZ is a promising therapeutic for NAFLD patients.
Collapse
Affiliation(s)
- Ziqi Chen
- Department of Organ Transplantation, ChangZheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Fang Liu
- Department of Organ Transplantation, ChangZheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Nanxin Zheng
- Department of Colorectal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Meng Guo
- Department of Organ Transplantation, ChangZheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Leilei Bao
- National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Yangyang Zhan
- National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Mingjian Zhang
- National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Yuanyu Zhao
- Department of Organ Transplantation, ChangZheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Wenyuan Guo
- Department of Organ Transplantation, ChangZheng Hospital, Second Military Medical University, Shanghai, 200003, China.
| | - Guoshan Ding
- Department of Organ Transplantation, ChangZheng Hospital, Second Military Medical University, Shanghai, 200003, China.
| |
Collapse
|
39
|
Abstract
The nuclear receptor peroxisome proliferator-activated receptor δ (PPARδ) can transcriptionally regulate target genes. PPARδ exerts essential regulatory functions in the heart, which requires constant energy supply. PPARδ plays a key role in energy metabolism, controlling not only fatty acid (FA) and glucose oxidation, but also redox homeostasis, mitochondrial biogenesis, inflammation, and cardiomyocyte proliferation. PPARδ signaling is impaired in the heart under various pathological conditions, such as pathological cardiac hypertrophy, myocardial ischemia/reperfusion, doxorubicin cardiotoxicity and diabetic cardiomyopathy. PPARδ deficiency in the heart leads to cardiac dysfunction, myocardial lipid accumulation, cardiac hypertrophy/remodeling and heart failure. This article provides an up-today overview of this research area and discusses the role of PPARδ in the heart in light of the complex mechanisms of its transcriptional regulation and its potential as a translatable therapeutic target for the treatment of cardiac disorders.
Collapse
Affiliation(s)
- Qinglin Yang
- Cardiovascular Center of Excellence, LSU Healther Science Center, 533 Bolivar St, New Orleans, LA 70112, USA
| | - Qinqiang Long
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| |
Collapse
|
40
|
Chen J, Montagner A, Tan NS, Wahli W. Insights into the Role of PPARβ/δ in NAFLD. Int J Mol Sci 2018; 19:ijms19071893. [PMID: 29954129 PMCID: PMC6073272 DOI: 10.3390/ijms19071893] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/13/2018] [Accepted: 06/23/2018] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue in developed countries. Although usually associated with obesity, NAFLD is also diagnosed in individuals with low body mass index (BMI) values, especially in Asia. NAFLD can progress from steatosis to non-alcoholic steatohepatitis (NASH), which is characterized by liver damage and inflammation, leading to cirrhosis and hepatocellular carcinoma (HCC). NAFLD development can be induced by lipid metabolism alterations; imbalances of pro- and anti-inflammatory molecules; and changes in various other factors, such as gut nutrient-derived signals and adipokines. Obesity-related metabolic disorders may be improved by activation of the nuclear receptor peroxisome proliferator-activated receptor (PPAR)β/δ, which is involved in metabolic processes and other functions. This review is focused on research findings related to PPARβ/δ-mediated regulation of hepatic lipid and glucose metabolism and NAFLD development. It also discusses the potential use of pharmacological PPARβ/δ activation for NAFLD treatment.
Collapse
Affiliation(s)
- Jiapeng Chen
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore.
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Alexandra Montagner
- ToxAlim, Research Center in Food Toxicology, National Institute for Agricultural Research (INRA), 180 Chemin de Tournefeuille, 31300 Toulouse, France.
- Institut National de La Santé et de La Recherche Médicale (INSERM), UMR1048, Institute of Metabolic and Cardiovascular Diseases, 31027 Toulouse, France.
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore.
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
- KK Research Centre, KK Women's and Children Hospital, 100 Bukit Timah Road, Singapore 229899, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science Technology & Research, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore.
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore.
- ToxAlim, Research Center in Food Toxicology, National Institute for Agricultural Research (INRA), 180 Chemin de Tournefeuille, 31300 Toulouse, France.
- Center for Integrative Genomics, University of Lausanne, Génopode, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
41
|
Jung TW, Park HS, Choi GH, Kim D, Jeong JH, Lee T. Chitinase‐3‐like protein 1 ameliorates atherosclerotic responses via PPARδ‐mediated suppression of inflammation and ER stress. J Cell Biochem 2018; 119:6795-6805. [DOI: 10.1002/jcb.26873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/21/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Tae Woo Jung
- Research Administration TeamSeoul National University Bundang HospitalSeongnamKorea
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
| | - Hyung Sub Park
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
| | - Geum Hee Choi
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
| | - Daehwan Kim
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
| | - Ji Hoon Jeong
- Department of PharmacologyCollege of MedicineChung‐Ang UniversitySeoulKorea
| | - Taeseung Lee
- Department of SurgerySeoul National University Bundang HospitalSeoul National University College of MedicineSeongnamKorea
- Department of SurgerySeoul National University College of MedicineSeoulKorea
| |
Collapse
|
42
|
Botta M, Audano M, Sahebkar A, Sirtori CR, Mitro N, Ruscica M. PPAR Agonists and Metabolic Syndrome: An Established Role? Int J Mol Sci 2018; 19:1197. [PMID: 29662003 PMCID: PMC5979533 DOI: 10.3390/ijms19041197] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/10/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022] Open
Abstract
Therapeutic approaches to metabolic syndrome (MetS) are numerous and may target lipoproteins, blood pressure or anthropometric indices. Peroxisome proliferator-activated receptors (PPARs) are involved in the metabolic regulation of lipid and lipoprotein levels, i.e., triglycerides (TGs), blood glucose, and abdominal adiposity. PPARs may be classified into the α, β/δ and γ subtypes. The PPAR-α agonists, mainly fibrates (including newer molecules such as pemafibrate) and omega-3 fatty acids, are powerful TG-lowering agents. They mainly affect TG catabolism and, particularly with fibrates, raise the levels of high-density lipoprotein cholesterol (HDL-C). PPAR-γ agonists, mainly glitazones, show a smaller activity on TGs but are powerful glucose-lowering agents. Newer PPAR-α/δ agonists, e.g., elafibranor, have been designed to achieve single drugs with TG-lowering and HDL-C-raising effects, in addition to the insulin-sensitizing and antihyperglycemic effects of glitazones. They also hold promise for the treatment of non-alcoholic fatty liver disease (NAFLD) which is closely associated with the MetS. The PPAR system thus offers an important hope in the management of atherogenic dyslipidemias, although concerns regarding potential adverse events such as the rise of plasma creatinine, gallstone formation, drug-drug interactions (i.e., gemfibrozil) and myopathy should also be acknowledged.
Collapse
Affiliation(s)
- Margherita Botta
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy.
| | - Matteo Audano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
| | - Cesare R Sirtori
- Centro Dislipidemie, Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy.
| | - Nico Mitro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy.
| | - Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy.
| |
Collapse
|
43
|
Inhibition of the transcriptional repressor complex Bcl-6/BCoR induces endothelial sprouting but does not promote tumor growth. Oncotarget 2018; 8:552-564. [PMID: 27880939 PMCID: PMC5352177 DOI: 10.18632/oncotarget.13477] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 11/14/2016] [Indexed: 01/01/2023] Open
Abstract
The oncogenic potential of the transcriptional repressor Bcl-6 (B-cell lymphoma 6) was originally discovered in non-Hodgkin patients and the soluble Bcl-6 inhibitor 79-6 was developed to treat diffuse large B-cell lymphomas with aberrant Bcl-6 expression. Since we found Bcl-6 and its co-repressor BCoR (Bcl-6 interacting co-repressor) to be regulated in human microvascular endothelium by colorectal cancer cells, we investigated their function in sprouting angiogenesis which is central to tumor growth. Based on Bcl-6/BCoR gene silencing we found that the transcriptional repressor complex in fact constitutes an endogenous inhibitor of vascular sprouting by supporting the stalk cell phenotype: control of Notch target genes (HES1, HEY1, DLL4) and cell cycle regulators (cyclin A and B1). Thus, when endothelial cells were transiently transfected with Bcl-6 and/or BCoR siRNA, vascular sprouting was prominently induced. Comparably, when the soluble Bcl-6 inhibitor 79-6 was applied in the mouse retina model of physiological angiogenesis, endothelial sprouting and branching were significantly enhanced. To address the question whether clinical treatment with 79-6 might therefore have detrimental therapeutic effects by promoting tumor angiogenesis, mouse xenograft models of colorectal cancer and diffuse large B-cell lymphoma were tested. Despite a tendency to increased tumor vessel density, 79-6 therapy did not enhance tumor expansion. In contrast, growth of colorectal carcinomas was significantly reduced which is likely due to a combined 79-6 effect on cancer cells and tumor stroma. These findings may provide valuable information regarding the future clinical development of Bcl-6 inhibitors.
Collapse
|
44
|
Banno A, Reddy AT, Lakshmi SP, Reddy RC. PPARs: Key Regulators of Airway Inflammation and Potential Therapeutic Targets in Asthma. NUCLEAR RECEPTOR RESEARCH 2017; 5. [PMID: 29450204 DOI: 10.11131/2018/101306] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Asthma affects approximately 300 million people worldwide, significantly impacting quality of life and healthcare costs. While current therapies are effective in controlling many patients' symptoms, a large number continue to experience exacerbations or treatment-related adverse effects. Alternative therapies are thus urgently needed. Accumulating evidence has shown that the peroxisome proliferator-activated receptor (PPAR) family of nuclear hormone receptors, comprising PPARα, PPARβ/δ, and PPARγ, is involved in asthma pathogenesis and that ligand-induced activation of these receptors suppresses asthma pathology. PPAR agonists exert their anti-inflammatory effects primarily by suppressing pro-inflammatory mediators and antagonizing the pro-inflammatory functions of various cell types relevant to asthma pathophysiology. Experimental findings strongly support the potential clinical benefits of PPAR agonists in the treatment of asthma. We review current literature, highlighting PPARs' key role in asthma pathogenesis and their agonists' therapeutic potential. With additional research and rigorous clinical studies, PPARs may become attractive therapeutic targets in this disease.
Collapse
Affiliation(s)
- Asoka Banno
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Aravind T Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| | - Sowmya P Lakshmi
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| | - Raju C Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| |
Collapse
|
45
|
Toral M, Jiménez R, Romero M, Robles-Vera I, Sánchez M, Salaices M, Sabio JM, Duarte J. Role of endoplasmic reticulum stress in the protective effects of PPARβ/δ activation on endothelial dysfunction induced by plasma from patients with lupus. Arthritis Res Ther 2017; 19:268. [PMID: 29208022 PMCID: PMC5717848 DOI: 10.1186/s13075-017-1478-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 11/20/2017] [Indexed: 01/01/2023] Open
Abstract
Background We tested whether GW0742, a peroxisome proliferator-activated receptor beta/delta (PPARβ/δ) agonist, improves endothelial dysfunction induced by plasma from patients with systemic lupus erythematosus (SLE) involving the inhibition of endoplasmic reticulum (ER) stress. Methods A total of 12 non-pregnant women with lupus and 5 non-pregnant healthy women (controls) participated in the study. Cytokines and double-stranded DNA autoantibodies (anti-dsDNA) were tested in plasma samples. Endothelial cells, isolated from human umbilical cord veins (HUVECs), were used to measure nitric oxide (NO), intracellular reactive oxygen species (ROS) production, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and ER stress markers. Results Interferon-γ, interleukin-6, and interleukin-12 levels were significantly increased in plasma from patients with SLE with active nephritis (AN), as compared to both patients with SLE with inactive nephritis (IN) and the control group. The NO production stimulated by both the calcium ionophore A23187 and insulin was significantly reduced in HUVECs incubated with plasma from patients with AN-SLE as compared with the control group. Plasma from patients with IN-SLE did not modify A23187-stimulated NO production. Increased ROS production and NADPH oxidase activity were found in HUVECs incubated with plasma from patients with AN-SLE, which were suppressed by the ER stress inhibitor 4-PBA and the NADPH oxidase inhibitors, apocynin and VAS2870. GW0742 incubation restored the impaired NO production, the increased ROS levels, and the increased ER stress markers induced by plasma from patients with AN-SLE. These protective effects were abolished by the PPARβ/δ antagonist GSK0660 and by silencing PPARβ/δ. Conclusions PPARβ/δ activation may be an important target to control endothelial dysfunction in patients with SLE.
Collapse
Affiliation(s)
- Marta Toral
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain.,CIBER of cardiovascular diseases (CIBERCV), Madrid, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain.,CIBER of cardiovascular diseases (CIBERCV), Madrid, Spain
| | - Iñaki Robles-Vera
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain
| | - Manuel Sánchez
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Mercedes Salaices
- CIBER of cardiovascular diseases (CIBERCV), Madrid, Spain.,Department of Pharmacology, School of Medicine, Autonomous University of Madrid, Research Institute Universitary Hospital La Paz (IdiPAZ), 28029, Madrid, Spain
| | - José Mario Sabio
- Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain.,Department of Internal Medicine, Virgen de las Nieves Universitary Hospital, Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy, University of Granada, 18071, Granada, Spain. .,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain. .,CIBER of cardiovascular diseases (CIBERCV), Madrid, Spain.
| |
Collapse
|
46
|
de Wit NM, Vanmol J, Kamermans A, Hendriks JJA, de Vries HE. Inflammation at the blood-brain barrier: The role of liver X receptors. Neurobiol Dis 2017; 107:57-65. [DOI: 10.1016/j.nbd.2016.09.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 09/11/2016] [Accepted: 09/17/2016] [Indexed: 02/05/2023] Open
|
47
|
Traditional Chinese Medicine Protects against Cytokine Production as the Potential Immunosuppressive Agents in Atherosclerosis. J Immunol Res 2017; 2017:7424307. [PMID: 29038791 PMCID: PMC5606136 DOI: 10.1155/2017/7424307] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/10/2017] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease caused by dyslipidemia and mediated by both innate and adaptive immune responses. Inflammation is a critical factor at all stages of atherosclerosis progression. Proinflammatory cytokines accelerate atherosclerosis progression, while anti-inflammatory cytokines ameliorate the disease. Accordingly, strategies to inhibit immune activation and impede immune responses towards anti-inflammatory activity are an alternative therapeutic strategy to conventional chemotherapy on cardiocerebrovascular outcomes. Since a number of Chinese medicinal plants have been used traditionally to prevent and treat atherosclerosis, it is reasonable to assume that the plants used for such disease may suppress the immune responses and the resultant inflammation. This review focuses on plants that have immunomodulatory effects on the production of inflammatory cytokine burst and are used in Chinese traditional medicine for the prevention and therapy of atherosclerosis.
Collapse
|
48
|
Wang S, Xie X, Lei T, Zhang K, Lai B, Zhang Z, Guan Y, Mao G, Xiao L, Wang N. Statins Attenuate Activation of the NLRP3 Inflammasome by Oxidized LDL or TNF α in Vascular Endothelial Cells through a PXR-Dependent Mechanism. Mol Pharmacol 2017; 92:256-264. [PMID: 28546421 DOI: 10.1124/mol.116.108100] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 05/22/2017] [Indexed: 12/11/2022] Open
Abstract
Excessive activation of the NLRP3 inflammasome is implicated in cardiovascular diseases. Statins exert an anti-inflammatory effect independent of their cholesterol-lowering effect. This study investigated the potential role of statins in the activation of the NLRP3 inflammasome in endothelial cells (ECs). Western blotting and quantitative reverse-transcription polymerase chain reaction showed that oxidized low-density lipoprotein (ox-LDL) or tumor necrosis factor α (TNFα) activated the NLRP3 inflammasome in ECs. Simvastatin or mevastatin significantly suppressed the effects of ox-LDL or TNFα Promoter reporter assays and small interfering RNA knockdown revealed that statins inhibit ox-LDL-mediated NLRP3 inflammasome activation via the pregnane X receptor (PXR). In addition, PXR agonists (rifampicin and SR12813) or overexpression of a constitutively active PXR markedly suppressed the NLRP3 inflammasome activation. Conversely, PXR knockdown abrogated the suppressive effect of rifampicin on NLRP3 inflammasome activation. Knockdown of lectin-like ox-LDL receptor or overexpression of IκBα-attenuated ox-LDL- or TNFα-triggered activation of the NLRP3 inflammasome. Chromatin immunoprecipitation assays indicated that mevastatin inhibited nuclear factor-κB binding to the promoter regions of the human NLRP3 gene. Collectively, these results demonstrate that the statin activation of PXR inhibits the activation of NLRP3 inflammasome in response to atherogenic stimuli such as ox-LDL and TNFα in ECs, providing a new mechanism for the cardiovascular benefit of statins.
Collapse
MESH Headings
- Cells, Cultured
- Endothelial Cells/metabolism
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Lipoproteins, LDL/antagonists & inhibitors
- Lipoproteins, LDL/pharmacology
- NF-kappa B/physiology
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/physiology
- Pregnane X Receptor
- Promoter Regions, Genetic
- Receptors, Steroid/agonists
- Receptors, Steroid/physiology
- Scavenger Receptors, Class E/physiology
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Shaolan Wang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Xinya Xie
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Ting Lei
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Kang Zhang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Baochang Lai
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Zihui Zhang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Youfei Guan
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Guangmei Mao
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Lei Xiao
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| | - Nanping Wang
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an , People's Republic of China (S.W., X.X., T.L., K.Z., B.L., Z.Z., L.X., N.W.); The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, People's Republic of China (Y.G., N.W.); and Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio (G.M.)
| |
Collapse
|
49
|
Lu H, Fan Y, Qiao C, Liang W, Hu W, Zhu T, Zhang J, Chen YE. TFEB inhibits endothelial cell inflammation and reduces atherosclerosis. Sci Signal 2017; 10:10/464/eaah4214. [PMID: 28143903 DOI: 10.1126/scisignal.aah4214] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Transcription factor EB (TFEB) is a master regulator of autophagy and lysosome biogenesis. We investigated the function of TFEB in vascular biology and pathophysiology and demonstrated that TFEB in endothelial cells inhibited inflammation and reduced atherosclerosis development. Laminar shear stress, which protects against atherosclerosis, increased TFEB abundance in cultured primary human endothelial cells. Furthermore, TFEB overexpression in these cells was anti-inflammatory, whereas TFEB knockdown aggravated inflammation. The anti-inflammatory effect of TFEB was, at least, partially due to reduced oxidative stress because TFEB overexpression in endothelial cells decreased the concentrations of reactive oxygen species and increased the expression of the antioxidant genes HO1 (which encodes heme oxygenase 1) and SOD2 (which encodes superoxide dismutase 2). In addition, transgenic mice with endothelial cell-specific expression of TFEB exhibited reduced leukocyte recruitment to endothelial cells and decreased atherosclerosis development. Our study suggests that TFEB is a protective transcription factor against endothelial cell inflammation and a potential target for treating atherosclerosis and associated cardiovascular diseases.
Collapse
Affiliation(s)
- Haocheng Lu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| | - Congzhen Qiao
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Wenying Liang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Wenting Hu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Tianqing Zhu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
50
|
Zhang Z, Jiang M, Xie X, Yang H, Wang X, Xiao L, Wang N. Oleanolic acid ameliorates high glucose-induced endothelial dysfunction via PPARδ activation. Sci Rep 2017; 7:40237. [PMID: 28067284 PMCID: PMC5220361 DOI: 10.1038/srep40237] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/02/2016] [Indexed: 02/07/2023] Open
Abstract
Oleanolic acid (3β-hydroxyolean-12-en-28-oic acid, OA) is a pentacyclic triterpenes widely distributed in food, medicinal plants and nutritional supplements. OA exhibits various pharmacological properties, such as hepatoprotective and anti-tumor effects. In this study, we analyzed the effect of OA on endothelial dysfunction induced by high glucose in human vascular endothelial cells (ECs). Western blotting showed that OA attenuated high glucose-reduced nitric production oxide (NO) as well as Akt-Ser473 and eNOS-Ser1177 phosphorylation in cultured human umbilical vein ECs (HUVECs). Next, luciferase reporter assay showed that OA activated peroxisome proliferators-activated receptor δ (PPARδ) activity. Quantitative reverse transcriptase PCR (qRT-PCR) demonstrated that OA increased the expressions of PPARδ target genes (PDK4, ADRP and ANGPTL4) in ECs. Meanwhile, the induced expressions of PDK4, ADRP and ANGPTL4 by OA were inhibited by GSK0660, a specific antagonist of PPARδ. In addition, inhibition of PPARδ abolished OA-induced the Akt-Ser473 and eNOS-Ser1177 phosphorylation, and NO production. Finally, by using Multi Myograph System, we showed that OA prevented high glucose-impaired vasodilation. This protective effect on vasodilation was inhibited in aortic rings pretreated with GSK0660. Collectively, we demonstrated that OA improved high glucose-impaired endothelial function via a PPARδ-mediated mechanism and through eNOS/Akt/NO pathway.
Collapse
Affiliation(s)
- Zihui Zhang
- Cardiovascular Research Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Manli Jiang
- Cardiovascular Research Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xinya Xie
- Cardiovascular Research Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Haixia Yang
- Cardiovascular Research Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xinfeng Wang
- Cardiovascular Research Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lei Xiao
- Cardiovascular Research Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Nanping Wang
- Cardiovascular Research Center, Xi'an Jiaotong University, Xi'an, 710061, China.,The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| |
Collapse
|