1
|
Sandilya V, El-Gameel D, Atashi M, Nguyen T, Fowowe M, Bhuiyan MMAA, Daramola O, Nwaiwu J, Hamdy NA, Ghanem M, El-Khordagui LK, Abdallah SM, El-Yazbi A, Mechref Y. LC-MS/MS-Profiling of Human Serum Unveils Significant Increase in Neuroinflammation and Carcinogenesis Following Chronic Organophosphate Exposure. J Proteome Res 2025; 24:1342-1355. [PMID: 39905624 DOI: 10.1021/acs.jproteome.4c00995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
The utilization of organophosphate pesticides (OPs) has escalated in response to the growing global food demand driven by a rapidly increasing population and the environmental disruptions caused by climate change. While acute exposure leads to cholinergic poisoning, chronic OP exposure has been linked to organ dysfunction, inflammation, and carcinogenesis. Serum samples from healthy individuals (n = 11), patients with acute OP exposure (n = 12), and those with chronic OP exposure (n = 31) were analyzed to discern the differentially expressed pathways after acute and chronic OP exposure. Differential expression analysis identified 132 proteins altered in chronic exposure vs control, 86 in acute exposure vs control, and 124 in chronic vs acute exposure. Pathway analysis revealed increased blood coagulation and reduced LXR/RXR activation and DCHR24 signaling in both acute and chronic exposures. Elevated levels of pro-inflammatory proteins, such as S100A8, VWF, and GPIBA, were observed, particularly in chronic exposure, highlighting significant inflammatory effects of OP exposure. These findings provide insights into the pathological mechanisms underlying chronic OP exposure and its contribution to inflammation and long-term health risks.
Collapse
Affiliation(s)
- Vishal Sandilya
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Dina El-Gameel
- Department of Pharmacy Practice, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mojgan Atashi
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Thu Nguyen
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Mojibola Fowowe
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | | | - Oluwatosin Daramola
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Judith Nwaiwu
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Noha A Hamdy
- Department of Pharmacy Practice, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Maha Ghanem
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Labiba K El-Khordagui
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Salwa M Abdallah
- Center of Excellence for Toxicological Testing, Department of Mammalian and Aquatic Toxicology, Central Agricultural Pesticides Lab (CAPL), Agricultural Research Center (ARC), Giza, Egypt
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria, Egypt
- Faculty of Pharmacy and Research & Innovation Hub, Alamein International University, Alamein, Egypt
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| |
Collapse
|
2
|
George M, Allerkamp HH, Koshenov Z, Oflaz FE, Tam-Amersdorfer C, Kolesnik T, Rittchen S, Lang M, Fröhlich E, Graier W, Strobl H, Wadsack C. Liver X receptor activation mitigates oxysterol-induced dysfunction in fetoplacental endothelial cells. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159466. [PMID: 38369253 DOI: 10.1016/j.bbalip.2024.159466] [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: 10/04/2023] [Revised: 01/19/2024] [Accepted: 02/12/2024] [Indexed: 02/20/2024]
Abstract
Maintaining the homeostasis of the placental vasculature is of paramount importance for ensuring normal fetal growth and development. Any disruption in this balance can lead to perinatal morbidity. Several studies have uncovered an association between high levels of oxidized cholesterol (oxysterols), and complications during pregnancy, including gestational diabetes mellitus (GDM) and preeclampsia (PE). These complications often coincide with disturbances in placental vascular function. Here, we investigate the role of two oxysterols (7-ketocholesterol, 7β-hydroxycholesterol) in (dys)function of primary fetoplacental endothelial cells (fpEC). Our findings reveal that oxysterols exert a disruptive influence on fpEC function by elevating the production of reactive oxygen species (ROS) and interfering with mitochondrial transmembrane potential, leading to its depolarization. Moreover, oxysterol-treated fpEC exhibited alterations in intracellular calcium (Ca2+) levels, resulting in the reorganization of cell junctions and a corresponding increase in membrane stiffness and vascular permeability. Additionally, we observed an enhanced adhesion of THP-1 monocytes to fpEC following oxysterol treatment. We explored the influence of activating the Liver X Receptor (LXR) with the synthetic agonist T0901317 (TO) on oxysterol-induced endothelial dysfunction in fpEC. Our results demonstrate that LXR activation effectively reversed oxysterol-induced ROS generation, monocyte adhesion, and cell junction permeability in fpEC. Although the effects on mitochondrial depolarization and calcium mobilization did not reach statistical significance, a strong trend towards stabilization of calcium mobilization was evident in LXR-activated cells. Taken together, our results suggest that high levels of systemic oxysterols link to placental vascular dysfunction and LXR agonists may alleviate their impact on fetoplacental vasculature.
Collapse
Affiliation(s)
- Meekha George
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria.
| | | | - Zhanat Koshenov
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; Department of Biochemistry, Weill Cornell Medicine, New York, USA
| | - Furkan E Oflaz
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Carmen Tam-Amersdorfer
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria
| | | | - Sonja Rittchen
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria; Department of Pharmacology, Medical University of Graz, Austria
| | - Magdalena Lang
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria
| | | | - Wolfgang Graier
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Herbert Strobl
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; BioTech-Med, 8010 Graz, Austria.
| |
Collapse
|
3
|
Petrov AM. Oxysterols in Central and Peripheral Synaptic Communication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:91-123. [PMID: 38036877 DOI: 10.1007/978-3-031-43883-7_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cholesterol is a key molecule for synaptic transmission, and both central and peripheral synapses are cholesterol rich. During intense neuronal activity, a substantial portion of synaptic cholesterol can be oxidized by either enzymatic or non-enzymatic pathways to form oxysterols, which in turn modulate the activities of neurotransmitter receptors (e.g., NMDA and adrenergic receptors), signaling molecules (nitric oxide synthases, protein kinase C, liver X receptors), and synaptic vesicle cycling involved in neurotransmitters release. 24-Hydroxycholesterol, produced by neurons in the brain, could directly affect neighboring synapses and change neurotransmission. 27-Hydroxycholesterol, which can cross the blood-brain barrier, can alter both synaptogenesis and synaptic plasticity. Increased generation of 25-hydroxycholesterol by activated microglia and macrophages could link inflammatory processes to learning and neuronal regulation. Amyloids and oxidative stress can lead to an increase in the levels of ring-oxidized sterols and some of these oxysterols (4-cholesten-3-one, 5α-cholestan-3-one, 7β-hydroxycholesterol, 7-ketocholesterol) have a high potency to disturb or modulate neurotransmission at both the presynaptic and postsynaptic levels. Overall, oxysterols could be used as "molecular prototypes" for therapeutic approaches. Analogs of 24-hydroxycholesterol (SGE-301, SGE-550, SAGE718) can be used for correction of NMDA receptor hypofunction-related states, whereas inhibitors of cholesterol 24-hydroxylase, cholestane-3β,5α,6β-triol, and cholest-4-en-3-one oxime (olesoxime) can be utilized as potential anti-epileptic drugs and (or) protectors from excitotoxicity.
Collapse
Affiliation(s)
- Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, RT, Russia.
- Kazan State Medial University, Kazan, RT, Russia.
- Kazan Federal University, Kazan, RT, Russia.
| |
Collapse
|
4
|
Yu L, Xu L, Chu H, Peng J, Sacharidou A, Hsieh HH, Weinstock A, Khan S, Ma L, Durán JGB, McDonald J, Nelson ER, Park S, McDonnell DP, Moore KJ, Huang LJS, Fisher EA, Mineo C, Huang L, Shaul PW. Macrophage-to-endothelial cell crosstalk by the cholesterol metabolite 27HC promotes atherosclerosis in male mice. Nat Commun 2023; 14:4101. [PMID: 37491347 PMCID: PMC10368733 DOI: 10.1038/s41467-023-39586-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Hypercholesterolemia and vascular inflammation are key interconnected contributors to the pathogenesis of atherosclerosis. How hypercholesterolemia initiates vascular inflammation is poorly understood. Here we show in male mice that hypercholesterolemia-driven endothelial activation, monocyte recruitment and atherosclerotic lesion formation are promoted by a crosstalk between macrophages and endothelial cells mediated by the cholesterol metabolite 27-hydroxycholesterol (27HC). The pro-atherogenic actions of macrophage-derived 27HC require endothelial estrogen receptor alpha (ERα) and disassociation of the cytoplasmic scaffolding protein septin 11 from ERα, leading to extranuclear ERα- and septin 11-dependent activation of NF-κB. Furthermore, pharmacologic inhibition of cyp27a1, which generates 27HC, affords atheroprotection by reducing endothelial activation and monocyte recruitment. These findings demonstrate cell-to-cell communication by 27HC, and identify a major causal linkage between the hypercholesterolemia and vascular inflammation that partner to promote atherosclerosis. Interventions interrupting this linkage may provide the means to blunt vascular inflammation without impairing host defense to combat the risk of atherosclerotic cardiovascular disease that remains despite lipid-lowering therapies.
Collapse
Affiliation(s)
- Liming Yu
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center and Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Haiyan Chu
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jun Peng
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Anastasia Sacharidou
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hsi-Hsien Hsieh
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ada Weinstock
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
- Department of Medicine, University of Chicago School of Medicine, Chicago, IL, 60637, USA
| | - Sohaib Khan
- University of Cincinnati Cancer Center, Cincinnati, OH, 45267, USA
| | - Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | | | - Jeffrey McDonald
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sunghee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kathryn J Moore
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Lily Jun-Shen Huang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Edward A Fisher
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Linzhang Huang
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200433, China.
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Fudan University, Shanghai, 200433, China.
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200433, China.
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
5
|
Sola IM, Karin-Kujundzic V, Paic F, Lijovic L, Glibo M, Serman N, Duic T, Skrtic A, Kuna K, Vranic S, Serman L. WNT5A, β‑catenin and SUFU expression patterns, and the significance of microRNA deregulation in placentas with intrauterine growth restriction. Mol Med Rep 2022; 27:28. [PMID: 36524356 PMCID: PMC9813565 DOI: 10.3892/mmr.2022.12914] [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: 07/07/2022] [Accepted: 10/05/2022] [Indexed: 12/15/2022] Open
Abstract
Placental insufficiency is a common cause of intrauterine growth restriction (IUGR). It affects ~10% of pregnancies and increases fetal and neonatal morbidity and mortality. Although Wnt and Hh pathways are crucial for embryonic development and placentation, their role in the pathology of IUGR is still not sufficiently explored. The present study analyzed the expression of positive regulators of the Wnt pathway, WNT5A and β‑catenin, and the expression of the Hh pathway negative regulator suppressor of fused (SUFU). Immunohistochemical and reverse transcription‑quantitative PCR (RT‑qPCR) assays were performed on 34 IUGR and 18 placental tissue samples from physiologic singleton‑term pregnancies. Epigenetic mechanisms of SUFU gene regulation were also investigated by methylation‑specific PCR analysis of its promoter and RT‑qPCR analysis of miR‑214‑3p and miR‑378a‑5p expression. WNT5A protein expression was higher in endothelial cells of placental villi from IUGR compared with control tissues. That was also the case for β‑catenin protein expression in trophoblasts and endothelial cells and SUFU protein expression in trophoblasts from IUGR placentas. The SUFU gene promoter remained unmethylated in all tissue samples, while miR‑214‑3p and miR‑378a‑5p were downregulated in IUGR. The present results suggested altered Wnt and Hh signaling in IUGR. DNA methylation did not appear to be a mechanism of SUFU regulation in the pathogenesis of IUGR, but its expression could be regulated by miRNA targeting.
Collapse
Affiliation(s)
- Ida Marija Sola
- Department of Obstetrics and Gynecology, University Hospital Sestre Milosrdnice, 10000 Zagreb, Croatia
| | - Valentina Karin-Kujundzic
- Department of Biology, University of Zagreb, 10000 Zagreb, Croatia,Centre of Excellence in Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia,Correspondence to: Dr Valentina Karin-Kujundzic, Department of Biology, School of Medicine, University of Zagreb, Salata 3, 10000 Zagreb, Croatia, E-mail:
| | - Frane Paic
- Department of Biology, University of Zagreb, 10000 Zagreb, Croatia
| | - Lada Lijovic
- Department of Anesthesiology and Critical Care, General Hospital Fra Mihovil Sučić, 80101 Livno, Bosnia and Herzegovina
| | - Mislav Glibo
- Department of Biology, University of Zagreb, 10000 Zagreb, Croatia
| | - Nikola Serman
- Zagreb Emergency Medicine Service, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Tihana Duic
- Department of Biology, University of Zagreb, 10000 Zagreb, Croatia
| | - Anita Skrtic
- Centre of Excellence in Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia,Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia,Department of Pathology, University Hospital Merkur, 10000 Zagreb, Croatia
| | - Krunoslav Kuna
- Department of Obstetrics and Gynecology, University Hospital Sestre Milosrdnice, 10000 Zagreb, Croatia
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar
| | - Ljiljana Serman
- Department of Biology, University of Zagreb, 10000 Zagreb, Croatia,Centre of Excellence in Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
6
|
Savla SR, Prabhavalkar KS, Bhatt LK. Liver X Receptor: a potential target in the treatment of atherosclerosis. Expert Opin Ther Targets 2022; 26:645-658. [PMID: 36003057 DOI: 10.1080/14728222.2022.2117610] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Liver X receptors (LXRs) are master regulators of atherogenesis. Their anti-atherogenic potential has been attributed to their role in the inhibition of macrophage-mediated inflammation and promotion of reverse cholesterol transport. Owing to the significance of their anti-atherogenic potential, it is essential to develop and test new generation LXR agonists, both synthetic and natural, to identify potential LXR-targeted therapeutics for the future. AREAS COVERED This review describes the role of LXRs in atherosclerotic development, provides a summary of LXR agonists and future directions for atherosclerosis research. We searched PubMed, Scopus and Google Scholar for relevant reports, from last 10 years, using atherosclerosis, liver X receptor, and LXR agonist as keywords. EXPERT OPINION LXRα has gained widespread recognition as a regulator of cholesterol homeostasis and expression of inflammatory genes. Further research using models of cell type-specific knockout and specific agonist-targeted LXR isoforms is warranted. Enthusiasm for therapeutic value of LXR agonists has been tempered due to LXRα-mediated induction of hepatic lipogenesis. LXRα agonism and LXRβ targeting, gut-specific inverse LXR agonists, investigations combining LXR agonists with other lipogenesis mitigating agents, like IDOL antagonists and synthetic HDL, and targeting ABCA1, M2 macrophages and LXRα phosphorylation, remain as promising possibilities.
Collapse
Affiliation(s)
- Shreya R Savla
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| | - Lokesh K Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India
| |
Collapse
|
7
|
Hayashi K, Sato K, Ochi S, Kawano S, Munesue S, Harashima A, Oshima Y, Kimura K, Kyoi T, Yamamoto Y. Inhibitory Effects of Saururus chinensis Extract on Receptor for Advanced Glycation End-Products-Dependent Inflammation and Diabetes-Induced Dysregulation of Vasodilation. Int J Mol Sci 2022; 23:ijms23105757. [PMID: 35628567 PMCID: PMC9147798 DOI: 10.3390/ijms23105757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
Advanced glycation end-products (AGEs) and the receptor for AGEs (RAGE) are implicated in inflammatory reactions and vascular complications in diabetes. Signaling pathways downstream of RAGE are involved in NF-κB activation. In this study, we examined whether ethanol extracts of Saururus chinensis (Lour.) Baill. (SE) could affect RAGE signaling and vascular relaxation in streptozotocin (STZ)-induced diabetic rats. Treatment with SE inhibited AGEs-modified bovine serum albumin (AGEs-BSA)-elicited activation of NF-κB and could compete with AGEs-BSA binding to RAGE in a dose-dependent manner. Tumor necrosis factor-α (TNF-α) secretion induced by lipopolysaccharide (LPS)-a RAGE ligand-was also reduced by SE treatment in wild-type Ager+/+ mice as well as in cultured peritoneal macrophages from Ager+/+ mice but not in Ager-/- mice. SE administration significantly ameliorated diabetes-related dysregulation of acetylcholine-mediated vascular relaxation in STZ-induced diabetic rats. These results suggest that SE would inhibit RAGE signaling and would be useful for the improvement of vascular endothelial dysfunction in diabetes.
Collapse
Affiliation(s)
- Kenjiro Hayashi
- Food Development Labs, Functional Food Division, Nippon Shinyaku Co., Ltd., Kyoto 601-8550, Japan; (K.H.); (K.S.); (S.O.); (S.K.); (T.K.)
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; (S.M.); (A.H.); (Y.O.); (K.K.)
| | - Koichi Sato
- Food Development Labs, Functional Food Division, Nippon Shinyaku Co., Ltd., Kyoto 601-8550, Japan; (K.H.); (K.S.); (S.O.); (S.K.); (T.K.)
| | - Seishi Ochi
- Food Development Labs, Functional Food Division, Nippon Shinyaku Co., Ltd., Kyoto 601-8550, Japan; (K.H.); (K.S.); (S.O.); (S.K.); (T.K.)
| | - Shuhei Kawano
- Food Development Labs, Functional Food Division, Nippon Shinyaku Co., Ltd., Kyoto 601-8550, Japan; (K.H.); (K.S.); (S.O.); (S.K.); (T.K.)
| | - Seiichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; (S.M.); (A.H.); (Y.O.); (K.K.)
| | - Ai Harashima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; (S.M.); (A.H.); (Y.O.); (K.K.)
| | - Yu Oshima
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; (S.M.); (A.H.); (Y.O.); (K.K.)
| | - Kumi Kimura
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; (S.M.); (A.H.); (Y.O.); (K.K.)
| | - Takashi Kyoi
- Food Development Labs, Functional Food Division, Nippon Shinyaku Co., Ltd., Kyoto 601-8550, Japan; (K.H.); (K.S.); (S.O.); (S.K.); (T.K.)
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; (S.M.); (A.H.); (Y.O.); (K.K.)
- Correspondence:
| |
Collapse
|
8
|
Gao YP, Zhou W, Huang PN, Liu HY, Bi XJ, Zhu Y, Sun J, Tang QY, Li L, Zhang J, Zhu WH, Cheng XQ, Liu YN, Deng YB. Persistent Endothelial Dysfunction in Coronavirus Disease-2019 Survivors Late After Recovery. Front Med (Lausanne) 2022; 9:809033. [PMID: 35237624 PMCID: PMC8882598 DOI: 10.3389/fmed.2022.809033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/10/2022] [Indexed: 12/26/2022] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) can result in an endothelial dysfunction in acute phase. However, information on the late vascular consequences of COVID-19 is limited. Methods Brachial artery flow-mediated dilation (FMD) examination were performed, and inflammatory biomarkers were assessed in 86 survivors of COVID-19 for 327 days (IQR 318–337 days) after recovery. Comparisons were made with 28 age-matched and sex-matched healthy controls and 30 risk factor-matched patients. Results Brachial artery FMD was significantly lower in the survivors of COVID-19 than in the healthy controls and risk factor-matched controls [median (IQR) 7.7 (5.1–10.7)% for healthy controls, 6.9 (5.5–9.4)% for risk factor-matched controls, and 3.5(2.2–4.6)% for COVID-19, respectively, p < 0.001]. The FMD was lower in 25 patients with elevated tumor necrosis factor (TNF)-α [2.7(1.2–3.9)] than in 61 patients without elevated TNF-α [3.8(2.6–5.3), p = 0.012]. Furthermore, FMD was inversely correlated with serum concentration of TNF-α (r = −0.237, p = 0.007). Conclusion Survivors of COVID-19 have a reduced brachial artery FMD, which is inversely correlated with increased serum concentration of TNF-α. Prospective studies on the association of endothelial dysfunction with long-term cardiovascular outcomes, especially the early onset of atherosclerosis, are warranted in survivors of COVID-19.
Collapse
|
9
|
Paredes A, Santos-Clemente R, Ricote M. Untangling the Cooperative Role of Nuclear Receptors in Cardiovascular Physiology and Disease. Int J Mol Sci 2021; 22:ijms22157775. [PMID: 34360540 PMCID: PMC8346021 DOI: 10.3390/ijms22157775] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The heart is the first organ to acquire its physiological function during development, enabling it to supply the organism with oxygen and nutrients. Given this early commitment, cardiomyocytes were traditionally considered transcriptionally stable cells fully committed to contractile function. However, growing evidence suggests that the maintenance of cardiac function in health and disease depends on transcriptional and epigenetic regulation. Several studies have revealed that the complex transcriptional alterations underlying cardiovascular disease (CVD) manifestations such as myocardial infarction and hypertrophy is mediated by cardiac retinoid X receptors (RXR) and their partners. RXRs are members of the nuclear receptor (NR) superfamily of ligand-activated transcription factors and drive essential biological processes such as ion handling, mitochondrial biogenesis, and glucose and lipid metabolism. RXRs are thus attractive molecular targets for the development of effective pharmacological strategies for CVD treatment and prevention. In this review, we summarize current knowledge of RXR partnership biology in cardiac homeostasis and disease, providing an up-to-date view of the molecular mechanisms and cellular pathways that sustain cardiomyocyte physiology.
Collapse
|
10
|
Yano M, Nishino M, Ukita K, Kawamura A, Nakamura H, Matsuhiro Y, Yasumoto K, Tsuda M, Okamoto N, Tanaka A, Matsunaga-Lee Y, Egami Y, Shutta R, Tanouchi J, Yamada T, Yasumura Y, Tamaki S, Hayashi T, Nakagawa A, Nakagawa Y, Suna S, Nakatani D, Hikoso S, Sakata Y. High density lipoprotein cholesterol / C reactive protein ratio in heart failure with preserved ejection fraction. ESC Heart Fail 2021; 8:2791-2801. [PMID: 33934563 PMCID: PMC8318396 DOI: 10.1002/ehf2.13350] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/16/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
AIMS The impacts of high density lipoprotein cholesterol (HDL-C) as an anti-inflammatory and C reactive protein (CRP) as inflammatory properties on the pathogenesis of heart failure were reported. At present, the clinical significance of the HDL-C/CRP ratio in heart failure with preserved ejection fraction (HFpEF) patients remains unknown. METHODS AND RESULTS We examined the data on 796 consecutive HFpEF (left ventricular ejection fraction ≥50%) patients hospitalized due to acute decompensated heart failure from the PURSUIT-HFpEF registry, a prospective, multicentre observational study. We calculated the HDL/CRP ratios and evaluated the relationship between the values and clinical outcomes, including degree of cardiac function. The mean follow-up duration was 420 ± 346 days. All-cause death occurred in 118 patients, of which 51 were cardiac deaths. HDL/CRP ≤ 4.05 was independently and significantly associated with all-cause death (odds ratio = 1.84, 95% CI: 1.06-3.20, P = 0.023), and HDL/CRP ≤ 3.14 was associated with cardiac death by multivariate Cox proportional hazard analysis (odds ratio = 2.86, 95% CI: 1.36-6.01, P = 0.003). HDL-C/CRP ratio significantly correlated with the product of the left atrial volume and left ventricular mass index as well as the tricuspid annular plane systolic excursion by multiple regression analysis (standardized beta-coefficient = -0.085, P = 0.034 and standardized beta-coefficient = 0.081, P = 0.044, respectively). CONCLUSIONS HDL-C/CRP ratio was a useful marker for predicting all-cause death and cardiac death and correlated with left ventricular diastolic function and right ventricular systolic function in HFpEF patients.
Collapse
Affiliation(s)
- Masamichi Yano
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Masami Nishino
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Kohei Ukita
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Akito Kawamura
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Hitoshi Nakamura
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Yutaka Matsuhiro
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Koji Yasumoto
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Masaki Tsuda
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Naotaka Okamoto
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Akihiro Tanaka
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Yasuharu Matsunaga-Lee
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Yasuyuki Egami
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Ryu Shutta
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Jun Tanouchi
- Division of Cardiology, Osaka Rosai Hospital, 3-1179 Nagasonecho, Kita-ku, Sakai, Osaka, 591-8025, Japan
| | - Takahisa Yamada
- Division of Cardiology, Osaka General Medical Center, Osaka, Japan
| | - Yoshio Yasumura
- Division of Cardiology, Amagasaki Chuo Hospital, Amagasaki, Japan
| | - Shunsuke Tamaki
- Division of Cardiology, Osaka General Medical Center, Osaka, Japan
| | | | - Akito Nakagawa
- Division of Cardiology, Amagasaki Chuo Hospital, Amagasaki, Japan.,Department of Medical Informatics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yusuke Nakagawa
- Division of Cardiology, Kawanishi City Hospital, Kawanishi, Japan
| | - Shinichiro Suna
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Daisaku Nakatani
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shungo Hikoso
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | | |
Collapse
|
11
|
Wang X, Chen S, Xiang H, Liang Z, Lu H. Role of sphingosine-1-phosphate receptors in vascular injury of inflammatory bowel disease. J Cell Mol Med 2021; 25:2740-2749. [PMID: 33595873 PMCID: PMC7957208 DOI: 10.1111/jcmm.16333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Sphingosine‐1‐phosphate receptors (S1PRs) have an impact on the intestinal inflammation of inflammatory bowel disease (IBD) by regulating lymphocyte migration and differentiation. S1PR modulators as an emerging therapeutic approach are being investigated for the treatment of IBD. However, the role of S1PRs in intestinal vessels has not drawn much attention. Intestinal vascular damage is one of the major pathophysiological features of IBD, characterized by increased vascular density and impaired barrier function. S1PRs have pleiotropic effects on vascular endothelial cells, including proliferation, migration, angiogenesis and barrier homeostasis. Mounting evidence shows that S1PRs are abnormally expressed on intestinal vascular endothelial cells in IBD. Unexpectedly, S1PR modulators may damage intestinal vasculature, for example increase intestinal bleeding; therefore, S1PRs are thought to be involved in the regulation of intestinal vascular function in IBD. However, little is understood about how S1PRs regulate intestinal vascular function and participate in the initiation and progression of IBD. In this review, we summarize the pathogenic role of S1PRs in and the underlying mechanisms behind the intestinal vascular injury in IBD in order for improving IBD practice including S1PR‐targeted therapies.
Collapse
Affiliation(s)
- Xuewen Wang
- Center for Experimental Medicine, the Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, China
| | - Hong Xiang
- Center for Experimental Medicine, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Ziwei Liang
- Department of Clinical laboratory, Yueyang Hospital Affiliated to Hunan Normal University, Yueyang, China
| | - Hongwei Lu
- Center for Experimental Medicine, the Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
12
|
Zhao L, Lei W, Deng C, Wu Z, Sun M, Jin Z, Song Y, Yang Z, Jiang S, Shen M, Yang Y. The roles of liver X receptor α in inflammation and inflammation-associated diseases. J Cell Physiol 2020; 236:4807-4828. [PMID: 33305467 DOI: 10.1002/jcp.30204] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/19/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
Liver X receptor α (LXRα; also known as NR1H3), an isoform of LXRs, is a member of the nuclear receptor family of transcription factors and plays essential roles in the transcriptional control of cholesterol homeostasis. Previous in-depth phenotypic analyses of mouse models with deficient LXRα have also demonstrated various physiological functions of this receptor within inflammatory responses. LXRα activation exerts a combination of metabolic and anti-inflammatory actions resulting in the modulation and the amelioration of inflammatory disorders. The tight "repercussions" between LXRα and inflammation, as well as cholesterol homeostasis, have suggested that LXRα could be pharmacologically targeted in pathologies such as atherosclerosis, acute lung injury, and Alzheimer's disease. This review gives an overview of the recent advances in understanding the roles of LXRα in inflammation and inflammation-associated diseases, which will help in the design of future experimental researches on the potential of LXRα and advance the investigation of LXRα as pharmacological inflammatory targets.
Collapse
Affiliation(s)
- Lin Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China.,Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wangrui Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhen Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| | - Meng Sun
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yanbin Song
- Department of Cardiology, Affiliated Hospital, Yan'an University, China
| | - Zhi Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| | - Shuai Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| | - Mingzhi Shen
- Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Sanya, Hainan, China.,Hainan Branch of National Clinical Reasearch Center of Geriatrics Disease, Sanya, Hainan, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education Life of Sciences, Northwest University, Xi'an, China
| |
Collapse
|
13
|
Camici PG, Tschöpe C, Di Carli MF, Rimoldi O, Van Linthout S. Coronary microvascular dysfunction in hypertrophy and heart failure. Cardiovasc Res 2020; 116:806-816. [PMID: 31999329 DOI: 10.1093/cvr/cvaa023] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/05/2019] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
Left ventricular (LV) hypertrophy (LVH) is a growth in left myocardial mass mainly caused by increased cardiomyocyte size. LVH can be a physiological adaptation to physical exercise or a pathological condition either primary, i.e. genetic, or secondary to LV overload. Patients with both primary and secondary LVH have evidence of coronary microvascular dysfunction (CMD). The latter is mainly due to capillary rarefaction and adverse remodelling of intramural coronary arterioles due to medial wall thickening with an increased wall/lumen ratio. An important feature of this phenomenon is the diffuse nature of this remodelling, which generally affects the coronary microvessels in the whole of the left ventricle. Patients with LVH secondary to arterial hypertension can develop both heart failure with preserved ejection fraction (HFpEF) and heart failure with reduced ejection fraction (HFrEF). These patients can develop HFrEF via a 'direct pathway' with an interval myocardial infarction and also in its absence. On the other hand, patients can develop HFpEF that can then progress to HFrEF with or without interval myocardial infarction. A similar evolution towards LV dysfunction and both HFpEF and HFrEF can occur in patients with hypertrophic cardiomyopathy, the most common genetic cardiomyopathy with a phenotype characterized by massive LVH. In this review article, we will discuss both the experimental and clinical studies explaining the mechanisms responsible for CMD in LVH as well as the evidence linking CMD with HFpEF and HFrEF.
Collapse
Affiliation(s)
- Paolo G Camici
- Vita Salute University and San Raffaele Hospital, Milano, Italy
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Marcelo F Di Carli
- Cardiovascular Imaging Program, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ornella Rimoldi
- Vita Salute University and San Raffaele Hospital, Milano, Italy.,CNR IBFM, Segrate, Italy
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
14
|
Demchenko SA, Koklin IS, Koklina NY. Role of Arginase 2 as a potential pharmacological target for the creation of new drugs to correct cardiovascular diseases. RESEARCH RESULTS IN PHARMACOLOGY 2020. [DOI: 10.3897/rrpharmacology.6.50942] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: The review provides relevant information about arginase 2, the role of this enzyme in the formation of endothelial dysfunction and, as a consequence, the development of cardiovascular diseases.
History of the discovery of arginase and its functions: The discovery of arginase took place long before its active study as a substance that affects the formation of endothelial dysfunction.
Role of arginase 2 in the development of a number of cardiovascular diseases: The role of NO synthase and arginase 2 in the formation of oxidative stress is determined. The pathophysiological mechanisms of the development of a number of cardiovascular diseases, such as coronary heart disease, atherosclerosis, and aortic aneurysm, are described. The modern possibilities of treatment of endothelial dysfunction in the pathology of the cardiovascular system and the possibility of creation of new drugs are considered. An increase in the activity of arginase 2 was proven to occur in the case of the development of coronary heart disease (CHD), hypertension, type II diabetes mellitus, hypercholesterolemia, as well as in the process of aging. According to the WHO, coronary heart disease and apoplectic attack have topped the list of causes of death worldwide over the past 15 years.
Arginase 2 as a potential pharmacological target: The purpose of this literature review is to determine the possibilities of use of arginase 2 as a new target for the pharmacological correction of cardiovascular diseases.
Collapse
|
15
|
Nasi M, Patrizi G, Pizzi C, Landolfo M, Boriani G, Dei Cas A, Cicero AFG, Fogacci F, Rapezzi C, Sisca G, Capucci A, Vitolo M, Galiè N, Borghi C, Berrettini U, Piepoli M, Mattioli AV. The role of physical activity in individuals with cardiovascular risk factors: an opinion paper from Italian Society of Cardiology-Emilia Romagna-Marche and SIC-Sport. J Cardiovasc Med (Hagerstown) 2020; 20:631-639. [PMID: 31436678 DOI: 10.2459/jcm.0000000000000855] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
: Regular physical activity is a cornerstone in the prevention and treatment of atherosclerotic cardiovascular disease (CVD) due to its positive effects in reducing several cardiovascular risk factors. Current guidelines on CVD suggest for healthy adults to perform at least 150 min/week of moderate intensity or 75 min/week of vigorous intensity aerobic physical activity. The current review explores the effects of physical activity on some risk factors, specifically: diabetes, dyslipidemia, hypertension and hyperuricemia. Physical activity induces an improvement in insulin sensitivity and in glucose control independently of weight loss, which may further contribute to ameliorate both diabetes-associated defects. The benefits of adherence to physical activity have recently proven to extend beyond surrogate markers of metabolic syndrome and diabetes by reducing hard endpoints such as mortality. In recent years, obesity has greatly increased in all countries. Weight losses in these patients have been associated with improvements in many cardiometabolic risk factors. Strategies against obesity included caloric restriction, however greater results have been obtained with association of diet and physical activity. Similarly, the beneficial effect of training on blood pressure via its action on sympathetic activity and on other factors such as improvement of endothelial function and reduction of oxidative stress can have played a role in preventing hypertension development in active subjects. The main international guidelines on prevention of CVD suggest to encourage and to increase physical activity to improve lipid pattern, hypertension and others cardiovascular risk factor. An active action is required to the National Society of Cardiology together with the Italian Society of Sports Cardiology to improve the prescription of organized physical activity in patients with CVD and/or cardiovascular risk factors.
Collapse
Affiliation(s)
- Milena Nasi
- Department of Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena
| | | | - Carmine Pizzi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum
| | - Matteo Landolfo
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna
| | - Giuseppe Boriani
- Division of Cardiology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena
| | - Alessandra Dei Cas
- Endocrinology and Metabolism, Department of Clinical and Experimental Medicine, University of Parma, Parma
| | - Arrigo F G Cicero
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna
| | - Federica Fogacci
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna
| | - Claudio Rapezzi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum
| | - Giovanbattista Sisca
- Bologna F.C.,FIFA Medical Centre of Excellence, Isokinetic Medical Group, Bologna
| | | | - Marco Vitolo
- Division of Cardiology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena
| | - Nazzareno Galiè
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum
| | - Claudio Borghi
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna
| | | | - Massimo Piepoli
- Heart Failure Unit, Cardiology, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Anna V Mattioli
- Department of Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena
| |
Collapse
|
16
|
Liu Y, Afzal J, Vakrou S, Greenland GV, Talbot CC, Hebl VB, Guan Y, Karmali R, Tardiff JC, Leinwand LA, Olgin JE, Das S, Fukunaga R, Abraham MR. Differences in microRNA-29 and Pro-fibrotic Gene Expression in Mouse and Human Hypertrophic Cardiomyopathy. Front Cardiovasc Med 2019; 6:170. [PMID: 31921893 PMCID: PMC6928121 DOI: 10.3389/fcvm.2019.00170] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Hypertrophic cardiomyopathy (HCM) is characterized by myocyte hypertrophy and fibrosis. Studies in two mouse models (R92W-TnT/R403Q-MyHC) at early HCM stage revealed upregulation of endothelin (ET1) signaling in both mutants, but TGFβ signaling only in TnT mutants. Dysregulation of miR-29 expression has been implicated in cardiac fibrosis. But it is unknown whether expression of miR-29a/b/c and profibrotic genes is commonly regulated in mouse and human HCM. Methods: In order to understand mechanisms underlying fibrosis in HCM, and examine similarities/differences in expression of miR-29a/b/c and several profibrotic genes in mouse and human HCM, we performed parallel studies in rat cardiac myocyte/fibroblast cultures, examined gene expression in two mouse models of (non-obstructive) HCM (R92W-TnT, R403Q-MyHC)/controls at early (5 weeks) and established (24 weeks) disease stage, and analyzed publicly available mRNA/miRNA expression data from obstructive-HCM patients undergoing septal myectomy/controls (unused donor hearts). Results: Myocyte cultures: ET1 increased superoxide/H2O2, stimulated TGFβ expression/secretion, and suppressed miR-29a expression in myocytes. The effect of ET1 on miR-29 and TGFβ expression/secretion was antagonized by N-acetyl-cysteine, a reactive oxygen species scavenger. Fibroblast cultures: ET1 had no effect on pro-fibrotic gene expression in fibroblasts. TGFβ1/TGFβ2 suppressed miR-29a and increased collagen expression, which was abolished by miR-29a overexpression. Mouse and human HCM: Expression of miR-29a/b/c was lower, and TGFB1/collagen gene expression was higher in TnT mutant-LV at 5 and 24 weeks; no difference was observed in expression of these genes in MyHC mutant-LV and in human myectomy tissue. TGFB2 expression was higher in LV of both mutant mice and human myectomy tissue. ACE2, a negative regulator of the renin-angiotensin-aldosterone system, was the most upregulated transcript in human myectomy tissue. Pathway analysis predicted upregulation of the anti-hypertrophic/anti-fibrotic liver X receptor/retinoid X receptor (LXR/RXR) pathway only in human myectomy tissue. Conclusions: Our in vitro studies suggest that activation of ET1 signaling in cardiac myocytes increases reactive oxygen species and stimulates TGFβ secretion, which downregulates miR-29a and increases collagen in fibroblasts, thus contributing to fibrosis. Our gene expression studies in mouse and human HCM reveal allele-specific differences in miR-29 family/profibrotic gene expression in mouse HCM, and activation of anti-hypertrophic/anti-fibrotic genes and pathways in human HCM.
Collapse
Affiliation(s)
- Yamin Liu
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States.,Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - Junaid Afzal
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States.,Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - Styliani Vakrou
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - Gabriela V Greenland
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States.,Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - C Conover Talbot
- Johns Hopkins School of Medicine, Institute for Basic Biomedical Sciences, Baltimore, MD, United States
| | - Virginia B Hebl
- Intermountain Medical Center, Intermountain Heart Institute, Murray, UT, United States
| | - Yufan Guan
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - Rehan Karmali
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States
| | - Jil C Tardiff
- Sarver Heart Center, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Leslie A Leinwand
- Molecular, Cellular and Developmental Biology, Biofrontiers Institute, University of Colorado, Boulder, CO, United States
| | - Jeffrey E Olgin
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States
| | - Samarjit Das
- Department of Anesthesia and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - M Roselle Abraham
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States.,Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
17
|
Van Linthout S, Tschöpe C. The Quest for Antiinflammatory and Immunomodulatory Strategies in Heart Failure. Clin Pharmacol Ther 2019; 106:1198-1208. [PMID: 31544235 DOI: 10.1002/cpt.1637] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022]
Abstract
Intensive research over the last 3 decades has unequivocally demonstrated the relevance of inflammation in heart failure (HF). Despite our current and ever increasing knowledge about inflammation, the clinical success of antiinflammatory and immunomodulatory therapies in HF is still limited. This review outlines the complexity and diversity of inflammation, its reciprocal interaction with HF, and addresses future perspectives, calling for immunomodulatory therapies that are specific for factors that activate the immune system without the risk of nonspecific immune suppression.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Campus Virchow Clinic, Charité University Medicine Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site, Berlin, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Campus Virchow Clinic, Charité University Medicine Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site, Berlin, Germany.,Department of Cardiology, Campus Virchow Klinikum, Charité, University Medicine Berlin, Berlin, Germany
| |
Collapse
|
18
|
Mukhutdinova KA, Kasimov MR, Zakyrjanova GF, Gumerova MR, Petrov AM. Oxysterol modulates neurotransmission via liver-X receptor/NO synthase-dependent pathway at the mouse neuromuscular junctions. Neuropharmacology 2019; 150:70-79. [DOI: 10.1016/j.neuropharm.2019.03.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 02/15/2019] [Accepted: 03/13/2019] [Indexed: 02/08/2023]
|
19
|
Simvastatin Modulates Interaction Between Vascular Smooth Muscle Cell/Macrophage and TNF-α-Activated Endothelial Cell. J Cardiovasc Pharmacol 2019; 71:268-274. [PMID: 29389739 DOI: 10.1097/fjc.0000000000000567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cellular interactions between endothelial cell (EC) and vascular smooth muscle cell (VSMC)/macrophages seem to be greatly changed under inflammatory conditions. Although simvastatin could regulate inflammatory transcription factors in EC and VSMC and also could inhibit leukocyte-endothelium interaction, whether it could modulate VSMC/macrophage functions that are induced by tumor necrosis factor-α (TNF-α)-activated EC remained unclear. The purpose of this study was to investigate the effects of simvastatin on VSMC/macrophage functions, which are induced by TNF-α-activated EC in coculture system in vitro. The results showed that under noncontacting conditions, simvastatin could reduce the proliferation, apoptosis, and TNF-α, IL-6, and vascular endothelial growth factor secretion both in VSMC and macrophage, which is induced by TNF-α-activated EC. And a hypothesis that simvastatin regulates the interactions and the soluble factors between EC and VSMC/macrophages could be drawn. And that might be a potential anti-atherosclerosis mechanism of simvastatin.
Collapse
|
20
|
Pentoxifylline ameliorates chronic stress/high-fat diet-induced vascular wall disease: the role of circulating endothelial progenitor cells. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:669-683. [DOI: 10.1007/s00210-019-01627-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/31/2019] [Indexed: 11/25/2022]
|
21
|
Norton GR, Peterson VR, Robinson C, Norman G, Libhaber CD, Libhaber E, Gomes M, Sareli P, Woodiwiss AJ. Independent of left ventricular mass, circulating inflammatory markers rather than pressure load are associated with concentric left ventricular remodelling. Int J Cardiol 2019; 274:342-347. [DOI: 10.1016/j.ijcard.2018.09.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 09/13/2018] [Accepted: 09/17/2018] [Indexed: 01/09/2023]
|
22
|
Fulop T, Witkowski JM, Olivieri F, Larbi A. The integration of inflammaging in age-related diseases. Semin Immunol 2018; 40:17-35. [PMID: 30287177 DOI: 10.1016/j.smim.2018.09.003] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023]
|
23
|
Han S, Bal NB, Sadi G, Usanmaz SE, Uludag MO, Demirel-Yilmaz E. The effects of LXR agonist GW3965 on vascular reactivity and inflammation in hypertensive rat aorta. Life Sci 2018; 213:287-293. [PMID: 30366037 DOI: 10.1016/j.lfs.2018.10.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/18/2018] [Accepted: 10/21/2018] [Indexed: 11/17/2022]
Abstract
AIMS Liver X receptors (LXRs) play an important role in the regulation of cholesterol, fatty acid and glucose metabolisms together with inflammatory processes. In the present study, the effects of LXR agonist GW3965 on vascular reactivity and expression of functional proteins in DOCA-Salt induced hypertension were examined. MAIN METHODS Hypertension was induced through unilateral nephrectomy and deoxycorticosterone-acetate (DOCA) injection (20 mg/kg, twice a week) for 6 weeks in male Wistar albino rats (8 weeks old). An LXR agonist GW3965 (10 mg/kg/day, i.p.) was administered to animals for last seven days. KEY FINDINGS GW3965 treatment reduced systolic blood pressures in hypertensive rats. Acetylcholine-induced endothelium-dependent and sodium nitroprusside-induced endothelium-independent vasorelaxations were decreased in hypertensive rats but not affected by GW3965. GW3965 treatment enhanced plasma nitrite levels in normotensive rats. KCl and phenylephrine (Phe)-induced vasocontractions were reduced in hypertensive groups and increased with GW3965 treatment. Decreased sarco/endoplasmic reticulum Ca2+-ATPase2 (SERCA2) expression in the hypertensive aorta was not changed by GW3965 treatment. Expression of inositoltrisphosphate receptor1 (IP3R1) was increased by GW3965 in normotensive animals. The nuclear factor kappaB (NF-κB) and tumor necrosis factor alpha (TNF-α) expressions were increased in hypertensive rats and reduced by GW3965 treatment. SIGNIFICANCE The results of study indicate that the LXR agonist, GW3965, exhibited a beneficial effect on increased blood pressure and improved hypertension-induced impairment in contractile activity of vessel and inflammatory markers in vascular tissue. Therefore, these effects of LXR agonists on vessel should be taken into account in experimental or therapeutic approaches to hypertension.
Collapse
Affiliation(s)
- Sevtap Han
- Gazi University, Faculty of Pharmacy, Department of Pharmacology, Etiler, 06330 Ankara, Turkey.
| | - Nur Banu Bal
- Gazi University, Faculty of Pharmacy, Department of Pharmacology, Etiler, 06330 Ankara, Turkey
| | - Gökhan Sadi
- Karamanoglu Mehmetbey University, K.Ö. Faculty of Science, Department of Biology, Karaman, Turkey
| | - Suzan Emel Usanmaz
- Ankara University, Faculty of Medicine, Department of Medical Pharmacology, Sıhhiye, 06100 Ankara, Turkey
| | - Mecit Orhan Uludag
- Gazi University, Faculty of Pharmacy, Department of Pharmacology, Etiler, 06330 Ankara, Turkey
| | - Emine Demirel-Yilmaz
- Ankara University, Faculty of Medicine, Department of Medical Pharmacology, Sıhhiye, 06100 Ankara, Turkey
| |
Collapse
|
24
|
Beyond the Foam Cell: The Role of LXRs in Preventing Atherogenesis. Int J Mol Sci 2018; 19:ijms19082307. [PMID: 30087224 PMCID: PMC6121590 DOI: 10.3390/ijms19082307] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic condition associated with cardiovascular disease. While largely identified by the accumulation of lipid-laden foam cells within the aorta later on in life, atherosclerosis develops over several stages and decades. During atherogenesis, various cell types of the aorta acquire a pro-inflammatory phenotype that initiates the cascade of signaling events facilitating the formation of these foam cells. The liver X receptors (LXRs) are nuclear receptors that upon activation induce the expression of transporters responsible for promoting cholesterol efflux. In addition to promoting cholesterol removal from the arterial wall, LXRs have potent anti-inflammatory actions via the transcriptional repression of key pro-inflammatory cytokines. These beneficial functions sparked an interest in the potential to target LXRs and the development of agonists as anti-atherogenic agents. These early studies focused on mediating the contributions of macrophages to the underlying pathogenesis. However, further evidence has since demonstrated that LXRs reduce atherosclerosis through their actions in multiple cell types apart from those monocytes/macrophages that infiltrate the lesion. LXRs and their target genes have profound effects on multiple other cells types of the hematopoietic system. Furthermore, LXRs can also mediate dysfunction within vascular cell types of the aorta including endothelial and smooth muscle cells. Taken together, these studies demonstrate the whole-body benefits of LXR activation with respect to anti-atherogenesis, and that LXRs remain a viable target for the treatment of atherosclerosis, with a reach which extends beyond plaque macrophages.
Collapse
|
25
|
|
26
|
Rasheed A, Tsai R, Cummins CL. Loss of the Liver X Receptors Disrupts the Balance of Hematopoietic Populations, With Detrimental Effects on Endothelial Progenitor Cells. J Am Heart Assoc 2018; 7:JAHA.117.007787. [PMID: 29739800 PMCID: PMC6015321 DOI: 10.1161/jaha.117.007787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background The liver X receptors (LXRs; α/β) are nuclear receptors known to regulate cholesterol homeostasis and the production of select hematopoietic populations. The objective of this study was to determine the importance of LXRs and a high‐fat high‐cholesterol diet on global hematopoiesis, with special emphasis on endothelial progenitor cells (EPCs), a vasoreparative cell type that is derived from bone marrow hematopoietic stem cells. Methods and Results Wild‐type and LXR double‐knockout (Lxrαβ−/−) mice were fed a Western diet (WD) to increase plasma cholesterol levels. In WD‐fed Lxrαβ−/− mice, flow cytometry and complete blood cell counts revealed that hematopoietic stem cells, a myeloid progenitor, and mature circulating myeloid cells were increased; EPC numbers were significantly decreased. Hematopoietic stem cells from WD‐fed Lxrαβ−/− mice showed increased cholesterol content, along with increased myeloid colony formation compared with chow‐fed mice. In contrast, EPCs from WD‐fed Lxrαβ−/− mice also demonstrated increased cellular cholesterol content that was associated with greater expression of the endothelial lineage markers Cd144 and Vegfr2, suggesting accelerated differentiation of the EPCs. Treatment of human umbilical vein endothelial cells with conditioned medium collected from these EPCs increased THP‐1 monocyte adhesion. Increased monocyte adhesion to conditioned medium–treated endothelial cells was recapitulated with conditioned medium from Lxrαβ−/−EPCs treated with cholesterol ex vivo, suggesting cholesterol is the main component of the WD inducing EPC dysfunction. Conclusions LXRs are crucial for maintaining the balance of hematopoietic cells in a hypercholesterolemic environment and for mitigating the negative effects of cholesterol on EPC differentiation/secretome changes that promote monocyte‐endothelial adhesion.
Collapse
Affiliation(s)
- Adil Rasheed
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | - Ricky Tsai
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada .,Banting and Best Diabetes Centre, Toronto, Ontario, Canada.,The Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Miteva K, Madonna R, De Caterina R, Van Linthout S. Innate and adaptive immunity in atherosclerosis. Vascul Pharmacol 2018; 107:S1537-1891(17)30464-0. [PMID: 29684642 DOI: 10.1016/j.vph.2018.04.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 02/03/2018] [Accepted: 04/15/2018] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is a chronic inflammatory disorder of the large and medium-size arteries characterized by the subendothelial accumulation of cholesterol, immune cells, and extracellular matrix. At the early onset of atherogenesis, endothelial dysfunction takes place. Atherogenesis is further triggered by the accumulation of cholesterol-carrying low-density lipoproteins, which acquire properties of damage-associated molecular patterns and thereby trigger an inflammatory response. Following activation of the innate immune response, mainly governed by monocytes and macrophages, the adaptive immune response is started which further promotes atherosclerotic plaque formation. In this review, an overview is given describing the role of damage-associated molecular patterns, NLRP3 inflammasome activation, and innate and adaptive immune cells in the atherogenesis process.
Collapse
Affiliation(s)
- Kapka Miteva
- Department of Biomedical Sciences, Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano, Milano, Italy
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Raffaele De Caterina
- Center of Aging Sciences and Translational Medicine - CESI-MeT, Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany; Department of Cardiology, Charité, University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
28
|
Schiattarella GG, Madonna R, Van Linthout S, Thum T, Schulz R, Ferdinandy P, Perrino C. Epigenetic modulation of vascular diseases: Assessing the evidence and exploring the opportunities. Vascul Pharmacol 2018; 107:S1537-1891(17)30468-8. [PMID: 29548901 DOI: 10.1016/j.vph.2018.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 01/27/2018] [Accepted: 02/22/2018] [Indexed: 02/09/2023]
Abstract
Vascular adaptations to either physiological or pathophysiological conditions commonly require gene expression modifications in the most represented cellular elements of the vessel wall, i.e. endothelial and smooth muscle cells. In addition to transcription factors, a number of mechanisms contribute to the regulation of gene expression in these cells including noncoding RNAs, histone and DNA modifications, collectively indicated as epigenetic modifications. Here, we summarize the state of art regarding the role of epigenetic changes in major vascular diseases, and discuss the potential diagnostic and therapeutic applications of epigenetic modulation in this context.
Collapse
Affiliation(s)
| | - Rosalinda Madonna
- Center for Aging Sciences and Translational Medicine - CESI-MeT, Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences, "G. D'Annunzio" University, Chiety, Italy; Center for Cardiovascular Biology and Atherosclerosis Research, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow, Berlin, Germany; Charité University Medicine Berlin, Campus Rudolf Virchow, Department of Cardiology, Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Giessen, Germany
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
| |
Collapse
|
29
|
Van Linthout S, Hamdani N, Miteva K, Koschel A, Müller I, Pinzur L, Aberman Z, Pappritz K, Linke WA, Tschöpe C. Placenta-Derived Adherent Stromal Cells Improve Diabetes Mellitus-Associated Left Ventricular Diastolic Performance. Stem Cells Transl Med 2017; 6:2135-2145. [PMID: 29024485 PMCID: PMC5702519 DOI: 10.1002/sctm.17-0130] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/25/2017] [Indexed: 12/23/2022] Open
Abstract
Left ventricular (LV) diastolic dysfunction is among others attributed to cardiomyocyte stiffness. Mesenchymal stromal cells (MSC) have cardiac-protective properties. We explored whether intravenous (i.v.) application of PLacenta-eXpanded (PLX) MSC-like cells (PLX) improves LV diastolic relaxation in streptozotocin (STZ)-induced diabetic mice and investigated underlying mechanisms. Diabetes mellitus was induced by STZ application (50 mg/kg body weight) during five subsequent days. One week after the first STZ injection, PLX or saline were i.v. applied. Two weeks later, mice were hemodynamically characterized and sacrificed. At this early stage of diabetic cardiomyopathy with low-grade inflammation and no cardiac fibrosis, PLX reduced LV vascular cell adhesion molecule-1, transforming growth factor-β1, and interferon-γ mRNA expression, induced the percentage of circulating regulatory T cells, and decreased the splenic pro-fibrotic potential in STZ mice. STZ + PLX mice exhibited higher LV vascular endothelial growth factor mRNA expression and arteriole density versus STZ mice. In vitro, hyperglycemic PLX conditioned medium restored the hyperglycemia-impaired tube formation and adhesion capacity of human umbelical vein endothelial cells (HUVEC) via increasing nitric oxide (NO) bioavailability. PLX further induced the diabetes-downregulated activity of the NO downstream protein kinase G, as well as of protein kinase A, in STZ mice, which was associated with a raise in phosphorylation of the titin isoforms N2BA and N2B. Concomitantly, the passive force was lower in single isolated cardiomyocytes from STZ + PLX versus from STZ mice, which led to an improvement of LV diastolic relaxation. We conclude that i.v. PLX injection improves diabetes mellitus-associated diastolic performance via decreasing cardiomyocyte stiffness. Stem Cells Translational Medicine 2017;6:2135-2145.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Charité - Universitätsmedizin Berlin, Germany.,Department of Cardiology, Charité - University Medicine Berlin, Campus Virchow, Berlin, Germany.,DZHK (German Center for Cardiovascular Research) partner site Berlin, Germany
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Kapka Miteva
- Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Charité - Universitätsmedizin Berlin, Germany.,Department of Cardiology, Charité - University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Annika Koschel
- Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Charité - Universitätsmedizin Berlin, Germany
| | - Irene Müller
- Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Charité - Universitätsmedizin Berlin, Germany.,DZHK (German Center for Cardiovascular Research) partner site Berlin, Germany
| | | | | | - Kathleen Pappritz
- Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Charité - Universitätsmedizin Berlin, Germany.,DZHK (German Center for Cardiovascular Research) partner site Berlin, Germany
| | | | - Carsten Tschöpe
- Berlin-Brandenburg Center for Regenerative Therapies, Campus Virchow Charité - Universitätsmedizin Berlin, Germany.,Department of Cardiology, Charité - University Medicine Berlin, Campus Virchow, Berlin, Germany.,DZHK (German Center for Cardiovascular Research) partner site Berlin, Germany
| |
Collapse
|
30
|
Pereiro P, Forn-Cuní G, Dios S, Coll J, Figueras A, Novoa B. Interferon-independent antiviral activity of 25-hydroxycholesterol in a teleost fish. Antiviral Res 2017; 145:146-159. [PMID: 28789986 DOI: 10.1016/j.antiviral.2017.08.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/21/2017] [Accepted: 08/04/2017] [Indexed: 01/06/2023]
Abstract
Oxysterols are a family of cholesterol oxygenated derivatives with diverse roles in many biological activities and have recently been linked with the induction of a cellular antiviral state. The antiviral effects of 25-hydroxycholesterol (25HC) extend to several mammalian enveloped and non-enveloped viruses. It has been reported that the expression of the gene encoding cholesterol 25-hydroxylase (CH25H) is induced by interferons (IFNs). In this work, five ch25h genes were identified in the zebrafish (Danio rerio) genome. The ch25h genes showed different tissue expression patterns and differed in their expression after immune stimulation with lipopolysaccharide (LPS), polyinosinic:polycytidylic acid (PolyI:C) and Spring Viremia Carp Virus (SVCV). Only one of the 5 genes, ch25hb, was overexpressed after the administration of the treatments. Synteny and phylogenetic analyses revealed that ch25hb is the putative homolog of mammalian Ch25h in zebrafish, while the remaining zebrafish ch25h genes are products of duplications within the teleost lineage. Interestingly, its modulation was not mediated by type I IFNs, contrasting previous reports on mammalian orthologs. Nevertheless, in vivo overexpression of ch25hb in zebrafish larvae significantly reduced mortality after SVCV challenge. Viral replication was also negatively affected by 25HC administration to the zebrafish cell line ZF4. In conclusion, the interferon-independent antiviral role of 25HC was extended to a non-mammalian species for the first time, and dual activity that both protects the cells and interacts with the virus cannot be discarded.
Collapse
Affiliation(s)
| | | | - Sonia Dios
- Instituto de Investigaciones Marinas (IIM-CSIC), Vigo, Spain
| | - Julio Coll
- Department of Biotechnology, Instituto Nacional Investigaciones Agrarias (INIA), Madrid, Spain
| | | | - Beatriz Novoa
- Instituto de Investigaciones Marinas (IIM-CSIC), Vigo, Spain.
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW With the intention to summarize the currently available evidence on the pathophysiological relevance of inflammation in heart failure, this review addresses the question whether inflammation is a cause or consequence of heart failure, or both. RECENT FINDINGS This review discusses the diversity (sterile, para-inflammation, chronic inflammation) and sources of inflammation and gives an overview of how inflammation (local versus systemic) can trigger heart failure. On the other hand, the review is outlined how heart failure-associated wall stress and signals released by stressed, malfunctioning, or dead cells (DAMPs: e.g., mitochondrial DNA, ATP, S100A8, matricellular proteins) induce cardiac sterile inflammation and how heart failure provokes inflammation in various peripheral tissues in a direct (inflammatory) and indirect (hemodynamic) manner. The crosstalk between the heart and peripheral organs (bone marrow, spleen, gut, adipose tissue) is outlined and the importance of neurohormonal mechanisms including the renin angiotensin aldosteron system and the ß-adrenergic nervous system in inflammation and heart failure is discussed. Inflammation and heart failure are strongly interconnected and mutually reinforce each other. This indicates the difficulty to counteract inflammation and heart failure once this chronic vicious circle has started and points out the need to control the inflammatory process at an early stage avoiding chronic inflammation and heart failure. The diversity of inflammation further addresses the need for a tailored characterization of inflammation enabling differentiation of inflammation and subsequent target-specific strategies. It is expected that the characterization of the systemic and/or cardiac immune profile will be part of precision medicine in the future of cardiology.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Tschöpe
- Berlin-Brandenburg Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Cardiology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
32
|
de Castro MRT, Ferreira APDO, Busanello GL, da Silva LRH, da Silveira Junior MEP, Fiorin FDS, Arrifano G, Crespo-López ME, Barcelos RP, Cuevas MJ, Bresciani G, González-Gallego J, Fighera MR, Royes LFF. Previous physical exercise alters the hepatic profile of oxidative-inflammatory status and limits the secondary brain damage induced by severe traumatic brain injury in rats. J Physiol 2017; 595:6023-6044. [PMID: 28726269 DOI: 10.1113/jp273933] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/19/2017] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS An early inflammatory response and oxidative stress are implicated in the signal transduction that alters both hepatic redox status and mitochondrial function after traumatic brain injury (TBI). Peripheral oxidative/inflammatory responses contribute to neuronal dysfunction after TBI Exercise training alters the profile of oxidative-inflammatory status in liver and protects against acute hyperglycaemia and a cerebral inflammatory response after TBI. Approaches such as exercise training, which attenuates neuronal damage after TBI, may have therapeutic potential through modulation of responses by metabolic organs. The vulnerability of the body to oxidative/inflammatory in TBI is significantly enhanced in sedentary compared to physically active counterparts. ABSTRACT Although systemic responses have been described after traumatic brain injury (TBI), little is known regarding potential interactions between brain and peripheral organs after neuronal injury. Accordingly, we aimed to investigate whether a peripheral oxidative/inflammatory response contributes to neuronal dysfunction after TBI, as well as the prophylactic role of exercise training. Animals were submitted to fluid percussion injury after 6 weeks of swimming training. Previous exercise training increased mRNA expression of X receptor alpha and ATP-binding cassette transporter, and decreased inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor (TNF)-α and interleukin (IL)-6 expression per se in liver. Interestingly, exercise training protected against hepatic inflammation (COX-2, iNOS, TNF-α and IL-6), oxidative stress (decreases in non-protein sulfhydryl and glutathione, as well as increases in 2',7'-dichlorofluorescein diacetate oxidation and protein carbonyl), which altered hepatic redox status (increases in myeloperoxidase and superoxide dismutase activity, as well as inhibition of catalase activity) mitochondrial function (decreases in methyl-tetrazolium and Δψ, as well as inhibition of citrate synthase activity) and ion gradient homeostasis (inhibition of Na+ ,K+ -ATPase activity inhibition) when analysed 24 h after TBI. Previous exercise training also protected against dysglycaemia, impaired hepatic signalling (increase in phosphorylated c-Jun NH2-terminal kinase, phosphorylated decreases in insulin receptor substrate and phosphorylated AKT expression), high levels of circulating and neuronal cytokines, the opening of the blood-brain barrier, neutrophil infiltration and Na+ ,K+ -ATPase activity inhibition in the ipsilateral cortex after TBI. Moreover, the impairment of protein function, neurobehavioural (neuromotor dysfunction and spatial learning) disability and hippocampal cell damage in sedentary rats suggests that exercise training also modulates peripheral oxidative/inflammatory pathways in TBI, which corroborates the ever increasing evidence regarding health-related outcomes with respect to a physically active lifestyle.
Collapse
Affiliation(s)
- Mauro Robson Torres de Castro
- Programa de Pós-graduação em Educação Física.,Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício
| | | | - Guilherme Lago Busanello
- Programa de Pós-graduação em Educação Física.,Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício
| | | | | | - Fernando da Silva Fiorin
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Gabriela Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Maria Elena Crespo-López
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas (ICB), Universidade Federal do Pará (UFPA), Belém, Brazil
| | - Rômulo Pillon Barcelos
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - María J Cuevas
- Institute of Biomedicine (IBIOMED) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), University of León, León, Spain
| | - Guilherme Bresciani
- Escuela de Educación Física, Pontificia Universidad Católica de Valparaiso (PUCV), Valparaiso, Chile
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED) and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), University of León, León, Spain
| | - Michele Rechia Fighera
- Programa de Pós-graduação em Educação Física.,Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício.,Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Luiz Fernando Freire Royes
- Programa de Pós-graduação em Educação Física.,Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício.,Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| |
Collapse
|
33
|
Gong Y, Yang Y, Wu Q, Gao G, Liu Y, Xiong Y, Huang C, Wu S. Activation of LXRα improves cardiac remodeling induced by pulmonary artery hypertension in rats. Sci Rep 2017; 7:6169. [PMID: 28733583 PMCID: PMC5522383 DOI: 10.1038/s41598-017-04640-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/17/2017] [Indexed: 12/25/2022] Open
Abstract
Inflammatory factors regulated by NF-κB play a significant role in PAH and myocardial hypertrophy. LXR activation may inhibit myocardial hypertrophy via suppressing inflammatory pathways; it is unknown whether LXR is also involved in PAH-induced myocardial hypertrophy or remodeling. To further explore the protective effect of LXR in PAH-induced cardiac hypertrophy and remodeling, a PAH model was developed, and T0901317, an agonist of LXR, was used to examine the effect of LXR activation. PAH rats demonstrated obvious cardiac hypertrophy and remodeling in the right ventricle, but significant improvement of cardiac hypertrophy and remodeling was observed in PAH rats treated with T0901317. Through RT-PCR, Western blot and ELISA examination, NF-κB, IL-6, TNF-α, and iNOS were found to be significantly reduced in PAH rats treated with T0901317 compared to PAH rats treated with DMSO. Apoptosis was also significantly reduced in PAH rats treated with T0901317. Thus, LXR activation may inhibit PAH-induced cardiac hypertrophy and remodeling by inhibiting NF-κB-mediated inflammatory pathways.
Collapse
Affiliation(s)
- Yibo Gong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qin Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ge Gao
- Faculty of Laboratory Medicine, Xiangya Medical College, Central South University, Changsha, China
| | - Yin Liu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yaoyao Xiong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Can Huang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Sijie Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
34
|
Tanaka T, Takei Y, Zaima N, Moriyama T, Yamanouchi D. Hyperglycemia Suppresses RANKL-Induced Osteoclast Differentiation through LXRβ Expression in RAW264.7 Cells. J Nutr Sci Vitaminol (Tokyo) 2017; 63:28-34. [PMID: 28367923 DOI: 10.3177/jnsv.63.28] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
There have been reports that hyperglycemia suppresses osteoclast (OCL) differentiation, although the underlying mechanism is poorly understood. Here we demonstrated that high glucose suppresses OCL differentiation through activation of liver X receptor (LXR) β, a recently reported glucose-sensing nuclear receptor. The effect of hyperglycemia on osteoclastogenesis was tested in RAW264.7 cells, a murine macrophage cell line. Cells were treated with receptor activator of NF-κB ligand (RANKL) under normoglycemic (5.5 mM glucose), normoglycemic with high osmotic pressure (5.5 mM glucose + 10.0 mM mannitol), and hyperglycemic (15.5 mM glucose) conditions. RANKL-induced osteoclastogenesis was significantly suppressed by high-glucose treatment. Mannitol treatment also significantly suppressed osteoclastogenesis, but the inhibitory effect was lower than for high-glucose treatment. The suppression of mRNA expression of Lxrβ by RANKL was significantly restored by high glucose, but not mannitol. Additionally, the deactivation of Lxrβ by siRNA attenuated high-glucose-induced suppression of osteoclastogenesis. Although further validation of the underlying pathway is necessary, targeting LXRβ is a potential therapeutic approach to treating osteoporosis.
Collapse
Affiliation(s)
- Teruyoshi Tanaka
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health
| | | | | | | | | |
Collapse
|
35
|
Regitz-Zagrosek V, Kararigas G. Mechanistic Pathways of Sex Differences in Cardiovascular Disease. Physiol Rev 2017; 97:1-37. [PMID: 27807199 DOI: 10.1152/physrev.00021.2015] [Citation(s) in RCA: 458] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Major differences between men and women exist in epidemiology, manifestation, pathophysiology, treatment, and outcome of cardiovascular diseases (CVD), such as coronary artery disease, pressure overload, hypertension, cardiomyopathy, and heart failure. Corresponding sex differences have been studied in a number of animal models, and mechanistic investigations have been undertaken to analyze the observed sex differences. We summarize the biological mechanisms of sex differences in CVD focusing on three main areas, i.e., genetic mechanisms, epigenetic mechanisms, as well as sex hormones and their receptors. We discuss relevant subtypes of sex hormone receptors, as well as genomic and nongenomic, activational and organizational effects of sex hormones. We describe the interaction of sex hormones with intracellular signaling relevant for cardiovascular cells and the cardiovascular system. Sex, sex hormones, and their receptors may affect a number of cellular processes by their synergistic action on multiple targets. We discuss in detail sex differences in organelle function and in biological processes. We conclude that there is a need for a more detailed understanding of sex differences and their underlying mechanisms, which holds the potential to design new drugs that target sex-specific cardiovascular mechanisms and affect phenotypes. The comparison of both sexes may lead to the identification of protective or maladaptive mechanisms in one sex that could serve as a novel therapeutic target in one sex or in both.
Collapse
Affiliation(s)
- Vera Regitz-Zagrosek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
36
|
Ma Y, Ma Z, Yin S, Yan X, Wang J. Arsenic and fluoride induce apoptosis, inflammation and oxidative stress in cultured human umbilical vein endothelial cells. CHEMOSPHERE 2017; 167:454-461. [PMID: 27750169 DOI: 10.1016/j.chemosphere.2016.10.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/06/2016] [Accepted: 10/07/2016] [Indexed: 06/06/2023]
Abstract
Excessive amount of inorganic arsenic (iAs) and fluoride (F) coexist in drinking water in many regions, which is associated with high risk of vascular diseases. However, the underlying mechanisms are not well studied. The present study was to evaluate the effects of iAs and F individual or combined exposure on endothelial activation and apoptosis in vitro. Primary human umbilical vein endothelial cells (HUVECs) were exposed to 5 μM As2O3 and/or 1 mM NaF. Changes in endothelial cell apoptosis, inflammation, oxidative stress and nitric oxide (NO) production were analyzed. The results showed that iAs and/or F induced significant increase in endothelial cell apoptosis and inflammation as indicated by the increase of mRNA and protein expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and pentraxin 3. Furthermore, iAs and/or F exposure induced intracellular reactive oxygen species and malondialdehyde generation. Results showed iAs and/or F exposure increased the activity of NADPH oxidase (NOX) and up-regulated the mRNA expression of NOX subunits p22phox. The results indicated that activation of NOX was related to oxidative stress induced by iAs and/or F. Also, iAs and/or F reduced NO production in HUVECs. The up-regulation of inflammation genes expression and oxidative stress in iAs and F co-exposed ECs were less pronounced as compared to single F-exposed cells, which showed an antagonistic effect between iAs and F. In conclusion, endothelial activation and apoptosis induced by iAs and/or F are potential mechanisms in their vascular toxicity. Oxidative stress and impaired NO production are involved in their pro-inflammatory and pro-apoptotic effects.
Collapse
Affiliation(s)
- Yanqin Ma
- College of Life Science, Shanxi Agricultural University, Taigu, Shanxi 030801, China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Zhenhua Ma
- College of Life Science, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Shuqin Yin
- College of Life Science, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xiaoyan Yan
- Health Toxicology Department, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jundong Wang
- Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| |
Collapse
|
37
|
Once and for all, LXRα and LXRβ are gatekeepers of the endocrine system. Mol Aspects Med 2016; 49:31-46. [DOI: 10.1016/j.mam.2016.04.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/08/2016] [Accepted: 04/10/2016] [Indexed: 01/08/2023]
|
38
|
Tanaka T, Takei Y, Yamanouchi D. Hyperglycemia Suppresses Calcium Phosphate-Induced Aneurysm Formation Through Inhibition of Macrophage Activation. J Am Heart Assoc 2016; 5:e003062. [PMID: 27021877 PMCID: PMC4943277 DOI: 10.1161/jaha.115.003062] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background The aim of this study was to elucidate aspects of diabetes mellitus–induced suppression of aneurysm. We hypothesized that high glucose suppresses aneurysm by inhibiting macrophage activation via activation of Nr1h2 (also known as liver X receptor β), recently characterized as a glucose‐sensing nuclear receptor. Methods and Results Calcium phosphate (CaPO4)–induced aneurysm formation was significantly suppressed in the arterial wall in type 1 and 2 diabetic mice. A murine macrophage cell line, RAW264.7, was treated with tumor necrosis factor α (TNF‐α) plus CaPO4 and showed a significant increase in matrix metalloproteinase 9 (Mmp9) mRNA and secreted protein expression compared with TNF‐α alone. Elevated Mmp9 expression was significantly suppressed by hyperglycemic conditions (15.5 mmol/L glucose) compared with normoglycemic conditions (5.5 mmol/L glucose) or normoglycemic conditions with high osmotic pressure (5.5 mmol/L glucose +10.0 mmol/L mannitol). Nr1h2 mRNA and protein expression were suppressed by treatment with TNF‐α plus CaPO4 but were restored by hyperglycemic conditions. Activation of Nr1h2 by the antagonist GW3965 during stimulation with TNF‐α plus CaPO4 mimicked hyperglycemic conditions and inhibited Mmp9 upregulation, whereas the deactivation of Nr1h2 by small interfering RNA (siRNA) under hyperglycemic conditions canceled the suppressive effect and restored Mmp9 expression induced by TNF‐α plus CaPO4. Moreover, Nr1h2 activation with GW3965 significantly suppressed CaPO4‐induced aneurysm in mice compared with vehicle‐injected control mice. Conclusions Our results show that hyperglycemia suppresses macrophage activation and aneurysmal degeneration through the activation of Nr1h2. Although further validation of the underlying pathway is necessary, targeting Nr1h2 is a potential therapeutic approach to treating aneurysm.
Collapse
Affiliation(s)
- Teruyoshi Tanaka
- Division of Vascular Surgery, Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Yuichiro Takei
- Division of Vascular Surgery, Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Dai Yamanouchi
- Division of Vascular Surgery, Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
39
|
Rohrmann S, Linseisen J, Allenspach M, von Eckardstein A, Müller D. Plasma Concentrations of Trimethylamine-N-oxide Are Directly Associated with Dairy Food Consumption and Low-Grade Inflammation in a German Adult Population. J Nutr 2016; 146:283-9. [PMID: 26674761 DOI: 10.3945/jn.115.220103] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/09/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Trimethylamine-N-oxide (TMAO) is a metabolite of carnitine, choline, and phosphatidylcholine, which is inversely associated with survival of cardiovascular disease (CVD) patients. OBJECTIVE We examined the associations of diet with plasma concentrations of TMAO, choline, and betaine and the associations of TMAO with plasma concentrations of various cytokines. METHODS Plasma TMAO, choline, and betaine concentrations were measured using LC-high resolution mass spectrometry in 271 participants, ≥18 y old, of the Second Bavarian Food Consumption Survey, conducted in 2002 and 2003. Food consumption was assessed using at least two 24-h dietary recalls. Cytokines were measured in plasma with enzyme-linked immunosorbent assays. Geometric mean concentrations of TMAO, choline, and betaine by categories of meat, dairy food, egg, and fish consumption were computed, adjusted for sex, age, and BMI. Multivariable-adjusted geometric mean concentrations of cytokines [tumor necrosis factor-α (TNF-α), soluble TNF receptors (sTNF-R) p55, sTNF-R p75, interleukin-6 (IL-6), and C-reactive protein (CRP)] were computed by quartiles of TMAO concentration using general linear models. RESULTS Meat, egg, or fish consumption was not associated with TMAO, choline, or betaine concentrations (all P-trend ≥ 0.05). With increases in milk and other dairy food consumption, the plasma TMAO concentration increased [geometric mean bottom quartile of milk consumption: 2.08 μM (95% CI: 1.69, 2.57 μM); compared with top quartile: 3.13 μM (95% CI: 2.56, 3.84 μM); P-trend = 0.008]. Participants in the top TMAO quartile had higher plasma concentrations of TNF-α, sTNF-R p55, and sTNF-R p75 than participants in the bottom quartile (percentage difference ranging between 14.4% and 17.3%; all P-trend < 0.05), but there were no differences in plasma concentrations of CRP and IL-6 (all P-trend ≥ 0.05). CONCLUSIONS Results of this study conducted among healthy adults from the general population do not indicate a strong effect of diet on plasma concentrations of TMAO, choline, or betaine, with the exception of a positive association between dairy food consumption and plasma TMAO concentrations. Also, plasma TMAO concentrations were positively associated with inflammation. Whether habitual diet is strongly linked to the plasma TMAO concentration, a potential marker of CVD risk, needs to be determined in further studies.
Collapse
Affiliation(s)
- Sabine Rohrmann
- Division of Chronic Disease Epidemiology, Epidemiology, Biostatistics and Prevention Institute, and
| | - Jakob Linseisen
- Institute of Epidemiology II, Helmholtz Centre Munich, Neuherberg, Germany
| | - Martina Allenspach
- Institute of Clinical Chemistry, University of Zurich, Zurich Switzerland; and
| | | | - Daniel Müller
- Institute of Clinical Chemistry, University of Zurich, Zurich Switzerland; and
| |
Collapse
|
40
|
Emerging role of liver X receptors in cardiac pathophysiology and heart failure. Basic Res Cardiol 2015; 111:3. [PMID: 26611207 PMCID: PMC4661180 DOI: 10.1007/s00395-015-0520-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/03/2015] [Indexed: 01/09/2023]
Abstract
Liver X receptors (LXRs) are master regulators of metabolism and have been studied for their pharmacological potential in vascular and metabolic disease. Besides their established role in metabolic homeostasis and disease, there is mounting evidence to suggest that LXRs may exert direct beneficial effects in the heart. Here, we aim to provide a conceptual framework to explain the broad mode of action of LXRs and how LXR signaling may be an important local and systemic target for the treatment of heart failure. We discuss the potential role of LXRs in systemic conditions associated with heart failure, such as hypertension, diabetes, and renal and vascular disease. Further, we expound on recent data that implicate a direct role for LXR activation in the heart, for its impact on cardiomyocyte damage and loss due to ischemia, and effects on cardiac hypertrophy, fibrosis, and myocardial metabolism. Taken together, the accumulating evidence supports the notion that LXRs may represent a novel therapeutic target for the treatment of heart failure.
Collapse
|
41
|
Fan A, Wang Q, Yuan Y, Cheng J, Chen L, Guo X, Li Q, Chen B, Huang X, Huang Q. Liver X receptor-α and miR-130a-3p regulate expression of sphingosine 1-phosphate receptor 2 in human umbilical vein endothelial cells. Am J Physiol Cell Physiol 2015; 310:C216-26. [PMID: 26669941 DOI: 10.1152/ajpcell.00102.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/23/2015] [Indexed: 02/07/2023]
Abstract
Recent studies have shown that activation of liver X receptors (LXRs) attenuates the development of atherosclerosis, not only by regulating lipid metabolism but also by suppressing inflammatory signaling. Sphingosine 1-phosphate receptor 2 (S1PR2), an important inflammatory gene product, plays a role in the development of various inflammatory diseases. It was proposed that S1PR2 might be regulated by LXR-α. In the present study, the effect of LXR-α on tumor necrosis factor-α (TNF-α)-induced S1PR2 expression in human umbilical vein endothelial cells (HUVECs) was investigated and the underlying mechanism was explored. The results demonstrated that TNF-α led to an increase in S1PR2 expression and triggered a downregulation of LXR-α expression in HUVECs as well. Downregulation of LXR-α with specific small interfering RNA (siRNA) remarkably enhanced the primary as well as TNF-α-induced expression of S1PR2 in HUVECs. Activation of LXR-α by agonist GW3965 inhibited both primary and TNF-α-induced S1PR2 expression. GW3965 also attenuated S1PR2-induced endothelial barrier dysfunction. The data further showed that TNF-α induced a significant decrease in miR-130a-3p expression. Overexpression of miR-130a-3p with mimic product reduced S1PR2 protein expression, and inhibition of miR-130a-3p by specific inhibitor resulted in an increase in S1PR2 protein expression. Furthermore, activation of LXRs with agonist enhanced the expression of miR-130a-3p, and knockdown of LXR-α by siRNA suppressed miR-130a-3p expression. These results suggest that LXR-α might downregulate S1PR2 expression via miR-130a-3p in quiescent HUVECs. Stimulation of TNF-α attenuates the activity of LXR-α and results in enhanced S1PR2 expression.
Collapse
Affiliation(s)
- Aihui Fan
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China; Department of Physiology, Guangdong Medical College, Dongguan, People's Republic of China; and
| | - Qian Wang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Yongjun Yuan
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Jilun Cheng
- Department of Pharmacology, Guangdong Medical College, Dongguan, People's Republic of China
| | - Lixian Chen
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaohua Guo
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Qiang Li
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Bo Chen
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Xuliang Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China;
| | - Qiaobing Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
42
|
Tschöpe C, Van Linthout S. New insights in (inter)cellular mechanisms by heart failure with preserved ejection fraction. Curr Heart Fail Rep 2015; 11:436-44. [PMID: 25189801 PMCID: PMC4221658 DOI: 10.1007/s11897-014-0219-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Recently, a new paradigm for the development of heart failure with preserved ejection fraction (HFpEF) has been proposed, which identifies a systemic pro-inflammatory state induced by comorbidities as the origin of microvascular endothelial cell inflammation and subsequent concentric cardiac remodeling and dysfunction. This review further discusses the pivotal role of the inflamed endothelium in the pathogenesis of HFpEF-specific cardiac remodeling. The potential importance of reciprocal interactions of the endothelium with cardiac fibroblasts and cardiomyocytes and with the cardiac neurohumoral response in this cardiac remodeling process is outlined.
Collapse
Affiliation(s)
- Carsten Tschöpe
- Department of Cardiology and Pneumology, Charité, University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany,
| | | |
Collapse
|
43
|
Yang M, Wang R, Sun J, Yu K, Chen B, Xu L, Zhao B, Wang H. The liver X receptor agonist TO901317 protects mice against cisplatin-induced kidney injury. Exp Biol Med (Maywood) 2015; 240:1717-27. [PMID: 26062799 DOI: 10.1177/1535370215589906] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/21/2015] [Indexed: 12/13/2022] Open
Abstract
Liver X receptors are in the nuclear receptor superfamily and are contained in the regulation of lipid and cholesterol metabolism. Besides, liver X receptors are considered crucial regulators of the inflammatory response and innate immunity. The current study evaluates the in vivo effects that the synthetic liver X receptor agonist TO901317 protects against cisplatin-induced kidney injury in mice. Mice received cisplatin administration through a single intraperitoneal injection (20 mg/kg in saline). And then the mice were treated with the TO901317 by daily gavage (10 mg/kg/day) 12 h postcisplatin administration, and cisplatin nephrotoxicity was evaluated. At 72 h after cisplatin treatment, elevated plasma urea and creatinine levels (P < 0.05) were evidenced which indicates the renal dysfunction of the vehicle-treated mice, consistent with tubular necrosis, protein cast, dilation of renal tubules, and desquamation of epithelial cells in renal tubules. In contrast, the severity of renal dysfunction and histological damage was reduced in TO901317 treated mice (P < 0.05). In accordance, circulating tumor necrosis factor alpha levels, renal tumor necrosis factor alpha, p47(phox), gp91(phox), and protein expression levels and COX-2 mRNA, renal monocyte chemoattractant protein 1, VACAM-1 mRNA and intercellular adhesion molecule-1 contents, and renal prostaglandin E2 amounts, were higher in samples from cisplatin-treated mice in comparison with controls (P < 0.05) but attenuated in the TO901317 treatment group (P < 0.05). Taken together, treatment with the liver X receptor agonist TO901317 ameliorated the inflammatory response and oxidative stress in cisplatin-induced kidney injury in mice.
Collapse
Affiliation(s)
- Meng Yang
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Rong Wang
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Jing Sun
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Kezhou Yu
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Bing Chen
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Liang Xu
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Bing Zhao
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Haiping Wang
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| |
Collapse
|
44
|
Engelhardt PF, Seklehner S, Brustmann H, Riedl C, Lusuardi L. Tumor necrosis factor-α expression in patients with obstructive benign prostatic hyperplasia is associated with a higher incidence of asymptomatic inflammatory prostatitis NIH category IV and prostatic calcification. Scand J Urol 2015; 49:472-478. [DOI: 10.3109/21681805.2015.1044560] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
45
|
Arginase as a Critical Prooxidant Mediator in the Binomial Endothelial Dysfunction-Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:924860. [PMID: 26064427 PMCID: PMC4434223 DOI: 10.1155/2015/924860] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/11/2014] [Indexed: 12/31/2022]
Abstract
Arginase is a metalloenzyme which hydrolyzes L-arginine to L-ornithine and urea. Since its discovery, in the early 1900s, this enzyme has gained increasing attention, as literature reports have progressively pointed to its critical participation in regulating nitric oxide bioavailability. Indeed, accumulating evidence in the following years would picture arginase as a key player in vascular health. Recent studies have highlighted the arginase regulatory role in the progression of atherosclerosis, the latter an essentially prooxidant state. Apart from the fact that arginase has been proven to impair different metabolic pathways, and also as a consequence of this, the repercussions of the actions of such enzyme go further than first thought. In fact, such metalloenzyme exhibits direct implications in multiple cardiometabolic diseases, among which are hypertension, type 2 diabetes, and hypercholesterolemia. Considering the epidemiological repercussions of these clinical conditions, arginase is currently seen under the spotlights of the search for developing specific inhibitors, in order to mitigate its deleterious effects. That said, the present review focuses on the role of arginase in endothelial function and its participation in the establishment of atherosclerotic lesions, discussing the main regulatory mechanisms of the enzyme, also highlighting the potential development of pharmacological strategies in related cardiovascular diseases.
Collapse
|
46
|
Tai LM, Ghura S, Koster KP, Liakaite V, Maienschein‐Cline M, Kanabar P, Collins N, Ben‐Aissa M, Lei AZ, Bahroos N, Green SJ, Hendrickson B, Van Eldik LJ, LaDu MJ. APOE-modulated Aβ-induced neuroinflammation in Alzheimer's disease: current landscape, novel data, and future perspective. J Neurochem 2015; 133:465-88. [PMID: 25689586 PMCID: PMC4400246 DOI: 10.1111/jnc.13072] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 01/12/2023]
Abstract
Chronic glial activation and neuroinflammation induced by the amyloid-β peptide (Aβ) contribute to Alzheimer's disease (AD) pathology. APOE4 is the greatest AD-genetic risk factor; increasing risk up to 12-fold compared to APOE3, with APOE4-specific neuroinflammation an important component of this risk. This editorial review discusses the role of APOE in inflammation and AD, via a literature review, presentation of novel data on Aβ-induced neuroinflammation, and discussion of future research directions. The complexity of chronic neuroinflammation, including multiple detrimental and beneficial effects occurring in a temporal and cell-specific manner, has resulted in conflicting functional data for virtually every inflammatory mediator. Defining a neuroinflammatory phenotype (NIP) is one way to address this issue, focusing on profiling the changes in inflammatory mediator expression during disease progression. Although many studies have shown that APOE4 induces a detrimental NIP in peripheral inflammation and Aβ-independent neuroinflammation, data for APOE-modulated Aβ-induced neuroinflammation are surprisingly limited. We present data supporting the hypothesis that impaired apoE4 function modulates Aβ-induced effects on inflammatory receptor signaling, including amplification of detrimental (toll-like receptor 4-p38α) and suppression of beneficial (IL-4R-nuclear receptor) pathways. To ultimately develop APOE genotype-specific therapeutics, it is critical that future studies define the dynamic NIP profile and pathways that underlie APOE-modulated chronic neuroinflammation. In this editorial review, we present data supporting the hypothesis that impaired apoE4 function modulates Aβ-induced effects on inflammatory receptor signaling, including amplification of detrimental (TLR4-p38α) and suppression of beneficial (IL-4R-nuclear receptor) pathways, resulting in an adverse NIP that causes neuronal dysfunction. NIP, Neuroinflammatory phenotype; P.I., pro-inflammatory; A.I., anti-inflammatory.
Collapse
Affiliation(s)
- Leon M. Tai
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Shivesh Ghura
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Kevin P. Koster
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | | | | | - Pinal Kanabar
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | - Nicole Collins
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Manel Ben‐Aissa
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Arden Zhengdeng Lei
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | - Neil Bahroos
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | | | - Bill Hendrickson
- UIC Research Resources CenterUniversity of IllinoisChicagoIllinoisUSA
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| |
Collapse
|
47
|
Ma Y, Li W, Yin Y, Li W. AST IV inhibits H₂O₂-induced human umbilical vein endothelial cell apoptosis by suppressing Nox4 expression through the TGF-β1/Smad2 pathway. Int J Mol Med 2015; 35:1667-74. [PMID: 25891879 DOI: 10.3892/ijmm.2015.2188] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/08/2015] [Indexed: 11/06/2022] Open
Abstract
Endothelial cell apoptosis plays an important role in the pathophysiological mechanisms of vascular complications in diabetes mellitus (DM). NADPH oxidase 4 (Nox4)-dependent reactive oxygen species (ROS) aggregation is the main cause of vascular endothelial cell apoptosis. The transforming growth factor-β1 (TGF-β1)/Smad2 signaling pathway is involved in the apoptosis of several types of cells. However, the association between vascular endothelial cell apoptosis and Nox4, and the involvement of the TGF-β1/Smad2 signaling pathway in vascular endothelial cell apoptosis remain unclear. In the present study, we aimed to investigate the role of Nox4-dependent ROS production and to determine the involvement of the TGF-β1/Smad2 signaling pathway in endothelial cell apoptosis induced by oxidative stress which causes vascular injury in DM. We demonstrated that hydrogen peroxide (H2O2) increased Nox4-dependent-ROS aggregation, as well as the expression of TGF-β1, Smad2, Bax and caspase-3, decreased Bcl-2 expression and increased the apoptosis of human umbilical vein endothelial cells (HUVECs). Treatment with diphenyliodonium (DPI), a specific inhibitor of Nox4 or astragaloside IV (AST IV), a monomer located in an extract of astragaloside, decreased Nox4 expression and the levels of ROS, decreased TGF-β1 and Smad2 expression, altered the expression of apoptosis-related genes and decreased the apoptosis of HUVECs. Treatment with LY2109761, a selective inhibitor of the TGF-β1/Smad2 pathway, produced results similar to those of DPI; however, LY2109761 had no effect on Nox4 expression and ROS levels. Taken together, the findings of the present study suggest that H2O2 contributes to HUVEC apoptosis by inducing Nox4-dependent ROS aggregation and activating the TGF-β1/Smad2 signaling pathway. Our data indicate that the protective effects of AST IV against vascular endothelial cell apoptosis in DM are mainly associated with the decrease in Nox4 expression through the TGF-β1/Smad2 signaling pathway. Furthermore, the inhibition of the activation of the TGF-β1/Smad2 signaling pathway may be another potential therapeutic strategy in the treatment of DM.
Collapse
Affiliation(s)
- Yuhong Ma
- Department of Pharmacology, College of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Weizu Li
- Department of Pharmacology, College of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yanyan Yin
- Department of Pharmacology, College of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Weiping Li
- Department of Pharmacology, College of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
48
|
Guo H, Cao J, Li J, Yang X, Jiang J, Feng J, Li S, Zhang J, Chen B. Lymphocytes from intermittent hypoxia-exposed rats increase the apoptotic signals in endothelial cells via oxidative and inflammatory injury in vitro. Sleep Breath 2015; 19:969-76. [DOI: 10.1007/s11325-015-1128-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 01/01/2015] [Accepted: 01/22/2015] [Indexed: 01/09/2023]
|
49
|
Vikram A, Kim YR, Kumar S, Naqvi A, Hoffman TA, Kumar A, Miller FJ, Kim CS, Irani K. Canonical Wnt signaling induces vascular endothelial dysfunction via p66Shc-regulated reactive oxygen species. Arterioscler Thromb Vasc Biol 2014; 34:2301-9. [PMID: 25147340 DOI: 10.1161/atvbaha.114.304338] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Reactive oxygen species regulate canonical Wnt signaling. However, the role of the redox regulatory protein p66(Shc) in the canonical Wnt pathway is not known. We investigated whether p66(Shc) is essential for canonical Wnt signaling in the endothelium and determined whether the canonical Wnt pathway induces vascular endothelial dysfunction via p66(Shc)-mediated oxidative stress. APPROACH AND RESULTS The canonical Wnt ligand Wnt3a induced phosphorylation (activation) of p66(Shc) in endothelial cells. Wnt3a-stimulated dephosphorylation of β-catenin, and β-catenin-dependent transcription, was inhibited by knockdown of p66(Shc). Exogenous H2O2-induced β-catenin dephosphorylation was also mediated by p66(Shc). Moreover, p66(Shc) overexpression dephosphorylated β-catenin and increased β-catenin-dependent transcription, independent of Wnt3a ligand. P66(Shc)-induced β-catenin dephosphorylation was inhibited by antioxidants N-acetyl cysteine and catalase. Wnt3a upregulated endothelial NADPH oxidase-4, and β-catenin dephosphorylation was suppressed by knocking down NADPH oxidase-4 and by antioxidants. Wnt3a increased H2O2 levels in endothelial cells and impaired endothelium-dependent vasorelaxation in mouse aortas, both of which were rescued by p66(Shc) knockdown. P66(Shc) knockdown also inhibited adhesion of monocytes to Wnt3a-stimulated endothelial cells. Furthermore, constitutively active β-catenin expression in the endothelium increased vascular reactive oxygen species and impaired endothelium-dependent vasorelaxation. In vivo, high-fat diet feeding-induced endothelial dysfunction in mice was associated with increased endothelial Wnt3a, dephosphorylated β-catenin, and phosphorylated p66(Shc). High-fat diet-induced dephosphorylation of endothelial β-catenin was diminished in mice in which p66(Shc) was knocked down. CONCLUSIONS p66(Shc) plays a vital part in canonical Wnt signaling in the endothelium and mediates Wnt3a-stimulated endothelial oxidative stress and dysfunction.
Collapse
Affiliation(s)
- Ajit Vikram
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.).
| | - Young-Rae Kim
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.)
| | - Santosh Kumar
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.)
| | - Asma Naqvi
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.)
| | - Timothy A Hoffman
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.)
| | - Ajay Kumar
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.)
| | - Francis J Miller
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.)
| | - Cuk-Seong Kim
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.)
| | - Kaikobad Irani
- From the Cardiovascular Division, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City (A.V., Y.-R.K., S.K., F.J.M., K.I.); Cardiovascular Institute, University of Pittsburgh, PA (A.N., A.K.); Department of Biochemistry and Molecular Biology, University of Louisville, KY (T.A.H.); and Department of Physiology, Chungnam National University, Daejeon, Korea (C.-S.K.).
| |
Collapse
|
50
|
Affiliation(s)
- Rhian M Touyz
- From the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|