1
|
Abdollahi-Najafabadi M, Farhadian S, Shareghi B, Asgharzadeh S. The investigation of the interaction determination between carbendazim and elastase, using both in vitro and in silico methods. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 319:124586. [PMID: 38833886 DOI: 10.1016/j.saa.2024.124586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/06/2024]
Abstract
Pesticides, including fungicides, are one of the important groups of environmental toxins that affect human and animal health. Studies have shown that these compounds are considered chemical pollutants. Carbendazim is a systemic fungicide. Unfortunately, excessive use of carbendazim has caused environmental pollution all over the world. In this study, the effect of carbendazim on the enzyme elastase (secreted from the endocrine gland of the pancreas) has been investigated. In a study, the performance and reaction of carbendazim with elastase were investigated using spectroscopic techniques. The stability and structure of elastase enzymes were studied under the influence of carbendazim. The results of fluorescence emission and UV-visible absorption spectrum showed that in the presence of carbendazim, there is an increase in UV-Vis absorption and a decrease in the intensity of the intrinsic fluorescence emission in the protein spectrum. Additionally, a decrease in the thermal stability of elastase was observed in the presence of carbendazim. The stability and structure of elastase enzyme were investigated in the presence of carbendazim. The results revealed that the UV-Vis absorption increased due to the presence of carbendazim, as indicated by the hyperchromic spectrum at 220 and 280 nm peaks. Additionally, the intrinsic fluorescence emission in the protein spectrum decreased with increasing carbendazim concentration at three different temperatures (298, 303, and 313 K). Moreover, the study demonstrated that the TM decreased from 2.59 to 4.58 with the increase of carbendazim, suggesting a decrease in the stability of the elastase structure in response to the elevated carbendazim concentration. According to the results of the research, the interaction between elastase and carbendazim has occurred, and changes have been made in the enzyme under the influence of carbendazim. The formation of the complex between elastase and carbendazim was consistent with the results obtained from molecular simulation and confirmed the thermodynamic data.
Collapse
Affiliation(s)
| | - Sadegh Farhadian
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P. O. Box 115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran.
| | - Behzad Shareghi
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P. O. Box 115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran
| | - Sanaz Asgharzadeh
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P. O. Box 115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
2
|
Konishi T, Kawakami R, Vozenilek AE, Ghosh SKB, Xu W, Grogan A, Shah P, Tanaka T, Sekimoto T, Shiraki T, Kawai K, Sato Y, Mori M, Sakamoto A, Hisadome H, Ashida K, Bellissard A, Williams D, Dryanovski D, Kutys R, Cheng Q, Romero M, Chahal D, Virmani R, Finn AV. Mechanisms of Medial Wall Thinning in Chronic Total Occlusion. JACC Cardiovasc Interv 2024; 17:1719-1728. [PMID: 38970581 DOI: 10.1016/j.jcin.2024.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/25/2024] [Accepted: 05/07/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND The success rate of percutaneous coronary intervention (PCI) for chronic total occlusion (CTO) is lower and the risk for complications higher compared with other non-CTO PCI. Although interventionalists focus on intimal plaque characteristics, the coronary media is an important (especially for techniques involving antegrade dissection and re-entry) but poorly understood structure in CTO PCI. OBJECTIVES The aim of the present study was to investigate coronary medial wall thinning in CTO lesions and determine how this thinning might affect CTO PCI. METHODS A total of 2,586 sections were investigated, from arteries with evidence of CTO from 54 subjects (1,383 sections) and arteries without evidence of CTO from 54 subjects with non-coronary-related deaths (1,203 sections) after matching for age, gender, body weight, and body height. RESULTS The medial thickness in subjects with CTO was lower than that in those with non-coronary-related death (P < 0.001). In subjects with CTO, CTO lesions had thinner medial walls compared with those with lower luminal narrowing (P < 0.001). At the CTO distal segments, the 6- to 12-mm distal segment from the distal end of the CTO had significantly less luminal narrowing (P < 0.001), and similar medial thickness, compared with the distal end of the CTO. Immunohistochemical analysis revealed that short-duration CTO had more cleaved caspase-3-positive cells in media and had significantly more CD3+, CD4+, CD8+, and CD4+CD28null T cells compared with long-duration CTO. CONCLUSIONS CTO lesions demonstrated coronary medial thinning compared with non-CTO lesions. Further investigation of the cause-and-effect relationship among inflammation, apoptosis, and coronary medial wall thinning is warranted in future mechanistic studies.
Collapse
Affiliation(s)
- Takao Konishi
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA; Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Saikat Kumar B Ghosh
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Weili Xu
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Alyssa Grogan
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Palak Shah
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Takamasa Tanaka
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Teruo Sekimoto
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Tatsuya Shiraki
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Masayuki Mori
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Atsushi Sakamoto
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Kazuhiro Ashida
- Cardiovascular Center, Seirei Yokohama Hospital, Yokohama, Japan
| | - Arielle Bellissard
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Desiree Williams
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Dilyan Dryanovski
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Robert Kutys
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Qi Cheng
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Maria Romero
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Diljon Chahal
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA; School of Medicine, University of Maryland, Baltimore, Maryland, USA.
| |
Collapse
|
3
|
Goyal RK, Rattan S. Role of mechanoregulation in mast cell-mediated immune inflammation of the smooth muscle in the pathophysiology of esophageal motility disorders. Am J Physiol Gastrointest Liver Physiol 2024; 326:G398-G410. [PMID: 38290993 PMCID: PMC11213482 DOI: 10.1152/ajpgi.00258.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/15/2024] [Accepted: 01/29/2024] [Indexed: 02/01/2024]
Abstract
Major esophageal disorders involve obstructive transport of bolus to the stomach, causing symptoms of dysphagia and impaired clearing of the refluxed gastric contents. These may occur due to mechanical constriction of the esophageal lumen or loss of relaxation associated with deglutitive inhibition, as in achalasia-like disorders. Recently, immune inflammation has been identified as an important cause of esophageal strictures and the loss of inhibitory neurotransmission. These disorders are also associated with smooth muscle hypertrophy and hypercontractility, whose cause is unknown. This review investigated immune inflammation in the causation of smooth muscle changes in obstructive esophageal bolus transport. Findings suggest that smooth muscle hypertrophy occurs above the obstruction and is due to mechanical stress on the smooth muscles. The mechanostressed smooth muscles release cytokines and other molecules that may recruit and microlocalize mast cells to smooth muscle bundles, so that their products may have a close bidirectional effect on each other. Acting in a paracrine fashion, the inflammatory cytokines induce genetic and epigenetic changes in the smooth muscles, leading to smooth muscle hypercontractility, hypertrophy, and impaired relaxation. These changes may worsen difficulty in the esophageal transport. Immune processes differ in the first phase of obstructive bolus transport, and the second phase of muscle hypertrophy and hypercontractility. Moreover, changes in the type of mechanical stress may change immune response and effect on smooth muscles. Understanding immune signaling in causes of obstructive bolus transport, type of mechanical stress, and associated smooth muscle changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.NEW & NOTEWORTHY Esophageal disorders such as esophageal stricture or achalasia, and diffuse esophageal spasm are associated with smooth muscle hypertrophy and hypercontractility, above the obstruction, yet the cause of such changes is unknown. This review suggests that smooth muscle obstructive disorders may cause mechanical stress on smooth muscle, which then secretes chemicals that recruit, microlocalize, and activate mast cells to initiate immune inflammation, producing functional and structural changes in smooth muscles. Understanding the immune signaling in these changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.
Collapse
Affiliation(s)
- Raj K Goyal
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Boston Healthcare System, West Roxbury, Massachusetts, United States
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts, United States
| | - Satish Rattan
- Department of Medicine, Division of Gastroenterology and Hepatology, Sidney Kummel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
4
|
Chakraborty P, Orvos H, Hermesz E. Molecular Study on Twin Cohort with Discordant Birth Weight. Antioxidants (Basel) 2023; 12:1370. [PMID: 37507909 PMCID: PMC10376082 DOI: 10.3390/antiox12071370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
The increased rate of twinning has pointed out newer challenges in clinical practices related to gestational complications, intrauterine growth restriction, perinatal mortality, and comorbidities. As a twin pregnancy progresses, the increased demand for oxygen supply can easily disrupt the redox homeostasis balance and further impose a greater challenge for the developing fetuses. A substantial birth-weight difference acts as an indicator of a deficit in oxygenation or blood flow to one of the fetuses, which might be related to a low bioavailable nitric oxide level. Therefore, in this study, we focused on networks involved in the adjustment of oxygen supply, like the activation of inducible and endothelial nitric oxide synthase (NOS3) along with free radical and lipid peroxide formation in mature twin pairs with high birth-weight differences. The selected parameters were followed by immunofluorescence staining, fluorescence-activated cell sorting analysis, and biochemical measurements in the umbilical cord vessels and fetal red blood cells. Based on our data set, it is clear that the lower-weight siblings are markedly exposed to persistent intrauterine hypoxic conditions, which are connected to a decreased level in NOS3 activation. Furthermore, the increased level of peroxynitrite aggravates lipid peroxidation and induces morphological and functional damage and loss in redox homeostasis.
Collapse
Affiliation(s)
- Payal Chakraborty
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, P.O. Box 533, H-6701 Szeged, Hungary
- Department of Pharmaceutical Technology, JIS University, 81, Nilgunj Road, Kolkata 700109, India
| | - Hajnalka Orvos
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Szeged, Semmelweis u. 1, H-6725 Szeged, Hungary
| | - Edit Hermesz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, P.O. Box 533, H-6701 Szeged, Hungary
| |
Collapse
|
5
|
Jreije A, Medlej-Hashim M, Hajal J, Saliba Y, Chacar S, Fares N, Khouzami L. Calcitriol Supplementation Protects Against Apoptosis and Alleviates the Severity of Abdominal Aortic Aneurysm Induced by Angiotensin II and Anti-TGFβ. J Cardiovasc Transl Res 2022; 15:1340-1351. [PMID: 35445935 DOI: 10.1007/s12265-022-10254-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/04/2022] [Indexed: 12/16/2022]
Abstract
The present study aims to assess the effect of vitamin D deficiency (VDD) and its supplementation on the severity of AAA in mice. AAA was induced by AngII and anti-TGF-β administration. Animals were divided into four groups: Sham, mice with AAA, mice with AAA, and VDD, and mice with AAA supplemented with calcitriol. Blood pressure, echocardiography, abdominal aortic tissues, and plasma samples were monitored for all groups. VDD was associated with enhanced activity of cleaved MMP-9 and elastin degradation and positively correlated with the severity of AAA. Calcitriol supplementation decreased the INFγ/IL-10 ratio and enhanced the Nrf2 pathway. Moreover, Cu/Zn-superoxide dismutase expression and catalase and neutral sphingomyelinase activity were exacerbated in AAA and VDD groups. Furthermore, calcitriol supplementation showed a significantly lower protein expression of caspase-8, caspase-3, Bid, and t-Bid, and prevented the apoptosis of VSMCs treated by AngII and anti-TGF-β. Calcitriol supplementation may alleviate AAA severity and could be of great interest in the clinical management of AAA. VDD enhances antioxidant enzymes activity and expression, whereas calcitriol supplementation alleviates AAA severity by re-activating Nrf2 and inhibiting apoptotic pathways.
Collapse
Affiliation(s)
- Afaf Jreije
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon
| | - Myrna Medlej-Hashim
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon
| | - Joelle Hajal
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Youakim Saliba
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Stephanie Chacar
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Nassim Fares
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon.
| | - Lara Khouzami
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon.
| |
Collapse
|
6
|
Liu H, Ning F, Lash GE. Contribution of vascular smooth muscle cell apoptosis to spiral artery remodeling in early human pregnancy. Placenta 2022; 120:10-17. [DOI: 10.1016/j.placenta.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/02/2021] [Accepted: 02/07/2022] [Indexed: 11/15/2022]
|
7
|
Mansouri A, Reiner Ž, Ruscica M, Tedeschi-Reiner E, Radbakhsh S, Bagheri Ekta M, Sahebkar A. Antioxidant Effects of Statins by Modulating Nrf2 and Nrf2/HO-1 Signaling in Different Diseases. J Clin Med 2022; 11:1313. [PMID: 35268403 PMCID: PMC8911353 DOI: 10.3390/jcm11051313] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Statins are competitive inhibitors of hydroxymethylglutaryl-CoA (HMG-CoA) reductase and have been used to treat elevated low-density lipoprotein cholesterol (LDL-C) for almost four decades. Antioxidant and anti-inflammatory properties which are independent of the lipid-lowering effects of statins, i.e., their pleiotropic effects, might be beneficial in the prevention or treatment of many diseases. This review discusses the antioxidant effects of statins achieved by modulating the nuclear factor erythroid 2 related factor 2/ heme oxygenase-1 (Nrf2/HO-1) pathway in different organs and diseases. Nrf2 and other proteins involved in the Nrf2/HO-1 signaling pathway have a crucial role in cellular responses to oxidative stress, which is a risk factor for ASCVD. Statins can significantly increase the DNA-binding activity of Nrf2 and induce the expression of its target genes, such as HO-1 and glutathione peroxidase) GPx, (thus protecting the cells against oxidative stress. Antioxidant and anti-inflammatory properties of statins, which are independent of their lipid-lowering effects, could be partly explained by the modulation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Atena Mansouri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Željko Reiner
- Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, 10000 Zagreb, Croatia;
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20100 Milan, Italy;
| | - Eugenia Tedeschi-Reiner
- University Hospital Center Sestre Milosrdnice, University of Osijek, Vinogradska Cesta 29, 10000 Zagreb, Croatia;
| | - Shabnam Radbakhsh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
- Department of Medical Biotechnology and Nanotechnology, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| | - Mariam Bagheri Ekta
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, A.P. Avtsyn Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia;
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
8
|
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall, characterized by the formation of plaques containing lipid, connective tissue and immune cells in the intima of large and medium-sized arteries. Over the past three decades, a substantial reduction in cardiovascular mortality has been achieved largely through LDL-cholesterol-lowering regimes and therapies targeting other traditional risk factors for cardiovascular disease, such as hypertension, smoking, diabetes mellitus and obesity. However, the overall benefits of targeting these risk factors have stagnated, and a huge global burden of cardiovascular disease remains. The indispensable role of immunological components in the establishment and chronicity of atherosclerosis has come to the forefront as a clinical target, with proof-of-principle studies demonstrating the benefit and challenges of targeting inflammation and the immune system in cardiovascular disease. In this Review, we provide an overview of the role of the immune system in atherosclerosis by discussing findings from preclinical research and clinical trials. We also identify important challenges that need to be addressed to advance the field and for successful clinical translation, including patient selection, identification of responders and non-responders to immunotherapies, implementation of patient immunophenotyping and potential surrogate end points for vascular inflammation. Finally, we provide strategic guidance for the translation of novel targets of immunotherapy into improvements in patient outcomes. In this Review, the authors provide an overview of the immune cells involved in atherosclerosis, discuss preclinical research and published and ongoing clinical trials assessing the therapeutic potential of targeting the immune system in atherosclerosis, highlight emerging therapeutic targets from preclinical studies and identify challenges for successful clinical translation.
Inflammation is an important component of the pathophysiology of cardiovascular disease; an imbalance between pro-inflammatory and anti-inflammatory processes drives chronic inflammation and the formation of atherosclerotic plaques in the vessel wall. Clinical trials assessing canakinumab and colchicine therapies in atherosclerotic cardiovascular disease have provided proof-of-principle of the benefits associated with therapeutic targeting of the immune system in atherosclerosis. The immunosuppressive adverse effects associated with the systemic use of anti-inflammatory drugs can be minimized through targeted delivery of anti-inflammatory drugs to the atherosclerotic plaque, defining the window of opportunity for treatment and identifying more specific targets for cardiovascular inflammation. Implementing immunophenotyping in clinical trials in patients with atherosclerotic cardiovascular disease will allow the identification of immune signatures and the selection of patients with the highest probability of deriving benefit from a specific therapy. Clinical stratification via novel risk factors and discovery of new surrogate markers of vascular inflammation are crucial for identifying new immunotherapeutic targets and their successful translation into the clinic.
Collapse
|
9
|
Bioremediation of Uranium- and Nitrate-Contaminated Groundwater after the In Situ Leach Mining of Uranium. WATER 2021. [DOI: 10.3390/w13223188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Uranium and nitrate are common groundwater pollutants near in situ leach uranium mines. However, we still lack techniques that can simultaneously immobilize uranium and reduce nitrate using a single bacterial species. In this study, the potential of simultaneous uranium immobilization and nitrate reduction by a single AFODN (anaerobic Fe(II) oxidizing denitrifier), Clostridium sp. PXL2, was investigated. Clostridium sp. PXL2 showed tolerance to U(VI) concentrations varying from 4.2 µM to 42 µM. The U(VI) immobilization and nitrate reduction rates in groundwater samples inoculated with this bacterium reached up to 75.1% and 55.7%, respectively, under neutral conditions. Exposure to oxidation conditions led to further U(VI) removal but did not show any noticeable effect on nitrate reduction. The U(VI) immobilization rate reached up to 85% with an increased Fe(II) initial concentration, but this inhibited nitrate reduction. SEM (scanning electron microscopy) coupled with EDS (energy dispersive spectroscopy) showed that the U(VI) immobilization was mainly due to sorption to amorphous ferric oxides. U(VI) and nitrate bioremediation by AFODNs, including Clostridium sp. PXL2, may provide a promising method for the treatment of uranium- and nitrate-contaminated groundwater after the in situ leach mining of uranium.
Collapse
|
10
|
O'Morain VL, Chan Y, Williams JO, Alotibi R, Alahmadi A, Rodrigues NP, Plummer SF, Hughes TR, Michael DR, Ramji DP. The Lab4P Consortium of Probiotics Attenuates Atherosclerosis in LDL Receptor Deficient Mice Fed a High Fat Diet and Causes Plaque Stabilization by Inhibiting Inflammation and Several Pro-Atherogenic Processes. Mol Nutr Food Res 2021; 65:e2100214. [PMID: 34216185 PMCID: PMC9373067 DOI: 10.1002/mnfr.202100214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/07/2021] [Indexed: 12/17/2022]
Abstract
SCOPE Previous studies show that Lab4 probiotic consortium plus Lactobacillus plantarum CUL66 (Lab4P) reduces diet-induced weight gain and plasma cholesterol levels in C57BL/6J mice fed a high fat diet (HFD). The effect of Lab4P on atherosclerosis is not known and is therefore investigated. METHODS AND RESULTS Atherosclerosis-associated parameters are analyzed in LDL receptor deficient mice fed HFD for 12 weeks alone or supplemented with Lab4P. Lab4P increases plasma HDL and triglyceride levels and decreases LDL/VLDL levels. Lab4P also reduces plaque burden and content of lipids and macrophages, indicative of dampened inflammation, and increases smooth muscle cell content, a marker of plaque stabilization. Atherosclerosis arrays show that Lab4P alters the liver expression of 19 key disease-associated genes. Lab4P also decreases the frequency of macrophages and T-cells in the bone marrow. In vitro assays using conditioned media from probiotic bacteria demonstrates attenuation of several atherosclerosis-associated processes in vitro such as chemokine-driven monocytic migration, proliferation of monocytes and macrophages, foam cell formation and associated changes in expression of key genes, and proliferation and migration of vascular smooth muscle cells. CONCLUSION This study provides new insights into the anti-atherogenic actions of Lab4P together with the underlying mechanisms and supports further assessments in human trials.
Collapse
Affiliation(s)
- Victoria L. O'Morain
- Cardiff School of BiosciencesCardiff UniversitySir Martin Evans Building, Museum AvenueCardiffCF10 3AXUK
| | - Yee‐Hung Chan
- Cardiff School of BiosciencesCardiff UniversitySir Martin Evans Building, Museum AvenueCardiffCF10 3AXUK
| | - Jessica O. Williams
- Cardiff School of BiosciencesCardiff UniversitySir Martin Evans Building, Museum AvenueCardiffCF10 3AXUK
| | - Reem Alotibi
- Cardiff School of BiosciencesCardiff UniversitySir Martin Evans Building, Museum AvenueCardiffCF10 3AXUK
| | - Alaa Alahmadi
- Cardiff School of BiosciencesCardiff UniversitySir Martin Evans Building, Museum AvenueCardiffCF10 3AXUK
| | - Neil P. Rodrigues
- European Cancer Stem Cell Research Institute, Cardiff School of BiosciencesCardiff UniversityHadyn Ellis Building, Maindy RoadCardiffCF24 4HQUK
| | - Sue F. Plummer
- Cultech LimitedUnit 2 Christchurch Road, Baglan Industrial ParkPort TalbotSA12 7BZUK
| | - Timothy R. Hughes
- Systems Immunity Research Institute, School of MedicineCardiff UniversityCardiffCF14 4XNUK
| | - Daryn R. Michael
- Cultech LimitedUnit 2 Christchurch Road, Baglan Industrial ParkPort TalbotSA12 7BZUK
| | - Dipak P. Ramji
- Cardiff School of BiosciencesCardiff UniversitySir Martin Evans Building, Museum AvenueCardiffCF10 3AXUK
| |
Collapse
|
11
|
Fetal oxygen supply can be improved by an effective cross-talk between fetal erythrocytes and vascular endothelium. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166243. [PMID: 34371111 DOI: 10.1016/j.bbadis.2021.166243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/24/2022]
Abstract
In twin/multiple pregnancy, siblings experience an adverse intrauterine environment which forms the major etiological factor leading to pathological conditions. The status of the developing fetus is highly determined by the nitric oxide (NO) level, that facilitates vasodilation which in turn modulates the oxygen and nutrition supply. As the umbilical cord (UC) lacks innervation, activation of the endothelial nitric oxide synthase (NOS3) is fundamental to maintain adequate NO production. Recent ground breaking fact showed that under stress conditions, circulating red blood cells (RBCs) can actively produces NO as a "rescue mechanism". Therefore, this study majorly focused on the molecular mechanisms that affected the redox environment by altering NOS3 activation - both in the UC arteries and vein endothelium and RBCs - that have impacts on developmental parameters, like birth weight. In connection to that, we pursued the communication efficiency between the vessels' endothelium and the circulating RBCs in demand of bioavailable NO. Our results indicated that twinning itself at stage 33-35 weeks, does not reduce the NOS3 level and its phosphorylation status in the cord vessels. However, RBC-NOS3 activation is highly upregulated during this period - providing additional evidence for the active regulatory role of fetal RBCs in the rate of blood flow - and this functional activity highly correlates with the birth weight of the fetuses. Detailed analysis on NOS3 signalling at different time points of gestation could establish a benchmark in understanding of the pathophysiological mechanisms involved in the process of developing neonatal vascular diseases.
Collapse
|
12
|
Zahorán S, Szántó PR, Bódi N, Bagyánszki M, Maléth J, Hegyi P, Sári T, Hermesz E. Sustained Maternal Smoking Triggers Endothelial-Mediated Oxidative Stress in the Umbilical Cord Vessels, Resulting in Vascular Dysfunction. Antioxidants (Basel) 2021; 10:antiox10040583. [PMID: 33918732 PMCID: PMC8069726 DOI: 10.3390/antiox10040583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/15/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO) bioavailability is fundamental in the regulation of redox balance and functionality of the endothelium, especially in the case of the umbilical cord (UC), which has no innervation. The analysis of UC vessel-related complications could serve as a useful tool in the understanding of the pathophysiological mechanisms leading to neonatal cardiovascular disorders. Therefore, the aim of this study was to characterize the mechanisms that rule the severity of prenatal endothelial dysfunction, induced by the long-term effect of maternal smoking. Our analysis describes the initiation and the consequences of endothelial nitric oxide synthase (NOS3) deactivation, along with the up-regulation of possible compensatory pathways, using structural, molecular and biochemical approaches. This study was carried out on both the UC arteries and veins originated from neonates born to non-smoking and heavy-smoking mothers. The alterations stimulated by maternal smoking are vessel-specific and proportional to the level of exposure to harmful materials passed through the placenta. Typically, in the primarily exposed veins, an increased formation of reactive oxygen species and an up-regulation of the highly-efficient NOS2-NO producing pathway were detected. Despite all the extensive structural and functional damages, the ex vivo heat and cadmium ion-treated UC vein pieces still support the potential for stress response.
Collapse
Affiliation(s)
- Szabolcs Zahorán
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (S.Z.); (P.R.S.)
| | - Péter R. Szántó
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (S.Z.); (P.R.S.)
| | - Nikolett Bódi
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (N.B.); (M.B.)
| | - Mária Bagyánszki
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (N.B.); (M.B.)
| | - József Maléth
- First Department of Medicine, University of Szeged, H-6701 Szeged, Hungary;
- HAS-USZ Momentum Epithel Cell Signalling and Secretion Research Group, H-6701 Szeged, Hungary
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, H-7601 Pécs, Hungary;
| | - Tamás Sári
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Szeged, H-6701 Szeged, Hungary;
| | - Edit Hermesz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (S.Z.); (P.R.S.)
- Correspondence: ; Tel.: +36-(62)-544-887
| |
Collapse
|
13
|
Boraldi F, Lofaro FD, Quaglino D. Apoptosis in the Extraosseous Calcification Process. Cells 2021; 10:cells10010131. [PMID: 33445441 PMCID: PMC7827519 DOI: 10.3390/cells10010131] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 12/13/2022] Open
Abstract
Extraosseous calcification is a pathologic mineralization process occurring in soft connective tissues (e.g., skin, vessels, tendons, and cartilage). It can take place on a genetic basis or as a consequence of acquired chronic diseases. In this last case, the etiology is multifactorial, including both extra- and intracellular mechanisms, such as the formation of membrane vesicles (e.g., matrix vesicles and apoptotic bodies), mitochondrial alterations, and oxidative stress. This review is an overview of extraosseous calcification mechanisms focusing on the relationships between apoptosis and mineralization in cartilage and vascular tissues, as these are the two tissues mostly affected by a number of age-related diseases having a progressively increased impact in Western Countries.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
- Correspondence:
| | - Francesco Demetrio Lofaro
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.D.L.); (D.Q.)
- Interuniversity Consortium for Biotechnologies (CIB), Italy
| |
Collapse
|
14
|
Ogata Y, Yamada T, Hasegawa S, Sanada A, Iwata Y, Arima M, Nakata S, Sugiura K, Akamatsu H. SASP-induced macrophage dysfunction may contribute to accelerated senescent fibroblast accumulation in the dermis. Exp Dermatol 2020; 30:84-91. [PMID: 33010063 DOI: 10.1111/exd.14205] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 12/31/2022]
Abstract
Recently, increasing attention has been paid to senescence-associated secretory phenotype (SASP), a phenomenon that senescent cells secrete molecules such as inflammatory cytokines and matrix metalloproteinases (MMPs), due to its noxious effects on the surrounding tissue. Senescent cells in the blood and liver are known to be properly depleted by macrophages. In the dermis, accumulation of senescent cells has been reported and is thought to be involved with skin ageing. In this study, to elucidate the clearance mechanism of senescent cells in the dermis, we focused on macrophage functions. Our co-culture experiments of senescent fibroblasts and macrophages revealed a two-step clearance mechanism: first, TNF-α secreted from macrophages induces apoptosis in senescent fibroblasts, and then, dead cells are phagocytosed by macrophages. Furthermore, it was suggested that SASP factors suppress both of the two steps of the senescent cell clearance by macrophages. From these findings, normally senescent cells in the dermis are thought to be removed by macrophages, but when senescent cells are excessively accumulated owing to oxidative stress, ultraviolet (UV) ray or other reasons, SASP was suggested to suppress the macrophage-dependent clearance functions and thereby cause further accumulation of senescent cells.
Collapse
Affiliation(s)
- Yuichiro Ogata
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan
| | - Takaaki Yamada
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan.,Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Toyoake, Japan.,Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Seiji Hasegawa
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan.,Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan.,Nagoya University-MENARD Collaborative Chair, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ayumi Sanada
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan
| | - Yohei Iwata
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masaru Arima
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Satoru Nakata
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan
| | - Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hirohiko Akamatsu
- Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
15
|
Aravani D, Foote K, Figg N, Finigan A, Uryga A, Clarke M, Bennett M. Cytokine regulation of apoptosis-induced apoptosis and apoptosis-induced cell proliferation in vascular smooth muscle cells. Apoptosis 2020; 25:648-662. [PMID: 32627119 PMCID: PMC7527356 DOI: 10.1007/s10495-020-01622-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are the main structural cell of blood vessels, and VSMC apoptosis occurs in vascular disease, after injury, and in vessel remodeling during development. Although VSMC apoptosis is viewed as silent, recent studies show that apoptotic cells can promote apoptosis-induced compensatory proliferation (AICP), apoptosis-induced apoptosis (AIA), and migration of both local somatic and infiltrating inflammatory cells. However, the effects of VSMC apoptosis on adjacent VSMCs, and their underlying signaling and mechanisms are unknown. We examined the consequences of VSMC apoptosis after activating extrinsic and intrinsic death pathways. VSMCs undergoing apoptosis through Fas/CD95 or the protein kinase inhibitor staurosporine transcriptionally activated interleukin 6 (IL-6) and granulocyte-macrophage colony stimulating factor (GM-CSF), leading to their secretion. Apoptosis induced activation of p38MAPK, JNK, and Akt, but neither p38 and JNK activation nor IL-6 or GM-CSF induction required caspase cleavage. IL-6 induction depended upon p38 activity, while Fas-induced GM-CSF expression required p38 and JNK. Conditioned media from apoptotic VSMCs induced VSMC apoptosis in vitro, and IL-6 and GM-CSF acted as pro-survival factors for AIA. VSMC apoptosis was studied in vivo using SM22α-DTR mice that express the diphtheria toxin receptor in VSMCs only. DT administration induced VSMC apoptosis and VSMC proliferation, and also signficantly induced IL-6 and GM-CSF. We conclude that VSMC apoptosis activates multiple caspase-independent intracellular signaling cascades, leading to release of soluble cytokines involved in regulation of both cell proliferation and apoptosis. VSMC AICP may ameliorate while AIA may amplify the effects of pro-apoptotic stimuli in vessel remodeling and disease.
Collapse
Affiliation(s)
- Dimitra Aravani
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Kirsty Foote
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Nichola Figg
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Alison Finigan
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Anna Uryga
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Murray Clarke
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK
| | - Martin Bennett
- Division of Cardiovascular Medicine, University of Cambridge, ACCI, Addenbrooke's Hospital, Box 110, CB2 0QQ, Cambridge, UK.
| |
Collapse
|
16
|
Rapp N, Evenepoel P, Stenvinkel P, Schurgers L. Uremic Toxins and Vascular Calcification-Missing the Forest for All the Trees. Toxins (Basel) 2020; 12:E624. [PMID: 33003628 PMCID: PMC7599869 DOI: 10.3390/toxins12100624] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/23/2022] Open
Abstract
The cardiorenal syndrome relates to the detrimental interplay between the vascular system and the kidney. The uremic milieu induced by reduced kidney function alters the phenotype of vascular smooth muscle cells (VSMC) and promotes vascular calcification, a condition which is strongly linked to cardiovascular morbidity and mortality. Biological mechanisms involved include generation of reactive oxygen species, inflammation and accelerated senescence. A better understanding of the vasotoxic effects of uremic retention molecules may reveal novel avenues to reduce vascular calcification in CKD. The present review aims to present a state of the art on the role of uremic toxins in pathogenesis of vascular calcification. Evidence, so far, is fragmentary and limited with only a few uremic toxins being investigated, often by a single group of investigators. Experimental heterogeneity furthermore hampers comparison. There is a clear need for a concerted action harmonizing and standardizing experimental protocols and combining efforts of basic and clinical researchers to solve the complex puzzle of uremic vascular calcification.
Collapse
MESH Headings
- Animals
- Cardio-Renal Syndrome/metabolism
- Cardio-Renal Syndrome/pathology
- Cardio-Renal Syndrome/physiopathology
- Cardio-Renal Syndrome/therapy
- Humans
- Kidney/metabolism
- Kidney/pathology
- Kidney/physiopathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Prognosis
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/physiopathology
- Renal Insufficiency, Chronic/therapy
- Toxins, Biological/metabolism
- Uremia/metabolism
- Uremia/pathology
- Uremia/physiopathology
- Uremia/therapy
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/physiopathology
- Vascular Calcification/therapy
Collapse
Affiliation(s)
- Nikolas Rapp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Pieter Evenepoel
- Laboratory of Nephrology, KU Leuven Department of Microbiology and Immunology, University Hospitals Leuven, 3000 Leuven, Belgium;
| | - Peter Stenvinkel
- Karolinska Institute, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, 141 86 Stockholm, Sweden;
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
17
|
Chakraborty P, Dugmonits KN, Orvos H, Hermesz E. Mature Twin Neonates Exhibit Oxidative Stress via Nitric Oxide Synthase Dysfunctionality: A Prognostic Stress Marker in the Red Blood Cells and Umbilical Cord Vessels. Antioxidants (Basel) 2020; 9:antiox9090845. [PMID: 32927592 PMCID: PMC7555925 DOI: 10.3390/antiox9090845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/07/2020] [Indexed: 11/22/2022] Open
Abstract
Intrauterine hypoxic condition increases the generation of reactive oxygen species and fetal oxidative stress. Multiple pregnancy always bears an additional oxidative stress condition with severe complications, such as prematurity, structural abnormalities, delayed development and low birthweight. The umbilical cord (UC) vessels, along with circulating fetal red blood cells (RBCs), highly determine the oxygenation status of fetus and regulate the feto-placental circulation. As UC lacks innervation, the activation of the endothelial nitric oxide synthase (NOS3) is fundamental for proper NO production. Therefore, we aimed to study the NOS3 activation pathways along with damages to macromolecules in the endothelium of UC vessels and RBCs of mature non-discordant twins, in connection to major differences in their birth weight. We provide evidence that, under severe hypoxic conditions such as twin pregnancy, the NOS3-related NO production pathways are altered both in UC vessels and RBCs; moreover, the extent of changes is highly birthweight-specific. Furthermore, macromolecular damages are prominent in the RBCs and arteries compared to the vein, with a similar increase in the Arginase1 level, which is believed to play a role in NOS3 functionality, resulting in endothelial dysfunctionality, which might have relevance to the major etiologies of cardiovascular diseases in later life.
Collapse
Affiliation(s)
- Payal Chakraborty
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, P.O.Box 533, H-6701 Szeged, Hungary; (P.C.); (K.N.D.)
| | - Krisztina N. Dugmonits
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, P.O.Box 533, H-6701 Szeged, Hungary; (P.C.); (K.N.D.)
| | - Hajnalka Orvos
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Szeged, P.O.Box 533, H-6701 Szeged, Hungary;
| | - Edit Hermesz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, P.O.Box 533, H-6701 Szeged, Hungary; (P.C.); (K.N.D.)
- Correspondence: ; Tel./Fax: +36-62-544887
| |
Collapse
|
18
|
Zhang Z, Xu MH, Wei FJ, Shang LN. Clinical study of different doses of atorvastatin combined with febuxostat in patients with gout and carotid atherosclerosis. Pak J Med Sci 2020; 36:1334-1338. [PMID: 32968404 PMCID: PMC7501040 DOI: 10.12669/pjms.36.6.2945] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective: To evaluate the efficacy of atorvastatin combined with febuxostat in the treatment of gout patients with carotid atherosclerosis and to observe the effects on serum tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and C-reactive protein (CRP) levels, carotid plaques, and the adverse reactions. Methods: Seventy patients with gout and carotid atherosclerosis admitted to Affiliated Hospital of Hebei University from January 2014 to June 2017 were randomly divided into a treatment group and a control group. The treatment group received oral febuxostat 40 mg/day combined with atorvastatin 40 mg/day. The control group was given 40 mg/day febuxostat combined with 20 mg/day atorvastatin for 90 days. The effects of treatment on TNF-α, IL-1β, and CRP levels and carotid plaques of the patients were observed. Results: After 90 days of treatment, serum TNF-α, IL-1β, and CRP levels, as well as HUA and total cholesterol (TC), decreased in both groups after treatment. There were significant differences observed (p < 0.05). The carotid artery plaques in the two groups were significantly smaller after treatment (P<0.05). There was no significant difference in adverse reactions between the two groups (P > 0.05). Conclusion: Double doses of atorvastatin combined with febuxostat can effectively reduce uric acid to improve the inflammatory state in patients and reduce carotid plaques without increasing the incidence of adverse reactions.
Collapse
Affiliation(s)
- Zheng Zhang
- Zheng Zhang, Department of Rheumatology and Immunology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P. R. China
| | - Ming-Hua Xu
- Ming-hua Xu, Department of Rheumatology and Immunology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P. R. China
| | - Feng-Ju Wei
- Feng-ju Wei, Department of Traditional Chinese Medicine Department, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P. R. China
| | - Li-Na Shang
- Li-na Shang, Department of Ultrasound, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P. R. China
| |
Collapse
|
19
|
Gurung R, Choong AM, Woo CC, Foo R, Sorokin V. Genetic and Epigenetic Mechanisms Underlying Vascular Smooth Muscle Cell Phenotypic Modulation in Abdominal Aortic Aneurysm. Int J Mol Sci 2020; 21:ijms21176334. [PMID: 32878347 PMCID: PMC7504666 DOI: 10.3390/ijms21176334] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) refers to the localized dilatation of the infra-renal aorta, in which the diameter exceeds 3.0 cm. Loss of vascular smooth muscle cells, degradation of the extracellular matrix (ECM), vascular inflammation, and oxidative stress are hallmarks of AAA pathogenesis and contribute to the progressive thinning of the media and adventitia of the aortic wall. With increasing AAA diameter, and left untreated, aortic rupture ensues with high mortality. Collective evidence of recent genetic and epigenetic studies has shown that phenotypic modulation of smooth muscle cells (SMCs) towards dedifferentiation and proliferative state, which associate with the ECM remodeling of the vascular wall and accompanied with increased cell senescence and inflammation, is seen in in vitro and in vivo models of the disease. This review critically analyses existing publications on the genetic and epigenetic mechanisms implicated in the complex role of SMCs within the aortic wall in AAA formation and reflects the importance of SMCs plasticity in AAA formation. Although evidence from the wide variety of mouse models is convincing, how this knowledge is applied to human biology needs to be addressed urgently leveraging modern in vitro and in vivo experimental technology.
Collapse
Affiliation(s)
- Rijan Gurung
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 9, Singapore 119228, Singapore; (R.G.); (R.F.)
- Genome Institute of Singapore, A*STAR, 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Andrew Mark Choong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 8, Singapore 119228, Singapore; (A.M.C.); (C.C.W.)
- Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 9, Singapore 119228, Singapore
| | - Chin Cheng Woo
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 8, Singapore 119228, Singapore; (A.M.C.); (C.C.W.)
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 9, Singapore 119228, Singapore; (R.G.); (R.F.)
- Genome Institute of Singapore, A*STAR, 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Vitaly Sorokin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 8, Singapore 119228, Singapore; (A.M.C.); (C.C.W.)
- Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, 1E Kent Ridge Road, NUHS Tower Block, Level 9, Singapore 119228, Singapore
- Correspondence: ; Tel.: +65-6779-5555
| |
Collapse
|
20
|
Geng YJ, Wei ZY, Qian HY, Huang J, Lodato R, Castriotta RJ. Pathophysiological characteristics and therapeutic approaches for pulmonary injury and cardiovascular complications of coronavirus disease 2019. Cardiovasc Pathol 2020; 47:107228. [PMID: 32375085 PMCID: PMC7162778 DOI: 10.1016/j.carpath.2020.107228] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 04/11/2020] [Indexed: 02/06/2023] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) has emerged as a major health crisis, with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) having infected over a million people around the world within a few months of its identification as a human pathogen. Initially, SARS-CoV-2 infects cells in the respiratory system and causes inflammation and cell death. Subsequently, the virus spreads out and damages other vital organs and tissues, triggering a complicated spectrum of pathophysiological changes and symptoms, including cardiovascular complications. Acting as the receptor for SARS-CoV entering mammalian cells, angiotensin converting enzyme-2 (ACE2) plays a pivotal role in the regulation of cardiovascular cell function. Diverse clinical manifestations and laboratory abnormalities occur in patients with cardiovascular injury in COVID-19, characterizing the development of this complication, as well as providing clues to diagnosis and treatment. This review provides a summary of the rapidly appearing laboratory and clinical evidence for the pathophysiology and therapeutic approaches to COVID-19 pulmonary and cardiovascular complications.
Collapse
Affiliation(s)
- Yong-Jian Geng
- Department of Internal Medicine, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Zhi-Yao Wei
- Department of Cardiology, Center for Coronary Heart Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai-Yan Qian
- Department of Cardiology, Center for Coronary Heart Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ji Huang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Robert Lodato
- Department of Internal Medicine, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Richard J Castriotta
- Department of Internal Medicine, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA; Division of Pulmonary, Critical and Sleep Medicine, University of South California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
21
|
St-Germain LE, Castellana B, Baltayeva J, Beristain AG. Maternal Obesity and the Uterine Immune Cell Landscape: The Shaping Role of Inflammation. Int J Mol Sci 2020; 21:E3776. [PMID: 32471078 PMCID: PMC7312391 DOI: 10.3390/ijms21113776] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is often equated to the physiological response to injury or infection. Inflammatory responses defined by cytokine storms control cellular mechanisms that can either resolve quickly (i.e., acute inflammation) or remain prolonged and unabated (i.e., chronic inflammation). Perhaps less well-appreciated is the importance of inflammatory processes central to healthy pregnancy, including implantation, early stages of placentation, and parturition. Pregnancy juxtaposed with disease can lead to the perpetuation of aberrant inflammation that likely contributes to or potentiates maternal morbidity and poor fetal outcome. Maternal obesity, a prevalent condition within women of reproductive age, associates with increased risk of developing multiple pregnancy disorders. Importantly, chronic low-grade inflammation is thought to underlie the development of obesity-related obstetric and perinatal complications. While diverse subsets of uterine immune cells play central roles in initiating and maintaining healthy pregnancy, uterine leukocyte dysfunction as a result of maternal obesity may underpin the development of pregnancy disorders. In this review we discuss the current knowledge related to the impact of maternal obesity and obesity-associated inflammation on uterine immune cell function, utero-placental establishment, and pregnancy health.
Collapse
Affiliation(s)
- Lauren E. St-Germain
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Barbara Castellana
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Jennet Baltayeva
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Alexander G. Beristain
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| |
Collapse
|
22
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
23
|
Shaffer RM, Liang R, Knight K, Carter-Brooks CM, Abramowitch S, Moalli PA. Impact of polypropylene prolapse mesh on vaginal smooth muscle in rhesus macaque. Am J Obstet Gynecol 2019; 221:330.e1-330.e9. [PMID: 31102587 DOI: 10.1016/j.ajog.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/02/2019] [Accepted: 05/10/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND The use of polypropylene prolapse mesh to treat pelvic organ prolapse has been limited by mesh-related complications. Gynemesh PS mesh, implanted via sacrocolpopexy in rhesus macaques, had a negative impact on the vagina with thinning of vaginal muscularis and decreased vaginal smooth muscle contractility. The negative effect was attenuated when a bioscaffold derived from urinary bladder extracellular matrix was used as a composite with Gynemesh PS. OBJECTIVE The objective of the study was to further elucidate the impact of Gynemesh PS polypropylene mesh and MatriStem extracellular matrix bioscaffolds on the vaginal smooth muscle in terms of micromorphology of vaginal smooth muscle (muscle bundles and individual myocytes), innervation, and nerve-mediated contractile function following their implantations in a rhesus macaque model via sacrocolpopexy. STUDY DESIGN Thirty-two middle-aged rhesus macaques were randomized to undergo either a sham surgery (sham, n = 8), or the implantation of Gynemesh PS alone (n = 8) vs composite mesh comprised of Gynemesh PS plus 2-ply MatriStem (n = 8) vs 6-ply MatriStem alone (n = 8) via sacrocolpopexy. The graft-vagina complexes were harvested 3 months later. Histomorphometrics of smooth muscle bundles and myocytes were performed by immunofluorescent labeling of alpha smooth muscle actin, caveolin-3 (membrane protein), and cell nuclei followed by confocal imaging. The cross-sectional diameters of smooth muscle bundles and individual myocytes were quantified using images randomly taken in at least 5 areas of each section of sample. Contractile proteins alpha smooth muscle actin and smoothelin were quantified by Western immunoblotting. Nerve density was measured by immunohistochemical labeling of a pan-neuron marker, PGP9.5. Nerve-mediated smooth muscle contractility was quantified using electrical field stimulation. One-way analysis of variance and appropriate post hoc tests were used for statistical comparisons. RESULTS Compared with sham, the implantation of Gynemesh PS alone resulted in a disorganized smooth muscle morphology with the number of small muscle bundles (cross-sectional diameter less than 20 μm) increased 67% (P = .004) and the myocyte diameter decreased 22% (P < .001). Levels of contractile proteins were all decreased vs sham with alpha smooth muscle actin decreased by 68% (P = .009), low-molecular-weight smoothelin by 51% (P = .014), and high-molecular-weight smoothelin by 40% (P = .015). Nerve density was decreased by 48% (P = .03 vs sham) paralleled by a 63% decrease of nerve-mediated contractility (P = .02). Following the implantation of composite mesh, the results of measurements were similar to sham (all P > .05), with a 39% increase in the myocyte diameter (P < .001) and a 2-fold increase in the level of alpha smooth muscle actin relative to Gynemesh (P = .045). Following the implantation of MatriStem alone, the number of small muscle bundles were increased 54% vs sham (P = .002), while the other parameters were not significantly different from sham (all P > .05). CONCLUSION The implantation of Gynemesh PS had a negative impact on the structural and functional integrity of vaginal smooth muscle evidenced by atrophic macro- and microscopic muscle morphology, decreased innervation, and impaired contractile property, consistent with a maladaptive remodeling response. The extracellular matrix bioscaffold (MatriStem), when used with Gynemesh PS as a composite (2 ply), attenuated the negative impact of Gynemesh PS; when used alone (6 ply), it induced adaptive remodeling as evidenced by an increased fraction of small smooth muscle bundles with normal contractility.
Collapse
Affiliation(s)
- Rebecca M Shaffer
- Department of Obstetrics and Gynecology, Larner College of Medicine at the University of Vermont, Burlington, VT
| | - Rui Liang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA
| | - Katrina Knight
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA
| | - Charelle M Carter-Brooks
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA
| | - Steven Abramowitch
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Pamela A Moalli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA.
| |
Collapse
|
24
|
Gene polymorphisms and circulating levels of the TNF-alpha are associated with ischemic stroke: A meta-analysis based on 19,873 individuals. Int Immunopharmacol 2019; 75:105827. [PMID: 31454695 DOI: 10.1016/j.intimp.2019.105827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
|
25
|
Abdolmaleki F, Gheibi Hayat SM, Bianconi V, Johnston TP, Sahebkar A. Atherosclerosis and immunity: A perspective. Trends Cardiovasc Med 2019; 29:363-371. [DOI: 10.1016/j.tcm.2018.09.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/09/2018] [Accepted: 09/25/2018] [Indexed: 01/11/2023]
|
26
|
Carpinus turczaninowii Extract May Alleviate High Glucose-Induced Arterial Damage and Inflammation. Antioxidants (Basel) 2019; 8:antiox8060172. [PMID: 31212679 PMCID: PMC6616550 DOI: 10.3390/antiox8060172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/02/2019] [Accepted: 06/07/2019] [Indexed: 01/06/2023] Open
Abstract
Hyperglycemia-induced oxidative stress triggers severe vascular damage and induces an inflammatory vascular state, and is, therefore, one of the main causes of atherosclerosis. Recently, interest in the natural compound Carpinus turczaninowii has increased because of its reported antioxidant and anti-inflammatory properties. We investigated whether a C. turczaninowii extract was capable of attenuating high glucose-induced inflammation and arterial damage using human aortic vascular smooth muscle cells (hASMCs). mRNA expression levels of proinflammatory response [interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α)], endoplasmic reticulum (ER) stress [CCAAT-enhancer-binding proteins (C/EBP) homologous protein (CHOP)], and adenosine monophosphate (AMP)-protein activated kinase α2 (AMPK α2)], and DNA damage [phosphorylated H2.AX (p-H2.AX)] were measured in hASMCs treated with the C. turczaninowii extracts (1 and 10 μg/mL) after being stimulated by high glucose (25 mM) or not. The C. turczaninowii extract attenuated the increased mRNA expression of IL-6, TNF-α, and CHOP in hASMCs under high glucose conditions. The expression levels of p-H2.AX and AMPK α2 induced by high glucose were also significantly decreased in response to treatment with the C. turczaninowii extract. In addition, 15 types of phenolic compounds including quercetin, myricitrin, and ellagic acid, which exhibit antioxidant and anti-inflammatory properties, were identified in the C. turczaninowii extract through ultra-performance liquid chromatography-quadrupole-time of flight (UPLC-Q-TOF) mass spectrometry. In conclusion, C. turczaninowii may alleviate high glucose-induced inflammation and arterial damage in hASMCs, and may have potential in the treatment of hyperglycemia-induced atherosclerosis.
Collapse
|
27
|
Liu X, Du H, Chen D, Yuan H, Chen W, Jia W, Wang X, Li X, Gao L. Cyclophilin D deficiency protects against the development of mitochondrial ROS and cellular inflammation in aorta. Biochem Biophys Res Commun 2019; 508:1202-1208. [PMID: 30554656 DOI: 10.1016/j.bbrc.2018.12.064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Inflammation and oxidative stress are closely correlated in the pathology of cardiovascular disease. Mitochondrial cyclophilin D (CypD), the important modulator for mPTP opening, is increasingly recognized as a key regulator of cellular ROS generation. Besides, its association with cell inflammation is also being discovered. However, the effects of CypD in modulating vascular inflammatory response is unknown. We sought to investigate whether CypD deficiency attenutes vascular inflammation under physical conditions. METHODS AND RESULTS We adopted CypD KO mouse and their littermate controls to observe the effects of CypD deficiency on aortic mitochondrial functions and vascular inflammation. As we found in our study, we confirmed that under physical conditions, CypD deficiency enhanced mouse whole body metabolic status, increased aortic mitochondrial complex III activity and decreased mitochondrial ROS generation. Functionally, CypD deficiency also attenuated inflammatory molecules expression, including VCAM-1, IL-6 and TNF-α in mouse aorta. CONCLUSIONS Our results review that mitochondrial CypD is involved in the regulation of inflammation in aorta and provide insights that blocking mitochondrial CypD enhances vascular resistance to inflammatory injuries.
Collapse
Affiliation(s)
- Xiaojing Liu
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, 250021, China
| | - Heng Du
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Dan Chen
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China; Department of Electrocardiographic, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China
| | - Hai Yuan
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China
| | - Wenbin Chen
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China
| | - Wenyu Jia
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, 250021, China
| | - Xiaolei Wang
- Deparment of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, 250021, China
| | - Xia Li
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong, 250021, China.
| | - Ling Gao
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, 250021, China; Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
28
|
Juhas M, Abutaleb N, Wang JT, Ye J, Shaikh Z, Sriworarat C, Qian Y, Bursac N. Incorporation of macrophages into engineered skeletal muscle enables enhanced muscle regeneration. Nat Biomed Eng 2018; 2:942-954. [PMID: 30581652 DOI: 10.1038/s41551-018-0290-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adult skeletal muscle has a robust capacity for self-repair, owing to synergies between muscle satellite cells and the immune system. In vitro models of muscle self-repair would facilitate the basic understanding of muscle regeneration and the screening of therapies for muscle disease. Here, we show that the incorporation of macrophages into muscle tissues engineered from adult-rat myogenic cells enables near-complete structural and functional repair after cardiotoxic injury in vitro. First, we show that-in contrast with injured neonatal-derived engineered muscle-adult-derived engineered muscle fails to properly self-repair after injury, even when treated with pro-regenerative cytokines. We then show that rat bone-marrow-derived macrophages or human blood-derived macrophages resident within the in vitro engineered tissues stimulate muscle satellite cell-mediated myogenesis while significantly limiting myofibre apoptosis and degeneration. Moreover, bone-marrow-derived macrophages within engineered tissues implanted in a mouse dorsal window-chamber model augmented blood vessel ingrowth, cell survival, muscle regeneration and contractile function.
Collapse
Affiliation(s)
- Mark Juhas
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nadia Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jason T Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jean Ye
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Zohaib Shaikh
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Ying Qian
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA. .,Regeneration Next, Duke University, Durham, NC, USA.
| |
Collapse
|
29
|
Liu W, Huang Q, Lin D, Zhao L, Ma J. Effect of lung protective ventilation on coronary heart disease patients undergoing lung cancer resection. J Thorac Dis 2018; 10:2760-2770. [PMID: 29997938 DOI: 10.21037/jtd.2018.04.90] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Mechanical ventilation, especially large tidal volume (Vt) one-lung ventilation (OLV), can cause ventilator-induced lung injury (VILI) that can stimulate cytokines. Meanwhile, cytokines are considered very important factor influencing coronary heart disease (CHD) patient prognosis. So minimization of pulmonary inflammatory responses by reduction of cytokine levels for CHD undergoing lung resection during OLV should be a priority. Because previous studies have demonstrated that lung-protective ventilation (LPV) reduced lung inflammation, this ventilation approach was studied for CHD patients undergoing lung resection here to evaluate the effects of LPV on pulmonary inflammatory responses. Methods This is a single center, randomized controlled trial. Primary endpoint of the study are plasma concentrations of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-10 and C-reactive protein (CRP). Secondary endpoints include respiratory variables and hemodynamic variables. 60 CHD patients undergoing video-assisted thoracoscopic lung resection were randomly divided into conventional ventilation group [10 mL/kg Vt and 0 cmH2O positive end-expiratory pressure (PEEP), C group] and protective ventilation group (6 mL/kg Vt and 6 cmH2O PEEP, P group; 30 patients/group). Hemodynamic variables, peak inspiratory pressure (Ppeak), dynamic compliance (Cdyn), arterial oxygen tension (PaO2) and arterial carbon dioxide tension (PaCO2) were recorded as test data at three time points: T1-endotracheal intubation for two-lung ventilation (TLV) when breathing and hemodynamics were stable; T2-after TLV was substituted with OLV when breathing and hemodynamics were stable; T3-OLV was substituted with TLV at the end of surgery when breathing and hemodynamics were stable. The concentrations of TNF-α, IL-6, IL-10 and CRP in patients' blood in both groups at the very beginning of OLV (beginning of OLV) and the end moment of the surgery (end of surgery) were measured. Results The P group exhibited greater PaO2, higher Cdyn and lower Ppeak than the C group at T2, T3 (P<0.05). At the end moment of the surgery, although the P group tended to exhibit higher TNF-α and IL-10 values than the C group, the differences did not reach statistical significance(P=0.0817, P=0.0635). Compared with C group at the end moment of the surgery, IL-6 and CRP were lower in P group, the differences were statistically significant (P=0.0093, P=0.0005). There were no significant differences in hemodynamic variables between the two groups (P>0.05). Conclusions LPV can effectively reduce the airway pressure, improve Cdyn and PaO2, reduce concentrations of IL-6 and CRP during lung resection of CHD patients.Trial registration: The trial was registered in the Chinese Clinical Trial Registry.
Collapse
Affiliation(s)
- Wenjun Liu
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qian Huang
- Department of Respiratory Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Duomao Lin
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Liyun Zhao
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jun Ma
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
30
|
The Impact of Uremic Toxins on Vascular Smooth Muscle Cell Function. Toxins (Basel) 2018; 10:toxins10060218. [PMID: 29844272 PMCID: PMC6024314 DOI: 10.3390/toxins10060218] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/26/2018] [Accepted: 05/27/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with profound vascular remodeling, which accelerates the progression of cardiovascular disease. This remodeling is characterized by intimal hyperplasia, accelerated atherosclerosis, excessive vascular calcification, and vascular stiffness. Vascular smooth muscle cell (VSMC) dysfunction has a key role in the remodeling process. Under uremic conditions, VSMCs can switch from a contractile phenotype to a synthetic phenotype, and undergo abnormal proliferation, migration, senescence, apoptosis, and calcification. A growing body of data from experiments in vitro and animal models suggests that uremic toxins (such as inorganic phosphate, indoxyl sulfate and advanced-glycation end products) may directly impact the VSMCs’ physiological functions. Chronic, low-grade inflammation and oxidative stress—hallmarks of CKD—are also strong inducers of VSMC dysfunction. Here, we review current knowledge about the impact of uremic toxins on VSMC function in CKD, and the consequences for pathological vascular remodeling.
Collapse
|
31
|
van Puijvelde GHM, Foks AC, van Bochove RE, Bot I, Habets KLL, de Jager SC, ter Borg MND, van Osch P, Boon L, Vos M, de Waard V, Kuiper J. CD1d deficiency inhibits the development of abdominal aortic aneurysms in LDL receptor deficient mice. PLoS One 2018; 13:e0190962. [PMID: 29346401 PMCID: PMC5773169 DOI: 10.1371/journal.pone.0190962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/22/2017] [Indexed: 11/19/2022] Open
Abstract
An abdominal aortic aneurysm (AAA) is a dilatation of the abdominal aorta leading to serious complications and mostly to death. AAA development is associated with an accumulation of inflammatory cells in the aorta including NKT cells. An important factor in promoting the recruitment of these inflammatory cells into tissues and thereby contributing to the development of AAA is angiotensin II (Ang II). We demonstrate that a deficiency in CD1d dependent NKT cells under hyperlipidemic conditions (LDLr-/-CD1d-/- mice) results in a strong decline in the severity of angiotensin II induced aneurysm formation when compared with LDLr-/- mice. In addition, we show that Ang II amplifies the activation of NKT cells both in vivo and in vitro. We also provide evidence that type I NKT cells contribute to AAA development by inducing the expression of matrix degrading enzymes in vSMCs and macrophages, and by cytokine dependently decreasing vSMC viability. Altogether, these data prove that CD1d-dependent NKT cells contribute to AAA development in the Ang II-mediated aneurysm model by enhancing aortic degradation, establishing that therapeutic applications which target NKT cells can be a successful way to prevent AAA development.
Collapse
Affiliation(s)
- Gijs H. M. van Puijvelde
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- * E-mail:
| | - Amanda C. Foks
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Rosemarie E. van Bochove
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Kim L. L. Habets
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Saskia C. de Jager
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mariëtte N. D. ter Borg
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Puck van Osch
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Mariska Vos
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
32
|
Xu YJ, Zheng L, Hu YW, Wang Q. Pyroptosis and its relationship to atherosclerosis. Clin Chim Acta 2018; 476:28-37. [DOI: 10.1016/j.cca.2017.11.005] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/31/2022]
|
33
|
Trittmann JK, Jin Y, Chicoine LG, Liu Y, Chen B, Nelin LD. An arginase-1 SNP that protects against the development of pulmonary hypertension in bronchopulmonary dysplasia enhances NO-mediated apoptosis in lymphocytes. Physiol Rep 2017; 4:4/22/e13041. [PMID: 27895230 PMCID: PMC5358007 DOI: 10.14814/phy2.13041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/20/2016] [Accepted: 10/26/2016] [Indexed: 11/24/2022] Open
Abstract
Arginase and nitric oxide synthase (NOS) share a common substrate, l‐arginine, and have opposing effects on vascular remodeling. Arginase is the first step in polyamine and proline synthesis necessary for cellular proliferation, while NO produced from NOS promotes apoptosis. Previously, we identified a single nucleotide polymorphism (SNP) in the arginase‐1 (ARG1) gene, rs2781666 (T‐allele) that was associated with a decreased risk for developing pulmonary hypertension (PH) in a cohort of infants with bronchopulmonary dysplasia (BPD). In this study, we utilized lymphocytes from neonates (the only readily available cells from these patients expressing the two genotypes of interest) with either the rs2781666 SNP (TT) or wild type (GG) to test the hypothesis that the protection of the ARG1 SNP against the development of PH in BPD would involve augmented NO production leading to more apoptosis. Lymphocytes were stimulated with IL‐4, IL‐13, and phorbol myristate acetate (PMA). We found that TT lymphocytes had similar levels of arginase I and arginase II expression, but there was a tendency for lower urea production (a surrogate marker of arginase activity), than in the GG lymphocytes. The TT lymphocytes also had significantly greater NO production than did GG lymphocytes despite no differences in iNOS expression between genotypes. Furthermore, the TT lymphocytes had lower numbers of viable cells, and higher levels of cleaved caspase‐3 than did GG lymphocytes. Inhibiting NOS activity using Nω‐Nitro‐l‐arginine methyl ester hydrochloride (l‐NAME) significantly decreased cleaved caspase‐3 levels in the TT lymphocytes. These data demonstrate that the TT genotype results in greater levels of NO production leading to more apoptosis, which is consistent with the concept that BPD patients with the TT genotype are protected against the development of PH by producing greater basal levels of endogenous NO.
Collapse
Affiliation(s)
- Jennifer K Trittmann
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio .,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio
| | - Louis G Chicoine
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Ohio State University, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
34
|
Vendrov AE, Stevenson MD, Alahari S, Pan H, Wickline SA, Madamanchi NR, Runge MS. Attenuated Superoxide Dismutase 2 Activity Induces Atherosclerotic Plaque Instability During Aging in Hyperlipidemic Mice. J Am Heart Assoc 2017; 6:e006775. [PMID: 29079564 PMCID: PMC5721769 DOI: 10.1161/jaha.117.006775] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/14/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Atherosclerosis progression during aging culminates in the development of vulnerable plaques, which may increase the risk of cardiovascular events. Increased generation and/or decreased scavenging of reactive oxygen species in the vascular wall are major contributors to atherogenesis. We previously showed that superoxide dismutase 2 deficiency increased vascular oxidative stress and reduced aortic compliance in aged wild-type mice and that young Apoe-/-/Sod2+/- had increased mitochondrial DNA damage and atherosclerosis versus young Apoe-/- mice. Here we investigated the effects of superoxide dismutase 2 deficiency on atherosclerosis progression and plaque morphology in middle-aged Apoe-/- mice. METHODS AND RESULTS Compared with Apoe-/-, middle-aged Apoe-/-/Sod2+/- mice had increased vascular wall reactive oxygen species (P<0.05) and higher atherosclerotic lesion area (P<0.001). The atherosclerotic plaques in middle-aged Apoe-/-/Sod2+/- mice had an increased necrotic core with higher inflammatory cell infiltration, a thinned fibrous cap with depleted smooth muscle content, and intraplaque hemorrhage. In addition, the plaque shoulder area had higher levels of calpain-2, caspase-3, and matrix metalloproteinase-2 in intimal smooth muscle cells and depleted fibrous cap collagen. Targeting mitochondrial reactive oxygen species with MitoTEMPO attenuated features of atherosclerotic plaque vulnerability in middle-aged Apoe-/-/Sod2+/- mice by lowering expression of calpain-2, caspase-3, and matrix metalloproteinase-2 and decreasing smooth muscle cell apoptosis and matrix degradation. CONCLUSIONS Enhanced mitochondrial oxidative stress under hyperlipidemic conditions in aging induces plaque instability, in part by increasing smooth muscle cell apoptosis, necrotic core expansion, and matrix degradation. Targeting mitochondrial reactive oxygen species or its effectors may be a viable therapeutic strategy to prevent aging-associated and oxidative stress-related atherosclerosis complications.
Collapse
MESH Headings
- Age Factors
- Aging/genetics
- Aging/metabolism
- Aging/pathology
- Animals
- Aorta/enzymology
- Aorta/pathology
- Aortic Diseases/blood
- Aortic Diseases/enzymology
- Aortic Diseases/genetics
- Aortic Diseases/pathology
- Apoptosis
- Apoptosis Regulatory Proteins/metabolism
- Atherosclerosis/blood
- Atherosclerosis/enzymology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Cells, Cultured
- DNA Damage
- Disease Models, Animal
- Extracellular Matrix Proteins/metabolism
- Fibrosis
- Genetic Predisposition to Disease
- Hyperlipidemias/blood
- Hyperlipidemias/enzymology
- Hyperlipidemias/genetics
- Male
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Mitochondria, Muscle/enzymology
- Mitochondria, Muscle/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Necrosis
- Oxidative Stress
- Phenotype
- Plaque, Atherosclerotic
- Proteolysis
- Rupture, Spontaneous
- Superoxide Dismutase/deficiency
- Superoxide Dismutase/genetics
- Vascular Remodeling
Collapse
Affiliation(s)
- Aleksandr E Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI
| | - Mark D Stevenson
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI
| | - Samthosh Alahari
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI
| | - Hua Pan
- Department of Cardiovascular Sciences, USF Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Samuel A Wickline
- Department of Cardiovascular Sciences, USF Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Nageswara R Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI
| | - Marschall S Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
35
|
Patel AK, Suri HS, Singh J, Kumar D, Shafique S, Nicolaides A, Jain SK, Saba L, Gupta A, Laird JR, Giannopoulos A, Suri JS. A Review on Atherosclerotic Biology, Wall Stiffness, Physics of Elasticity, and Its Ultrasound-Based Measurement. Curr Atheroscler Rep 2017; 18:83. [PMID: 27830569 DOI: 10.1007/s11883-016-0635-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Functional and structural changes in the common carotid artery are biomarkers for cardiovascular risk. Current methods for measuring functional changes include pulse wave velocity, compliance, distensibility, strain, stress, stiffness, and elasticity derived from arterial waveforms. The review is focused on the ultrasound-based carotid artery elasticity and stiffness measurements covering the physics of elasticity and linking it to biological evolution of arterial stiffness. The paper also presents evolution of plaque with a focus on the pathophysiologic cascade leading to arterial hardening. Using the concept of strain, and image-based elasticity, the paper then reviews the lumen diameter and carotid intima-media thickness measurements in combined temporal and spatial domains. Finally, the review presents the factors which influence the understanding of atherosclerotic disease formation and cardiovascular risk including arterial stiffness, tissue morphological characteristics, and image-based elasticity measurement.
Collapse
Affiliation(s)
- Anoop K Patel
- Department of Computer Engineering, NIT, Kurukshetra, India
| | | | - Jaskaran Singh
- Department of Computer Engineering, NIT, Kurukshetra, India
| | - Dinesh Kumar
- Point-of-Care Devices, Global Biomedical Technologies, Inc., Roseville, CA, USA
| | | | | | - Sanjay K Jain
- Department of Computer Engineering, NIT, Kurukshetra, India
| | - Luca Saba
- Department of Radiology, University of Cagliari, Cagliari, Italy
| | - Ajay Gupta
- Radiology Department, Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - John R Laird
- UC Davis Vascular Center, University of California, Davis, CA, USA
| | | | - Jasjit S Suri
- Vascular Diagnostic Center, University of Cyprus, Nicosia, Cyprus. .,Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, USA. .,Department of Electrical Engineering, University of Idaho (Affl.), Moscow, ID, USA. .,Diagnosis and Stroke Monitoring Division, AtheroPoint™, Roseville, CA, USA.
| |
Collapse
|
36
|
Ping S, Liu S, Zhou Y, Li Z, Li Y, Liu K, Bardeesi AS, Wang L, Chen J, Deng L, Wang J, Wang H, Chen D, Zhang Z, Sheng P, Li C. Protein disulfide isomerase-mediated apoptosis and proliferation of vascular smooth muscle cells induced by mechanical stress and advanced glycosylation end products result in diabetic mouse vein graft atherosclerosis. Cell Death Dis 2017; 8:e2818. [PMID: 28542133 PMCID: PMC5520728 DOI: 10.1038/cddis.2017.213] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/12/2017] [Accepted: 04/05/2017] [Indexed: 01/08/2023]
Abstract
Protein disulfide isomerase (PDI) involves cell survival and death. Whether PDI mediates mechanical stretch stress (SS) and/or advanced glycosylation end products (AGEs) -triggered simultaneous increases in proliferation and apoptosis of vascular smooth muscle cells (VSMCs) is unknown. Here, we hypothesized that different expression levels of PDI trigger completely opposite cell fates among the different VSMC subtypes. Mouse veins were grafted into carotid arteries of non-diabetic and diabetic mice for 8 weeks; the grafted veins underwent simultaneous increases in proliferation and apoptosis, which triggered vein graft arterializations in non-diabetic or atherosclerosis in diabetic mice. A higher rate of proliferation and apoptosis was seen in the diabetic group. SS and/or AGEs stimulated the quiescent cultured VSMCs, resulting in simultaneous increases in proliferation and apoptosis; they could induce increased PDI activation and expression. Both in vivo and in vitro, the proliferating VSMCs indicated weak co-expression of PDI and SM-α-actin while apoptotic or dead cells showed strong co-expression of both. Either SS or AGEs rapidly upregulated the expression of PDI, NOX1 and ROS, and their combination had synergistic effects. Inhibiting PDI simultaneously suppressed the proliferation and apoptosis of VSMCs, while inhibition of SM-α-actin with cytochalasin D led to increased apoptosis and cleaved caspases-3 but had no effect on proliferation. In conclusion, different expression levels of PDI in VSMCs induced by SS and/or AGEs triggered a simultaneous increase in proliferation and apoptosis, accelerated vein graft arterializations or atherosclerosis, leading us to propose PDI as a novel target for the treatment of vascular remodeling and diseases.
Collapse
Affiliation(s)
- Suning Ping
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuhuan Zhou
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ziqing Li
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuhuang Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kefeng Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Adham Sa Bardeesi
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linli Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingbo Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lie Deng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingjing Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hong Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dadi Chen
- Experimental Center for Basic Medical Teaching, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhengyu Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Department of Histology and Embryology, School of Basic Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
37
|
Tzeng HP, Lan KC, Yang TH, Chung MN, Liu SH. Benzo[a]pyrene activates interleukin-6 induction and suppresses nitric oxide-induced apoptosis in rat vascular smooth muscle cells. PLoS One 2017; 12:e0178063. [PMID: 28531207 PMCID: PMC5439712 DOI: 10.1371/journal.pone.0178063] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/08/2017] [Indexed: 11/18/2022] Open
Abstract
Benzo[a]pyrene, a ubiquitous environmental pollutant, has been suggested to be capable of initiating and/or accelerating atherosclerosis. Accumulation of vascular smooth muscle cells (VSMCs) in vessel intima is a hallmark of atherosclerosis. Nitric oxide (NO) can suppress VSMCs proliferation and induce VSMCs apoptosis. NO plays a compensatory role in the vascular lesions to reduce proliferation and/or accelerate apoptosis of VSMCs. The aim of this study was to investigate whether benzo[a]pyrene can affect VSMCs growth and apoptosis induced by NO. Benzo[a]pyrene (1–30 μmol/L) did not affect the cell number and cell cycle distribution in VSMCs under serum deprivation condition. Sodium nitroprusside (SNP), a NO donor, decreased cell viability and induced apoptosis in VSMCs. Benzo[a]pyrene significantly suppressed SNP-induced cell viability reduction and apoptosis. VSMCs cultured in conditioned medium from cells treated with benzo[a]pyrene could also prevent SNP-induced apoptosis. Benzo[a]pyrene was capable of inducing the activation of nuclear factor (NF)-κB and phosphorylation of p38 mitogen-activated protein kinase (MAPK) in VSMCs. Both NF-κB inhibitor and p38 MAPK inhibitor significantly reversed the anti-apoptotic effect of benzo[a]pyrene on SNP-treated VSMCs. Incubation of VSMCs with benzo[a]pyrene significantly and dose-dependently increased interleukin (IL)-6 production. A neutralizing antibody to IL-6 effectively reversed the anti-apoptotic effect of benzo[a]pyrene on SNP-treated VSMCs. Taken together, these results demonstrate for the first time that benzo[a]pyrene activates IL-6 induction and protects VSMCs from NO-induced apoptosis. These findings propose a new mechanism for the atherogenic effect of benzo[a]pyrene.
Collapse
Affiliation(s)
- Huei-Ping Tzeng
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ting-Hua Yang
- Department of Otolaryngology, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan
| | - Min-Ni Chung
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shing Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
38
|
Yan X, Wu L, Li B, Meng X, Dai H, Zheng Y, Fu J. Cyanidin-3-O-glucoside Induces Apoptosis and Inhibits Migration of Tumor Necrosis Factor-α-Treated Rat Aortic Smooth Muscle Cells. Cardiovasc Toxicol 2017; 16:251-9. [PMID: 26138096 DOI: 10.1007/s12012-015-9333-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Blueberries are rich in anthocyanins (ACNs), which have recently been noted to protect against atherosclerosis development in mice. Cyanidin-3-O-glucoside (C3G), a member of blueberry ACN family, can inhibit the tumor necrosis factor-α (TNF-α)-induced proliferation of vascular smooth muscle cells (VSMCs). However, the effects of C3G on VSMC apoptosis and migration remain unclear. This study was thus conducted to examine whether and how C3G affected the apoptosis and migration of rat aortic smooth muscle cells (RASMCs) challenged by TNF-α. Primary cultured RASMCs were pretreated with C3G (25, 50 or 100 μM) for 2 h and then stimulated with TNF-α (10 ng/ml) for additional 24 h. Our results illustrated that C3G pretreatment induced significant apoptosis in TNF-α-stimulated RASMCs in a dose-dependent way, which was accompanied with increased cleaved caspase-3, caspase-9 and Bax and decreased Bcl-2. Moreover, RASMC migration was enhanced by TNF-α, but markedly suppressed by C3G pretreatment. The expressions and activities of matrix metalloproteinase-2 (MMP-2) and MMP-9 were inhibited by C3G. In addition, TNF-α-enhanced nuclear translocation of nuclear factor kappa B (NF-κB) subunit p65 and phosphorylation of NF-κB inhibitor α (IκBα) in RASMCs were attenuated by C3G. In summary, our study reveals that C3G can induce significant apoptosis in TNF-α-treated RASMCs and markedly inhibit their migration.
Collapse
Affiliation(s)
- Xuerui Yan
- Plant Protection College, Shenyang Agricultural University, 120 Dongling Road, Shenyang, 110161, Liaoning, People's Republic of China
| | - Lin Wu
- Jilin Province Pulan High-Tech Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Bin Li
- Food College, Shenyang Agricultural University, Shenyang, 110161, Liaoning, People's Republic of China
| | - Xianjun Meng
- Food College, Shenyang Agricultural University, Shenyang, 110161, Liaoning, People's Republic of China
| | - Hanping Dai
- Horticulture College, Shenyang Agricultural University, Shenyang, 110161, Liaoning, People's Republic of China
| | - Yanan Zheng
- College of Forestry, Shenyang Agricultural University, Shenyang, 110161, Liaoning, People's Republic of China
| | - Junfan Fu
- Plant Protection College, Shenyang Agricultural University, 120 Dongling Road, Shenyang, 110161, Liaoning, People's Republic of China.
| |
Collapse
|
39
|
Hayat U, Thondapu V, Tsay T, Barlis P. Atherogenesis and Inflammation. Interv Cardiol 2016. [DOI: 10.1002/9781118983652.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Umair Hayat
- Melbourne Medical School; The University of Melbourne; Australia
| | - Vikas Thondapu
- Melbourne Medical School; The University of Melbourne; Australia
| | - Tim Tsay
- Melbourne Medical School; The University of Melbourne; Australia
| | | |
Collapse
|
40
|
Chernyavskiy I, Veeranki S, Sen U, Tyagi SC. Atherogenesis: hyperhomocysteinemia interactions with LDL, macrophage function, paraoxonase 1, and exercise. Ann N Y Acad Sci 2016; 1363:138-54. [PMID: 26849408 DOI: 10.1111/nyas.13009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/23/2015] [Accepted: 01/05/2016] [Indexed: 12/13/2022]
Abstract
Despite great strides in understanding the atherogenesis process, the mechanisms are not entirely known. In addition to diet, cigarette smoking, genetic predisposition, and hypertension, hyperhomocysteinemia (HHcy), an accumulation of the noncoding sulfur-containing amino acid homocysteine (Hcy), is a significant contributor to atherogenesis. Although exercise decreases HHcy and increases longevity, the complete mechanism is unclear. In light of recent evidence, in this review, we focus on the effects of HHcy on macrophage function, differentiation, and polarization. Though there is need for further evidence, it is most likely that HHcy-mediated alterations in macrophage function are important contributors to atherogenesis, and HHcy-countering strategies, such as nutrition and exercise, should be included in the combinatorial regimens for effective prevention and regression of atherosclerotic plaques. Therefore, we also included a discussion on the effects of exercise on the HHcy-mediated atherogenic process.
Collapse
Affiliation(s)
- Ilya Chernyavskiy
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Sudhakar Veeranki
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Utpal Sen
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
41
|
Moss JWE, Ramji DP. Interferon-γ: Promising therapeutic target in atherosclerosis. World J Exp Med 2015; 5:154-159. [PMID: 26309816 PMCID: PMC4543809 DOI: 10.5493/wjem.v5.i3.154] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/21/2015] [Accepted: 05/08/2015] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder of the vasculature and is the primary cause of cardiovascular disease (CVD). CVD is currently the world’s leading cause of death and the numbers are predicted to rise further because of a global increase in risk factors such as diabetes and obesity. Current therapies such as statins have had a major impact in reducing mortality from CVD. However, there is a marked residual CVD risk in patients on statin therapy. It is therefore important to understand the molecular basis of this disease in detail and to develop alternative novel therapeutics. Interferon-γ (IFN-γ) is a pro-inflammatory cytokine that is often regarded as a master regulator of atherosclerosis development. IFN-γ is able to influence several key steps during atherosclerosis development, including pro-inflammatory gene expression, the recruitment of monocytes from the blood to the activated arterial endothelium and plaque stability. This central role of IFN-γ makes it a promising therapeutic target. The purpose of this editorial is to describe the key role IFN-γ plays during atherosclerosis development, as well as discuss potential strategies to target it therapeutically.
Collapse
|
42
|
Batchu SN, Xia J, Ko KA, Doyley MM, Abe JI, Morrell CN, Korshunov VA. Axl modulates immune activation of smooth muscle cells in vein graft remodeling. Am J Physiol Heart Circ Physiol 2015; 309:H1048-58. [PMID: 26276821 DOI: 10.1152/ajpheart.00495.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/06/2015] [Indexed: 12/13/2022]
Abstract
The pathophysiological mechanisms of the immune activation of smooth muscle cells are not well understood. Increased expression of Axl, a receptor tyrosine kinase, was recently found in arteries from patients after coronary bypass grafts. In the present study, we hypothesized that Axl-dependent immune activation of smooth muscle cells regulates vein graft remodeling. We observed a twofold decrease in intimal thickening after vascular and systemic depletion of Axl in vein grafts. Local depletion of Axl had the greatest effect on immune activation, whereas systemic deletion of Axl reduced intima due to an increase in apoptosis in vein grafts. Primary smooth muscle cells isolated from Axl knockout mice had reduced proinflammatory responses by prevention of the STAT1 pathway. The absence of Axl increased suppressor of cytokine signaling (SOCS)1 expression in smooth muscle cells, a major inhibitory protein for STAT1. Ultrasound imaging suggested that vascular depletion of Axl reduced vein graft stiffness. Axl expression determined the STAT1-SOCS1 balance in vein graft intima and progression of the remodeling. The results of this investigation demonstrate that Axl promotes STAT1 signaling via inhibition of SOCS1 in activated smooth muscle cells in vein graft remodeling.
Collapse
Affiliation(s)
- Sri N Batchu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Jixiang Xia
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Kyung Ae Ko
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Marvin M Doyley
- Department of Electrical and Computer Engineering, University of Rochester and Hajim School of Engineering and Applied Sciences, Rochester, New York; and
| | - Jun-Ichi Abe
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Craig N Morrell
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Vyacheslav A Korshunov
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, New York;
| |
Collapse
|
43
|
Fraser R, Whitley GSJ, Thilaganathan B, Cartwright JE. Decidual natural killer cells regulate vessel stability: implications for impaired spiral artery remodelling. J Reprod Immunol 2015; 110:54-60. [PMID: 26004035 PMCID: PMC4502446 DOI: 10.1016/j.jri.2015.04.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 04/08/2015] [Accepted: 04/21/2015] [Indexed: 11/18/2022]
Abstract
Decidual NK (dNK) cells are present during uterine spiral artery remodelling, an event that is crucial for successful placentation and the provision of an adequate blood supply to the developing fetus. Spiral artery remodelling is impaired in the pregnancy complication pre-eclampsia. Although dNK cells are known to play active roles at the maternal-fetal interface, little is known about their effect on endothelial integrity, an important component of vessel stability. We present a study in which we have modelled dNK-endothelium interactions, using first-trimester dNK cells isolated from both normal pregnancies and those with impaired spiral artery remodelling. dNK cells were isolated from first-trimester pregnancies, screened by uterine artery Doppler ultrasound to determine resistance indices (RI) that relate to the extent of spiral artery remodelling. dNK culture supernatant from normal-RI pregnancies (but not high-RI pregnancies) destabilised endothelial tube-like structures in Matrigel, and normal-RI dNK cells induced endothelial intercellular adhesion molecule-1 and tumour necrosis factor-α expression to a greater extent than high-RI dNK cells. We have established a functional role for dNK cells in the disruption of endothelial structures and have suggested how impairment of this process may be contributing to the reduced vessel remodelling in pregnancies with a high uterine artery resistance index. These findings have implications for our understanding of the pathology of pre-eclampsia and other pregnancy disorders where remodelling is impaired.
Collapse
Affiliation(s)
- Rupsha Fraser
- Institute of Cardiovascular and Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, United Kingdom
| | - Guy St J Whitley
- Institute of Cardiovascular and Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, United Kingdom
| | - Baskaran Thilaganathan
- Fetal Medicine Unit, St. George's Hospital, Blackshaw Road, London SW17 0QT, United Kingdom
| | - Judith E Cartwright
- Institute of Cardiovascular and Cell Sciences, St. George's, University of London, Cranmer Terrace, London SW17 0RE, United Kingdom.
| |
Collapse
|
44
|
Tivesten Å, Pinthus JH, Clarke N, Duivenvoorden W, Nilsson J. Cardiovascular risk with androgen deprivation therapy for prostate cancer: potential mechanisms. Urol Oncol 2015; 33:464-75. [PMID: 26141678 DOI: 10.1016/j.urolonc.2015.05.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 05/27/2015] [Accepted: 05/27/2015] [Indexed: 10/23/2022]
Abstract
Androgen deprivation therapy (ADT) is frequently used for the treatment of advanced prostate cancer. ADT is associated with numerous side effects related to its mode of action, namely the suppression of testosterone to castrate levels. Recently, several large retrospective studies have also reported an increased risk of diabetes and cardiovascular disease in men receiving ADT, although these risks have not been confirmed by prospective randomized trials. We review the literature to consider the risk of cardiovascular disease with different forms of ADT and examine in detail potential mechanisms by which any such risk could be mediated. Mechanisms discussed include the metabolic syndrome resulting from low testosterone level and the potential roles of testosterone flare, gonadotropin-releasing hormone receptors outside the pituitary gland, and altered levels of follicle-stimulating hormone. Finally, the clinical implications for men prescribed ADT for the treatment of advanced prostate cancer are considered.
Collapse
Affiliation(s)
- Åsa Tivesten
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Sahlgrenska University Hospital, Göteborg, Sweden.
| | - Jehonathan H Pinthus
- Department of Surgery, Division of Urology, McMaster University, Hamilton, Ontario, Canada
| | - Noel Clarke
- Department of Urology, The Christie and Salford Royal Hospitals, Manchester, UK
| | | | - Jan Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
45
|
Duque JC, Martinez L, Mesa A, Wei Y, Tabbara M, Salman LH, Vazquez-Padron RI. CD4(+) lymphocytes improve venous blood flow in experimental arteriovenous fistulae. Surgery 2015; 158:529-36. [PMID: 25999254 DOI: 10.1016/j.surg.2015.02.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/16/2015] [Accepted: 02/21/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND The role of immune cells in arteriovenous fistulae (AVF) maturation is poorly understood and has received, until quite recently, little attention. This study examines the function of T lymphocytes in AVF vascular remodeling. METHODS Experimental fistulae were created in athymic rnu nude rats lacking mature T lymphocytes and euthymic control animals by anastomosing the left superior epigastric vein to the nearby femoral artery. Blood flow rates, wall morphology, and histologic changes were assessed in AVF 21 days after creation. The effect of CD4(+) lymphocytes on AVF maturation in athymic animals was analyzed by adoptive transfer of cells after fistula creation. RESULTS The absence of T lymphocytes compromised blood flow in experimental fistulae. Histopathologic inspection of AVF from athymic rats revealed that T-cell immunodeficiency negatively affected venous vascular remodeling, as evidenced by a reduced lumen, a thick muscular layer, and a low number of inflammatory cells compared with control animals. Adoptive transfer of CD4(+) lymphocytes from euthymic rats into athymic animals after fistula creation improved blood flow and reduced intima-media thickness. CONCLUSION These results point at the protective role of CD4(+) lymphocytes in the remodeling of the AVF vascular wall.
Collapse
Affiliation(s)
- Juan C Duque
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Annia Mesa
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Yuntao Wei
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Marwan Tabbara
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL
| | - Loay H Salman
- Section of Interventional Nephrology, University of Miami Miller School of Medicine, Miami, FL
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL.
| |
Collapse
|
46
|
Han X, Boisvert WA. Interleukin-10 protects against atherosclerosis by modulating multiple atherogenic macrophage function. Thromb Haemost 2014; 113:505-12. [PMID: 25373619 DOI: 10.1160/th14-06-0509] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/22/2014] [Indexed: 01/15/2023]
Abstract
Atherosclerosis is primarily a disorder of lipid metabolism, but there is also a prominent chronic inflammatory component that drives the atherosclerotic lesion progression in the artery wall. During hyperlipidaemic conditions, there is a rapid influx of circulating monocytes into the atherosclerosis-prone areas of the arterial intima. These infiltrated monocytes differentiate into macrophages and take up the atherogenic lipoproteins in the intima of the vessel wall that have been modified within the lesion environment. Interleukin (IL)-10 is a prototypic anti-inflammatory cytokine made primarily by the macrophages and Th2 subtype T lymphocytes. In terms of atherosclerosis its major roles include inhibition of macrophage activation as well as inhibition of matrix metalloproteinase, pro-inflammatory cytokines and cyclooxygenase-2 expression in lipid-loaded and activated macrophage foam cells. Recent discoveries suggest another important role of IL-10 in atherosclerosis: its ability to alter lipid metabolism in macrophages. The current review will highlight the present knowledge on multiple ways in which IL-10 mediates atherosclerosis. As macrophages play a critical role in all stages of atherosclerosis, the review will concentrate on how IL-10 regulates the activities of macrophages that are especially important in the development of atherosclerosis.
Collapse
Affiliation(s)
| | - William A Boisvert
- William A. Boisvert, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, USA, Tel.: +1 808 692 1567, Fax: +1 808 692 1973, E-mail:
| |
Collapse
|
47
|
Segedy AK, Pyle AL, Li B, Zhang Y, Babaev VR, Jat P, Fazio S, Atkinson JB, Linton MF, Young PP. Identification of small proline-rich repeat protein 3 as a novel atheroprotective factor that promotes adaptive Akt signaling in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2014; 34:2527-36. [PMID: 25278290 DOI: 10.1161/atvbaha.114.303644] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Atherosclerosis is the primary driver of cardiovascular disease, the leading cause of death worldwide. Identification of naturally occurring atheroprotective genes has become a major goal for the development of interventions that will limit atheroma progression and associated adverse events. To this end, we have identified small proline-rich repeat protein (SPRR3) as selectively upregulated in vascular smooth muscle cells (VSMCs) of atheroma-bearing arterial tissue versus healthy arterial tissue. In this study, we sought to determine the role of SPRR3 in atheroma pathophysiology. APPROACH AND RESULTS We found that atheroprone apolipoprotein E-null mice lacking SPRR3 developed significantly greater atheroma burden. To determine the cellular driver(s) of this increase, we evaluated SPRR3-dependent changes in bone marrow-derived cells, endothelial cells, and VSMCs. Bone marrow transplant of SPRR3-expressing cells into SPRR3(-/-)apolipoprotein E-deficient recipients failed to rescue atheroma burden. Similarly, endothelial cells did not exhibit a response to SPRR3 loss. However, atheromas from SPRR3-deficient mice exhibited increased TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling)-positive VSMCs compared with control. Cell death in SPRR3-deficient VSMCs was significantly increased in vitro. Conversely, SPRR3-overexpressing VSMCs exhibited reduced apoptosis compared with control. We also observed a PI3K (phosphatidylinositol 3-kinase)/Akt-dependent positive association between SPRR3 expression and levels of active Akt in VSMCs. The survival advantage seen in SPRR3-overexpressing VSMCs was abrogated after the addition of a PI3K/Akt pathway inhibitor. CONCLUSIONS These results indicate that SPRR3 protects the lesion from VSMC loss by promoting survival signaling in plaque VSMCs, thereby significantly decreasing atherosclerosis progression. As the first identified atheroma-specific VSMC prosurvival factor, SPRR3 represents a potential target for lesion-specific modulation of VSMC survival.
Collapse
Affiliation(s)
- Amanda K Segedy
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Amy L Pyle
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Bin Li
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Youmin Zhang
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Vladimir R Babaev
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Parmjit Jat
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Sergio Fazio
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - James B Atkinson
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - MacRae F Linton
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.)
| | - Pampee P Young
- From the Department of Pathology, Microbiology, and Immunology (A.K.S., B.L., S.F., J.B.A., P.P.Y.) and Departments of Veterans Affairs Medical Center (J.B.A., P.P.Y.), Pharmacology (M.F.L.), and Medicine (Y.Z., V.R.B., S.F., M.F.L., P.P.Y.), Vanderbilt University Medical Center, Nashville, TN; Department of Neurodegenerative Diseases, Institute of Neurology, University College London, Queen Square, London, United Kingdom (P.J.); Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH (A.L.P.); and Department of Pathology, The Ohio State University, Columbus (A.L.P.).
| |
Collapse
|
48
|
Jung I, Choi J, Jin J, Jeong S, Jeon S, Lim C, Lee M, Yoo J, Sonn S, Kim YH, Choi BK, Kwon BS, Seoh J, Lee CW, Kim D, Oh GT. CD137‐inducing factors from T cells and macrophages accelerate the destabilization of atherosclerotic plaques in hyperlipidemic mice. FASEB J 2014; 28:4779-91. [DOI: 10.1096/fj.14-253732] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- In‐Hyuk Jung
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
- Department of Veterinary PathologyCollege of Veterinary MedicineSeoul National UniversitySeoulKorea
| | - Jae‐Hoon Choi
- Department of Life ScienceCollege of Natural SciencesHanyang UniversitySeoulKorea
| | - Jing Jin
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
| | - Se‐Jin Jeong
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
| | - Sejin Jeon
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
| | - Chaeji Lim
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
| | - Mi‐Ran Lee
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
| | - Ji‐Young Yoo
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
| | - Seong‐Keun Sonn
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
| | - Young Ho Kim
- Immune Cell Production UnitProgram for Immunotherapeutic ResearchNational Cancer CenterGoyangKorea
| | - Beom Kyu Choi
- Cancer Immunology BranchDivision of Cancer BiologyNational Cancer CenterGoyangKorea
| | - Byoung S. Kwon
- Cancer Immunology BranchDivision of Cancer BiologyNational Cancer CenterGoyangKorea
| | - Ju‐Young Seoh
- Department of MicrobiologyGraduate School of MedicineEwha Womans UniversitySeoulKorea
| | - Cheol Whan Lee
- Department of MedicineAsan Medical CenterUniversity of UlsanSeoulKorea
| | - Dae‐Yong Kim
- Department of Veterinary PathologyCollege of Veterinary MedicineSeoul National UniversitySeoulKorea
| | - Goo Taeg Oh
- Department of Life SciencesGraduate School of MedicineEwha Womans UniversitySeoulKorea
- GT5 ProgramGraduate School of MedicineEwha Womans UniversitySeoulKorea
| |
Collapse
|
49
|
Baek MW, Seong KJ, Jeong YJ, Kim GM, Park HJ, Kim SH, Chung HJ, Kim WJ, Jung JY. Nitric oxide induces apoptosis in human gingival fibroblast through mitochondria-dependent pathway and JNK activation. Int Endod J 2014; 48:287-97. [DOI: 10.1111/iej.12314] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 05/19/2014] [Indexed: 01/04/2023]
Affiliation(s)
- M.-W. Baek
- Department of Oral Physiology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| | - K.-J. Seong
- Department of Oral Physiology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| | - Y.-J. Jeong
- Department of Oral Physiology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| | - G.-M. Kim
- Department of Oral Physiology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| | - H.-J. Park
- Department of Oral and Maxillofacial Surgery; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| | - S.-H. Kim
- Department of Oral Anatomy; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| | - H.-J. Chung
- Department of Periodontology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| | - W.-J. Kim
- Department of Oral Physiology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| | - J.-Y. Jung
- Department of Oral Physiology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju South Korea
| |
Collapse
|
50
|
Inflammation and atherosclerosis: disease modulating therapies. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2013; 15:681-95. [PMID: 23979859 DOI: 10.1007/s11936-013-0268-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OPINION STATEMENT Advances in the mechanistic understanding of atheroma initiation, repair, progression, and rupture have solidified the pivotal role played by the immune system in the pathophysiology of atherosclerotic vascular disease. These mechanistic findings have been extended into humans, with a strong evidence basis for the independent association between elevated blood markers of inflammation and future cardiovascular (CV) events. Investigations with statins as well as more conventional anti-inflammatory medications provide indirect evidence to support the concept that modifying immune responses can improve CV outcomes; however, robust evidence to support the use of anti-inflammatory treatment strategies to manage atherosclerotic vascular disease is still lacking. Such evidence may emerge from a new wave of clinical trials directly exploring the effects of targeted immune modulation on CV risk. These trials will provide key additional insights into atherosclerosis and will help determine the fate of immune modulation as a new treatment strategy in atherosclerotic vascular disease.
Collapse
|