1
|
Sinoglu B, Ersoy A. Effects of smoking on controlled hypotension with nitroglycerin during ear-nose-throat surgery. Niger J Clin Pract 2023; 26:657-665. [PMID: 37470636 DOI: 10.4103/njcp.njcp_1311_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Background and Aim In this study, the aim was to research the effects of smoking habits on controlled hypotension administered with nitroglycerin during ear-nose-throat surgery. Materials and Methods This study administered controlled hypotension with nitroglycerin and total intravenous anesthesia to a total of 80 patients undergoing septoplasty operations. The patients were divided into two groups of 40 non-smokers (Group 1) and 40 smokers (Group 2). Intravenous propofol infusion was used for anesthesia maintenance. Nitroglycerin with 0.25-1 μg/kg/min dose was titrated to provide controlled hypotension. During this process, the hemodynamic parameters of patients, total propofol and nitroglycerin amounts used, operation duration, and duration of controlled hypotension were recorded at the end of the operation. At the end of the operation, the surgeon assessed the lack of blood in the surgical field with Fromme Scale. Results Fromme scale values were significantly higher in Group 2 compared to Group 1. The MAP values at 10, 20, 30 min, and end of operation were lower, while 10- and 20-min heart rate values were higher in Group 2 compared to Group 1. Conclusion Nitroglycerin, chosen for controlled hypotension to reduce hemorrhage in the surgical field during nasal surgery, was shown to cause more pronounced hypotension and reflex tachycardia due to endothelial dysfunction linked to nicotine in patients who smoke. Despite lower pressure values in the smoking group, the negative effects of nicotine on platelet functions combined with similar effects of nitroglycerin to increase bleeding amounts.
Collapse
Affiliation(s)
- B Sinoglu
- Anesthesiology and Intensive Care Department, Kelkit State Hospital, Gümüshane, Turkey
| | - A Ersoy
- Anesthesiology and Intensive Care Department, Sultan 2, Abdülhamit Han Education and Reseach Hospital, Universty of Health Sciences, Istanbul, Turkey
| |
Collapse
|
2
|
Ohlrogge AH, Frost L, Schnabel RB. Harmful Impact of Tobacco Smoking and Alcohol Consumption on the Atrial Myocardium. Cells 2022; 11:2576. [PMID: 36010652 PMCID: PMC9406618 DOI: 10.3390/cells11162576] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Tobacco smoking and alcohol consumption are widespread exposures that are legal and socially accepted in many societies. Both have been widely recognized as important risk factors for diseases in all vital organ systems including cardiovascular diseases, and with clinical manifestations that are associated with atrial dysfunction, so-called atrial cardiomyopathy, especially atrial fibrillation and stroke. The pathogenesis of atrial cardiomyopathy, atrial fibrillation, and stroke in context with smoking and alcohol consumption is complex and multifactorial, involving pathophysiological mechanisms, environmental, and societal aspects. This narrative review summarizes the current literature regarding alterations in the atrial myocardium that is associated with smoking and alcohol.
Collapse
Affiliation(s)
- Amelie H. Ohlrogge
- Department of Cardiology, University Heart and Vascular Centre Hamburg, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Lars Frost
- Diagnostic Centre, University Clinic for Development of Innovative Patient Pathways, Silkeborg Regional Hospital, 8600 Silkeborg, Denmark
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Renate B. Schnabel
- Department of Cardiology, University Heart and Vascular Centre Hamburg, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
3
|
Trifunovic S, Smiljanić K, Sickmann A, Solari FA, Kolarevic S, Divac Rankov A, Ljujic M. Electronic cigarette liquids impair metabolic cooperation and alter proteomic profiles in V79 cells. Respir Res 2022; 23:191. [PMID: 35840976 PMCID: PMC9285873 DOI: 10.1186/s12931-022-02102-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/02/2022] [Indexed: 11/20/2022] Open
Abstract
Background Although still considered a safer alternative to classical cigarettes, growing body of work points to harmful effects of electronic cigarettes (e-cigarettes) affecting a range of cellular processes. The biological effect of e-cigarettes needs to be investigated in more detail considering their widespread use. Methods In this study, we treated V79 lung fibroblasts with sub-cytotoxic concentration of e-cigarette liquids, with and without nicotine. Mutagenicity was evaluated by HPRT assay, genotoxicity by comet assay and the effect on cellular communication by metabolic cooperation assay. Additionally, comprehensive proteome analysis was performed via high resolution, parallel accumulation serial fragmentation-PASEF mass spectrometry. Results E-cigarette liquid concentration used in this study showed no mutagenic or genotoxic effect, however it negatively impacted metabolic cooperation between V79 cells. Both e-cigarette liquids induced significant depletion in total number of proteins and impairment of mitochondrial function in treated cells. The focal adhesion proteins were upregulated, which is in accordance with the results of metabolic cooperation assay. Increased presence of posttranslational modifications (PTMs), including carbonylation and direct oxidative modifications, was observed. Data are available via ProteomeXchange with identifier PXD032071. Conclusions Our study revealed impairment of metabolic cooperation as well as significant proteome and PTMs alterations in V79 cells treated with e-cigarette liquid warranting future studies on e-cigarettes health impact. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02102-w.
Collapse
Affiliation(s)
- Sara Trifunovic
- Biology of Robustness Group, Mediterranean Institute for Life Sciences, Split, Croatia.
| | - Katarina Smiljanić
- Department of Biochemistry and Centre of Excellence for Molecular Food Sciences, University of Belgrade, Faculty of Chemistry, Studentski Trg 12-14, 11000, Belgrade, Serbia
| | - Albert Sickmann
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., Bunsen-Kirchhoff-Straße 11, Dortmund, Germany.,Medizinische Fakultät, Medizinisches Proteom-Center (MPC), Ruhr-Universität Bochum, 44801, Bochum, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, AB243FX, Scotland, UK
| | - Fiorella A Solari
- Leibniz-Institut Für Analytische Wissenschaften - ISAS - E.V., Bunsen-Kirchhoff-Straße 11, Dortmund, Germany
| | - Stoimir Kolarevic
- Department of Hydroecology and Water Protection, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Divac Rankov
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Mila Ljujic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
4
|
Fu X, Zong T, Yang P, Li L, Wang S, Wang Z, Li M, Li X, Zou Y, Zhang Y, Htet Aung LH, Yang Y, Yu T. Nicotine: Regulatory roles and mechanisms in atherosclerosis progression. Food Chem Toxicol 2021; 151:112154. [PMID: 33774093 DOI: 10.1016/j.fct.2021.112154] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/22/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
Smoking is an independent risk factor for atherosclerosis. The smoke produced by tobacco burning contains more than 7000 chemicals, among which nicotine is closely related to the occurrence and development of atherosclerosis. Nicotine, a selective cholinergic agonist, accelerates the formation of atherosclerosis by stimulating nicotinic acetylcholine receptors (nAChRs) located in neuronal and non-neuronal tissues. This review introduces the pathogenesis of atherosclerosis and the mechanisms involving nicotine and its receptors. Herein, we focus on the various roles of nicotine in atherosclerosis, such as upregulation of growth factors, inflammation, and the dysfunction of endothelial cells, vascular smooth muscle cells (VSMC) as well as macrophages. In addition, nicotine can stimulate the generation of reactive oxygen species, cause abnormal lipid metabolism, and activate immune cells leading to the onset and progression of atherosclerosis. Exosomes, are currently a research hotspot, due to their important connections with macrophages and the VSMC, and may represent a novel application into future preventive treatment to promote the prevention of smoking-related atherosclerosis. In this review, we will elaborate on the regulatory mechanism of nicotine on atherosclerosis, as well as the effects of interference with nicotine receptors and the use of exosomes to prevent atherosclerosis development.
Collapse
Affiliation(s)
- Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Tingyu Zong
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Panyu Yang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Lin Li
- Department of Vascular Surgery, The Qingdao Hiser Medical Center, Qingdao, Shandong Province, China
| | - Shizhong Wang
- The Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 66000, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Yulin Zou
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Ying Zhang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Lynn Htet Htet Aung
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao, 266021, People's Republic of China.
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| |
Collapse
|
5
|
The Role of Connexin 43 in Lung Disease. Life (Basel) 2020; 10:life10120363. [PMID: 33352732 PMCID: PMC7766413 DOI: 10.3390/life10120363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 01/10/2023] Open
Abstract
The term lung disease describes a broad category of disorders that impair lung function. More than 35 million Americans have a preventable chronic lung disease with high mortality rates due to limited treatment efficacy. The recent increase in patients with lung disease highlights the need to increase our understanding of mechanisms driving lung inflammation. Connexins, gap junction proteins, and more specifically connexin 43 (Cx43), are abundantly expressed in the lung and are known to play a role in lung diseases. This review focuses on the role of Cx43 in pathology associated with acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD) and asthma. Additionally, we discuss the role of Cx43 in preventing disease through the transfer of mitochondria between cells. We aim to highlight the need to better understand what cell types are expressing Cx43 and how this expression influences lung disease.
Collapse
|
6
|
Nijjar S, Maddison D, Meigh L, de Wolf E, Rodgers T, Cann MJ, Dale N. Opposing modulation of Cx26 gap junctions and hemichannels by CO 2. J Physiol 2020; 599:103-118. [PMID: 33022747 DOI: 10.1113/jp280747] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/02/2020] [Indexed: 01/21/2023] Open
Abstract
KEY POINTS A moderate increase in P C O 2 (55 mmHg) closes Cx26 gap junctions. This effect of CO2 is independent of changes in intra- or extracellular pH. The CO2 -dependent closing effect depends on the same residues (K125 and R104) that are required for the CO2 -dependent opening of Cx26 hemichannels. Pathological mutations of Cx26 abolish the CO2 -dependent closing of the gap junction. Elastic network modelling suggests that the effect of CO2 on Cx26 hemichannels and gap junctions is mediated through changes in the lowest entropy state of the protein. ABSTRACT Cx26 hemichannels open in response to moderate elevations of CO2 ( P C O 2 55 mmHg) via a carbamylation reaction that depends on residues K125 and R104. Here we investigate the action of CO2 on Cx26 gap junctions. Using a dye transfer assay, we found that an elevated P C O 2 of 55 mmHg greatly delayed the permeation of a fluorescent glucose analogue (NBDG) between HeLa cells coupled by Cx26 gap junctions. However, the mutations K125R or R104A abolished this effect of CO2 . Whole cell recordings demonstrated that elevated CO2 reduced the Cx26 gap junction conductance (median reduction 66.7%, 95% CI, 50.5-100.0%) but had no effect on Cx26K125R or Cx31 gap junctions. CO2 can cause intracellular acidification. Using 30 mm propionate, we found that acidification in the absence of a change in P C O 2 caused a median reduction in the gap junction conductance of 41.7% (95% CI, 26.6-53.7%). This effect of propionate was unaffected by the K125R mutation (median reduction 48.1%, 95% CI, 28.0-86.3%). pH-dependent and CO2 -dependent closure of the gap junction are thus mechanistically independent. Mutations of Cx26 associated with the keratitis ichthyosis deafness syndrome (N14K, A40V and A88V), in combination with the mutation M151L, also abolished the CO2 -dependent gap junction closure. Elastic network modelling suggests that the lowest entropy state when CO2 is bound is the closed configuration for the gap junction but the open state for the hemichannel. The opposing actions of CO2 on Cx26 gap junctions and hemichannels thus depend on the same residues and presumed carbamylation reaction.
Collapse
Affiliation(s)
- Sarbjit Nijjar
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Daniel Maddison
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Louise Meigh
- School of Life Sciences, University of Warwick, Coventry, UK
| | | | - Thomas Rodgers
- School of Chemical Engineering and Analytical Science, University of Manchester, Manchester, UK
| | - Martin J Cann
- Department of Biosciences, Durham University, Durham, UK
| | - Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
7
|
Kenmochi H, Yamasaki T, Koizumi S, Sameshima T, Namba H. Nicotine does not affect stem cell properties requisite for suicide gene therapy against glioma. Neurol Res 2020; 42:818-827. [PMID: 32588772 DOI: 10.1080/01616412.2020.1782123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE: Glioblastoma is one of the most lethal tumors in adult central nervous system with a median survival of a year and half and effective therapeutic strategy is urgently needed. For that reason, stem cell-based suicide gene therapies have attracted much interest because of potent tumor tropism of stem cells and bystander effect. In this current clinical situation, stem cells are promising delivery tool of suicide genes for glioma therapy. Since habitual cigarette smoking still prevails worldwide, we investigated the effect of nicotine on stem cell tropism toward glioma and gap junctional intercellular communication (GJIC) function between glioma and stem cells, both of which are important for suicide gene therapies. Methods: Mouse induced pluripotent stem cell-derived neural stem cells (iPS-NSCs) and human dental pulp mesenchymal stem cells (DPSCs) were used. The effect of nicotine on tumor tropism to glioma-conditioned medium (CM) at a non-cytotoxic concentration was assessed with Matrigel invasion assay. Nicotine effect on GJIC was assessed with the scrape loading/dye transfer (SL/DT) assay for co-culture of glioma and stem cells and the parachute assay among glioma cells using high-content analysis. Results: Tumor tropism of iPS-NSCs toward GL261-CM and DPSCs toward U251-CM was not affected by nicotine (0.1 and 1 µM). Nicotine at the concentrations equivalent to habitual smoking (1 µM) did not affect GJIC of iPS-NSC/GL261 and DPSC/U251 and GJIC among each glioma cells. Conclusions: The study demonstrated that non-cytotoxic concentrations of nicotine did not significantly change the stem cell properties requisite for stem cell-based suicide gene therapy.
Collapse
Affiliation(s)
- Hiroaki Kenmochi
- Department of Neurosurgery, Hamamatsu University School of Medicine , Higashi-ku, Hamamatsu, Japan
| | - Tomohiro Yamasaki
- Department of Neurosurgery, Hamamatsu University School of Medicine , Higashi-ku, Hamamatsu, Japan
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine , Higashi-ku, Hamamatsu, Japan
| | - Tetsuro Sameshima
- Department of Neurosurgery, Hamamatsu University School of Medicine , Higashi-ku, Hamamatsu, Japan
| | - Hiroki Namba
- Department of Neurosurgery, Hamamatsu University School of Medicine , Higashi-ku, Hamamatsu, Japan
| |
Collapse
|
8
|
Shaito A, Saliba J, Husari A, El-Harakeh M, Chhouri H, Hashem Y, Shihadeh A, El-Sabban M. Electronic Cigarette Smoke Impairs Normal Mesenchymal Stem Cell Differentiation. Sci Rep 2017; 7:14281. [PMID: 29079789 PMCID: PMC5660168 DOI: 10.1038/s41598-017-14634-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022] Open
Abstract
Electronic cigarettes (e-cigarettes) are promoted as low-risk alternatives to combustible cigarettes. However, the effects of chronic inhalation of potential toxicants emitted by ecigarettes remain largely unexamined. It is conceivable that smoking-induced chronic diseases result in cellular injury, in the absence of effective repair by stem cells. This study evaluates the effect of cigarette and e-cigarette aerosol extracts on the survival and differentiation of bone marrow-derived mesenchymal stem cells (MSCs). MSC growth and osteogenic differentiation were examined after exposure to smoke extracts. Data revealed detrimental effects of both cigarette and e-cigarette extracts on MSC morphology and growth. Levels and activity of alkaline phosphatase, an osteogenic marker, decreased and induction of osteoblastic differentiation was impaired. Both smoke extracts prevented osteogenic differentiation from progressing, evident by decreased expression of terminal osteogenic markers and mineralization. Elevated levels of reactive oxygen species (ROS) were detected in cells exposed to smoke extracts. Moreover, decreased differentiation potential was concomitant with severe down-regulation of Connexin 43 expression, leading to the loss of gap junction-mediated communication, which together with elevated ROS levels, could explain decreased proliferation and loss of differentiation potential. Hence, e-cigarettes present similar risk as combustible cigarettes with respect to tissue repair impairment.
Collapse
Affiliation(s)
- A Shaito
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | - J Saliba
- Department of Biology, Faculty of Science, Lebanese University, Beirut, Lebanon
| | - A Husari
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - M El-Harakeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - H Chhouri
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Y Hashem
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - A Shihadeh
- Department of Mechanical Engineering, Faculty of Engineering, American University of Beirut, Beirut, Lebanon
| | - M El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
9
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
10
|
Corsini S, Tortora M, Rauti R, Nistri A. Nicotine protects rat hypoglossal motoneurons from excitotoxic death via downregulation of connexin 36. Cell Death Dis 2017; 8:e2881. [PMID: 28617431 PMCID: PMC5520892 DOI: 10.1038/cddis.2017.232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 04/21/2017] [Accepted: 04/26/2017] [Indexed: 01/01/2023]
Abstract
Motoneuron disease including amyotrophic lateral sclerosis may be due, at an early stage, to deficit in the extracellular clearance of the excitatory transmitter glutamate. A model of glutamate-mediated excitotoxic cell death based on pharmacological inhibition of its uptake was used to investigate how activation of neuronal nicotinic receptors by nicotine may protect motoneurons. Hypoglossal motoneurons (HMs) in neonatal rat brainstem slices were exposed to the glutamate uptake blocker DL-threo-β-benzyloxyaspartate (TBOA) that evoked large Ca2+ transients time locked among nearby HMs, whose number fell by about 30% 4 h later. As nicotine or the gap junction blocker carbenoxolone suppressed bursting, we studied connexin 36 (Cx36), which constitutes gap junctions in neurons and found it largely expressed by HMs. Cx36 was downregulated when nicotine or carbenoxolone was co-applied with TBOA. Expression of Cx36 was preferentially observed in cytosolic rather than membrane fractions after nicotine and TBOA, suggesting protein redistribution with no change in synthesis. Nicotine raised the expression of heat shock protein 70 (Hsp70), a protective factor that binds the apoptotic-inducing factor (AIF) whose nuclear translocation is a cause of cell death. TBOA increased intracellular AIF, an effect blocked by nicotine. These results indicate that activation of neuronal nicotinic receptors is an early tool for protecting motoneurons from excitotoxicity and that this process is carried out via the combined decrease in Cx36 activity, overexpression of Hsp70 and fall in AIF translocation. Thus, retarding or inhibiting HM death may be experimentally achieved by targeting one of these processes leading to motoneuron death.
Collapse
Affiliation(s)
- Silvia Corsini
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Maria Tortora
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Rossana Rauti
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Andrea Nistri
- Department of Neuroscience, International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
11
|
Autocrine control of angiogenesis by endogenous acetylcholine in an in vitro model using human endothelial cells: evidence for an autocrine cholinergic system in endothelial cells. J Cardiovasc Pharmacol 2016; 65:508-15. [PMID: 25636069 DOI: 10.1097/fjc.0000000000000221] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We wanted to elucidate whether acetylcholine as the endogenous ligand at cholinoceptors (ChRs) may have effects on angiogenesis and whether they are transduced through muscarinic or nicotinic ChRs. Human umbilical vein endothelial cells were cultured until confluence and thereafter seeded in Matrigel in vitro angiogenesis assays for 18 hours. During the entire cell culture and angiogenesis period, cells were treated with vehicle, eserine (1 μM), in the absence or presence of additional atropine (1 μM) or mecamylamine (1 μM). Finally, the resulting angiogenetic network was investigated histologically. Eserine significantly enhanced acetylcholine formation. When acetylcholine acted through muscarinic ChRs (eserine + mecamylamine), we observed enhanced complexity of the angiogenic network pattern with increased tube length and cell number. In contrast, when acting through nicotinic ChRs (eserine + atropine), we found reduced complexity of pattern with less branches, shorter tubes, and reduced cell number. If acting on both types of ChRs (eserine alone), there were only very small effects. Using α-bungarotoxin, lobeline, and dihydro-β-erythroidine, we also could show that these effects to various degrees involve α7, α3/β2, and α4/β2 n-ChRs. In conclusion, our results support the hypothesis that human umbilical vein endothelial cells possess an autocrine nonneuronal cholinergic system regulating angiogenesic branch formation through the partially opposing effects of n-ChRs and m-ChRs.
Collapse
|
12
|
Yuan D, Sun G, Zhang R, Luo C, Ge M, Luo G, Hei Z. Connexin 43 expressed in endothelial cells modulates monocyte‑endothelial adhesion by regulating cell adhesion proteins. Mol Med Rep 2015; 12:7146-52. [PMID: 26324251 DOI: 10.3892/mmr.2015.4273] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 08/04/2015] [Indexed: 11/05/2022] Open
Abstract
Adhesion between circulating monocytes and vascular endothelial cells is a key initiator of atherosclerosis. In our previous studies, it was demonstrated that the expression of connexin (Cx)43 in monocytes modulates cell adhesion, however, the effects of the expression of Cx43 in endothelial cells remains to be elucidated. Therefore, the present study investigated the role of the expression of Cx43 in endothelial cells in the process of cell adhesion. A total of four different methods with distinct mechanisms were used to change the function and expression of Cx43 channels in human umbilical vein endothelial cells: Cx43 channel inhibitor (oleamide), enhancer (retinoic acid), overexpression of Cx43 by transfection with pcDNA‑Cx43 and knock‑down of the expression of Cx43 by small interfering RNA against Cx43. The results indicated that the upregulation of the expression of Cx43 enhanced monocyte‑endothelial adhesion and this was markedly decreased by downregulation of Cx43. This mechanism was associated with Cx43‑induced expression of vascular cell adhesion molecule‑1 and intercellular cell adhesion molecule‑1. The effects of Cx43 in endothelial cells was independent of Cx37 or Cx40. These experiments suggested that local regulation of endothelial Cx43 expression within the vasculature regulates monocyte‑endothelial adhesion, a critical event in the development of atherosclerosis and other inflammatory pathologies, with baseline adhesion set by the expression of Cx43. This balance may be crucial in controlling leukocyte involvement in inflammatory cascades.
Collapse
Affiliation(s)
- Dongdong Yuan
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Guoliang Sun
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Rui Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Chenfang Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Mian Ge
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Gangjian Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
13
|
Ginkgolide B Inhibits JAM-A, Cx43, and VE-Cadherin Expression and Reduces Monocyte Transmigration in Oxidized LDL-Stimulated Human Umbilical Vein Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:907926. [PMID: 26246869 PMCID: PMC4515296 DOI: 10.1155/2015/907926] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/21/2015] [Indexed: 12/02/2022]
Abstract
Aim. To investigate the effect of ginkgolide B on junction proteins and the reduction of monocyte migration in oxidized low-density lipoprotein- (ox-LDL-) treated endothelial cells. Methods. Human umbilical vein endothelial cells (HUVECs) were used in the present study. Immunofluorescence and Western blot were performed to determine the expression of junctional adhesion molecule-A (JAM-A), connexin 43 (Cx43), and vascular endothelial cadherin (VE-cadherin). Monocyte migration was detected by the Transwell assay. Results. ox-LDL stimulation increased JAM-A expression by 35%, Cx43 expression by 24%, and VE-cadherin expression by 37% in HUVECs. Ginkgolide B (0.2, 0.4, and 0.6 mg/mL) dose-dependently abolished the expression of these junction proteins. The monocyte transmigration experiments showed that the level of monocyte migration was sixfold higher in the ox-LDL-treated group than in the control group. Ginkgolide B (0.6 mg/mL) nearly completely abolished monocyte migration. Both ginkgolide B and LY294002 suppressed Akt phosphorylation and the expression of these junction proteins in ox-LDL-treated endothelial cells. These results suggest that the ginkgolide B-induced inhibition of junction protein expression is associated with blockade of the PI3K/Akt pathway. Conclusion. Ginkgolide B suppressed junction protein expression and reduced monocyte transmigration that was induced by ox-LDL. Ginkgolide B may improve vascular permeability in atherosclerosis.
Collapse
|
14
|
Dhein S, Gaertner C, Georgieff C, Salameh A, Schlegel F, Mohr FW. Effects of isoprenaline on endothelial connexins and angiogenesis in a human endothelial cell culture system. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2015; 388:101-8. [PMID: 25358823 DOI: 10.1007/s00210-014-1059-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 10/10/2014] [Indexed: 10/24/2022]
Abstract
Downregulation of endothelial connexins has been shown to result in impaired angiogenesis. Isoprenaline is known to upregulate Cx43 in cardiomyocytes. Effects of isoprenaline on endothelial connexins are unknown. We wanted to investigate whether isoprenaline might induce upregulation of connexins Cx37, Cx40, or Cx43 in human endothelial cells and whether it may promote angiogenesis. Human umbilical vein endothelial cells (HUVECs) were cultured until confluence (5 days) and subsequently seeded in Matrigel in vitro angiogenesis assays for 18 h. During the entire cell culture and angiogenesis period, cells were treated with vehicle or isoprenaline (100 nM). Finally, the resulting angiogenetic network was investigated (immuno)histologically. Moreover, expression of Cx37, Cx40, and Cx43 was determined by Western blot. In addition, we measured functional intercellular gap junction coupling by dye injection using patch clamp technique. Isoprenaline resulted in significantly enhanced expression of endothelial Cx43 and to a lower degree of Cx40 and Cx37. The number of coupling cells was significantly increased. Regarding angiogenesis, we observed significantly enhanced formation of branches and a higher complexity of the tube networks with more branches/length. Isoprenaline increases endothelial connexin expression and intercellular coupling and promotes tube formation.
Collapse
Affiliation(s)
- Stefan Dhein
- Clinic for Cardiac Surgery, Heart Center Leipzig, University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany,
| | | | | | | | | | | |
Collapse
|
15
|
Nicotine effects on human endothelial intercellular communication via α4β2 and α3β2 nicotinic acetylcholine receptor subtypes. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2012; 385:621-32. [DOI: 10.1007/s00210-012-0738-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 02/01/2012] [Indexed: 11/25/2022]
|
16
|
Gödecke S, Roderigo C, Rose CR, Rauch BH, Gödecke A, Schrader J. Thrombin-induced ATP release from human umbilical vein endothelial cells. Am J Physiol Cell Physiol 2011; 302:C915-23. [PMID: 22159088 DOI: 10.1152/ajpcell.00283.2010] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ATP and its degradation products play an important role as signaling molecules in the vascular system, and endothelial cells are considered to be an important source of nucleotide release. To investigate the mechanism and physiological significance of endothelial ATP release, we compared different pharmacological stimuli for their ability to evoke ATP release from first passage cultivated human umbilical vein endothelial cells (HUVECs). Agonists known to increase intracellular Ca(2+) levels (A23187, histamine, thrombin) induced a stable, non-lytic ATP release. Since thrombin proved to be the most robust and reproducible stimulus, the molecular mechanism of thrombin-mediated ATP release from HUVECs was further investigated. ATP rapidly increased with thrombin (1 U/ml) and reached a steady-state level after 4 min. Loading the cells with BAPTA-AM to capture intracellular calcium suppressed ATP release. The thrombin-specific, protease-activated receptor 1 (PAR-1)-specific agonist peptide TFLLRN (10 μM) fully mimicked thrombin action on ATP release. To identify the nature of the ATP-permeable pathway, we tested various inhibitors of potential ATP channels for their ability to inhibit the thrombin response. Carbenoxolone, an inhibitor of connexin hemichannels and pannexin channels, as well as Gd(3+) were highly effective in blocking the thrombin-mediated ATP release. Specifically targeting connexin43 (Cx43) and pannexin1 (Panx1) revealed that reducing Panx1 expression significantly reduced ATP release, while downregulating Cx43 was ineffective. Our study demonstrates that thrombin at physiological concentrations is a potent stimulus of endothelial ATP release involving PAR-1 receptor activation and intracellular calcium mobilization. ATP is released by a carbenoxolone- and Gd(3+)- sensitive pathway, most likely involving Panx1 channels.
Collapse
Affiliation(s)
- Stefanie Gödecke
- Department of Cardiovascular Physiology, Heinrich-Heine-University, Düsseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
17
|
Gärtner C, Ziegelhöffer B, Kostelka M, Stepan H, Mohr FW, Dhein S. Knock-down of endothelial connexins impairs angiogenesis. Pharmacol Res 2011; 65:347-57. [PMID: 22154844 DOI: 10.1016/j.phrs.2011.11.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 11/22/2011] [Accepted: 11/22/2011] [Indexed: 02/07/2023]
Abstract
Connexins (Cx) are suggested to play important roles in growth and differentiation. Aim of our study was to investigate the role of endothelial Cx in the angiogenic process. Several parameters of angiogenesis were assessed in 18 h Matrigel in vitro angiogenesis assays with human umbilical vein endothelial cells (HUVEC). Prior to culture on Matrigel cells were treated with nicotine or the gap junction inhibitor palmitoleic acid (PA), or siRNA-knock-down of either Cx37, Cx40 or Cx43 was performed. Changes in Cx expression and their effects on gap-junctional communication were investigated using immunofluorescence microscopy, Western blot and Lucifer Yellow dye transfer. Knock-down of each Cx-isoform significantly reduced the amount of specific Cx protein in HUVEC. Cx-knock-down as well as treatment with PA impaired intercellular communication via gap junctions and diminished significantly the number of capillary branches. Knock-down of Cx43 and Cx40 or treatment with PA reduced complexity pattern in the angiogenesis assay. Nicotine significantly reduced expression of Cx43 and Cx37 as well as average length of capillary branches, number of branches and pattern in the Matrigel assay. We can conclude that connexins are involved in angiogenesis, in particular in branch formation. This can partly explain the changes in angiogenesis seen under nicotine treatment.
Collapse
Affiliation(s)
- Christiane Gärtner
- Department of Cardiac Surgery, Heart Centre Leipzig, University of Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Down-regulation of connexin43 gap junction by serum deprivation in human endothelial cells was improved by (-)-Epigallocatechin gallate via ERK MAP kinase pathway. Biochem Biophys Res Commun 2010; 404:217-22. [PMID: 21110950 DOI: 10.1016/j.bbrc.2010.11.096] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/22/2010] [Indexed: 11/23/2022]
Abstract
Intercellular communication through gap junctions (GJIC) plays an essential role in maintaining the functional integrity of vascular endothelium. Despite emerging evidence suggests that (-)-Epigallocatechin gallate (EGCG) may improve endothelial function. However, its effect on Cx43 gap junction in endothelial cells remains unexplored. Here we investigated the effect of EGCG on connexin43 (Cx43) gap junction in endothelial cells. The levels of Cx43 protein in human umbilical vein endothelial cells (HUVECs) cultured under serum-deprivation 48 h decreased about 50%, accompanied by decreased GJIC. This reduction can be reversed by treatments with EGCG. In addition, EGCG activated ERK, P38, and JNK mitogen-activated protein kinases (MAPKs), which were supposed to participate in the regulation of Cx43. A MEK inhibitor PD98059, but not SB203580 (a p38 kinase inhibitor) or SP600125 (a JNK kinase inhibitor), abolished the effects of EGCG on Cx43 expression and GJIC. Moreover, although both Akt and eNOS phosphorylation were time-dependently augmented by EGCG, neither PI3K inhibitor LY294002 nor eNOS inhibitor L-NAME blocked the effects of EGCG on Cx43 gap junctions. Thus, EGCG attenuated Cx43 down-regulation and impaired GJIC induced by serum deprivation, ERK MAPK Signal transduction pathway appears to be involved in these processes.
Collapse
|
19
|
Zheng YF, Dai DZ, Dai Y. NaHS ameliorates diabetic vascular injury by correcting depressed connexin 43 and 40 in the vasculature in streptozotocin-injected rats. J Pharm Pharmacol 2010; 62:883-9. [DOI: 10.1211/jpp.62.05.0009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
20
|
Inhibitory Effect of Ghrelin on Nicotine-induced VCAM-1 Expression in Human Umbilical Vein Endothelial Cells. J Cardiovasc Pharmacol 2009; 53:241-5. [DOI: 10.1097/fjc.0b013e31819c74dc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Yang G, Zhang A, Xu LX. Experimental study of intracellular ice growth in human umbilical vein endothelial cells. Cryobiology 2008; 58:96-102. [PMID: 18950613 DOI: 10.1016/j.cryobiol.2008.10.123] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 07/22/2008] [Accepted: 10/01/2008] [Indexed: 10/21/2022]
Abstract
Study of the intracellular ice formation (IIF) and growth is essential to the mechanistic understanding of cellular damage through freezing. In the aid of high speed and high resolution cryo-imaging technology, the transient intracellular ice formation and growth processes of the attached human umbilical vein endothelial cells (HUVEC) were successfully captured during freezing. It was found that the intracellular ice nucleation site was on the cell membrane closer to the nucleus. The ice growth was directional and toward the nucleus, which covered the whole nucleus before growing into the cytoplasm. The crystal growth rate in the nucleus was much larger than that in the cytoplasm, and its morphology was influenced by the cooling rate. During the thawing process, small crystals fused into larger ones inside the nucleus. Moreover, the cumulative fraction of the HUVEC with IIF was mainly dependent on the cooling rate not the confluence of the cells attached.
Collapse
Affiliation(s)
- Geer Yang
- Med-X Research Institute, Shanghai Jiao Tong University, PR China; Department of Biomedical Engineering, School of Life Science and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai 200240, PR China
| | - Aili Zhang
- Med-X Research Institute, Shanghai Jiao Tong University, PR China; Department of Biomedical Engineering, School of Life Science and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai 200240, PR China
| | - Lisa X Xu
- Med-X Research Institute, Shanghai Jiao Tong University, PR China; Department of Biomedical Engineering, School of Life Science and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai 200240, PR China.
| |
Collapse
|