1
|
Wei L, Aitchison JD, Kaushansky A, Mast FD. Systems-level reconstruction of kinase phosphosignaling networks regulating endothelial barrier integrity using temporal data. NPJ Syst Biol Appl 2024; 10:134. [PMID: 39548089 DOI: 10.1038/s41540-024-00468-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Phosphosignaling networks control cellular processes. We built kinase-mediated regulatory networks elicited by thrombin stimulation of brain endothelial cells using two computational strategies: Temporal Pathway Synthesizer (TPS), which uses phosphoproteomics data as input, and Temporally REsolved KInase Network Generation (TREKING), which uses kinase inhibitor screens. TPS and TREKING predicted overlapping barrier-regulatory kinases connected with unique network topology. Each strategy effectively describes regulatory signaling networks and is broadly applicable across biological systems.
Collapse
Affiliation(s)
- Ling Wei
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA.
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98105, USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98105, USA
| | - Alexis Kaushansky
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98105, USA
- Department of Global Health, University of Washington, Seattle, WA, 98105, USA
| | - Fred D Mast
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, 98105, USA.
| |
Collapse
|
2
|
Wei L, Aitchison JD, Kaushansky A, Mast FD. Systems-level reconstruction of kinase phosphosignaling networks regulating endothelial barrier integrity using temporal data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606198. [PMID: 39149238 PMCID: PMC11326140 DOI: 10.1101/2024.08.01.606198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Phosphosignaling networks control cellular processes. We built kinase-mediated regulatory networks elicited by thrombin stimulation of brain endothelial cells using two computational strategies: Temporal Pathway Synthesizer (TPS), which uses phosphoproetiomics data as input, and Temporally REsolved KInase Network Generation (TREKING), which uses kinase inhibitor screens. TPS and TREKING predicted overlapping barrier-regulatory kinases connected with unique network topology. Each strategy effectively describes regulatory signaling networks and is broadly applicable across biological systems.
Collapse
|
3
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [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] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
4
|
Prüschenk S, Majer M, Schlossmann J. Novel Functional Features of cGMP Substrate Proteins IRAG1 and IRAG2. Int J Mol Sci 2023; 24:9837. [PMID: 37372987 DOI: 10.3390/ijms24129837] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
The inositol triphosphate-associated proteins IRAG1 and IRAG2 are cGMP kinase substrate proteins that regulate intracellular Ca2+. Previously, IRAG1 was discovered as a 125 kDa membrane protein at the endoplasmic reticulum, which is associated with the intracellular Ca2+ channel IP3R-I and the PKGIβ and inhibits IP3R-I upon PKGIβ-mediated phosphorylation. IRAG2 is a 75 kDa membrane protein homolog of IRAG1 and was recently also determined as a PKGI substrate. Several (patho-)physiological functions of IRAG1 and IRAG2 were meanwhile elucidated in a variety of human and murine tissues, e.g., of IRAG1 in various smooth muscles, heart, platelets, and other blood cells, of IRAG2 in the pancreas, heart, platelets, and taste cells. Hence, lack of IRAG1 or IRAG2 leads to diverse phenotypes in these organs, e.g., smooth muscle and platelet disorders or secretory deficiency, respectively. This review aims to highlight the recent research regarding these two regulatory proteins to envision their molecular and (patho-)physiological tasks and to unravel their functional interplay as possible (patho-)physiological counterparts.
Collapse
Affiliation(s)
- Sally Prüschenk
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| | - Michael Majer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| | - Jens Schlossmann
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| |
Collapse
|
5
|
Engelbrecht E, Kooistra T, Knipe RS. The Vasculature in Pulmonary Fibrosis. CURRENT TISSUE MICROENVIRONMENT REPORTS 2022; 3:83-97. [PMID: 36712832 PMCID: PMC9881604 DOI: 10.1007/s43152-022-00040-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 02/02/2023]
Abstract
Purpose of Review The current paradigm of idiopathic pulmonary fibrosis (IPF) pathogenesis involves recurrent injury to a sensitive alveolar epithelium followed by impaired repair responses marked by fibroblast activation and deposition of extracellular matrix. Multiple cell types are involved in this response with potential roles suggested by advances in single-cell RNA sequencing and lung developmental biology. Notably, recent work has better characterized the cell types present in the pulmonary endothelium and identified vascular changes in patients with IPF. Recent Findings Lung tissue from patients with IPF has been examined at single-cell resolution, revealing reductions in lung capillary cells and expansion of a population of vascular cells expressing markers associated with bronchial endothelium. In addition, pre-clinical models have demonstrated a fundamental role for aging and vascular permeability in the development of pulmonary fibrosis. Summary Mounting evidence suggests that the endothelium undergoes changes in the context of fibrosis, and these changes may contribute to the development and/or progression of pulmonary fibrosis. Additional studies will be needed to further define the functional role of these vascular changes.
Collapse
Affiliation(s)
| | - Tristan Kooistra
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Rachel S. Knipe
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Shimauchi T, Numaga-Tomita T, Kato Y, Morimoto H, Sakata K, Matsukane R, Nishimura A, Nishiyama K, Shibuta A, Horiuchi Y, Kurose H, Kim SG, Urano Y, Ohshima T, Nishida M. A TRPC3/6 Channel Inhibitor Promotes Arteriogenesis after Hind-Limb Ischemia. Cells 2022; 11:cells11132041. [PMID: 35805125 PMCID: PMC9266111 DOI: 10.3390/cells11132041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/04/2022] Open
Abstract
Retarded revascularization after progressive occlusion of large conductance arteries is a major cause of bad prognosis for peripheral artery disease (PAD). However, pharmacological treatment for PAD is still limited. We previously reported that suppression of transient receptor potential canonical (TRPC) 6 channel activity in vascular smooth muscle cells (VSMCs) facilitates VSMC differentiation without affecting proliferation and migration. In this study, we found that 1-benzilpiperadine derivative (1-BP), a selective inhibitor for TRPC3 and TRPC6 channel activities, induced VSMC differentiation. 1-BP-treated mice showed increased capillary arterialization and improvement of peripheral circulation and skeletal muscle mass after hind-limb ischemia (HLI) in mice. 1-BP had no additive effect on the facilitation of blood flow recovery after HLI in TRPC6-deficient mice, suggesting that suppression of TRPC6 underlies facilitation of the blood flow recovery by 1-BP. 1-BP also improved vascular nitric oxide bioavailability and blood flow recovery after HLI in hypercholesterolemic mice with endothelial dysfunction, suggesting the retrograde interaction from VSMCs to endothelium. These results suggest that 1-BP becomes a potential seed for PAD treatments that target vascular TRPC6 channels.
Collapse
Affiliation(s)
- Tsukasa Shimauchi
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8585, Japan; (T.S.); (T.N.-T.); (A.N.)
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Takuro Numaga-Tomita
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8585, Japan; (T.S.); (T.N.-T.); (A.N.)
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Molecular Pharmacology, Shinshu University School of Medicine and Health Sciences, Matsumoto 390-8621, Japan
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Hiroyuki Morimoto
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Kosuke Sakata
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Ryosuke Matsukane
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8585, Japan; (T.S.); (T.N.-T.); (A.N.)
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8585, Japan
| | - Kazuhiro Nishiyama
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Atsushi Shibuta
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Yutoku Horiuchi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Hitoshi Kurose
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Sang Geon Kim
- College of Pharmacy, Dongguk University-Seoul, Goyang-si 10326, Gyeonggi-Do, Korea;
| | - Yasuteru Urano
- Laboratory of Chemistry and Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan;
| | - Takashi Ohshima
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
| | - Motohiro Nishida
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8585, Japan; (T.S.); (T.N.-T.); (A.N.)
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.K.); (H.M.); (K.S.); (R.M.); (K.N.); (A.S.); (Y.H.); (H.K.); (T.O.)
- Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8585, Japan
- Correspondence: ; Tel./Fax: +81-92-642-6556
| |
Collapse
|
7
|
Regional Diversities in Fibrogenesis Weighed as a Key Determinant for Atrial Arrhythmogenesis. Biomedicines 2021; 9:biomedicines9121900. [PMID: 34944715 PMCID: PMC8698388 DOI: 10.3390/biomedicines9121900] [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: 11/21/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 11/18/2022] Open
Abstract
Atrial fibrosis plays a key role in atrial myopathy, resulting in the genesis of atrial fibrillation (AF). The abnormal distribution of fibrotic tissue, electrical coupling, paracrine interactions, and biomechanical–electrical interactions have all been suggested as causes of fibrosis-related arrhythmogenesis. Moreover, the regional difference in fibrogenesis, specifically the left atrium (LA) exhibiting a higher arrhythmogenesis and level of fibrosis than the right atrium (RA) in AF, is a key contributor to atrial arrhythmogenesis. LA fibroblasts have greater profibrotic cellular activities than RA fibroblasts, but knowledge about the regional diversity of atrial regional fibrogenesis remains limited. This article provides a comprehensive review of research findings on the association between fibrogenesis and arrhythmogenesis from laboratory to clinical evidence and updates the current understanding of the potential mechanism underlying the difference in fibrogenesis between the LA and RA.
Collapse
|
8
|
Cordes S, Mokhtari Z, Bartosova M, Mertlitz S, Riesner K, Shi Y, Mengwasser J, Kalupa M, McGeary A, Schleifenbaum J, Schrezenmeier J, Bullinger L, Diaz-Ricart M, Palomo M, Carrreras E, Beutel G, Schmitt CP, Beilhack A, Penack O. Endothelial damage and dysfunction in acute graft-versus-host disease. Haematologica 2021; 106:2147-2160. [PMID: 32675225 PMCID: PMC8327719 DOI: 10.3324/haematol.2020.253716] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Indexed: 12/20/2022] Open
Abstract
Clinical studies suggested that endothelial dysfunction and damage could be involved in the development and severity of acute graft-versus-host disease (aGVHD). Accordingly, we found increased percentage of apoptotic Casp3+ blood vessels in duodenal and colonic mucosa biopsies of patients with severe aGVHD. In murine experimental aGVHD, we detected severe microstructural endothelial damage and reduced endothelial pericyte coverage accompanied by reduced expression of endothelial tight junction proteins leading to increased endothelial leakage in aGVHD target organs. During intestinal aGVHD, colonic vasculature structurally changed, reflected by increased vessel branching and vessel diameter. Because recent data demonstrated an association of endothelium-related factors and steroid refractory aGVHD (SR-aGVHD), we analyzed human biopsies and murine tissues from SR-aGVHD. We found extensive tissue damage but low levels of alloreactive T cell infiltration in target organs, providing the rationale for T-cell independent SR-aGVHD treatment strategies. Consequently, we tested the endothelium-protective PDE5 inhibitor sildenafil, which reduced apoptosis and improved metabolic activity of endothelial cells in vitro. Accordingly, sildenafil treatment improved survival and reduced target organ damage during experimental SR-aGVHD. Our results demonstrate extensive damage, structural changes, and dysfunction of the vasculature during aGVHD. Therapeutic intervention by endothelium-protecting agents is an attractive approach for SR-aGVHD complementing current anti-inflammatory treatment options.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Shi
- Charité Universitätsmedizin Berlin
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Hofmann F. The cGMP system: components and function. Biol Chem 2021; 401:447-469. [PMID: 31747372 DOI: 10.1515/hsz-2019-0386] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022]
Abstract
The cyclic guanosine monophosphate (cGMP) signaling system is one of the most prominent regulators of a variety of physiological and pathophysiological processes in many mammalian and non-mammalian tissues. Targeting this pathway by increasing cGMP levels has been a very successful approach in pharmacology as shown for nitrates, phosphodiesterase (PDE) inhibitors and stimulators of nitric oxide-guanylyl cyclase (NO-GC) and particulate GC (pGC). This is an introductory review to the cGMP signaling system intended to introduce those readers to this system, who do not work in this area. This article does not intend an in-depth review of this system. Signal transduction by cGMP is controlled by the generating enzymes GCs, the degrading enzymes PDEs and the cGMP-regulated enzymes cyclic nucleotide-gated ion channels, cGMP-dependent protein kinases and cGMP-regulated PDEs. Part A gives a very concise introduction to the components. Part B gives a very concise introduction to the functions modulated by cGMP. The article cites many recent reviews for those who want a deeper insight.
Collapse
Affiliation(s)
- Franz Hofmann
- Pharmakologisches Institut, Technische Universität München, Biedersteiner Str. 29, D-80802 München, Germany
| |
Collapse
|
10
|
Chi Y, Liu X, Chai J. A narrative review of changes in microvascular permeability after burn. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:719. [PMID: 33987417 PMCID: PMC8106041 DOI: 10.21037/atm-21-1267] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective We aimed to review and discuss some of the latest research results related to post-burn pathophysiological changes and provide some clues for future study. Background Burns are one of the most common and serious traumas and consist of a series of pathophysiological changes of thermal injury. Accompanied by thermal damage to skin and soft tissues, inflammatory mediators are released in large quantities. Changes in histamine, bradykinin, and cytokines such as vascular endothelial growth factor (VEGF), metabolic factors such as adenosine triphosphate (ATP), and activated neutrophils all affect the body’s vascular permeability. Methods We searched articles with subject words “microvascular permeability”, “burn” “endothelium”, and “endothelial barrier” in PubMed in English published from the beginning of database to Dec, 2020. Conclusions The essence of burn shock is the rapid and extensive fluid transfer in burn and non-burn tissue. After severe burns, the local and systemic vascular permeability increase, causing intravascular fluid extravasation, leading to a progressive decrease in effective circulation volume, an increase in systemic vascular resistance, a decrease in cardiac output, peripheral tissue edema, multiple organ failure, and even death. There are many cells, tissues, mediators and structures involved in the pathophysiological process of the damage to vascular permeability. Ulinastatin is a promising agent for this problem.
Collapse
Affiliation(s)
- Yunfei Chi
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| | - Xiangyu Liu
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| | - Jiake Chai
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Shen Y, Ding W. Therapeutic Hypothermia Mitigates the Sepsis-Increased Permeability in EA. hy926 Cells by Preserving Rap1 Expression. Ther Hypothermia Temp Manag 2021; 11:201-207. [PMID: 33709787 DOI: 10.1089/ther.2020.0037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To determine the effect and potential mechanisms of therapeutic hypothermia (TH) on the permeability of septic cells. Human EA. hy926 cells were transfected with, or without, control or ras-proximate-1 (Rap1)-specific siRNA and treated with 2 μg/mL of lipopolysaccharide (LPS). The cells were cultured in normal temperature (NT) or a temporary TH for 10 hours. The cellular permeability of each group of cells was determined by transwell permeability assay. The relative levels of Rap1, RhoA (a small GTP enzyme of the Rho family), VE-cadherin expression, and myosin light chain (MLC) phosphorylation were quantified by Western blot and immunofluorescent assays. Compared with the control group, LPS stimulation increased cellular permeability in EA. hy926 cells under an NT condition, but significantly mitigated by TH. The effect of TH decreased after Rap1 silencing. Furthermore, LPS upregulated RhoA expression and MLC phosphorylation, but reduced Rap1 and VE-cadherin expression, which were also enhanced by Rap1 silencing, but significantly mitigated by TH. Immunofluorescent analyses indicated that LPS significantly increased phosphorylated MLC, but decreased VE-cadherin expression, which were further deteriorated by Rap1 silencing, but significantly mitigated by TH in EA. hy926 cells. TH significantly mitigated the sepsis-increased permeability of EA. hy926 cells by enhancing the Rap1 expression to attenuate the RhoA/MLC signaling.
Collapse
Affiliation(s)
- Yuehong Shen
- Department of Burns, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wu Ding
- Department of Emergency Medicine, Second Hospital of Zhejiang University School of Medicine and Research Institute of Emergency Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Bellis A, Mauro C, Barbato E, Trimarco B, Morisco C. The Rationale for Angiotensin Receptor Neprilysin Inhibitors in a Multi-Targeted Therapeutic Approach to COVID-19. Int J Mol Sci 2020; 21:ijms21228612. [PMID: 33203141 PMCID: PMC7696732 DOI: 10.3390/ijms21228612] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) disease (COVID-19) determines the angiotensin converting enzyme 2 (ACE2) down-regulation and related decrease in angiotensin II degradation. Both these events trigger “cytokine storm” leading to acute lung and cardiovascular injury. A selective therapy for COVID-19 has not yet been identified. Clinical trials with remdesivir gave discordant results. Thus, healthcare systems have focused on “multi-targeted” therapeutic strategies aiming at relieving systemic inflammation and thrombotic complications. No randomized clinical trial has demonstrated the efficacy of renin angiotensin system antagonists in reducing inflammation related to COVID-19. Dexamethasone and tocilizumab showed encouraging data, but their use needs to be further validated. The still-controversial efficacy of these treatments highlighted the importance of organ injury prevention in COVID-19. Neprilysin (NEP) might be an interesting target for this purpose. NEP expression is increased by cytokines on lung fibroblasts surface. NEP activity is elevated in acute respiratory distress syndrome and it is conceivable that it is also high in COVID-19. NEP is implicated in the degradation of natriuretic peptides, bradykinin, substance P, adrenomedullin, and apelin that account for prevention of organ injury. Thus, NEP/angiotensin receptor type 1 (AT1R) inhibitor sacubitril/valsartan (SAC/VAL) may increase levels of these molecules and block AT1Rs required for ACE2 endocytosis in SARS-CoV-2 infection. Moreover, SAC/VAL has a positive impact on acute heart failure that is very frequently observed in deceased COVID-19 patients. The current review aims to summarize actual therapeutic strategies for COVID-19 and to examine the data supporting the potential benefits of SAC/VAL in COVID-19 treatment.
Collapse
Affiliation(s)
- Alessandro Bellis
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica-Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy; (A.B.); (C.M.)
| | - Ciro Mauro
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica-Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy; (A.B.); (C.M.)
| | - Emanuele Barbato
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (E.B.); (B.T.)
| | - Bruno Trimarco
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (E.B.); (B.T.)
| | - Carmine Morisco
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (E.B.); (B.T.)
- Correspondence: ; Tel.: +39-081-746-2253; Fax: +39-081-746-2256
| |
Collapse
|
13
|
Sadek MS, Cachorro E, El-Armouche A, Kämmerer S. Therapeutic Implications for PDE2 and cGMP/cAMP Mediated Crosstalk in Cardiovascular Diseases. Int J Mol Sci 2020; 21:E7462. [PMID: 33050419 PMCID: PMC7590001 DOI: 10.3390/ijms21207462] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Phosphodiesterases (PDEs) are the principal superfamily of enzymes responsible for degrading the secondary messengers 3',5'-cyclic nucleotides cAMP and cGMP. Their refined subcellular localization and substrate specificity contribute to finely regulate cAMP/cGMP gradients in various cellular microdomains. Redistribution of multiple signal compartmentalization components is often perceived under pathological conditions. Thereby PDEs have long been pursued as therapeutic targets in diverse disease conditions including neurological, metabolic, cancer and autoimmune disorders in addition to numerous cardiovascular diseases (CVDs). PDE2 is a unique member of the broad family of PDEs. In addition to its capability to hydrolyze both cAMP and cGMP, PDE2 is the sole isoform that may be allosterically activated by cGMP increasing its cAMP hydrolyzing activity. Within the cardiovascular system, PDE2 serves as an integral regulator for the crosstalk between cAMP/cGMP pathways and thereby may couple chronically adverse augmented cAMP signaling with cardioprotective cGMP signaling. This review provides a comprehensive overview of PDE2 regulatory functions in multiple cellular components within the cardiovascular system and also within various subcellular microdomains. Implications for PDE2- mediated crosstalk mechanisms in diverse cardiovascular pathologies are discussed highlighting the prospective use of PDE2 as a potential therapeutic target in cardiovascular disorders.
Collapse
Affiliation(s)
| | | | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| | - Susanne Kämmerer
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| |
Collapse
|
14
|
Bellis A, Mauro C, Barbato E, Di Gioia G, Sorriento D, Trimarco B, Morisco C. The Rationale of Neprilysin Inhibition in Prevention of Myocardial Ischemia-Reperfusion Injury during ST-Elevation Myocardial Infarction. Cells 2020; 9:cells9092134. [PMID: 32967374 PMCID: PMC7565478 DOI: 10.3390/cells9092134] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
During the last three decades, timely myocardial reperfusion using either thrombolytic therapy or primary percutaneous intervention (pPCI) has allowed amazing improvements in outcomes with a more than halving in 1-year ST-elevation myocardial infarction (STEMI) mortality. However, mortality and left ventricle (LV) remodeling remain substantial in these patients. As such, novel therapeutic interventions are required to reduce myocardial infarction size, preserve LV systolic function, and improve survival in reperfused-STEMI patients. Myocardial ischemia-reperfusion injury (MIRI) prevention represents the main goal to reach in order to reduce STEMI mortality. There is currently no effective therapy for MIRI prevention in STEMI patients. A significant reason for the weak and inconsistent results obtained in this field may be the presence of multiple, partially redundant, mechanisms of cell death during ischemia-reperfusion, whose relative importance may depend on the conditions. Therefore, it is always more recognized that it is important to consider a "multi-targeted cardioprotective therapy", defined as an additive or synergistic cardioprotective agents or interventions directed to distinct targets with different timing of application (before, during, or after pPCI). Given that some neprilysin (NEP) substrates (natriuretic peptides, angiotensin II, bradykinin, apelins, substance P, and adrenomedullin) exert a cardioprotective effect against ischemia-reperfusion injury, it is conceivable that antagonism of proteolytic activity by this enzyme may be considered in a multi-targeted strategy for MIRI prevention. In this review, by starting from main pathophysiological mechanisms promoting MIRI, we discuss cardioprotective effects of NEP substrates and the potential benefit of NEP pharmacological inhibition in MIRI prevention.
Collapse
Affiliation(s)
- Alessandro Bellis
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica—Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy;
| | - Ciro Mauro
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica—Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy;
| | - Emanuele Barbato
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Giuseppe Di Gioia
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Cardiac Catheterization Laboratory, Montevergine Clinic, 83013 Mercogliano (AV), Italy
| | - Daniela Sorriento
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Bruno Trimarco
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Carmine Morisco
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Correspondence: ; Tel.: +39-081-746-2253; Fax: +39-081-746-2256
| |
Collapse
|
15
|
Formoso K, Susperreguy S, Freichel M, Birnbaumer L. RNA-seq analysis reveals TRPC genes to impact an unexpected number of metabolic and regulatory pathways. Sci Rep 2020; 10:7227. [PMID: 32350291 PMCID: PMC7190874 DOI: 10.1038/s41598-020-61177-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
The seven-member transient receptor potential canonical genes (TRPC1-7) encode cation channels linked to several human diseases. There is little understanding of the participation of each TRPC in each pathology, considering functional redundancy. Also, most of the inhibitors available are not specific. Thus, we developed mice that lack all of the TRPCs and performed a transcriptome analysis in eight tissues. The aim of this research was to address the impact of the absence of all TRPC channels on gene expression. We obtained a total of 4305 differentially expressed genes (DEGs) in at least one tissue where spleen showed the highest number of DEGs (1371). Just 21 genes were modified in all the tissues. Performing a pathway enrichment analysis, we found that many important signaling pathways were modified in more than one tissue, including PI3K (phosphatidylinositol 3-kinase/protein kinase-B) signaling pathway, cytokine-cytokine receptor interaction, extracellular matrix (ECM)-receptor interaction and circadian rhythms. We describe for the first time the changes at the transcriptome level due to the lack of all TRPC proteins in a mouse model and provide a starting point to understand the function of TRPC channels and their possible roles in pathologies.
Collapse
Affiliation(s)
- Karina Formoso
- Institute for Biomedical Research (BIOMED UCA-CONICET). School of Medical Sciences, Catholic University of Argentina (UCA), Buenos Aires, C1107AFF, Argentina
| | - Sebastian Susperreguy
- Institute for Biomedical Research (BIOMED UCA-CONICET). School of Medical Sciences, Catholic University of Argentina (UCA), Buenos Aires, C1107AFF, Argentina
| | - Marc Freichel
- Institute of Physiology and Pathophysiology, Heidelberg University, 69120, Heidelberg, Germany
| | - Lutz Birnbaumer
- Institute for Biomedical Research (BIOMED UCA-CONICET). School of Medical Sciences, Catholic University of Argentina (UCA), Buenos Aires, C1107AFF, Argentina. .,Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, North Carolina, 27709, USA.
| |
Collapse
|
16
|
Jackson WF. Introduction to ion channels and calcium signaling in the microcirculation. CURRENT TOPICS IN MEMBRANES 2020; 85:1-18. [PMID: 32402636 DOI: 10.1016/bs.ctm.2020.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The microcirculation is the network of feed arteries, arterioles, capillaries and venules that supply and drain blood from every tissue and organ in the body. It is here that exchange of heat, oxygen, carbon dioxide, nutrients, hormones, water, cytokines, and immune cells takes place; essential functions necessary to maintenance of homeostasis throughout the life span. This chapter will outline the structure and function of each microvascular segment highlighting the critical roles played by ion channels in the microcirculation. Feed arteries upstream from the true microcirculation and arterioles within the microcirculation contribute to systemic vascular resistance and blood pressure control. They also control total blood flow to the downstream microcirculation with arterioles being responsible for distribution of blood flow within a tissue or organ dependent on the metabolic needs of the tissue. Terminal arterioles control blood flow and blood pressure to capillary units, the primary site of diffusional exchange between blood and tissues due to their large surface area. Venules collect blood from capillaries and are important sites for fluid exchange and immune cell trafficking. Ion channels in microvascular smooth muscle cells, endothelial cells and pericytes importantly contribute to all of these functions through generation of intracellular Ca2+ and membrane potential signals in these cells.
Collapse
Affiliation(s)
- William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
17
|
Wang H, Cheng X, Tian J, Xiao Y, Tian T, Xu F, Hong X, Zhu MX. TRPC channels: Structure, function, regulation and recent advances in small molecular probes. Pharmacol Ther 2020; 209:107497. [PMID: 32004513 DOI: 10.1016/j.pharmthera.2020.107497] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/14/2020] [Indexed: 02/08/2023]
Abstract
Transient receptor potential canonical (TRPC) channels constitute a group of receptor-operated calcium-permeable nonselective cation channels of the TRP superfamily. The seven mammalian TRPC members, which can be further divided into four subgroups (TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7) based on their amino acid sequences and functional similarities, contribute to a broad spectrum of cellular functions and physiological roles. Studies have revealed complexity of their regulation involving several components of the phospholipase C pathway, Gi and Go proteins, and internal Ca2+ stores. Recent advances in cryogenic electron microscopy have provided several high-resolution structures of TRPC channels. Growing evidence demonstrates the involvement of TRPC channels in diseases, particularly the link between genetic mutations of TRPC6 and familial focal segmental glomerulosclerosis. Because TRPCs were discovered by the molecular identity first, their pharmacology had lagged behind. This is rapidly changing in recent years owning to great efforts from both academia and industry. A number of potent tool compounds from both synthetic and natural products that selective target different subtypes of TRPC channels have been discovered, including some preclinical drug candidates. This review will cover recent advancements in the understanding of TRPC channel regulation, structure, and discovery of novel TRPC small molecular probes over the past few years, with the goal of facilitating drug discovery for the study of TRPCs and therapeutic development.
Collapse
Affiliation(s)
- Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaoding Cheng
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yuling Xiao
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Tian Tian
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Fuchun Xu
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Xuechuan Hong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China; Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China.
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
18
|
Nishida M, Tanaka T, Mangmool S, Nishiyama K, Nishimura A. Canonical Transient Receptor Potential Channels and Vascular Smooth Muscle Cell Plasticity. J Lipid Atheroscler 2020; 9:124-139. [PMID: 32821726 PMCID: PMC7379077 DOI: 10.12997/jla.2020.9.1.124] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) play a pivotal role in the stability and tonic regulation of vascular homeostasis. VSMCs can switch back and forth between highly proliferative (synthetic) and fully differentiated (contractile) phenotypes in response to changes in the vessel environment. Abnormal phenotypic switching of VSMCs is a distinctive characteristic of vascular disorders, including atherosclerosis, pulmonary hypertension, stroke, and peripheral artery disease; however, how the control of VSMC phenotypic switching is dysregulated under pathological conditions remains obscure. Canonical transient receptor potential (TRPC) channels have attracted attention as a key regulator of pathological phenotype switching in VSMCs. Several TRPC subfamily member proteins—especially TRPC1 and TRPC6—are upregulated in pathological VSMCs, and pharmacological inhibition of TRPC channel activity has been reported to improve hypertensive vascular remodeling in rodents. This review summarizes the current understanding of the role of TRPC channels in cardiovascular plasticity, including our recent finding that TRPC6 participates in aberrant VSMC phenotype switching under ischemic conditions, and discusses the therapeutic potential of TRPC channels.
Collapse
Affiliation(s)
- Motohiro Nishida
- National Institute for Physiological Sciences and Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan.,Center for Novel Science Initiatives (CNSI), NINS, Tokyo 105-0001, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomohiro Tanaka
- National Institute for Physiological Sciences and Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan.,Center for Novel Science Initiatives (CNSI), NINS, Tokyo 105-0001, Japan
| | | | - Kazuhiro Nishiyama
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akiyuki Nishimura
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
19
|
Post-Translational Modification and Natural Mutation of TRPC Channels. Cells 2020; 9:cells9010135. [PMID: 31936014 PMCID: PMC7016788 DOI: 10.3390/cells9010135] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Transient Receptor Potential Canonical (TRPC) channels are homologues of Drosophila TRP channel first cloned in mammalian cells. TRPC family consists of seven members which are nonselective cation channels with a high Ca2+ permeability and are activated by a wide spectrum of stimuli. These channels are ubiquitously expressed in different tissues and organs in mammals and exert a variety of physiological functions. Post-translational modifications (PTMs) including phosphorylation, N-glycosylation, disulfide bond formation, ubiquitination, S-nitrosylation, S-glutathionylation, and acetylation play important roles in the modulation of channel gating, subcellular trafficking, protein-protein interaction, recycling, and protein architecture. PTMs also contribute to the polymodal activation of TRPCs and their subtle regulation in diverse physiological contexts and in pathological situations. Owing to their roles in the motor coordination and regulation of kidney podocyte structure, mutations of TRPCs have been implicated in diseases like cerebellar ataxia (moonwalker mice) and focal and segmental glomerulosclerosis (FSGS). The aim of this review is to comprehensively integrate all reported PTMs of TRPCs, to discuss their physiological/pathophysiological roles if available, and to summarize diseases linked to the natural mutations of TRPCs.
Collapse
|
20
|
Chen W, Werner F, Illerhaus A, Knopp T, Völker K, Potapenko T, Hofmann U, Frantz S, Baba HA, Rösch M, Zernecke A, Karbach S, Wenzel P, Kuhn M. Stabilization of Perivascular Mast Cells by Endothelial CNP (C-Type Natriuretic Peptide). Arterioscler Thromb Vasc Biol 2020; 40:682-696. [PMID: 31893950 DOI: 10.1161/atvbaha.119.313702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Activated perivascular mast cells (MCs) participate in different cardiovascular diseases. Many factors provoking MC degranulation have been described, while physiological counterregulators are barely known. Endothelial CNP (C-type natriuretic peptide) participates in the maintenance of vascular barrier integrity, but the target cells and mechanisms are unclear. Here, we studied whether MCs are regulated by CNP. Approach and Results: In cultured human and murine MCs, CNP activated its specific GC (guanylyl cyclase)-B receptor and cyclic GMP signaling. This enhanced cyclic GMP-dependent phosphorylation of the cytoskeleton-associated VASP (vasodilator-stimulated phosphoprotein) and inhibited ATP-evoked degranulation. To elucidate the relevance in vivo, mice with a floxed GC-B (Npr2) gene were interbred with a Mcpt5-CreTG line to generate mice lacking GC-B in connective tissue MCs (MC GC-B knockout). In anesthetized mice, acute ischemia-reperfusion of the cremaster muscle microcirculation provoked extensive MC degranulation and macromolecule extravasation. Superfusion of CNP markedly prevented MC activation and endothelial barrier disruption in control but not in MC GC-B knockout mice. Notably, already under resting conditions, such knockout mice had increased numbers of degranulated MCs in different tissues, together with elevated plasma chymase levels. After transient coronary occlusion, their myocardial areas at risk and with infarction were enlarged. Moreover, MC GC-B knockout mice showed augmented perivascular neutrophil infiltration and deep vein thrombosis in a model of inferior vena cava ligation. CONCLUSIONS CNP, via GC-B/cyclic GMP signaling, stabilizes resident perivascular MCs at baseline and prevents their excessive activation under pathological conditions. Thereby CNP contributes to the maintenance of vascular integrity in physiology and disease.
Collapse
Affiliation(s)
- Wen Chen
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.).,Comprehensive Heart Failure Center (W.C., U.H., S.F., M.K.), University Hospital Würzburg, Germany
| | - Franziska Werner
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.)
| | - Anja Illerhaus
- Institute of Experimental Biomedicine (M.R., A.Z.), University Hospital Würzburg, Germany
| | - Tanja Knopp
- Department of Dermatology, University of Cologne, Germany (A.I.)
| | - Katharina Völker
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.)
| | - Tamara Potapenko
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.)
| | - Ulrich Hofmann
- Comprehensive Heart Failure Center (W.C., U.H., S.F., M.K.), University Hospital Würzburg, Germany
| | - Stefan Frantz
- Comprehensive Heart Failure Center (W.C., U.H., S.F., M.K.), University Hospital Würzburg, Germany
| | - Hideo A Baba
- Center of Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Germany (T.K., S.K., P.W.)
| | - Melanie Rösch
- Institute of Experimental Biomedicine (M.R., A.Z.), University Hospital Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine (M.R., A.Z.), University Hospital Würzburg, Germany
| | - Susanne Karbach
- Department of Dermatology, University of Cologne, Germany (A.I.).,Institute of Pathology, University Hospital Essen, University Duisburg-Essen (H.A.B.)
| | - Philip Wenzel
- Department of Dermatology, University of Cologne, Germany (A.I.).,Institute of Pathology, University Hospital Essen, University Duisburg-Essen (H.A.B.)
| | - Michaela Kuhn
- From the Institute of Physiology, University of Würzburg, Germany (W.C., F.W., K.V., T.P., M.K.).,Comprehensive Heart Failure Center (W.C., U.H., S.F., M.K.), University Hospital Würzburg, Germany
| |
Collapse
|
21
|
Špiranec K, Chen W, Werner F, Nikolaev VO, Naruke T, Koch F, Werner A, Eder-Negrin P, Diéguez-Hurtado R, Adams RH, Baba HA, Schmidt H, Schuh K, Skryabin BV, Movahedi K, Schweda F, Kuhn M. Endothelial C-Type Natriuretic Peptide Acts on Pericytes to Regulate Microcirculatory Flow and Blood Pressure. Circulation 2019; 138:494-508. [PMID: 29626067 DOI: 10.1161/circulationaha.117.033383] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Peripheral vascular resistance has a major impact on arterial blood pressure levels. Endothelial C-type natriuretic peptide (CNP) participates in the local regulation of vascular tone, but the target cells remain controversial. The cGMP-producing guanylyl cyclase-B (GC-B) receptor for CNP is expressed in vascular smooth muscle cells (SMCs). However, whereas endothelial cell-specific CNP knockout mice are hypertensive, mice with deletion of GC-B in vascular SMCs have unaltered blood pressure. METHODS We analyzed whether the vasodilating response to CNP changes along the vascular tree, ie, whether the GC-B receptor is expressed in microvascular types of cells. Mice with a floxed GC-B ( Npr2) gene were interbred with Tie2-Cre or PDGF-Rβ-Cre ERT2 lines to develop mice lacking GC-B in endothelial cells or in precapillary arteriolar SMCs and capillary pericytes. Intravital microscopy, invasive and noninvasive hemodynamics, fluorescence energy transfer studies of pericyte cAMP levels in situ, and renal physiology were combined to dissect whether and how CNP/GC-B/cGMP signaling modulates microcirculatory tone and blood pressure. RESULTS Intravital microscopy studies revealed that the vasodilatatory effect of CNP increases toward small-diameter arterioles and capillaries. CNP consistently did not prevent endothelin-1-induced acute constrictions of proximal arterioles, but fully reversed endothelin effects in precapillary arterioles and capillaries. Here, the GC-B receptor is expressed both in endothelial and mural cells, ie, in pericytes. It is notable that the vasodilatatory effects of CNP were preserved in mice with endothelial GC-B deletion, but abolished in mice lacking GC-B in microcirculatory SMCs and pericytes. CNP, via GC-B/cGMP signaling, modulates 2 signaling cascades in pericytes: it activates cGMP-dependent protein kinase I to phosphorylate downstream targets such as the cytoskeleton-associated vasodilator-activated phosphoprotein, and it inhibits phosphodiesterase 3A, thereby enhancing pericyte cAMP levels. These pathways ultimately prevent endothelin-induced increases of pericyte calcium levels and pericyte contraction. Mice with deletion of GC-B in microcirculatory SMCs and pericytes have elevated peripheral resistance and chronic arterial hypertension without a change in renal function. CONCLUSIONS Our studies indicate that endothelial CNP regulates distal arteriolar and capillary blood flow. CNP-induced GC-B/cGMP signaling in microvascular SMCs and pericytes is essential for the maintenance of normal microvascular resistance and blood pressure.
Collapse
Affiliation(s)
- Katarina Špiranec
- Institute of Physiology, University of Würzburg and Comprehensive Heart Failure Center, University Hospital Würzburg, Germany (K. Špiranec, W.C., S.C., F.W., T.N., F.K., P.E.-N., K. Schuh, M.K.)
| | - Wen Chen
- Institute of Physiology, University of Würzburg and Comprehensive Heart Failure Center, University Hospital Würzburg, Germany (K. Špiranec, W.C., S.C., F.W., T.N., F.K., P.E.-N., K. Schuh, M.K.)
| | - Franziska Werner
- Institute of Physiology, University of Würzburg and Comprehensive Heart Failure Center, University Hospital Würzburg, Germany (K. Špiranec, W.C., S.C., F.W., T.N., F.K., P.E.-N., K. Schuh, M.K.)
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany (V.O.N.)
| | - Takashi Naruke
- Institute of Physiology, University of Würzburg and Comprehensive Heart Failure Center, University Hospital Würzburg, Germany (K. Špiranec, W.C., S.C., F.W., T.N., F.K., P.E.-N., K. Schuh, M.K.)
| | - Franziska Koch
- Institute of Physiology, University of Würzburg and Comprehensive Heart Failure Center, University Hospital Würzburg, Germany (K. Špiranec, W.C., S.C., F.W., T.N., F.K., P.E.-N., K. Schuh, M.K.)
| | - Andrea Werner
- Institute of Physiology, University of Regensburg, Germany (A.W., F.S.)
| | - Petra Eder-Negrin
- Institute of Physiology, University of Würzburg and Comprehensive Heart Failure Center, University Hospital Würzburg, Germany (K. Špiranec, W.C., S.C., F.W., T.N., F.K., P.E.-N., K. Schuh, M.K.)
| | - Rodrigo Diéguez-Hurtado
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis (R.D.-H., R.H.A.)
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis (R.D.-H., R.H.A.)
| | - Hideo A Baba
- Faculty of Medicine, University of Münster, Germany. Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Germany (H.A.B.)
| | - Hannes Schmidt
- Interfaculty Institute of Biochemistry, University of Tübingen, Germany (H.S.)
| | - Kai Schuh
- Institute of Physiology, University of Würzburg and Comprehensive Heart Failure Center, University Hospital Würzburg, Germany (K. Špiranec, W.C., S.C., F.W., T.N., F.K., P.E.-N., K. Schuh, M.K.)
| | - Boris V Skryabin
- Core Facility Transgenic Animal and genetic engineering Models (B.V.S.)
| | - Kiavash Movahedi
- Myeloid Cell Immunology Lab, Vesalius Research Center, Center for Inflammation Research, and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium (K.M.)
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Germany (A.W., F.S.)
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg and Comprehensive Heart Failure Center, University Hospital Würzburg, Germany (K. Špiranec, W.C., S.C., F.W., T.N., F.K., P.E.-N., K. Schuh, M.K.)
| |
Collapse
|
22
|
Matsuo A, Nagai-Okatani C, Nishigori M, Kangawa K, Minamino N. Natriuretic peptides in human heart: Novel insight into their molecular forms, functions, and diagnostic use. Peptides 2019; 111:3-17. [PMID: 30120963 DOI: 10.1016/j.peptides.2018.08.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/12/2018] [Accepted: 08/13/2018] [Indexed: 01/16/2023]
Abstract
Among the three natriuretic peptides, atrial/A-type natriuretic peptide (ANP) and brain/B-type natriuretic peptide (BNP) are primarily produced by, and secreted from, heart tissue. They maintain cardiovascular homeostasis by binding to natriuretic peptide receptor-A. Since plasma ANP and BNP concentrations, as well as expression, are elevated in response to increased body fluid volume and pressure load on the heart wall, these peptides are widely utilized as diagnostic biomarkers for evaluating heart failure. Regardless of their high utility, differences in their molecular forms between healthy and diseased subjects and how these relate to pathophysiology have not well been examined. Recent studies have shown that the circulating molecular forms of ANP and BNP are not uniform; bioactive α-ANP is the major ANP form, whereas the weakly active proBNP is the major BNP form. The relative ratios of the different molecular forms are altered under different pathophysiological conditions. These facts indicate that detailed measurements of each form may provide useful information on the pathophysiological state of heart tissue. Here, we revisit the relationship between the molecular forms of, and pathophysiological alterations in, human ANP and BNP and discuss the possible utility of the measurement of each of the molecular forms. The third peptide, C-type natriuretic peptide, activates natriuretic peptide receptor-B, but little is known about its production and function in the heart because of its extremely low levels. However, through recent studies, its role in the heart is gradually becoming clear. Here, we summarize its molecular forms, assay systems, and functions in the heart.
Collapse
Affiliation(s)
- Ayaka Matsuo
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Chiaki Nagai-Okatani
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-8568, Japan
| | - Mitsuhiro Nishigori
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Kenji Kangawa
- Research Institute, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Naoto Minamino
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan.
| |
Collapse
|
23
|
Ramirez GA, Coletto LA, Sciorati C, Bozzolo EP, Manunta P, Rovere-Querini P, Manfredi AA. Ion Channels and Transporters in Inflammation: Special Focus on TRP Channels and TRPC6. Cells 2018; 7:E70. [PMID: 29973568 PMCID: PMC6070975 DOI: 10.3390/cells7070070] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/27/2018] [Accepted: 06/29/2018] [Indexed: 12/14/2022] Open
Abstract
Allergy and autoimmune diseases are characterised by a multifactorial pathogenic background. Several genes involved in the control of innate and adaptive immunity have been associated with diseases and variably combine with each other as well as with environmental factors and epigenetic processes to shape the characteristics of individual manifestations. Systemic or local perturbations in salt/water balance and in ion exchanges between the intra- and extracellular spaces or among tissues play a role. In this field, usually referred to as elementary immunology, novel evidence has been recently acquired on the role of members of the transient potential receptor (TRP) channel family in several cellular mechanisms of potential significance for the pathophysiology of the immune response. TRP canonical channel 6 (TRPC6) is emerging as a functional element for the control of calcium currents in immune-committed cells and target tissues. In fact, TRPC6 influences leukocytes’ tasks such as transendothelial migration, chemotaxis, phagocytosis and cytokine release. TRPC6 also modulates the sensitivity of immune cells to apoptosis and influences tissue susceptibility to ischemia-reperfusion injury and excitotoxicity. Here, we provide a view of the interactions between ion exchanges and inflammation with a focus on the pathogenesis of immune-mediated diseases and potential future therapeutic implications.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Lavinia A Coletto
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Clara Sciorati
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Enrica P Bozzolo
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Paolo Manunta
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Nephrology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Patrizia Rovere-Querini
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Angelo A Manfredi
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
- Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
24
|
Fitzakerley JL, Trachte GJ. Genetics of guanylyl cyclase pathways in the cochlea and their influence on hearing. Physiol Genomics 2018; 50:780-806. [PMID: 29958079 DOI: 10.1152/physiolgenomics.00056.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although hearing loss is the most common sensory deficit in Western societies, there are no successful pharmacological treatments for this disorder. Recent experiments have demonstrated that manipulation of intracellular cyclic guanosine monophosphate (cGMP) concentrations can have both beneficial and harmful effects on hearing. In this review, we will examine the role of cGMP as a key second messenger involved in many aspects of cochlear function and discuss the known functions of downstream effectors of cGMP in sound processing. The nitric oxide-stimulated soluble guanylyl cyclase system (sGC) and the two natriuretic peptide-stimulated particulate GCs (pGCs) will be more extensively covered because they have been studied most thoroughly. The cochlear GC systems are attractive targets for medical interventions that improve hearing while simultaneously representing an under investigated source of sensorineural hearing loss.
Collapse
Affiliation(s)
- Janet L Fitzakerley
- Department of Biomedical Sciences, University of Minnesota Medical School , Duluth, Minnesota
| | - George J Trachte
- Department of Biomedical Sciences, University of Minnesota Medical School , Duluth, Minnesota
| |
Collapse
|
25
|
Abstract
Natriuretic peptides are structurally related, functionally diverse hormones. Circulating atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) are delivered predominantly by the heart. Two C-type natriuretic peptides (CNPs) are paracrine messengers, notably in bone, brain, and vessels. Natriuretic peptides act by binding to the extracellular domains of three receptors, NPR-A, NPR-B, and NPR-C of which the first two are guanylate cyclases. NPR-C is coupled to inhibitory proteins. Atrial wall stress is the major regulator of ANP secretion; however, atrial pressure changes plasma ANP only modestly and transiently, and the relation between plasma ANP and atrial wall tension (or extracellular volume or sodium intake) is weak. Absence and overexpression of ANP-related genes are associated with modest blood pressure changes. ANP augments vascular permeability and reduces vascular contractility, renin and aldosterone secretion, sympathetic nerve activity, and renal tubular sodium transport. Within the physiological range of plasma ANP, the responses to step-up changes are unimpressive; in man, the systemic physiological effects include diminution of renin secretion, aldosterone secretion, and cardiac preload. For BNP, the available evidence does not show that cardiac release to the blood is related to sodium homeostasis or body fluid control. CNPs are not circulating hormones, but primarily paracrine messengers important to ossification, nervous system development, and endothelial function. Normally, natriuretic peptides are not powerful natriuretic/diuretic hormones; common conclusions are not consistently supported by hard data. ANP may provide fine-tuning of reno-cardiovascular relationships, but seems, together with BNP, primarily involved in the regulation of cardiac performance and remodeling. © 2017 American Physiological Society. Compr Physiol 8:1211-1249, 2018.
Collapse
Affiliation(s)
- Peter Bie
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
26
|
Komarova YA, Kruse K, Mehta D, Malik AB. Protein Interactions at Endothelial Junctions and Signaling Mechanisms Regulating Endothelial Permeability. Circ Res 2017; 120:179-206. [PMID: 28057793 DOI: 10.1161/circresaha.116.306534] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/31/2022]
Abstract
The monolayer of endothelial cells lining the vessel wall forms a semipermeable barrier (in all tissue except the relatively impermeable blood-brain and inner retinal barriers) that regulates tissue-fluid homeostasis, transport of nutrients, and migration of blood cells across the barrier. Permeability of the endothelial barrier is primarily regulated by a protein complex called adherens junctions. Adherens junctions are not static structures; they are continuously remodeled in response to mechanical and chemical cues in both physiological and pathological settings. Here, we discuss recent insights into the post-translational modifications of junctional proteins and signaling pathways regulating plasticity of adherens junctions and endothelial permeability. We also discuss in the context of what is already known and newly defined signaling pathways that mediate endothelial barrier leakiness (hyperpermeability) that are important in the pathogenesis of cardiovascular and lung diseases and vascular inflammation.
Collapse
Affiliation(s)
- Yulia A Komarova
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Kevin Kruse
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Dolly Mehta
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Asrar B Malik
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago.
| |
Collapse
|
27
|
Duan CY, Zhang J, Wu HL, Li T, Liu LM. Regulatory mechanisms, prophylaxis and treatment of vascular leakage following severe trauma and shock. Mil Med Res 2017; 4:11. [PMID: 28361006 PMCID: PMC5370457 DOI: 10.1186/s40779-017-0117-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/06/2017] [Indexed: 02/08/2023] Open
Abstract
Vascular leakage, or increased vascular permeability, is a common but important pathological process for various critical diseases, including severe trauma, shock, sepsis, and multiple organ dysfunction syndrome (MODS), and has become one of the most important causes of death for intensive care units (ICU) patients. Currently, although there has been some progress in knowledge of the pathogenesis of these vascular disorders, the detailed mechanisms remain unclear, and effective prophylaxis and treatment are still lacking. In this study, we aimed to provide a review of the literature regarding the regulatory mechanisms and prophylaxis as well as the treatment of vascular leakage in critical diseases such as severe trauma and shock, which could be beneficial for the overall clinical treatment of vascular leakage disorders.
Collapse
Affiliation(s)
- Chen-Yang Duan
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Jie Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Hui-Ling Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| |
Collapse
|
28
|
Zhang B, Jiang J, Yue Z, Liu S, Ma Y, Yu N, Gao Y, Sun S, Chen S, Liu P. Store-Operated Ca2+ Entry (SOCE) contributes to angiotensin II-induced cardiac fibrosis in cardiac fibroblasts. J Pharmacol Sci 2016; 132:171-180. [DOI: 10.1016/j.jphs.2016.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/22/2016] [Accepted: 05/23/2016] [Indexed: 02/01/2023] Open
|
29
|
Piwkowska A. Role of Protein Kinase G and Reactive Oxygen Species in the Regulation of Podocyte Function in Health and Disease. J Cell Physiol 2016; 232:691-697. [PMID: 27662602 DOI: 10.1002/jcp.25613] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/22/2016] [Indexed: 11/09/2022]
Abstract
Podocytes and their foot processes form an important cellular layer of the glomerular barrier involved in the regulation of glomerular permeability. Disturbing the function of podocytes plays a central role in the development of proteinuria in diabetic nephropathy. Retraction of the podocyte foot processes that form slit diaphragms is a common feature of proteinuria; although, the correlation between these events in not well understood. Notably, it is unclear whether podocyte foot processes are able to regulate slit diaphragm permeability and glomerular ultrafiltration. The occurrence of reactive oxygen species generation, insulin resistance, and hyperglycemia characterizes early stages of type 2 diabetes. Protein kinase G type I alpha (PKGIα) is an intracellular target for vasorelaxant factors. It is activated in both cGMP-dependent and cGMP-independent manners. Recently, we demonstrated a relationship between oxidative stress, PKGIα activation, actin reorganization, and changes in the permeability of the filtration barrier. This review discusses how redox imbalance affects both the activity of PKGIα and PKGI-dependent signaling pathways in podocytes. J. Cell. Physiol. 232: 691-697, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Agnieszka Piwkowska
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland
| |
Collapse
|
30
|
Curry FRE, Clark JF, Jiang Y, Kim MH, Adamson RH, Simon SI. The role of atrial natriuretic peptide to attenuate inflammation in a mouse skin wound and individually perfused rat mesenteric microvessels. Physiol Rep 2016; 4:4/18/e12968. [PMID: 27670406 PMCID: PMC5037917 DOI: 10.14814/phy2.12968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 01/05/2023] Open
Abstract
We tested the hypothesis that the anti‐inflammatory actions of atrial natriuretic peptide (ANP) result from the modulation of leukocyte adhesion to inflamed endothelium and not solely ANP ligation of endothelial receptors to stabilize endothelial barrier function. We measured vascular permeability to albumin and accumulation of fluorescent neutrophils in a full‐thickness skin wound on the flank of LysM‐EGFP mice 24 h after formation. Vascular permeability in individually perfused rat mesenteric microvessels was also measured after leukocytes were washed out of the vessel lumen. Thrombin increased albumin permeability and increased the accumulation of neutrophils. The thrombin‐induced inflammatory responses were attenuated by pretreating the wound with ANP (30 min). During pretreatment ANP did not lower permeability, but transiently increased baseline albumin permeability concomitant with the reduction in neutrophil accumulation. ANP did not attenuate acute increases in permeability to histamine and bradykinin in individually perfused rat microvessels. The hypothesis that anti‐inflammatory actions of ANP depend solely on endothelial responses that stabilize the endothelial barrier is not supported by our results in either individually perfused microvessels in the absence of circulating leukocytes or the more chronic skin wound model. Our results conform to the alternate hypothesis that ANP modulates the interaction of leukocytes with the inflamed microvascular wall of the 24 h wound. Taken together with our previous observations that ANP reduces deformability of neutrophils and their strength of attachment, rolling, and transvascular migration, these observations provide the basis for additional investigations of ANP as an anti‐inflammatory agent to modulate leukocyte–endothelial cell interactions.
Collapse
Affiliation(s)
- Fitz-Roy E Curry
- Department of Physiology & Membrane Biology, University of California, Davis, Davis, California
| | - Joyce F Clark
- Department of Physiology & Membrane Biology, University of California, Davis, Davis, California
| | - Yanyan Jiang
- Department of Physiology & Membrane Biology, University of California, Davis, Davis, California
| | - Min-Ho Kim
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Roger H Adamson
- Department of Physiology & Membrane Biology, University of California, Davis, Davis, California
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| |
Collapse
|
31
|
Chen W, Spitzl A, Mathes D, Nikolaev VO, Werner F, Weirather J, Špiranec K, Röck K, Fischer JW, Kämmerer U, Stegner D, Baba HA, Hofmann U, Frantz S, Kuhn M. Endothelial Actions of ANP Enhance Myocardial Inflammatory Infiltration in the Early Phase After Acute Infarction. Circ Res 2016; 119:237-48. [PMID: 27142162 DOI: 10.1161/circresaha.115.307196] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 03/03/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE In patients after acute myocardial infarction (AMI), the initial extent of necrosis and inflammation determine clinical outcome. One early event in AMI is the increased cardiac expression of atrial natriuretic peptide (NP) and B-type NP, with their plasma levels correlating with severity of ischemia. It was shown that NPs, via their cGMP-forming guanylyl cyclase-A (GC-A) receptor and cGMP-dependent kinase I (cGKI), strengthen systemic endothelial barrier properties in acute inflammation. OBJECTIVE We studied whether endothelial actions of local NPs modulate myocardial injury and early inflammation after AMI. METHODS AND RESULTS Necrosis and inflammation after experimental AMI were compared between control mice and littermates with endothelial-restricted inactivation of GC-A (knockout mice with endothelial GC-A deletion) or cGKI (knockout mice with endothelial cGKI deletion). Unexpectedly, myocardial infarct size and neutrophil infiltration/activity 2 days after AMI were attenuated in knockout mice with endothelial GC-A deletion and unaltered in knockout mice with endothelial cGKI deletion. Molecular studies revealed that hypoxia and tumor necrosis factor-α, conditions accompanying AMI, reduce the endothelial expression of cGKI and enhance cGMP-stimulated phosphodiesterase 2A (PDE2A) levels. Real-time cAMP measurements in endothelial microdomains using a novel fluorescence resonance energy transfer biosensor revealed that PDE2 mediates NP/cGMP-driven decreases of submembrane cAMP levels. Finally, intravital microscopy studies of the mouse cremaster microcirculation showed that tumor necrosis factor-α-induced endothelial NP/GC-A/cGMP/PDE2 signaling impairs endothelial barrier functions. CONCLUSIONS Hypoxia and cytokines, such as tumor necrosis factor-α, modify the endothelial postreceptor signaling pathways of NPs, with downregulation of cGKI, induction of PDE2A, and altered cGMP/cAMP cross talk. Increased expression of PDE2 can mediate hyperpermeability effects of paracrine endothelial NP/GC-A/cGMP signaling and facilitate neutrophil extravasation during the early phase after MI.
Collapse
Affiliation(s)
- Wen Chen
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Annett Spitzl
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Denise Mathes
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Viacheslav O Nikolaev
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Franziska Werner
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Johannes Weirather
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Katarina Špiranec
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Katharina Röck
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Jens W Fischer
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Ulrike Kämmerer
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - David Stegner
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Hideo A Baba
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Ulrich Hofmann
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Stefan Frantz
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.)
| | - Michaela Kuhn
- From the Institute of Physiology (W.C., A.S., F.W., K.Š., M.K.), Comprehensive Heart Failure Center (D.M., J.W., U.H., S.F., M.K.), and Department of Experimental Biomedicine and Rudolf Virchow Center for Experimental Biomedicine (D.S.), University of Würzburg, Würzburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (V.O.N.); Institut für Pharmakologie und Klinische Pharmakologie und CARID, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (K.R., J.W.F.); Department of Obstetrics and Gynecology, University Hospital Würzburg, Würzburg, Germany (U.K.); Institute of Pathology, University Duisburg-Essen, Essen, Germany (H.A.B.); and Universitätsklinik und Poliklinik für Innere Medizin III, Universitätsklinikum Halle (Saale), Halle (Saale), Germany (U.H., S.F.).
| |
Collapse
|
32
|
Kuebler WM, Wittenberg C, Lee WL, Reppien E, Goldenberg NM, Lindner K, Gao Y, Winoto-Morbach S, Drab M, Mühlfeld C, Dombrowsky H, Ochs M, Schütze S, Uhlig S. Thrombin stimulates albumin transcytosis in lung microvascular endothelial cells via activation of acid sphingomyelinase. Am J Physiol Lung Cell Mol Physiol 2016; 310:L720-32. [PMID: 26851257 DOI: 10.1152/ajplung.00157.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 01/22/2016] [Indexed: 01/12/2023] Open
Abstract
Transcellular albumin transport occurs via caveolae that are abundant in lung microvascular endothelial cells. Stimulation of albumin transcytosis by proinflammatory mediators may contribute to alveolar protein leak in lung injury, yet the regulation of albumin transport and its underlying molecular mechanisms are so far incompletely understood. Here we tested the hypothesis that thrombin may stimulate transcellular albumin transport across lung microvascular endothelial cells in an acid-sphingomyelinase dependent manner. Thrombin increased the transport of fluorescently labeled albumin across confluent human lung microvascular endothelial cell (HMVEC-L) monolayers to an extent that markedly exceeds the rate of passive diffusion. Thrombin activated acid sphingomyelinase (ASM) and increased ceramide production in HMVEC-L, but not in bovine pulmonary artery cells, which showed little albumin transport in response to thrombin. Thrombin increased total caveolin-1 (cav-1) content in both whole cell lysates and lipid rafts from HMVEC-L, and this effect was blocked by inhibition of ASM or de novo protein biosynthesis. Thrombin-induced uptake of albumin into lung microvascular endothelial cells was confirmed in isolated-perfused lungs by real-time fluorescence imaging and electron microscopy of gold-labeled albumin. Inhibition of ASM attenuated thrombin-induced albumin transport both in confluent HMVEC-L and in intact lungs, whereas HMVEC-L treatment with exogenous ASM increased albumin transport and enriched lipid rafts in cav-1. Our findings indicate that thrombin stimulates transcellular albumin transport in an acid sphingomyelinase-dependent manner by inducing de novo synthesis of cav-1 and its recruitment to membrane lipid rafts.
Collapse
Affiliation(s)
- Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Heart Institute Berlin, Berlin, Germany; The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada; Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Claudia Wittenberg
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Warren L Lee
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada; Interdepartmental Division of Critical Care, Departments of Medicine and Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Eike Reppien
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Neil M Goldenberg
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada
| | - Karsten Lindner
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Yizhuo Gao
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada
| | | | - Marek Drab
- Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DLZ), Hannover, Germany; Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany; and
| | - Heike Dombrowsky
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DLZ), Hannover, Germany; Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany; and
| | - Stefan Schütze
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Stefan Uhlig
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany; Institute of Pharmacology and Toxicology, RWTH Aachen, Aachen, Germany
| |
Collapse
|
33
|
Chen W, Völker K, Gaßner B, Werner F, Rabenhorst A, Hartmann K, Kuhn M. C-type natriuretic peptide prevents activation of perivascular mast cells and inflammation in the postischemic microvasculature. BMC Pharmacol Toxicol 2015. [PMCID: PMC4565523 DOI: 10.1186/2050-6511-16-s1-a41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
34
|
Curry FRE, Clark JF, Adamson RH. Microperfusion Technique to Investigate Regulation of Microvessel Permeability in Rat Mesentery. J Vis Exp 2015. [PMID: 26436435 DOI: 10.3791/53210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Experiments to measure the permeability properties of individually perfused microvessels provide a bridge between investigation of molecular and cellular mechanisms regulating vascular permeability in cultured endothelial cell monolayers and the functional exchange properties of whole microvascular beds. A method to cannulate and perfuse venular microvessels of rat mesentery and measure the hydraulic conductivity of the microvessel wall is described. The main equipment needed includes an intravital microscope with a large modified stage that supports micromanipulators to position three different microtools: (1) a beveled glass micropipette to cannulate and perfuse the microvessel; (2) a glass micro-occluder to transiently block perfusion and enable measurement of transvascular water flow movement at a measured hydrostatic pressure, and (3) a blunt glass rod to stabilize the mesenteric tissue at the site of cannulation. The modified Landis micro-occlusion technique uses red cells suspended in the artificial perfusate as markers of transvascular fluid movement, and also enables repeated measurements of these flows as experimental conditions are changed and hydrostatic and colloid osmotic pressure difference across the microvessels are carefully controlled. Measurements of hydraulic conductivity first using a control perfusate, then after re-cannulation of the same microvessel with the test perfusates enable paired comparisons of the microvessel response under these well-controlled conditions. Attempts to extend the method to microvessels in the mesentery of mice with genetic modifications expected to modify vascular permeability were severely limited because of the absence of long straight and unbranched microvessels in the mouse mesentery, but the recent availability of the rats with similar genetic modifications using the CRISPR/Cas9 technology is expected to open new areas of investigation where the methods described herein can be applied.
Collapse
Affiliation(s)
- Fitz-Roy E Curry
- Department of Physiology & Membrane Biology, University of California Davis
| | - Joyce F Clark
- Department of Physiology & Membrane Biology, University of California Davis
| | - Roger H Adamson
- Department of Physiology & Membrane Biology, University of California Davis;
| |
Collapse
|
35
|
Friedland K, Harteneck C. Hyperforin: To Be or Not to Be an Activator of TRPC(6). Rev Physiol Biochem Pharmacol 2015; 169:1-24. [DOI: 10.1007/112_2015_25] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
36
|
Hoffmann LS, Chen HH. cGMP: transition from bench to bedside: a report of the 6th International Conference on cGMP Generators, Effectors and Therapeutic Implications. Naunyn Schmiedebergs Arch Pharmacol 2014; 387:707-18. [PMID: 24927824 DOI: 10.1007/s00210-014-0999-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 05/30/2014] [Indexed: 02/06/2023]
Abstract
Essential physiological homeostatic processes such as vascular tone, fluid balance, cardiorenal function, and sensory processes are regulated by the second messenger cyclic guanosine 3', 5'-monophosphate (cGMP). Dysregulation of cGMP-dependent pathways plays an important role in cardiovascular diseases such as hypertension, pulmonary hypertension, heart failure, or erectile dysfunction. Thus, the cGMP pathway consisting of the cGMP-generating guanylyl cyclases, protein kinases, and phosphodiesterases (PDE) has evolved to an important drug target and is the focus of a wide variety of research fields ranging from unraveling mechanisms on the molecular level to understanding the regulation of physiological and pathophysiological processes by cGMP. Based on the results from basic and preclinical research, therapeutic drugs have been developed which modulate the cGMP pathway and are investigated in clinical trials. Riociguat, a nitric oxide (NO)-independent soluble guanylyl cyclase stimulator; recombinant B-type natriuretic peptide (BNP); or recombinant atrial natriuretic peptide (ANP) and PDE5 inhibitors are cGMP-modulating drugs that are already available for the treatment of pulmonary hypertension, acute heart failure, and erectile dysfunction, respectively. The latest results from basic to clinical research on cGMP were presented on the 6th International Conference on cGMP in Erfurt, Germany, and are summarized in this article.
Collapse
Affiliation(s)
- Linda S Hoffmann
- Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, Bonn, Germany,
| | | |
Collapse
|
37
|
The role of canonical transient receptor potential channels in seizure and excitotoxicity. Cells 2014; 3:288-303. [PMID: 24722470 PMCID: PMC4092853 DOI: 10.3390/cells3020288] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/29/2014] [Accepted: 04/02/2014] [Indexed: 12/11/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels are a family of polymodal cation channels with some degree of Ca2+ permeability. Although initially thought to be channels mediating store-operated Ca2+ influx, TRPC channels can be activated by stimulation of Gq-coupled G-protein coupled receptors, or by an increase in intracellular free Ca2+ concentration. Thus, activation of TRPC channels could be a common downstream event of many signaling pathways that contribute to seizure and excitotoxicity, such as N-methyl-D-aspartate (NMDA) receptor-mediated Ca2+ influx, or metabotropic glutamate receptor activation. Recent studies with genetic ablation of various TRPC family members have demonstrated that TRPC channels, in particular heteromeric TRPC1/4 channels and homomeric TRPC5 channels, play a critical role in both pilocarpine-induced acute seizures and neuronal cell death. However, exact underlying mechanisms remain to be fully elucidated, and selective TRPC modulators and antibodies with better specificity are urgently needed for future research.
Collapse
|
38
|
Abstract
TRPC6 is a non-selective cation channel 6 times more permeable to Ca(2+) than to Na(+). Channel homotetramers heterologously expressed have a characteristic doubly rectifying current-voltage relationship and are directly activated by the second messenger diacylglycerol (DAG). TRPC6 proteins are also regulated by specific tyrosine or serine phosphorylation and phosphoinositides. Given its specific expression pattern, TRPC6 is likely to play a number of physiological roles which are confirmed by the analysis of a Trpc6 (-/-) mouse model. In smooth muscle Na(+) influx through TRPC6 channels and activation of voltage-gated Ca(2+) channels by membrane depolarisation is the driving force for contraction. Permeability of pulmonary endothelial cells depends on TRPC6 and induces ischaemia-reperfusion oedema formation in the lungs. TRPC6 was also identified as an essential component of the slit diaphragm architecture of kidney podocytes and plays an important role in the protection of neurons after cerebral ischaemia. Other functions especially in immune and blood cells remain elusive. Recently identified TRPC6 blockers may be helpful for therapeutic approaches in diseases with highly activated TRPC6 channel activity.
Collapse
Affiliation(s)
- Alexander Dietrich
- Walther-Straub-Institute for Pharmacology and Toxicology, School of Medicine, LM-University of Munich, 80336, Munich, Germany,
| | | |
Collapse
|