1
|
Wang B, Li Y, Ouyang Q, Xu MT, Wang YY, Fu SJ, Liu WQ, Liu XT, Ling H, Zhang X, Xiu RJ, Liu MM. Strain- and sex-dependent variability in hepatic microcirculation and liver function in mice. World J Gastroenterol 2025; 31:101058. [PMID: 40309233 PMCID: PMC12038547 DOI: 10.3748/wjg.v31.i15.101058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/02/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The integrity and functionality of the hepatic microcirculation are essential for maintaining liver health, which is influenced by sex and genetic background. Understanding these variations is crucial for addressing disparities in liver disease outcomes. AIM To investigate the sexual dimorphism and genetic heterogeneity of liver microcirculatory function in mice. METHODS We assessed hepatic microhemodynamics in BALB/c, C57BL/6J, and KM mouse strains using laser Doppler flowmetry and wavelet analysis. We analyzed the serum levels of alanine transaminase, glutamic acid aminotransferase, total bile acid, total protein, alkaline phosphatase, and glucose. Histological and immunohistochemical staining were employed to quantify microvascular density and the expression levels of cluster of differentiation (CD) 31, and estrogen receptor α, and β. Statistical analyses, including the Mantel test and Pearson correlation, were conducted to determine the relationships among hepatic function, microcirculation, and marcocirculation between different sexes and across genetic backgrounds. RESULTS We identified sex-based disparities in hepatic microhemodynamics across all strains, with males exhibiting higher microvascular perfusion and erythrocyte concentration, but lower blood velocity. Strain-specific differences were evident, particularly in the endothelial oscillatory characteristics of the erythrocyte concentration. No sex-dependent differences in estrogen receptor expression were observed, while significant variations in CD31 expression and microvascular density were observed. The correlations highlighted relationships between hepatic microhemodynamics and liver function indicators. CONCLUSION Our findings indicate the influence of genetic and sex differences on hepatic microcirculation and liver function, highlighting the necessity of incorporating both genetic background and sex into hepatic physiology studies and potential liver disease management strategies.
Collapse
Affiliation(s)
- Bing Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yuan Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Qin Ouyang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Science, Beijing 100102, China
| | - Meng-Ting Xu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Ying-Yu Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Sun-Jing Fu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Wei-Qi Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xue-Ting Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Hao Ling
- Department of Radiology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, Hunan Province, China
| | - Xu Zhang
- Laboratory of Electron Microscopy, Ultrastructural Pathology Center, Peking University First Hospital, Beijing 100034, China
| | - Rui-Juan Xiu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Ming-Ming Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
2
|
Sallam NA, Laher I. Regional heterogeneity in vascular contractile dysfunction in diabetic mice. Mol Cell Biochem 2025:10.1007/s11010-025-05257-4. [PMID: 40208461 DOI: 10.1007/s11010-025-05257-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/16/2025] [Indexed: 04/11/2025]
Abstract
Oxidative stress underlies many diabetic complications, including diabetic vasculopathy. It is unclear if oxidative stress has different effects in regionally distant arteries. We compared the contractile function of three arteries from diabetic mice and elucidated the mechanisms underlying their differential adaptation. We examined responses of the aorta, carotid and femoral arteries, isolated from the same diabetic (db/db) or normoglycemic control mice, to different vasoconstrictors in the presence and absence of indomethacin, apocynin, sulfaphenazole, L-NAME or a reactive oxygen species generating system to identify the enzyme(s) contributing to vascular dysfunction. Expression of superoxide dismutase (SOD) isoforms was measured. db/db aortae showed augmented contractile responses to KCl, phenylephrine, A23197 and U-46619 likely due to activated cyclooxygenases and hypersensitivity to thromboxane A2. Contractile responses of db/db carotid arteries were unaltered, likely due to higher SOD3 and SOD1 levels compared to the aortae. Femoral arteries were more vulnerable to oxidative stress, lacked SOD3 expression, and showed higher basal potassium channels activity. Phenylephrine contractions in femoral arteries were dependent on extracellular calcium entry; while contractions in aortae were dependent on extracellular calcium entry and intracellular calcium release. Femoral arteries from db/db mice exhibited higher basal potassium channels activity and attenuated contractility compared to control mice likely due to lower SOD levels. Heterogeneity exists between the three arteries at functional and molecular levels due to different signalling pathways and antioxidant defense mechanisms. Understanding regional differences in vasomotor control coupled with advanced delivery systems can help in developing therapies targeting specific vascular beds.
Collapse
Affiliation(s)
- Nada A Sallam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-aini Street, Cairo, 11562, Egypt.
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Abstract
Mechanical forces influence different cell types in our bodies. Among the earliest forces experienced in mammals is blood movement in the vascular system. Blood flow starts at the embryonic stage and ceases when the heart stops. Blood flow exposes endothelial cells (ECs) that line all blood vessels to hemodynamic forces. ECs detect these mechanical forces (mechanosensing) through mechanosensors, thus triggering physiological responses such as changes in vascular diameter. In this review, we focus on endothelial mechanosensing and on how different ion channels, receptors, and membrane structures detect forces and mediate intricate mechanotransduction responses. We further highlight that these responses often reflect collaborative efforts involving several mechanosensors and mechanotransducers. We close with a consideration of current knowledge regarding the dysregulation of endothelial mechanosensing during disease. Because hemodynamic disruptions are hallmarks of cardiovascular disease, studying endothelial mechanosensing holds great promise for advancing our understanding of vascular physiology and pathophysiology.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA;
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA;
| |
Collapse
|
4
|
Aitken C, Mehta V, Schwartz MA, Tzima E. Mechanisms of endothelial flow sensing. NATURE CARDIOVASCULAR RESEARCH 2023; 2:517-529. [PMID: 39195881 DOI: 10.1038/s44161-023-00276-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/14/2023] [Indexed: 08/29/2024]
Abstract
Fluid shear stress plays a key role in sculpting blood vessels during development, in adult vascular homeostasis and in vascular pathologies. During evolution, endothelial cells evolved several mechanosensors that convert physical forces into biochemical signals, a process termed mechanotransduction. This Review discusses our understanding of endothelial flow sensing and suggests important questions for future investigation.
Collapse
Affiliation(s)
- Claire Aitken
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Vedanta Mehta
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA.
| | - Ellie Tzima
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
5
|
ShamsEldeen AM, Mehesen MN, Aboulhoda BE, Rashed LA, Elsebaie MM, Mohamed EA, Gamal MM. Prenatal intake of omega-3 promotes Wnt/β-catenin signaling pathway, and preserves integrity of the blood-brain barrier in preeclamptic rats. Physiol Rep 2021; 9:e14925. [PMID: 34174018 PMCID: PMC8234480 DOI: 10.14814/phy2.14925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Preeclampsia is a systemic, multi-organ endotheliopathy, associated with oxidative injury to the blood-brain barrier (BBB). Preeclampsia initiates a cascade of events that include neuroinflammation. Recently, it was documented that Wnt/β-catenin signaling pathway exerts neuroprotective effects and maintain BBB integrity. We investigate the protective effect of omega-3 against neurovascular complication of preeclampsia and its relation to Wnt/β-catenin signaling pathway. METHODOLOGY After confirmation of day 0 pregnancy (G0), 24 adult pregnant female Wistar rats were divided into four groups control pregnant, pregnant supplemented with omega-3, preeclampsia (PE); female rats received N (ω)-nitro-L-arginine methyl ester (L-NAME) (50 mg/kg/day SC from day 7 to day 16 of pregnancy for induction of preeclampsia) and PE rats supplemented with omega-3. The intake of omega-3 started on day zero (0) of pregnancy until the end of the study (144 mg/kg\day orally). RESULTS We found that omega-3 supplementation significantly improved cognitive functions and EEG amplitude, decreased blood pressure, water contents of brain tissues, sFlt-1, oxidative stress, proteinuria, and enhanced Wnt\β-catenin proteins. Histological examination showed improved cerebral microangiopathy, increased expression of claudin-1 and -3, CD31, and VEGF in the cerebral cortical microvasculature and choroid plexus in PE rats treated with omega-3. A positive correlation between protein expression level of Wnt \β-catenin and cognitive functions, and a negative correlation between claudin-5 relative expression, claudin-1 and -3 area % from one side and water content of the brain tissues from the other side were observed. CONCLUSION Wnt/β-catenin signaling pathway suspected to have an important role to improve BBB integrity. Neuroprotective, antioxidant, and anti-inflammatory effects of omega-3 were observed and can be suggested as protective supplementation for preeclampsia.
Collapse
Affiliation(s)
| | - Marwa Nagi Mehesen
- Department of Medical PharmacologyFaculty of MedicineCairo UniversityCairoEgypt
| | - Basma Emad Aboulhoda
- Department of Anatomy and EmbryologyFaculty of MedicineCairo UniversityCairoEgypt
| | - Laila Ahmed Rashed
- Department of Biochemistry and Molecular BiologyFaculty of MedicineCairo UniversityCairoEgypt
| | | | | | | |
Collapse
|
6
|
Abstract
The potential of CD31 as a therapeutic target in atherosclerosis has been considered ever since its cloning in the 1990s, but the exact role played by this molecule in the biologic events underlying atherosclerosis has remained controversial, resulting in the stalling of any therapeutic perspective. Due to the supposed cell adhesive properties of CD31, specific monoclonal antibodies and recombinant proteins were regarded as blocking agents because their use prevented the arrival of leukocytes at sites of acute inflammation. However, the observed effect of those compounds likely resulted from the engagement of the immunomodulatory function of CD31 signaling. This was acknowledged only later though, upon the discovery of CD31's 2 intracytoplasmic tyrosine residues called immunoreceptor tyrosine inhibitory motifs. A growing body of evidence currently points at a therapeutic potential for CD31 agonists in atherothrombosis. Clinical observations show that CD31 expression is altered at the surface of leukocytes infiltrating unhealed atherothrombotic lesions and that the physiological immunomodulatory functions of CD31 are lost at the surface of blood leukocytes in patients with acute coronary syndromes. On the contrary, translational studies using candidate therapeutic molecules in laboratory animals have provided encouraging results: synthetic peptides administered to atherosclerotic mice as systemic drugs in the acute phases of atherosclerotic complications favor the healing of wounded arteries, whereas the immobilization of CD31 agonist peptides onto coronary stents implanted in farm pigs favors their peaceful integration within the coronary arterial wall.
Collapse
Affiliation(s)
- Giuseppina Caligiuri
- From the Laboratory for Vascular Translational Science, Inserm U1148, Université de Paris, France; and Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Nord Val-de-Seine, Site Bichat, France
| |
Collapse
|
7
|
Caligiuri G. Mechanotransduction, immunoregulation, and metabolic functions of CD31 in cardiovascular pathophysiology. Cardiovasc Res 2019; 115:1425-1434. [DOI: 10.1093/cvr/cvz132] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
Abstract
Biomechanical changes in the heart and vessels drive rapid and dynamic regulation of blood flow, a vital process for meeting the changing metabolic needs of the peripheral tissues at any given point in time. The fluid movement of the blood exerts haemodynamic stress upon the solid elements of the cardiovascular system: the heart, vessels, and cellular components of the blood. Cardiovascular diseases can lead to prolonged mechanical stress, such as cardiac remodelling during heart failure or vascular stiffening in atherosclerosis. This can lead to a significantly reduced or increasingly turbulent blood supply, inducing a shift in cellular metabolism that, amongst other effects, can trigger the release of reactive oxygen species and initiate a self-perpetuating cycle of inflammation and oxidative stress. CD31 is the most abundant constitutive co-signalling receptor glycoprotein on endothelial cells, which line the cardiovascular system and form the first-line of cellular contact with the blood. By associating with most endothelial receptors involved in mechanosensing, CD31 regulates the response to biomechanical stimuli. In addition, by relocating in the lipid rafts of endothelial cells as well as of cells stably interacting with the endothelium, including leucocytes and platelets, CD31–CD31 trans-homophilic engagement guides and restrains platelet and immune cell accumulation and activation and at sites of damage. In this way, CD31 is at the centre of mediating mechanical, metabolic, and immunological changes within the circulation and provides a single target that may have pleiotropic beneficial effects.
Collapse
Affiliation(s)
- Giuseppina Caligiuri
- Université de Paris, Cardiovascular Immunobiology, UMRS1148, INSERM, Paris, France
- Cardiology Department and Physiology Departments, AP-HP, University Hospital Xavier Bichat, 46 Rue Henri Huchard, Paris, France
| |
Collapse
|
8
|
Tanner MJ, Wang J, Ying R, Suboc TB, Malik M, Couillard A, Branum A, Puppala V, Widlansky ME. Dynamin-related protein 1 mediates low glucose-induced endothelial dysfunction in human arterioles. Am J Physiol Heart Circ Physiol 2016; 312:H515-H527. [PMID: 27923790 DOI: 10.1152/ajpheart.00499.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 11/22/2022]
Abstract
Intensive glycemic regulation has resulted in an increased incidence of hypoglycemia. Hypoglycemic burden correlates with adverse cardiovascular complications and contributes acutely and chronically to endothelial dysfunction. Prior data indicate that mitochondrial dysfunction contributes to hypoglycemia-induced endothelial dysfunction, but the mechanisms behind this linkage remain unknown. We attempt to determine whether clinically relevant low-glucose (LG) exposures acutely induce endothelial dysfunction through activation of the mitochondrial fission process. Characterization of mitochondrial morphology was carried out in cultured endothelial cells by using confocal microscopy. Isolated human arterioles were used to explore the effect LG-induced mitochondrial fission has on the formation of detrimental reactive oxygen species (ROS), bioavailability of nitric oxide (NO), and endothelial-dependent vascular relaxation. Fluorescence microscopy was employed to visualize changes in mitochondrial ROS and NO levels and videomicroscopy applied to measure vasodilation response. Pharmacological disruption of the profission protein Drp1 with Mdivi-1 during LG exposure reduced mitochondrial fragmentation among vascular endothelial cells (LG: 0.469; LG+Mdivi-1: 0.276; P = 0.003), prevented formation of vascular ROS (LG: 2.036; LG+Mdivi-1: 1.774; P = 0.005), increased the presence of NO (LG: 1.352; LG+Mdivi-1: 1.502; P = 0.048), and improved vascular dilation response to acetylcholine (LG: 31.6%; LG+Mdivi-1; 78.5% at maximum dose; P < 0.001). Additionally, decreased expression of Drp1 via siRNA knockdown during LG conditions also improved vascular relaxation. Exposure to LG imparts endothelial dysfunction coupled with altered mitochondrial phenotypes among isolated human arterioles. Disruption of Drp1 and subsequent mitochondrial fragmentation events prevents impaired vascular dilation, restores mitochondrial phenotype, and implicates mitochondrial fission as a primary mediator of LG-induced endothelial dysfunction.NEW & NOTEWORTHY Acute low-glucose exposure induces mitochondrial fragmentation in endothelial cells via Drp1 and is associated with impaired endothelial function in human arterioles. Targeting of Drp1 prevents fragmentation, improves vasofunction, and may provide a therapeutic target for improving cardiovascular complications among diabetics.Listen to this article's corresponding podcast @ http://ajpheart.podbean.com/e/mitochondrial-dynamics-impact-endothelial-function/.
Collapse
Affiliation(s)
- Michael J Tanner
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jingli Wang
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Rong Ying
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Tisha B Suboc
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mobin Malik
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allison Couillard
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Amberly Branum
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Venkata Puppala
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael E Widlansky
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
9
|
Potential Harmful Effects of PM2.5 on Occurrence and Progression of Acute Coronary Syndrome: Epidemiology, Mechanisms, and Prevention Measures. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13080748. [PMID: 27463723 PMCID: PMC4997434 DOI: 10.3390/ijerph13080748] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/13/2016] [Accepted: 07/20/2016] [Indexed: 12/18/2022]
Abstract
The harmful effects of particulate matter with an aerodynamic diameter of <2.5 µm (PM2.5) and its association with acute coronary syndrome (ACS) has gained increased attention in recent years. Significant associations between PM2.5 and ACS have been found in most studies, although sometimes only observed in specific subgroups. PM2.5-induced detrimental effects and ACS arise through multiple mechanisms, including endothelial injury, an enhanced inflammatory response, oxidative stress, autonomic dysfunction, and mitochondria damage as well as genotoxic effects. These effects can lead to a series of physiopathological changes including coronary artery atherosclerosis, hypertension, an imbalance between energy supply and demand to heart tissue, and a systemic hypercoagulable state. Effective strategies to prevent the harmful effects of PM2.5 include reducing pollution sources of PM2.5 and population exposure to PM2.5, and governments and organizations publicizing the harmful effects of PM2.5 and establishing air quality standards for PM2.5. PM2.5 exposure is a significant risk factor for ACS, and effective strategies with which to prevent both susceptible and healthy populations from an increased risk for ACS have important clinical significance in the prevention and treatment of ACS.
Collapse
|
10
|
Malik IA, Stange I, Martius G, Cameron S, Rave-Fränk M, Hess CF, Ellenrieder V, Wolff HA. Role of PECAM-1 in radiation-induced liver inflammation. J Cell Mol Med 2015; 19:2441-52. [PMID: 26177067 PMCID: PMC4594685 DOI: 10.1111/jcmm.12630] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/13/2015] [Indexed: 12/25/2022] Open
Abstract
Platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31) is known to play an important role in hepatic inflammation. Therefore, we investigated the role of PECAM-1 in wild-type (WT) and knock-out (KO)-mice after single-dose liver irradiation (25 Gy). Both, at mRNA and protein level, a time-dependent decrease in hepatic PECAM-1, corresponding to an increase in intercellular cell adhesion molecule-1 (ICAM-1) (6 hrs) was detected in WT-mice after irradiation. Immunohistologically, an increased number of neutrophil granulocytes (NG) (but not of mononuclear phagocytes) was observed in the liver of WT and PECAM-1-KO mice at 6 hrs after irradiation. The number of recruited NG was higher and prolonged until 24 hrs in KO compared to WT-mice. Correspondingly, a significant induction of hepatic tumour necrosis factor (TNF)-α and CXC-chemokines (KC/CXCL1 interleukin-8/CXCL8) was detected together with an elevation of serum liver transaminases (6–24 hrs) in WT and KO-mice. Likewise, phosphorylation of signal transducer and activator of transcription-3 (STAT-3) was observed in both animal groups after irradiation. The level of all investigated proteins as well as of the liver transaminases was significantly higher in KO than WT-mice. In the cell-line U937, irradiation led to a reduction in PECAM-1 in parallel to an increased ICAM-1 expression. TNF-α-blockage by anti-TNF-α prevented this change in both proteins in cell culture. Radiation-induced stress conditions induce a transient accumulation of granulocytes within the liver by down-regulation/absence of PECAM-1. It suggests that reduction/lack in PECAM-1 may lead to greater and prolonged inflammation which can be prevented by anti-TNFα.
Collapse
Affiliation(s)
- Ihtzaz Ahmed Malik
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Ina Stange
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Gesa Martius
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Silke Cameron
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Margret Rave-Fränk
- Department of Radiotherapy and Radiooncology, University Medical Center Goettingen, Goettingen, Germany
| | - Clemens Friedrich Hess
- Department of Radiotherapy and Radiooncology, University Medical Center Goettingen, Goettingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Hendrik Andreas Wolff
- Department of Radiotherapy and Radiooncology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
11
|
Wang S, Iring A, Strilic B, Albarrán Juárez J, Kaur H, Troidl K, Tonack S, Burbiel JC, Müller CE, Fleming I, Lundberg JO, Wettschureck N, Offermanns S. P2Y₂ and Gq/G₁₁ control blood pressure by mediating endothelial mechanotransduction. J Clin Invest 2015; 125:3077-86. [PMID: 26168216 DOI: 10.1172/jci81067] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022] Open
Abstract
Elevated blood pressure is a key risk factor for developing cardiovascular diseases. Blood pressure is largely determined by vasodilatory mediators, such as nitric oxide (NO), that are released from the endothelium in response to fluid shear stress exerted by the flowing blood. Previous work has identified several mechanotransduction signaling processes that are involved in fluid shear stress-induced endothelial effects, but how fluid shear stress initiates the response is poorly understood. Here, we evaluated human and bovine endothelial cells and found that the purinergic receptor P2Y2 and the G proteins Gq/G11 mediate fluid shear stress-induced endothelial responses, including [Ca2+]i transients, activation of the endothelial NO synthase (eNOS), phosphorylation of PECAM-1 and VEGFR-2, as well as activation of SRC and AKT. In response to fluid shear stress, endothelial cells released ATP, which activates the purinergic P2Y2 receptor. Mice with induced endothelium-specific P2Y2 or Gq/G11 deficiency lacked flow-induced vasodilation and developed hypertension that was accompanied by reduced eNOS activation. Together, our data identify P2Y2 and Gq/G11 as a critical endothelial mechanosignaling pathway that is upstream of previously described mechanotransduction processes and demonstrate that P2Y2 and Gq/G11 are required for basal endothelial NO formation, vascular tone, and blood pressure.
Collapse
|
12
|
Collins C, Tzima E, Patterson C. Inflammation. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
13
|
McCormick ME, Collins C, Makarewich CA, Chen Z, Rojas M, Willis MS, Houser SR, Tzima E. Platelet endothelial cell adhesion molecule-1 mediates endothelial-cardiomyocyte communication and regulates cardiac function. J Am Heart Assoc 2015; 4:e001210. [PMID: 25600142 PMCID: PMC4330051 DOI: 10.1161/jaha.114.001210] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Dilated cardiomyopathy is characterized by impaired contractility of cardiomyocytes, ventricular chamber dilatation, and systolic dysfunction. Although mutations in genes expressed in the cardiomyocyte are the best described causes of reduced contractility, the importance of endothelial‐cardiomyocyte communication for proper cardiac function is increasingly appreciated. In the present study, we investigate the role of the endothelial adhesion molecule platelet endothelial cell adhesion molecule (PECAM‐1) in the regulation of cardiac function. Methods and Results Using cell culture and animal models, we show that PECAM‐1 expressed in endothelial cells (ECs) regulates cardiomyocyte contractility and cardiac function via the neuregulin‐ErbB signaling pathway. Conscious echocardiography revealed left ventricular (LV) chamber dilation and systolic dysfunction in PECAM‐1−/− mice in the absence of histological abnormalities or defects in cardiac capillary density. Despite deficits in global cardiac function, cardiomyocytes isolated from PECAM‐1−/− hearts displayed normal baseline and isoproterenol‐stimulated contractility. Mechanistically, absence of PECAM‐1 resulted in elevated NO/ROS signaling and NRG‐1 release from ECs, which resulted in augmented phosphorylation of its receptor ErbB2. Treatment of cardiomyocytes with conditioned media from PECAM‐1−/− ECs resulted in enhanced ErbB2 activation, which was normalized by pre‐treatment with an NRG‐1 blocking antibody. To determine whether normalization of increased NRG‐1 levels could correct cardiac function, PECAM‐1−/− mice were treated with the NRG‐1 blocking antibody. Echocardiography showed that treatment significantly improved cardiac function of PECAM‐1−/− mice, as revealed by increased ejection fraction and fractional shortening. Conclusions We identify a novel role for PECAM‐1 in regulating cardiac function via a paracrine NRG1‐ErbB pathway. These data highlight the importance of tightly regulated cellular communication for proper cardiac function.
Collapse
Affiliation(s)
- Margaret E. McCormick
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC (M.E.M.C., C.C., Z.C., E.T.)
| | - Caitlin Collins
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC (M.E.M.C., C.C., Z.C., E.T.)
| | - Catherine A. Makarewich
- Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (C.A.M., S.R.H.)
| | - Zhongming Chen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC (M.E.M.C., C.C., Z.C., E.T.)
| | - Mauricio Rojas
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC (M.R., E.T.)
| | - Monte S. Willis
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (M.S.W.)
| | - Steven R. Houser
- Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (C.A.M., S.R.H.)
| | - Ellie Tzima
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC (M.E.M.C., C.C., Z.C., E.T.)
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC (M.R., E.T.)
| |
Collapse
|
14
|
Li Y, Zhao YJ, Zou QY, Zhang K, Wu YM, Zhou C, Wang K, Zheng J. Preeclampsia does not alter vascular growth and expression of CD31 and vascular endothelial cadherin in human placentas. J Histochem Cytochem 2014; 63:22-31. [PMID: 25362142 DOI: 10.1369/0022155414558063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Preeclampsia is characterized by maternal endothelial dysfunction (e.g., increased maternal vascular permeability caused by the disassembly of endothelial junction proteins). However, it is unclear if preeclampsia is associated with impaired vascular growth and expression of endothelial junction proteins in human placentas. Herein, we examined vascular growth in placentas from women with normal term (NT) and preeclamptic (PE) pregnancies using two endothelial junction proteins as endothelial markers: CD31 and vascular endothelial-cadherin (VE-Cad). We also compared protein and mRNA expression of CD31 and VE-Cad between NT and PE placentas, and determined the alternatively spliced expression of CD31 using PCR. We found that CD31 and VE-Cad were immunolocalized predominantly in villous endothelial cells. However, capillary number density (total capillary number per unit villous area) and capillary area density (total capillary lumen area per unit villous area) as well as CD31 and VE-Cad protein and mRNA levels were similar between NT and PE placentas. PCR in combination with sequence analysis revealed a single, full-length CD31, suggesting that there are no alternatively spliced isoform of CD31 expressed in placentas. These data indicate that preeclampsia does not significantly affect vascular growth or the expression of endothelial junction proteins in human placentas.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ)
| | - Ying-Jie Zhao
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ),Department of Rheumatology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China (YJZ)
| | - Qing-Yun Zou
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ)
| | - Kevin Zhang
- Department of Biological Sciences, Dartmouth College, Hanover, NH (KZ)
| | - Yan-Ming Wu
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China (YMW, KW)
| | - Chi Zhou
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ)
| | - Kai Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China (YMW, KW)
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ),Department of Cardiovascular Medicine, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, P.R. China (JZ)
| |
Collapse
|
15
|
Cassuto J, Dou H, Czikora I, Szabo A, Patel VS, Kamath V, Belin de Chantemele E, Feher A, Romero MJ, Bagi Z. Peroxynitrite disrupts endothelial caveolae leading to eNOS uncoupling and diminished flow-mediated dilation in coronary arterioles of diabetic patients. Diabetes 2014; 63:1381-93. [PMID: 24353182 PMCID: PMC3964507 DOI: 10.2337/db13-0577] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 12/10/2013] [Indexed: 12/18/2022]
Abstract
Peroxynitrite (ONOO(-)) contributes to coronary microvascular dysfunction in diabetes mellitus (DM). We hypothesized that in DM, ONOO(-) interferes with the function of coronary endothelial caveolae, which plays an important role in nitric oxide (NO)-dependent vasomotor regulation. Flow-mediated dilation (FMD) of coronary arterioles was investigated in DM (n = 41) and non-DM (n = 37) patients undergoing heart surgery. NO-mediated coronary FMD was significantly reduced in DM patients, which was restored by ONOO(-) scavenger, iron-(III)-tetrakis(N-methyl-4'pyridyl)porphyrin-pentachloride, or uric acid, whereas exogenous ONOO(-) reduced FMD in non-DM subjects. Immunoelectron microscopy demonstrated an increased 3-nitrotyrosine formation (ONOO(-)-specific protein nitration) in endothelial plasma membrane in DM, which colocalized with caveolin-1 (Cav-1), the key structural protein of caveolae. The membrane-localized Cav-1 was significantly reduced in DM and also in high glucose-exposed coronary endothelial cells. We also found that DM patients exhibited a decreased number of endothelial caveolae, whereas exogenous ONOO(-) reduced caveolae number. Correspondingly, pharmacological (methyl-β-cyclodextrin) or genetic disruption of caveolae (Cav-1 knockout mice) abolished coronary FMD, which was rescued by sepiapterin, the stable precursor of NO synthase (NOS) cofactor, tetrahydrobiopterin. Sepiapterin also restored coronary FMD in DM patients. Thus, we propose that ONOO(-) selectively targets and disrupts endothelial caveolae, which contributes to NOS uncoupling, and, hence, reduced NO-mediated coronary vasodilation in DM patients.
Collapse
Affiliation(s)
- James Cassuto
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Huijuan Dou
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Istvan Czikora
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Andras Szabo
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Vijay S. Patel
- Department of Surgery, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Vinayak Kamath
- Department of Surgery, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | | | - Attila Feher
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Maritza J. Romero
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Zsolt Bagi
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA
| |
Collapse
|
16
|
Privratsky JR, Newman PJ. PECAM-1: regulator of endothelial junctional integrity. Cell Tissue Res 2014; 355:607-19. [PMID: 24435645 DOI: 10.1007/s00441-013-1779-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 12/09/2013] [Indexed: 12/15/2022]
Abstract
PECAM-1 (also known as CD31) is a cellular adhesion and signaling receptor comprising six extracellular immunoglobulin (Ig)-like homology domains, a short transmembrane domain and a 118 amino acid cytoplasmic domain that becomes serine and tyrosine phosphorylated upon cellular activation. PECAM-1 expression is restricted to blood and vascular cells. In circulating platelets and leukocytes, PECAM-1 functions largely as an inhibitory receptor that, via regulated sequential phosphorylation of its cytoplasmic domain, limits cellular activation responses. PECAM-1 is also highly expressed at endothelial cell intercellular junctions, where it functions as a mechanosensor, as a regulator of leukocyte trafficking and in the maintenance of endothelial cell junctional integrity. In this review, we will describe (1) the functional domains of PECAM-1 and how they contribute to its barrier-enhancing properties, (2) how the physical properties of PECAM-1 influence its subcellular localization and its ability to influence endothelial cell barrier function, (3) various stimuli that initiate PECAM-1 signaling and/or function at the endothelial junction and (4) cross-talk of PECAM-1 with other junctional molecules, which can influence endothelial cell function.
Collapse
Affiliation(s)
- Jamie R Privratsky
- Blood Research Institute, BloodCenter of Wisconsin, P.O. Box 2178, 638N. 18th Street, Milwaukee, WI, 53201, USA
| | | |
Collapse
|
17
|
Gokina NI, Chan SL, Chapman AC, Oppenheimer K, Jetton TL, Cipolla MJ. Inhibition of PPARγ during rat pregnancy causes intrauterine growth restriction and attenuation of uterine vasodilation. Front Physiol 2013; 4:184. [PMID: 23888144 PMCID: PMC3719025 DOI: 10.3389/fphys.2013.00184] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 06/26/2013] [Indexed: 12/12/2022] Open
Abstract
Decreased peroxisome proliferator-activated receptor gamma (PPARγ) activity is thought to have a major role in preeclampsia through abnormal placental development. However, the role of PPARγ in adaptation of the uteroplacental vasculature that may lead to placental hypoperfusion and fetal growth restriction during pregnancy is not known. Here, pregnant Sprague-Dawley rats (n = 11/group) were treated during the second half of pregnancy with the PPARγ inhibitor GW9662 (10 mg/kg/day in food) or vehicle. Pregnancy outcome and PPARγ mRNA, vasodilation and structural remodeling were determined in maternal uterine and mesenteric arteries. PPARγ was expressed in uterine vascular tissue of both non-pregnant and pregnant rats with ~2-fold greater expression in radial vs. main uterine arteries. PPARγ mRNA levels were significantly higher in uterine compared to mesenteric arteries. GW9662 treatment during pregnancy did not affect maternal physiology (body weight, glucose, blood pressure), mesenteric artery vasodilation or structural remodeling of uterine and mesenteric vessels. Inhibition of PPARγ for the last 10 days of gestation caused decreased fetal weights on both day 20 and 21 of gestation that was associated with impaired vasodilation of radial uterine arteries in response to acetylcholine and sodium nitroprusside. These results define an essential role of PPARγ in the control of uteroplacental vasodilatory function during pregnancy, an important determinant of blood flow to the placenta and fetus. Strategies that target PPARγ activation in the uterine circulation could have important therapeutic potential in treatment of pregnancies complicated by hypertension, diabetes or preeclampsia.
Collapse
Affiliation(s)
- Natalia I Gokina
- Department of Obstetrics, Gynecology and Reproductive Sciences, College of Medicine, University of Vermont Burlington, VT, USA
| | | | | | | | | | | |
Collapse
|
18
|
Kvietys PR, Granger DN. Role of reactive oxygen and nitrogen species in the vascular responses to inflammation. Free Radic Biol Med 2012; 52:556-592. [PMID: 22154653 PMCID: PMC3348846 DOI: 10.1016/j.freeradbiomed.2011.11.002] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/04/2011] [Accepted: 11/04/2011] [Indexed: 12/23/2022]
Abstract
Inflammation is a complex and potentially life-threatening condition that involves the participation of a variety of chemical mediators, signaling pathways, and cell types. The microcirculation, which is critical for the initiation and perpetuation of an inflammatory response, exhibits several characteristic functional and structural changes in response to inflammation. These include vasomotor dysfunction (impaired vessel dilation and constriction), the adhesion and transendothelial migration of leukocytes, endothelial barrier dysfunction (increased vascular permeability), blood vessel proliferation (angiogenesis), and enhanced thrombus formation. These diverse responses of the microvasculature largely reflect the endothelial cell dysfunction that accompanies inflammation and the central role of these cells in modulating processes as varied as blood flow regulation, angiogenesis, and thrombogenesis. The importance of endothelial cells in inflammation-induced vascular dysfunction is also predicated on the ability of these cells to produce and respond to reactive oxygen and nitrogen species. Inflammation seems to upset the balance between nitric oxide and superoxide within (and surrounding) endothelial cells, which is necessary for normal vessel function. This review is focused on defining the molecular targets in the vessel wall that interact with reactive oxygen species and nitric oxide to produce the characteristic functional and structural changes that occur in response to inflammation. This analysis of the literature is consistent with the view that reactive oxygen and nitrogen species contribute significantly to the diverse vascular responses in inflammation and supports efforts that are directed at targeting these highly reactive species to maintain normal vascular health in pathological conditions that are associated with acute or chronic inflammation.
Collapse
Affiliation(s)
- Peter R Kvietys
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA.
| |
Collapse
|
19
|
McCormick ME, Goel R, Fulton D, Oess S, Newman D, Tzima E. Platelet-endothelial cell adhesion molecule-1 regulates endothelial NO synthase activity and localization through signal transducers and activators of transcription 3-dependent NOSTRIN expression. Arterioscler Thromb Vasc Biol 2010; 31:643-9. [PMID: 21183735 DOI: 10.1161/atvbaha.110.216200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND NO produced by the endothelial NO synthase (eNOS) is an important regulator of cardiovascular physiological and pathological features. eNOS is activated by numerous stimuli, and its activity is tightly regulated. Platelet-endothelial cell adhesion molecule-1 (PECAM-1) has been implicated in regulating eNOS activity in response to shear stress. The current study was conducted to determine the role of PECAM-1 in the regulation of basal eNOS activity. METHODS AND RESULTS We demonstrate that PECAM-1-knockout ECs have increased basal eNOS activity and NO production. Mechanistically, increased eNOS activity is associated with a decrease in the inhibitory interaction of eNOS with caveolin-1, impaired subcellular localization of eNOS, and decreased eNOS traffic inducer (NOSTRIN) expression in the absence of PECAM-1. Furthermore, we demonstrate that activation of blunted signal transducers and activators of transcription 3 (STAT3) in the absence of PECAM-1 results in decreased NOSTRIN expression via direct binding of the signal transducers and activators of transcription 3 to the NOSTRIN promoter. CONCLUSIONS Our results reveal an elegant mechanism of eNOS regulation by PECAM-1 through signal transducers and activators of transcription 3-mediated transcriptional control of NOSTRIN.
Collapse
Affiliation(s)
- Margaret E McCormick
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
RATIONALE Hemodynamic forces caused by the altered blood flow in response to an occlusion lead to the induction of collateral remodeling and arteriogenesis. Previous work showed that platelet endothelial cell adhesion molecule (PECAM)-1 is a component of a mechanosensory complex that mediates endothelial cell responses to shear stress. OBJECTIVE We hypothesized that PECAM-1 plays an important role in arteriogenesis and collateral remodeling. METHODS AND RESULTS PECAM-1 knockout (KO) and wild-type littermates underwent femoral artery ligation. Surprisingly, tissue perfusion and collateral-dependent blood flow were significantly increased in the KO mice immediately after surgery. Histology confirmed larger caliber of preexisting collaterals in the KO mice. Additionally, KO mice showed blunted recovery of perfusion from hindlimb ischemia and reduced collateral remodeling, because of deficits in shear stress-induced signaling, including activation of the nuclear factor κB pathway and inflammatory cell accumulation. Partial recovery was associated with normal responses to circumferential wall tension in the absence of PECAM-1, as evidenced by the upregulation of ephrin B2 and monocyte chemoattractant protein-1, which are 2 stretch-induced regulators of arteriogenesis, both in vitro and in vivo. CONCLUSIONS Our findings suggest a novel role for PECAM-1 in arteriogenesis and collateral remodeling. Furthermore, we identify PECAM-1 as the first molecule that determines preexisting collateral diameter.
Collapse
Affiliation(s)
- Zhongming Chen
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
21
|
Prieto D, Kaminski PM, Bagi Z, Ahmad M, Wolin MS. Hypoxic relaxation of penile arteries: involvement of endothelial nitric oxide and modulation by reactive oxygen species. Am J Physiol Heart Circ Physiol 2010; 299:H915-24. [PMID: 20581086 DOI: 10.1152/ajpheart.00382.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although obesity-related cardiovascular disease and hypoxia are associated with erectile dysfunction, little is known about the direct effects of hypoxia on penile arteries. In the present study, the effects of acute hypoxia (Po(2) = approximately 10 Torr, 20 min) were investigated in isolated penile arteries to determine the influence of endothelium removal, nitric oxide (NO) synthase (NOS), cyclooxygenase (COX), NADPH oxidase, changes in reactive oxygen species (ROS), and a high-fat diet. Hypoxia-relaxed penile arteries contracted with phenylephrine by approximately 50%. Relaxation to hypoxia and acetylcholine was reduced by endothelium removal and by inhibition of NOS (N(omega)-nitro-l-arginine) and COX (indomethacin) but was enhanced by Tempol and by NADPH oxidase inhibition with apocynin and gp91ds-tat. Basal superoxide levels detected by lucigenin chemiluminescence were reduced by Tempol and gp91ds-tat and were enhanced by NOS blockade. Hypoxic relaxant responses were enhanced by catalase and ebselen. Exogenous peroxide evoked relaxations of penile arteries, which were partially inhibited by endothelium removal and by the inhibition of COX and extracellular signal-regulated mitogen-activated protein kinase (MAPK) but enhanced by p38 MAPK blockade. The NO-dependent component of relaxation to hypoxia was impaired in penile arteries from high-fat diet-fed, obese rats associated with increased superoxide production. Thus hypoxic relaxation of penile arteries is partially mediated by endothelial NO in a manner that is normally attenuated by endogenous ROS production. Obesity further increases superoxide production and impairs the influence of NO. Therefore, cardiovascular disease involving decreased NO bioavailability and/or enhanced ROS generation may contribute to erectile dysfunction through impairing the relaxation of penile arteries to hypoxia.
Collapse
Affiliation(s)
- Dolores Prieto
- Dept. of Physiology, Faculty of Pharmacy, Universidad Complutense de Madrid, 28040-Madrid, Spain.
| | | | | | | | | |
Collapse
|
22
|
Privratsky JR, Tourdot BE, Newman DK, Newman PJ. The anti-inflammatory actions of platelet endothelial cell adhesion molecule-1 do not involve regulation of endothelial cell NF-kappa B. THE JOURNAL OF IMMUNOLOGY 2010; 184:3157-63. [PMID: 20173029 DOI: 10.4049/jimmunol.0901944] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PECAM-1 is a cell adhesion and signaling receptor that is expressed on many hematopoietic cells and at endothelial cell-cell junctions. Accumulating evidence from a number of in vitro and in vivo model systems suggests that PECAM-1 suppresses cytokine production and vascular permeability induced by a wide range of inflammatory stimuli. In several of these models of inflammatory disease, endothelial, and not leukocyte or platelet, PECAM-1 conferred protection against inflammatory insult. However, the mechanism by which endothelial PECAM-1 functions as an anti-inflammatory protein is poorly understood. It was recently suggested that PECAM-1 exerts its anti-inflammatory effects in endothelial cells by inhibiting the activity of NF-kappaB, a proinflammatory transcription factor. To confirm and extend these observations, we examined the effect of engaging, cross-linking, or expressing PECAM-1 on NF-kappaB activation in a variety of human cells. PECAM-1 had no effect on the phosphorylation of the NF-kappaB inhibitory protein, IkappaBalpha; on the nuclear translocation of NF-kappaB; on the suppression of cytokine-induced transcriptional activation of an NF-kappaB luciferase reporter plasmid; or on the cytokine-stimulated upregulation of ICAM-1, an NF-kappaB target gene, in endothelial cells. Taken together, these studies strongly suggest that the anti-inflammatory actions of PECAM-1 in endothelial cells are not likely to involve its regulation of NF-kappaB.
Collapse
Affiliation(s)
- Jamie R Privratsky
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201, USA
| | | | | | | |
Collapse
|
23
|
Chen Z, Tzima E. PECAM-1 is necessary for flow-induced vascular remodeling. Arterioscler Thromb Vasc Biol 2009; 29:1067-73. [PMID: 19390054 DOI: 10.1161/atvbaha.109.186692] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular remodeling is a physiological process that occurs in response to long-term changes in hemodynamic conditions, but may also contribute to the pathophysiology of intima-media thickening (IMT) and vascular disease. Shear stress detection by the endothelium is thought to be an important determinant of vascular remodeling. Previous work showed that platelet endothelial cell adhesion molecule-1 (PECAM-1) is a component of a mechanosensory complex that mediates endothelial cell (EC) responses to shear stress. METHODS AND RESULTS We tested the hypothesis that PECAM-1 contributes to vascular remodeling by analyzing the response to partial carotid artery ligation in PECAM-1 knockout mice and wild-type littermates. PECAM-1 deficiency resulted in impaired vascular remodeling and significantly reduced IMT in areas of low flow. Inward remodeling was associated with PECAM-1-dependent NFkappaB activation, surface adhesion molecule expression, and leukocyte infiltration as well as Akt activation and vascular cell proliferation. CONCLUSIONS PECAM-1 plays a crucial role in the activation of the NFkappaB and Akt pathways and inflammatory cell accumulation during vascular remodeling and IMT. Elucidation of some of the signals that drive vascular remodeling represent pharmacologically tractable targets for the treatment of restenosis after balloon angioplasty or stent placement.
Collapse
Affiliation(s)
- Zhongming Chen
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, NC 27599, USA
| | | |
Collapse
|
24
|
Bergom C, Paddock C, Gao C, Holyst T, Newman DK, Newman PJ. An alternatively spliced isoform of PECAM-1 is expressed at high levels in human and murine tissues, and suggests a novel role for the C-terminus of PECAM-1 in cytoprotective signaling. J Cell Sci 2008; 121:1235-42. [PMID: 18388311 DOI: 10.1242/jcs.025163] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Ig-ITIM family member PECAM-1 is expressed in vascular and endothelial cells, and its functions include suppression of mitochondria-dependent apoptosis. Previous studies have identified distinct PECAM-1 cytoplasmic domain splice variants at the mRNA, but not protein, level. Several relatively abundant mRNA isoforms lack exon 15 (Delta15) and would theoretically encode a protein with a truncated cytoplasmic domain and a unique C-terminal sequence. Using a novel rabbit polyclonal antibody that specifically recognizes Delta15 PECAM-1, we found that the Delta15 PECAM-1 isoform was expressed in human tissues, including brain, testes and ovary. This isoform was also expressed on the cell surface of human platelets, human umbilical vein endothelial cells (HUVECs) and the Jurkat T-cell leukemia, human erythroleukemia (HEL) and U937 histiocytic lymphoma cell lines. Furthermore, murine platelets and lung lysates demonstrated abundant amounts of exon-15-deficient PECAM-1. Functional studies revealed that Delta15 PECAM-1 retains both its homophilic binding capacity and its ability to signal by means of its immunoreceptor tyrosine-based inhibitory motif (ITIM) domains. Delta15 PECAM-1 was unable, however, to protect against apoptosis induced by overexpression of Bax or treatment with the chemotherapy agent etoposide. These studies suggest a novel role for the PECAM-1 C-terminus in cytoprotective signaling and highlight a need for further characterization of expression of PECAM-1 isoforms in normal and malignant tissues.
Collapse
Affiliation(s)
- Carmen Bergom
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Road, Milwaukee, WI 53201, USA
| | | | | | | | | | | |
Collapse
|
25
|
Goel R, Schrank BR, Arora S, Boylan B, Fleming B, Miura H, Newman PJ, Molthen RC, Newman DK. Site-specific effects of PECAM-1 on atherosclerosis in LDL receptor-deficient mice. Arterioscler Thromb Vasc Biol 2008; 28:1996-2002. [PMID: 18669884 DOI: 10.1161/atvbaha.108.172270] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Atherosclerosis is a vascular disease that involves lesion formation at sites of disturbed flow under the influence of genetic and environmental factors. Endothelial expression of adhesion molecules that enable infiltration of immune cells is important for lesion development. Platelet/endothelial cell adhesion molecule-1 (PECAM-1; CD31) is an adhesion and signaling receptor expressed by many cells involved in atherosclerotic lesion development. PECAM-1 transduces signals required for proinflammatory adhesion molecule expression at atherosusceptible sites; thus, it is predicted to be proatherosclerotic. PECAM-1 also inhibits inflammatory responses, on which basis it is predicted to be atheroprotective. METHODS AND RESULTS We evaluated herein the effect of PECAM-1 deficiency on development of atherosclerosis in LDL receptor-deficient mice. We found that PECAM-1 has both proatherosclerotic and atheroprotective effects, but that the former dominate in the inner curvature of the aortic arch whereas the latter dominate in the aortic sinus, branching arteries, and descending aorta. Endothelial cell expression of PECAM-1 was sufficient for its atheroprotective effects in the aortic sinus but not in the descending aorta, where the atheroprotective effects of PECAM-1 also required its expression on bone marrow-derived cells. CONCLUSIONS We conclude that PECAM-1 influences initiation and progression of atherosclerosis both positively and negatively, and that it does so in a site-specific manner.
Collapse
Affiliation(s)
- Reema Goel
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Attenuation of retinal vascular development and neovascularization in PECAM-1-deficient mice. Dev Biol 2008; 315:72-88. [PMID: 18206868 DOI: 10.1016/j.ydbio.2007.12.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 11/20/2007] [Accepted: 12/04/2007] [Indexed: 11/20/2022]
Abstract
Platelet-endothelial cell adhesion molecule-1 (PECAM-1/CD31) is expressed on the surface of endothelial cells (EC) at high levels with important roles in angiogenesis and inflammation. However, the physiological role PECAM-1 plays during vascular development and angiogenesis remains largely unknown. Here we determined the role of PECAM-1 in the postnatal development of retinal vasculature and retinal neovascularization during oxygen-induced ischemic retinopathy (OIR) using PECAM-1-deficient (PECAM-1-/-) mice. A significant decrease in retinal vascular density was observed in PECAM-1-/- mice compared with PECAM-1+/+ mice. This was attributed to a decreased number of EC in the retinas of PECAM-1-/- mice. An increase in the rate of apoptosis was observed in retinal vessels of PECAM-1-/- mice, which was compensated, in part, by an increase in the rate of proliferation. However, the development and regression of hyaloid vasculature were not affected in the absence of PECAM-1. We did not observe a significant defect in astrocytes, the number of endothelial tip cell filopodias, and the rate of developing retinal vasculature progression in PECAM-1-/- mice. However, we observed aberrant organization of arterioles and venules, decreased secondary branching, and dilated vessels in retinal vasculature of PECAM-1-/- mice. In addition, retinal neovascularization was attenuated in PECAM-1-/- mice during OIR despite an expression of VEGF similar to that of PECAM-1+/+ mice. Mechanistically, these changes were associated with an increase in EphB4 and ephrin B2, and a decrease in eNOS, expression in retinal vasculature of PECAM-1-/- mice. These results suggest that PECAM-1 expression and its potential interactions with EphB4/ephrin B2 and eNOS are important for survival, migration, and functional organization of EC during retinal vascular development and angiogenesis.
Collapse
|
27
|
Vouyouka AG, Jiang Y, Rastogi R, Basson MD. Ambient pressure upregulates nitric oxide synthase in a phosphorylated-extracellular regulated kinase- and protein kinase C-dependent manner. J Vasc Surg 2006; 44:1076-1084. [PMID: 17098545 DOI: 10.1016/j.jvs.2006.06.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Accepted: 06/27/2006] [Indexed: 10/23/2022]
Abstract
PURPOSE Using endothelial cell/smooth muscle cell (SMC) cocultures, we have demonstrated that pressurized endothelial cell coculture inhibits SMC proliferation and promotes apoptosis, and that this effect is transferable through pressurized endothelial medium. We now hypothesized that endothelial nitric oxide synthase (eNOS) plays a significant role in mediating these pressure-induced effects. METHODS Conditioned media from endothelial cells and SMCs exposed to ambient and increased pressure were transferred to recipient SMCs. We counted cells after 5 days of incubation with these media and evaluated eNOS and inducible NOS (iNOS) levels by Western blot. RESULTS Conditioned media from pressurized endothelial cells significantly decreased recipient SMC counts. This effect was sustained when N-nitro-L-arginine-methyl ester (L-NAME) was added to recipient cells but abolished when L-NAME was added to donor cells. SMCs were then exposed to control and pressurized conditions in monoculture or in coculture with endothelial cells. Pressure and coculture caused similar increase in iNOS levels but had no additive effect in combination. Finally, endothelial cells were exposed to control and pressurized environments. Pressure caused a 24% +/- 1.6% increase in eNOS protein (P = .04, n = 12). This effect was sustained when cells were treated with L-NAME (32% +/- 1.6% increase, P = .02) but abolished when endothelial cells were treated with calphostin C or PD98059 to block protein kinase C (PKC) or extracellular regulated kinase (ERK). Pressure also increased endothelial phosphorylated ERK (p-ERK) by 1.8-fold to 2.6-fold compared with control conditions after exposure of 2, 4, and 6 hours (P = .02, n = 4). This increase was sustained after pretreatment with calphostin C. CONCLUSION Pressure modulates endothelial cell effects on SMC growth by increasing eNOS in an ERK-dependent and PKC-dependent manner. CLINICAL RELEVANCE Intimal hyperplasia is the main cause for restenosis that complicates 10% to 30% of all such vascular procedures and 30% to 40% of endovascular procedures. This article provides some novel information about smooth muscle cell/endothelial cell interaction, one of the main regulators of vascular remodeling and intimal hyperplasia. The role of endothelial cell/smooth muscle cell interaction cannot be studied well in vivo because these interactions cannot be distinguished from other factors that coexist in vivo, such as flow dynamics, matrix proteins, inflammatory factors, and interactions with other cells in the vascular wall and in the bloodstream. In this work, we use pressure as a triggering stimulus to alter in vitro endothelial behavior and identify important changes in endothelial regulation of smooth muscle cell biology. The pathways involved in this process and discussed in this article could ultimately be used to manipulate endothelial cell/smooth muscle cell interaction in clinical disease.
Collapse
MESH Headings
- Animals
- Apoptosis
- Blotting, Western
- Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors
- Cell Proliferation
- Cells, Cultured
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Enzyme Inhibitors/pharmacology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Flavonoids/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- NG-Nitroarginine Methyl Ester/pharmacology
- Naphthalenes/pharmacology
- Nitric Oxide Synthase Type II/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Phosphorylation
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C/metabolism
- Rats
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Angela G Vouyouka
- Division of Vascular and General Surgery, John D. Dingell VA Medical Center and Wayne State University School of Medicine, Detroit, MI 48201-1932, USA.
| | | | | | | |
Collapse
|
28
|
Bai N, Khazaei M, van Eeden SF, Laher I. The pharmacology of particulate matter air pollution-induced cardiovascular dysfunction. Pharmacol Ther 2006; 113:16-29. [PMID: 16920197 DOI: 10.1016/j.pharmthera.2006.06.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Accepted: 06/14/2006] [Indexed: 01/30/2023]
Abstract
Since the London fog of 1952, in which more than 4000 people were killed in 4 days, the combined efforts of scientists from several disciplines, including those from the environmental health, clinical and biomedical disciplines, have raised serious concerns about the impact of air pollutants on human health. These environmental pollutants are rapidly being recognized as important and independent risk factors for several diseases such as asthma, chronic obstructive pulmonary disease, lung cancer, atherosclerosis, ischemic heart disease and stroke. Although the relative effects of particulate matter air pollution (aerodynamic diameter <10 microm, or PM(10)) are greater for respiratory than for cardiovascular deaths, the number of deaths attributable to PM(10) is much larger for cardiovascular than for respiratory reasons due to the higher prevalence of cardiovascular disease in the general population. This review summarizes current understanding of the mechanisms underlying the associations between PM(10) exposure and cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Ni Bai
- University of British Columbia, Department of Pharmacology and Therapeutics, Faculty of Medicine, 2176 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| | | | | | | |
Collapse
|