1
|
Blaha V, Rathouska JU, Langrova H, Blaha M, Studnicka J, Andrys C, Loefflerova V, Lanska M, Vejrazkova E, Nachtigal P, Stepanov A. Soluble endoglin as a biomarker of successful rheopheresis treatment in patients with age-related macular degeneration. Sci Rep 2024; 14:28902. [PMID: 39572693 PMCID: PMC11582623 DOI: 10.1038/s41598-024-80375-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
Age-related macular degeneration (AMD) is a progressive chronic disease causing visual impairment or central vision loss in the elderly. We hypothesized that successful rheopheresis would be associated with positive changes in soluble endoglin (sENG), PSCK9, alpha-2-macroglobulin (A2M), and hs-CRP levels. 31 elderly patients with the dry form of AMD, treated with rheopheresis with a follow-up period of at least 5 years and an average age of 68 ± 4 years, were evaluated. Each treated patient received a series of 8 procedures in 10 weeks and, after the 2-year period, another 2 procedures within 1 week. Then, the patients were followed up every 6 months and divided into the successfully treated and therapeutic failure group according to best-corrected visual acuity (BCVA), size of the drusen area, and the drusenoid pigment epithelium detachment (DPED). Based on the ophthalmological assessment, rheopheresis treatment was successful in 73% of AMD patients. The therapy was associated with a significant decrease in total cholesterol, LDL-C, HDL-C, apoprotein B, lipoprotein (a) levels, and rheologically important parameters, irrespective of the therapy's success or failure. The success of rheopheresis therapy was exclusively related to a significant decrease in sENG and A2M levels. Over the long term, rheopheresis prevented the decline of BCVA, reduced the DPED and area of macular drusen, and improved the preservation of an intact photoreceptor ellipsoid zone in most patients. Moreover, we showed for the first time that sENG and A2M could be potentially sensitive biomarkers of successful rheopheresis procedure, irrespective of lipid parameters changes.
Collapse
Affiliation(s)
- Vladimir Blaha
- Faculty of Medicine in Hradec Kralove, 3rd Department of Internal Medicine - Metabolism and Gerontology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Jana Urbankova Rathouska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05, Hradec Kralove, Czech Republic
| | - Hana Langrova
- Faculty of Medicine in Hradec Kralove, Department of Ophthalmology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Milan Blaha
- Faculty of Medicine in Hradec Kralove, 4th Department of Internal Medicine - Hematology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Jan Studnicka
- Faculty of Medicine in Hradec Kralove, Department of Ophthalmology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Ctirad Andrys
- Department of Immunology and Allergology, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | | | - Miriam Lanska
- Faculty of Medicine in Hradec Kralove, 4th Department of Internal Medicine - Hematology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Eva Vejrazkova
- Faculty of Medicine in Hradec Kralove, 4th Department of Internal Medicine - Hematology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05, Hradec Kralove, Czech Republic.
| | - Alexandr Stepanov
- Faculty of Medicine in Hradec Kralove, Department of Ophthalmology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
2
|
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024; 12:1795. [PMID: 39200259 PMCID: PMC11351371 DOI: 10.3390/biomedicines12081795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) substantially increase the risk for intracerebral hemorrhage (ICH), which is associated with significant morbidity and mortality. However, the treatment options for bAVMs are severely limited, primarily relying on invasive methods that carry their own risks for intraoperative hemorrhage or even death. Currently, there are no pharmaceutical agents shown to treat this condition, primarily due to a poor understanding of bAVM pathophysiology. For the last decade, bAVM research has made significant advances, including the identification of novel genetic mutations and relevant signaling in bAVM development. However, bAVM pathophysiology is still largely unclear. Further investigation is required to understand the detailed cellular and molecular mechanisms involved, which will enable the development of safer and more effective treatment options. Endothelial cells (ECs), the cells that line the vascular lumen, are integral to the pathogenesis of bAVMs. Understanding the fundamental role of ECs in pathological conditions is crucial to unraveling bAVM pathophysiology. This review focuses on the current knowledge of bAVM-relevant signaling pathways and dysfunctions in ECs, particularly the endothelial-to-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
| | | | | | | | | | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.Y.J.); (A.E.B.); (B.H.F.); (P.R.C.); (E.S.P.)
| |
Collapse
|
3
|
Khan MAO, Suvvari TK, Harooni SAS, Khan AA, Anees S, Bushra. Assessment of soluble thrombomodulin and soluble endoglin as endothelial dysfunction biomarkers in seriously ill surgical septic patients: correlation with organ dysfunction and disease severity. Eur J Trauma Emerg Surg 2024; 50:897-904. [PMID: 37741913 DOI: 10.1007/s00068-023-02369-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Sepsis, a complex condition characterized by dysregulated immune response and organ dysfunction, is a leading cause of mortality in ICU patients. Current diagnostic and prognostic approaches primarily rely on non-specific biomarkers and illness severity scores, despite early endothelial activation being a key feature of sepsis. This study aimed to evaluate the levels of soluble thrombomodulin and soluble endoglin in seriously ill surgical septic patients and explore their association with organ dysfunction and disease severity. METHODOLOGY A case control study was conducted from March 2022 to November 2022, involving seriously ill septic surgical patients. Baseline clinical and laboratory data were collected within 24 h of admission to the Surgical Intensive Care Unit. This included information such as age, sex, hemodynamic parameters, blood chemistry, SOFA score, qSOFA score, and APACHE-II score. A proforma was filled out to record these details. The outcome of each patient was noted at the time of discharge. RESULTS The study found significantly elevated levels of soluble thrombomodulin and soluble endoglin in seriously ill surgical septic patients. The RTqPCR analysis revealed a positive correlation between soluble thrombomodulin and soluble endoglin levels with the qSOFA score, as well as, there was a positive association between RTqPCR soluble thrombomodulin and the SOFA score. These findings indicate a correlation between these biomarkers and organ dysfunction and disease severity. CONCLUSION The study concludes that elevated levels of soluble thrombomodulin and soluble endoglin can serve as endothelial biomarkers for early diagnosis and prognostication in seriously ill surgical septic patients.
Collapse
Affiliation(s)
| | - Tarun Kumar Suvvari
- Rangaraya Medical College, Kakinada, India
- Squad Medicine and Research (SMR), Vizag, Andhra Pradesh, India
| | | | - Aleem Ahmed Khan
- Central Laboratory for Stem Cell Research and Translational Medicine, Deccan College of Medical Sciences, Hyderabad, Telangana, India
| | - Syyeda Anees
- Department of Biochemistry, Deccan College of Medical Sciences, Hyderabad, Telangana, India
| | - Bushra
- Central Laboratory for Stem Cell Research and Translational Medicine, Deccan College of Medical Sciences, Hyderabad, Telangana, India
| |
Collapse
|
4
|
Nemeckova I, Eissazadeh S, Rathouska JU, Silhavy J, Malinska H, Pravenec M, Nachtigal P. Transgenic human C-reactive protein affects oxidative stress but not inflammation biomarkers in the aorta of spontaneously hypertensive rats. BMC Cardiovasc Disord 2024; 24:211. [PMID: 38627621 PMCID: PMC11020172 DOI: 10.1186/s12872-024-03870-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/30/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND C-reactive protein (CRP) is an acute inflammatory protein detected in obese patients with metabolic syndrome. Moreover, increased CRP levels have been linked with atherosclerotic disease, congestive heart failure, and ischemic heart disease, suggesting that it is not only a biomarker but also plays an active role in the pathophysiology of cardiovascular diseases. Since endothelial dysfunction plays an essential role in various cardiovascular pathologies and is characterized by increased expression of cell adhesion molecules and inflammatory markers, we aimed to detect specific markers of endothelial dysfunction, inflammation, and oxidative stress in spontaneously hypertensive rats (SHR) expressing human CRP. This model is genetically predisposed to the development of the metabolic syndrome. METHODS Transgenic SHR male rats (SHR-CRP) and non-transgenic SHR (SHR) at the age of 8 months were used. Metabolic profile (including serum and tissue triglyceride (TAG), serum insulin concentrations, insulin-stimulated incorporation of glucose, and serum non-esterified fatty acids (NEFA) levels) was measured. In addition, human serum CRP, MCP-1 (monocyte chemoattractant protein-1), and adiponectin were evaluated by means of ELISA, histological analysis was used to study morphological changes in the aorta, and western blot analysis of aortic tissue was performed to detect expression of endothelial, inflammatory, and oxidative stress markers. RESULTS The presence of human CRP was associated with significantly decreased insulin-stimulated glycogenesis in skeletal muscle, increased muscle and hepatic accumulation of TAG and decreased plasmatic cGMP concentrations, reduced adiponectin levels, and increased monocyte chemoattractant protein-1 (MCP-1) levels in the blood, suggesting pro-inflammatory and presence of multiple features of metabolic syndrome in SHR-CRP animals. Histological analysis of aortic sections did not reveal any visible morphological changes in animals from both SHR and SHR-CRP rats. Western blot analysis of the expression of proteins related to the proper function of endothelium demonstrated significant differences in the expression of p-eNOS/eNOS in the aorta, although endoglin (ENG) protein expression remained unaffected. In addition, the presence of human CRP in SHR in this study did not affect the expression of inflammatory markers, namely p-NFkB, P-selectin, and COX2 in the aorta. On the other hand, biomarkers related to oxidative stress, such as HO-1 and SOD3, were significantly changed, indicating the induction of oxidative stress. CONCLUSIONS Our findings demonstrate that CRP alone cannot fully induce the expression of endothelial dysfunction biomarkers, suggesting other risk factors of cardiovascular disorders are necessary to be involved to induce endothelial dysfunction with CRP.
Collapse
Affiliation(s)
- Ivana Nemeckova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 05, Czech Republic
| | - Samira Eissazadeh
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 05, Czech Republic
| | - Jana Urbankova Rathouska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 05, Czech Republic
| | - Jan Silhavy
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Hana Malinska
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Michal Pravenec
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 05, Czech Republic.
| |
Collapse
|
5
|
Gallo G, Savoia C. New Insights into Endothelial Dysfunction in Cardiometabolic Diseases: Potential Mechanisms and Clinical Implications. Int J Mol Sci 2024; 25:2973. [PMID: 38474219 DOI: 10.3390/ijms25052973] [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: 01/30/2024] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
The endothelium is a monocellular layer covering the inner surface of blood vessels. It maintains vascular homeostasis regulating vascular tone and permeability and exerts anti-inflammatory, antioxidant, anti-proliferative, and anti-thrombotic functions. When the endothelium is exposed to detrimental stimuli including hyperglycemia, hyperlipidemia, and neurohormonal imbalance, different biological pathways are activated leading to oxidative stress, endothelial dysfunction, increased secretion of adipokines, cytokines, endothelin-1, and fibroblast growth factor, and reduced nitric oxide production, leading eventually to a loss of integrity. Endothelial dysfunction has emerged as a hallmark of dysmetabolic vascular impairment and contributes to detrimental effects on cardiac metabolism and diastolic dysfunction, and to the development of cardiovascular diseases including heart failure. Different biomarkers of endothelial dysfunction have been proposed to predict cardiovascular diseases in order to identify microvascular and macrovascular damage and the development of atherosclerosis, particularly in metabolic disorders. Endothelial dysfunction also plays an important role in the development of severe COVID-19 and cardiovascular complications in dysmetabolic patients after SARS-CoV-2 infection. In this review, we will discuss the biological mechanisms involved in endothelial dysregulation in the context of cardiometabolic diseases as well as the available and promising biomarkers of endothelial dysfunction in clinical practice.
Collapse
Affiliation(s)
- Giovanna Gallo
- Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035-1039, 00189 Rome, Italy
| | - Carmine Savoia
- Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1035-1039, 00189 Rome, Italy
| |
Collapse
|
6
|
Saita E, Kishimoto Y, Aoyama M, Ohmori R, Kondo K, Momiyama Y. Low Plasma Levels of Soluble Endoglin and Cardiovascular Events in Patients Undergoing Coronary Angiography. Biomedicines 2023; 11:2975. [PMID: 38001975 PMCID: PMC10669441 DOI: 10.3390/biomedicines11112975] [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: 09/25/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
TGF-β is recognized as playing a protective role against atherosclerosis. Endoglin is a receptor for TGF-β, and its expression is upregulated in atherosclerotic plaques. Endoglin is secreted from the cell membrane into the circulation as a soluble form (sEng). We previously reported that plasma sEng levels were low in patients with coronary artery disease (CAD). However, the prognostic value of sEng levels has not been clarified. We investigated the association between plasma sEng levels and cardiovascular events in 403 patients who had an elective coronary angiography and were then followed up. Cardiovascular events were defined as cardiovascular death, myocardial infarction, unstable angina, heart failure, stroke, or coronary revascularization. Of the 403 patients, 209 (52%) had CAD. Plasma sEng levels were lower in patients with CAD than in those without CAD (median 4.26 vs. 4.41 ng/mL, p < 0.025). During a mean follow-up period of 7.5 ± 4.5 years, cardiovascular events occurred in 79 patients. Compared with 324 patients without events, 79 with events had lower sEng levels (3.95 vs. 4.39 ng/mL) and more often had an sEng level < 3.9 ng/mL (47% vs. 28%) (p < 0.02). A Kaplan-Meier analysis showed lower event-free survival in patients with sEng < 3.9 ng/mL than in those with ≥3.9 ng/mL (p < 0.02). In a multivariate Cox proportional hazards analysis, the sEng level (<3.9 ng/mL) was an independent predictor of cardiovascular events (hazard ratio: 1.59; 95%CI: 1.01-2.49). Furthermore, only among the 209 patients with CAD, the sEng level was also a predictor of further cardiovascular events (hazard ratio: 2.07; 95%CI: 1.24-3.45). Thus, low plasma sEng levels were found to be associated with an increased risk of cardiovascular events in patients with CAD and patients undergoing coronary angiography.
Collapse
Affiliation(s)
- Emi Saita
- Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Yoshimi Kishimoto
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Setsunan University, 45-1 Na-gaotouge-cho, Hirakata 573-0101, Japan
| | - Masayuki Aoyama
- Department of Cardiovascular Medicine, Toho University Graduate School of Medicine, 5-21-16 Omorinishi, Ota-ku, Tokyo 143-8540, Japan
- Department of Cardiology, National Hospital Organization Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo 152-8902, Japan
| | - Reiko Ohmori
- Faculty of Regional Design, Utsunomiya University, 350 Minecho, Tochigi 321-8505, Japan
| | - Kazuo Kondo
- Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| | - Yukihiko Momiyama
- Department of Cardiology, National Hospital Organization Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo 152-8902, Japan
| |
Collapse
|
7
|
Grossini E, Smirne C, Venkatesan S, Tonello S, D'Onghia D, Minisini R, Cantaluppi V, Sainaghi PP, Comi C, Tanzi A, Bussolati B, Pirisi M. Plasma Pattern of Extracellular Vesicles Isolated from Hepatitis C Virus Patients and Their Effects on Human Vascular Endothelial Cells. Int J Mol Sci 2023; 24:10197. [PMID: 37373343 DOI: 10.3390/ijms241210197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Hepatitis C virus (HCV) patients are at increased risk of cardiovascular disease (CVD). In this study, we aimed to evaluate the role of extracellular vesicles (EVs) as pathogenic factors for the onset of HCV-related endothelial dysfunction. Sixty-five patients with various stages of HCV-related chronic liver disease were enrolled in this case series. Plasma EVs were characterized and used to stimulate human vascular endothelial cells (HUVEC), which were examined for cell viability, mitochondrial membrane potential, and reactive oxygen species (ROS) release. The results showed that EVs from HCV patients were mainly of endothelial and lymphocyte origin. Moreover, EVs were able to reduce cell viability and mitochondrial membrane potential of HUVEC, while increasing ROS release. Those harmful effects were reduced by the pretreatment of HUVEC with the NLR family pyrin domain containing 3 (NLRP3)/AMP-activated protein kinase and protein kinase B blockers. In conclusion, in HCV patients, we could highlight a circulating pattern of EVs capable of inducing damage to the endothelium. These data represent a novel possible pathogenic mechanism underlying the reported increase of CVD occurrence in HCV infection and could be of clinical relevance also in relation to the widespread use of antiviral drugs.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Carlo Smirne
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Sakthipriyan Venkatesan
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Stelvio Tonello
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Davide D'Onghia
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Rosalba Minisini
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Vincenzo Cantaluppi
- Nephrology Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Pier Paolo Sainaghi
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- CAAD-Center for Autoimmune and Allergic Diseases, and IRCAD-Interdisciplinary Research Center for Autoimmune Diseases, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Adele Tanzi
- Molecular Biotechnology Center "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Turin, Italy
| | - Benedetta Bussolati
- Molecular Biotechnology Center "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Turin, Italy
| | - Mario Pirisi
- Internal Medicine Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Maggiore della Carità Hospital, 28100 Novara, Italy
| |
Collapse
|
8
|
Peyronnel C, Totoson P, Martin H, Demougeot C. Relevance of circulating markers of endothelial activation for cardiovascular risk assessment in rheumatoid arthritis: a narrative review. Life Sci 2023; 314:121264. [PMID: 36470540 DOI: 10.1016/j.lfs.2022.121264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is associated with excessive cardiovascular mortality secondary to premature atherosclerosis, in which endothelial activation (EA) plays a central role. EA is characterized by loss of vascular integrity, expression of leucocyte adhesion molecules, transition from antithrombotic to prothrombotic phenotype, cytokines production, shedding of membrane microparticles and recruitment of endothelial progenitor cells. As EA is an early event in atherogenesis, circulating markers of EA are putative markers of vascular pathology and cardiovascular (CV) risk. After a presentation of biology of EA, the present review analyzed the available data regarding changes in EA markers in RA in link with the vascular pathology and CV events, discussed their relevance as biomarkers of CV risk and proposed future directions.
Collapse
Affiliation(s)
- Célian Peyronnel
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Perle Totoson
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Hélène Martin
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Céline Demougeot
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France.
| |
Collapse
|
9
|
Weng J, Chen M, Shi B, Liu D, Weng S, Guo R. Konjac glucomannan defends against high-fat diet-induced atherosclerosis in rabbits by promoting the PI3K/Akt pathway. Heliyon 2023; 9:e13682. [PMID: 36852043 PMCID: PMC9957759 DOI: 10.1016/j.heliyon.2023.e13682] [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: 10/10/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Atherosclerosis (AS) is the main cause of cardiovascular disease and cerebral infarction, which seriously endanger human health. This study aimed to investigate konjac glucomannan (KGM) defends against high-fat diet-induced AS in rabbits by promoting the PI3K/Akt pathway. KGM administration reduced the degree of AS indicated by reducing the plaques and foam cells, the tunica intima thickness, and the tunica intima/tunica media thickness ratio in the aorta, and enlarging the lumen of the aorta. In addition, KGM administration regulated blood lipids, ameliorated inflammation indicated by reducing the levels of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, CRP, and VCAM-1, and attenuated endothelial injury, simultaneously mitigated oxidative stress indicated by decreasing MPO activity and the concentrations of MDA and increasing the GSH-Px and SOD concentrations. Moreover, KGM promotes the phosphorylation of PI3K and AKT. However, these effects of KGM on rabbits with high-fat diet-induced AS were blocked by LY294002. In conclusion, KGM defends against high-fat diet-induced AS in rabbits by promoting the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Junting Weng
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China
| | - Min Chen
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China
| | - Bingbing Shi
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China
| | - Danjuan Liu
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China
| | - Shuoyun Weng
- School of Wenzhou Medical University, Wenzhou 325035, China
| | - Rongjie Guo
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian 351100, China
| |
Collapse
|
10
|
Zhong M, Huang J, Wu Z, Chan KG, Wang L, Li J, Lee LH, Law JWF. Potential Roles of Selectins in Periodontal Diseases and Associated Systemic Diseases: Could They Be Targets for Immunotherapy? Int J Mol Sci 2022; 23:14280. [PMID: 36430760 PMCID: PMC9698067 DOI: 10.3390/ijms232214280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
Periodontal diseases are predisposing factors to the development of many systemic disorders, which is often initiated via leukocyte infiltration and vascular inflammation. These diseases could significantly affect human health and quality of life. Hence, it is vital to explore effective therapies to prevent disease progression. Periodontitis, which is characterized by gingival bleeding, disruption of the gingival capillary's integrity, and irreversible destruction of the periodontal supporting bone, appears to be caused by overexpression of selectins in periodontal tissues. Selectins (P-, L-, and E-selectins) are vital members of adhesion molecules regulating inflammatory and immune responses. They are mainly located in platelets, leukocytes, and endothelial cells. Furthermore, selectins are involved in the immunopathogenesis of vascular inflammatory diseases, such as cardiovascular disease, diabetes, cancers, and so on, by mediating leukocyte recruitment, platelet activation, and alteration of endothelial barrier permeability. Therefore, selectins could be new immunotherapeutic targets for periodontal disorders and their associated systemic diseases since they play a crucial role in immune regulation and endothelium dysfunction. However, the research on selectins and their association with periodontal and systemic diseases remains limited. This review aims to discuss the critical roles of selectins in periodontitis and associated systemic disorders and highlights the potential of selectins as therapeutic targets.
Collapse
Affiliation(s)
- Mei Zhong
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Jiangyong Huang
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Zhe Wu
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Kok-Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China
| | - Lijing Wang
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Vascular Biology Research Institute, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiang Li
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| | - Jodi Woan-Fei Law
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
11
|
Tomášková V, Mýtniková A, Hortová Kohoutková M, Mrkva O, Skotáková M, Šitina M, Helánová K, Frič J, Pařenica J, Šrámek V, Helán M. Prognostic value of soluble endoglin in patients with septic shock and severe COVID-19. Front Med (Lausanne) 2022; 9:972040. [PMID: 36117974 PMCID: PMC9470754 DOI: 10.3389/fmed.2022.972040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/15/2022] [Indexed: 12/01/2022] Open
Abstract
Sepsis is a clinical syndrome characterized by a dysregulated response to infection. It represents a leading cause of mortality in ICU patients worldwide. Although sepsis is in the point of interest of research for several decades, its clinical management and patient survival are improving slowly. Monitoring of the biomarkers and their combinations could help in early diagnosis, estimation of prognosis and patient's stratification and response to the treatment. Circulating soluble endoglin (sEng) is the cleaved extracellular part of transmembrane glycoprotein endoglin. As a biomarker, sEng has been tested in several pathologic conditions where its elevation was associated with endothelial dysfunction. In this study we have tested the ability of sEng to predict mortality and its correlation with other clinical characteristics in the cohort of septic shock patients (n = 37) and patients with severe COVID-19 (n = 40). In patients with COVID-19 sEng did not predict mortality or correlate with markers of organ dysfunction. In contrast, in septic shock the level of sEng was significantly higher in patients with early mortality (p = 0.019; AUC = 0.801). Moreover, sEng levels correlated with signs of circulatory failure (required dose of noradrenalin and lactate levels; p = 0.002 and 0.016, respectively). The predominant clinical problem in patients with COVID-19 was ARDS, and although they often showed signs of other organ dysfunction, circulatory failure was exceptional. This potentially explains the difference between sEng levels in COVID-19 and septic shock. In conclusion, we have confirmed that sEng may reflect the extent of the circulatory failure in septic shock patients and thus could be potentially used for the early identification of patients with the highest degree of endothelial dysfunction who would benefit from endothelium-targeted individualized therapy.
Collapse
Affiliation(s)
- Veronika Tomášková
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Alexandra Mýtniková
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | | | - Ondřej Mrkva
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Monika Skotáková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Michal Šitina
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Kateřina Helánová
- Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Cardiology, University Hospital Brno, Brno, Czechia
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
- Department of Modern Immunotherapy Research, Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Jiří Pařenica
- Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Cardiology, University Hospital Brno, Brno, Czechia
| | - Vladimír Šrámek
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Martin Helán
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
- *Correspondence: Martin Helán
| |
Collapse
|
12
|
Bai T, Yu S, Feng J. Advances in the Role of Endothelial Cells in Cerebral Small Vessel Disease. Front Neurol 2022; 13:861714. [PMID: 35481273 PMCID: PMC9035937 DOI: 10.3389/fneur.2022.861714] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) poses a serious socio-economic burden due to its high prevalence and severe impact on the quality of life of elderly patients. Pathological changes in CSVD mainly influence small cerebral arteries, microarteries, capillaries, and small veins, which are usually caused by multiple vascular risk factors. CSVD is often identified on brain magnetic resonance imaging (MRI) by recent small subcortical infarcts, white matter hyperintensities, lacune, cerebral microbleeds (CMBs), enlarged perivascular spaces (ePVSs), and brain atrophy. Endothelial cell (EC) dysfunction is earlier than clinical symptoms. Immune activation, inflammation, and oxidative stress may be potential mechanisms of EC injury. ECs of the blood–brain–barrier (BBB) are the most important part of the neurovascular unit (NVU) that ensures constant blood flow to the brain. Impaired cerebral vascular autoregulation and disrupted BBB cause cumulative brain damage. This review will focus on the role of EC injury in CSVD. Furthermore, several specific biomarkers will be discussed, which may be useful for us to assess the endothelial dysfunction and explore new therapeutic directions.
Collapse
|
13
|
Soluble Endoglin Stimulates Inflammatory and Angiogenic Responses in Microglia That Are Associated with Endothelial Dysfunction. Int J Mol Sci 2022; 23:ijms23031225. [PMID: 35163148 PMCID: PMC8835690 DOI: 10.3390/ijms23031225] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/16/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Increased soluble endoglin (sENG) has been observed in human brain arteriovenous malformations (bAVMs). In addition, the overexpression of sENG in concurrence with vascular endothelial growth factor (VEGF)-A has been shown to induce dysplastic vessel formation in mouse brains. However, the underlying mechanism of sENG-induced vascular malformations is not clear. The evidence suggests the role of sENG as a pro-inflammatory modulator, and increased microglial accumulation and inflammation have been observed in bAVMs. Therefore, we hypothesized that microglia mediate sENG-induced inflammation and endothelial cell (EC) dysfunction in bAVMs. In this study, we confirmed that the presence of sENG along with VEGF-A overexpression induced dysplastic vessel formation. Remarkably, we observed increased microglial activation around dysplastic vessels with the expression of NLRP3, an inflammasome marker. We found that sENG increased the gene expression of VEGF-A, pro-inflammatory cytokines/inflammasome mediators (TNF-α, IL-6, NLRP3, ASC, Caspase-1, and IL-1β), and proteolytic enzyme (MMP-9) in BV2 microglia. The conditioned media from sENG-treated BV2 (BV2-sENG-CM) significantly increased levels of angiogenic factors (Notch-1 and TGFβ) and pERK1/2 in ECs but it decreased the level of IL-17RD, an anti-angiogenic mediator. Finally, the BV2-sENG-CM significantly increased EC migration and tube formation. Together, our study demonstrates that sENG provokes microglia to express angiogenic/inflammatory molecules which may be involved in EC dysfunction. Our study corroborates the contribution of microglia to the pathology of sENG-associated vascular malformations.
Collapse
|
14
|
Zakrzewski PK. Canonical TGFβ Signaling and Its Contribution to Endometrial Cancer Development and Progression-Underestimated Target of Anticancer Strategies. J Clin Med 2021; 10:3900. [PMID: 34501347 PMCID: PMC8432036 DOI: 10.3390/jcm10173900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023] Open
Abstract
Endometrial cancer is one of the leading gynecological cancers diagnosed among women in their menopausal and postmenopausal age. Despite the progress in molecular biology and medicine, no efficient and powerful diagnostic and prognostic marker is dedicated to endometrial carcinogenesis. The canonical TGFβ pathway is a pleiotropic signaling cascade orchestrating a variety of cellular and molecular processes, whose alterations are responsible for carcinogenesis that originates from different tissue types. This review covers the current knowledge concerning the canonical TGFβ pathway (Smad-dependent) induced by prototypical TGFβ isoforms and the involvement of pathway alterations in the development and progression of endometrial neoplastic lesions. Since Smad-dependent signalization governs opposed cellular processes, such as growth arrest, apoptosis, tumor cells growth and differentiation, as well as angiogenesis and metastasis, TGFβ cascade may act both as a tumor suppressor or tumor promoter. However, the final effect of TGFβ signaling on endometrial cancer cells depends on the cancer disease stage. The multifunctional role of the TGFβ pathway indicates the possible utilization of alterations in the TGFβ cascade as a potential target of novel anticancer strategies.
Collapse
Affiliation(s)
- Piotr K Zakrzewski
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
15
|
Vicen M, Igreja Sá IC, Tripská K, Vitverová B, Najmanová I, Eissazadeh S, Micuda S, Nachtigal P. Membrane and soluble endoglin role in cardiovascular and metabolic disorders related to metabolic syndrome. Cell Mol Life Sci 2021; 78:2405-2418. [PMID: 33185696 PMCID: PMC11072708 DOI: 10.1007/s00018-020-03701-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/05/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
Membrane endoglin (Eng, CD105) is a transmembrane glycoprotein essential for the proper function of vascular endothelium. It might be cleaved by matrix metalloproteinases to form soluble endoglin (sEng), which is released into the circulation. Metabolic syndrome comprises conditions/symptoms that usually coincide (endothelial dysfunction, arterial hypertension, hyperglycemia, obesity-related insulin resistance, and hypercholesterolemia), and are considered risk factors for cardiometabolic disorders such as atherosclerosis, type II diabetes mellitus, and liver disorders. The purpose of this review is to highlight current knowledge about the role of Eng and sEng in the disorders mentioned above, in vivo and in vitro extent, where we can find a wide range of contradictory results. We propose that reduced Eng expression is a hallmark of endothelial dysfunction development in chronic pathologies related to metabolic syndrome. Eng expression is also essential for leukocyte transmigration and acute inflammation, suggesting that Eng is crucial for the regulation of endothelial function during the acute phase of vascular defense reaction to harmful conditions. sEng was shown to be a circulating biomarker of preeclampsia, and we propose that it might be a biomarker of metabolic syndrome-related symptoms and pathologies, including hypercholesterolemia, hyperglycemia, arterial hypertension, and diabetes mellitus as well, despite the fact that some contradictory findings have been reported. Besides, sEng can participate in the development of endothelial dysfunction and promote the development of arterial hypertension, suggesting that high levels of sEng promote metabolic syndrome symptoms and complications. Therefore, we suggest that the treatment of metabolic syndrome should take into account the importance of Eng in the endothelial function and levels of sEng as a biomarker and risk factor of related pathologies.
Collapse
Affiliation(s)
- Matej Vicen
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Ivone Cristina Igreja Sá
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Katarína Tripská
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Barbora Vitverová
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Iveta Najmanová
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Samira Eissazadeh
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Stanislav Micuda
- Faculty of Medicine in Hradec Kralove, Department of Pharmacology, Charles University, Simkova 870, Hradec Kralove, 500 03, Czech Republic
| | - Petr Nachtigal
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic.
| |
Collapse
|
16
|
Víšek J, Bláha M, Bláha V, Lášticová M, Lánska M, Andrýs C, Tebbens JD, Igreja E Sá IC, Tripská K, Vicen M, Najmanová I, Nachtigal P. Monitoring of up to 15 years effects of lipoprotein apheresis on lipids, biomarkers of inflammation, and soluble endoglin in familial hypercholesterolemia patients. Orphanet J Rare Dis 2021; 16:110. [PMID: 33640001 PMCID: PMC7913462 DOI: 10.1186/s13023-021-01749-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Background Lipoprotein apheresis (LA) is considered as an add-on therapy for patients with familial hypercholesterolemia (FH). We aimed to analyze the data collected in the last 15 years from FH patients treated with LA, to elucidate the benefit of this procedure with respect to plasma lipids, biomarkers of inflammation, and endothelial dysfunction and soluble endoglin. Results 14 patients (10 heterozygous FH patients (HeFH), 4 homozygous FH patients (HoFH)) were treated by long-term lipoprotein apheresis. Lipid levels were examined, and ELISA detected biomarkers of inflammation and soluble endoglin. Paired tests were used for intergroup comparisons, and a linear regression model served to estimate the influence of the number of days patients were treated with LA on the studied parameters. LA treatment was associated with a significant decrease of total cholesterol (TC), LDL-C, HDL-C, and apoB, in both HeFH and HoFH patients, after single apheresis and in a long-term period during the monitored interval of 15 years. Biomarkers of inflammation and endothelial dysfunction were reduced for soluble endoglin, hsCRP, and MCP-1, and sP-selectin after each procedure in some HeFH and HoFH patients. Conclusions LA treatment up to 15 years, reduced cholesterol levels, levels of biomarkers related to endothelial dysfunction, and inflammation not only after each procedure but also in the long-term evaluation in FH patients. We propose that long-term LA treatment improves lipid profile and endothelial dysfunction in familial hypercholesterolemia patients, suggesting a promising improvement in cardiovascular prognosis in most FH patients.
Collapse
Affiliation(s)
- J Víšek
- Metabolism and Gerontology, 3rd Department of Internal Medicine, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové and Charles University, Hradec Králové, Czech Republic
| | - M Bláha
- 4th Department of Medicine - Hematology, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové and Charles University, Hradec Králové, Czech Republic
| | - V Bláha
- Metabolism and Gerontology, 3rd Department of Internal Medicine, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové and Charles University, Hradec Králové, Czech Republic
| | - M Lášticová
- Metabolism and Gerontology, 3rd Department of Internal Medicine, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové and Charles University, Hradec Králové, Czech Republic
| | - M Lánska
- 4th Department of Medicine - Hematology, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové and Charles University, Hradec Králové, Czech Republic
| | - C Andrýs
- Department of Immunology and Allergology, Faculty of Medicine in Hradec Králové, University Hospital Hradec Králové and Charles University, Hradec Králové, Czech Republic
| | - J Duintjer Tebbens
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Ivone Cristina Igreja E Sá
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - K Tripská
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - M Vicen
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - I Najmanová
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - P Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic.
| |
Collapse
|
17
|
Meyer N, Brodowski L, Richter K, von Kaisenberg CS, Schröder-Heurich B, von Versen-Höynck F. Pravastatin Promotes Endothelial Colony-Forming Cell Function, Angiogenic Signaling and Protein Expression In Vitro. J Clin Med 2021; 10:E183. [PMID: 33419165 PMCID: PMC7825508 DOI: 10.3390/jcm10020183] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/28/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022] Open
Abstract
Endothelial dysfunction is a primary feature of several cardiovascular diseases. Endothelial colony-forming cells (ECFCs) represent a highly proliferative subtype of endothelial progenitor cells (EPCs), which are involved in neovascularization and vascular repair. Statins are known to improve the outcome of cardiovascular diseases via pleiotropic effects. We hypothesized that treatment with the 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitor pravastatin increases ECFCs' functional capacities and regulates the expression of proteins which modulate endothelial health in a favourable manner. Umbilical cord blood derived ECFCs were incubated with different concentrations of pravastatin with or without mevalonate, a key intermediate in cholesterol synthesis. Functional capacities such as migration, proliferation and tube formation were addressed in corresponding in vitro assays. mRNA and protein levels or phosphorylation of protein kinase B (AKT), endothelial nitric oxide synthase (eNOS), heme oxygenase-1 (HO-1), vascular endothelial growth factor A (VEGF-A), placental growth factor (PlGF), soluble fms-like tyrosine kinase-1 (sFlt-1) and endoglin (Eng) were analyzed by real time PCR or immunoblot, respectively. Proliferation, migration and tube formation of ECFCs were enhanced after pravastatin treatment, and AKT- and eNOS-phosphorylation were augmented. Further, expression levels of HO-1, VEGF-A and PlGF were increased, whereas expression levels of sFlt-1 and Eng were decreased. Pravastatin induced effects were reversible by the addition of mevalonate. Pravastatin induces beneficial effects on ECFC function, angiogenic signaling and protein expression. These effects may contribute to understand the pleiotropic function of statins as well as to provide a promising option to improve ECFCs' condition in cell therapy in order to ameliorate endothelial dysfunction.
Collapse
Affiliation(s)
- Nadia Meyer
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (N.M.); (L.B.); (K.R.); (B.S.-H.)
| | - Lars Brodowski
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (N.M.); (L.B.); (K.R.); (B.S.-H.)
- Department of Obstetrics and Gynecology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany;
| | - Katja Richter
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (N.M.); (L.B.); (K.R.); (B.S.-H.)
| | - Constantin S. von Kaisenberg
- Department of Obstetrics and Gynecology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany;
| | - Bianca Schröder-Heurich
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (N.M.); (L.B.); (K.R.); (B.S.-H.)
| | - Frauke von Versen-Höynck
- Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany; (N.M.); (L.B.); (K.R.); (B.S.-H.)
- Department of Obstetrics and Gynecology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany;
| |
Collapse
|
18
|
Sri Iswari R, Dafip M, Purwantoyo E. Malondialdehyde (MDA) Production in Atherosclerosis Supplemented with Steamed Tomato. Pak J Biol Sci 2021; 24:319-325. [PMID: 34486316 DOI: 10.3923/pjbs.2021.319.325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background and Objective Malondialdehyde (MDA) may increase influenced by free radicals due to lipid oxidation. Tomato induction considers able to prevent free radical damage and atherosclerosis. Therefore, this study aims to understand the effect of steamed-tomato extracts on MDA and its potential as an early diagnosis of atherosclerosis. Materials and Methods A total of 24 healthy 12 weeks-old male-rats were divided into four treatment groups, equally. A normal control group (K1) was rats with placebo treatment. A negative control group (K2) was the rats supplemented with 2 mL kg-1 b.wt. per day of cholesterol until cholesterol. A K3 group was atherosclerosis rats given with 20 mg kg-1 b.wt. per day of atorvastatin and a K4 was atherosclerotic rats supplemented with 16 mg kg-1 b.wt. per day of tomato extract. All treatments were carried out for 60 consecutive days. Results Tomato extract in the K4 group was succeeded in lowering MDA production. Carotenoid compounds in tomato extract are well known to be prevention agents against lipid oxidation and inhibit free radicals. MDA levels have increased significantly in atherosclerosis conditions, making it potentially noticeable during early atherosclerotic, therefore, potentially developed as biomarkers. Conclusion MDA levels increase significantly and simultaneously after high cholesterol diets and in line with lipid parameters and damaged blood vessels. The steamed-tomato extract can reduce MDA, lipids levels and protect endothelial from lipid oxidation. More research should be conducted to breakdown the MDA function in the molecular pathway, including MDA correlation to microRNA expression and cell signaling.
Collapse
|
19
|
Pérez-Roque L, Núñez-Gómez E, Rodríguez-Barbero A, Bernabéu C, López-Novoa JM, Pericacho M. Pregnancy-Induced High Plasma Levels of Soluble Endoglin in Mice Lead to Preeclampsia Symptoms and Placental Abnormalities. Int J Mol Sci 2020; 22:ijms22010165. [PMID: 33375253 PMCID: PMC7795873 DOI: 10.3390/ijms22010165] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
Preeclampsia is a pregnancy-specific disease of high prevalence characterized by the onset of hypertension, among other maternal or fetal signs. Its etiopathogenesis remains elusive, but it is widely accepted that abnormal placentation results in the release of soluble factors that cause the clinical manifestations of the disease. An increased level of soluble endoglin (sEng) in plasma has been proposed to be an early diagnostic and prognostic biomarker of this disease. A pathogenic function of sEng involving hypertension has also been reported in several animal models with high levels of plasma sEng not directly dependent on pregnancy. The aim of this work was to study the functional effect of high plasma levels of sEng in the pathophysiology of preeclampsia in a model of pregnant mice, in which the levels of sEng in the maternal blood during pregnancy replicate the conditions of human preeclampsia. Our results show that wild type pregnant mice carrying human sEng-expressing transgenic fetuses (fWT(hsEng+)) present high plasma levels of sEng with a timing profile similar to that of human preeclampsia. High plasma levels of human sEng (hsEng) are associated with hypertension, proteinuria, fetal growth restriction, and the release of soluble factors to maternal plasma. In addition, fWT(hsEng+) mice also present placental alterations comparable to those caused by the poor remodeling of the spiral arteries characteristic of preeclampsia. In vitro and ex vivo experiments, performed in a human trophoblast cell line and human placental explants, show that sEng interferes with trophoblast invasion and the associated pseudovasculogenesis, a process by which cytotrophoblasts switch from an epithelial to an endothelial phenotype, both events being related to remodeling of the spiral arteries. Our findings provide a novel and useful animal model for future research in preeclampsia and reveal a much more relevant role of sEng in preeclampsia than initially proposed.
Collapse
Affiliation(s)
- Lucía Pérez-Roque
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Elena Núñez-Gómez
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Alicia Rodríguez-Barbero
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain;
| | - José M. López-Novoa
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Miguel Pericacho
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
- Correspondence:
| |
Collapse
|
20
|
Pan Y, Wang L, Xie Y, Tan Y, Chang C, Qiu X, Li X. Characterization of differentially expressed plasma proteins in patients with acute myocardial infarction. J Proteomics 2020; 227:103923. [PMID: 32736138 DOI: 10.1016/j.jprot.2020.103923] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 07/12/2020] [Accepted: 07/24/2020] [Indexed: 12/11/2022]
Abstract
Acute myocardial infarction (AMI) remains a leading cause of morbidity and mortality worldwide. Novel biomarkers are needed to identify NSTEMI in AMI patients. The study objective was to use proteomics to identify novel plasma biomarkers for STEMI and NSTEMI patients. iTRAQ analysis was performed on pooled samples from 8 healthy controls and 12 STEMI and 12 NSTEMI patients. Bioinformatics analysis identified 95 differentially expressed proteins that were differentially expressed in the plasma of AMI patients and healthy controls; 28 of these proteins were found in STEMI/Con (22 upregulated and 6 downregulated), 48 in NSTEMI/Con (12 upregulated and 36 downregulated), and 44 in NSTEMI/STEMI (11 upregulated and 33 downregulated). Protein network analysis was then performed using STRING software. Functional analysis revealed that the identified plasma proteins were mainly involved with carbon metabolism, toll-like receptor signaling pathway, and hypertrophic cardiomyopathy. Nine of the proteins (SSA1, MDH1, FCN2, GPI, S100A8, LBP, vinculin, VDBP, and RBP4) that changed levels during AMI progression were further validated by ELISA. The constructed plasma proteome could reflect the AMI pathogenesis molecular mechanisms and provide a method for the early identification of NSTEMI in AMI patients. SIGNIFICANCE: The aim of this study was to use proteomics to identify novel predictive plasma biomarkers for patients with acute myocardial infarction (AMI), which would allow for either identification of individuals at risk of an infarction, and early identification of NSTEMI in patients with AMI. Using an approach that combined iTRAQ with LC-MS/MS, we found 95 proteins that showed significant differences in expression levels among the AMI patients and healthy controls. The proteins SSA1, MDH1, FCN2, GPI, S100A8, LBP, vinculin, VDBP, and RBP4 were found to play crucial roles in the pathogenesis of AMI. Using bioinformatics analysis, we found that dysregulation of carbon metabolism, toll-like receptor signaling pathway, and hypertrophic cardiomyopathy may be the major driving forces for cardiac damage during myocardial infarction. However, further investigations are needed to verify the mechanisms involved in the development of AMI especially NSTEMI. Taken together, our findings lay the foundation for understanding the molecular mechanisms underlying the pathogenic processes of AMI, and suggest potential applications for specific biomarkers in early diagnosis and determination of prognosis.
Collapse
Affiliation(s)
- Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Linlin Wang
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Yaofeng Xie
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Yuan Tan
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cheng Chang
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Xueshan Qiu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China.
| |
Collapse
|
21
|
Vitverova B, Najmanova I, Vicen M, Tripska K, Sa ICI, Hyspler R, Pericacho M, Nachtigal P. Long term effects of soluble endoglin and mild hypercholesterolemia in mice hearts. PLoS One 2020; 15:e0233725. [PMID: 32470058 PMCID: PMC7259503 DOI: 10.1371/journal.pone.0233725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/11/2020] [Indexed: 01/15/2023] Open
Abstract
Soluble endoglin (sEng) released into the circulation was suggested to be related to cardiovascular based pathologies. It was demonstrated that a combination of high sEng levels and long-term exposure (six months) to high fat diet (HFD) resulted in aggravation of endothelial dysfunction in the aorta. Thus, in this study, we hypothesized that a similar experimental design would affect the heart morphology, TGFβ signaling, inflammation, fibrosis, oxidative stress and eNOS signaling in myocardium in transgenic mice overexpressing human sEng. Three-month-old female transgenic mice overexpressing human sEng in plasma (Sol-Eng+ high) and their age-matched littermates with low levels of human sEng (Sol-Eng+ low) were fed a high-fat diet containing 1.25% of cholesterol and 40% of fat for six months. A blood analysis was performed, and the heart samples were analyzed by qRT-PCR and Western blot. The results of this study showed no effects of sEng and HFD on myocardial morphology/hypertrophy/fibrosis. However, the expression of pSmad2/3 and p-eNOS was reduced in Sol-Eng+ high mice. On the other hand, sEng and HFD did not significantly affect the expression of selected members of TGFβ signaling (membrane endoglin, TGFβRII, ALK-5, ALK-1, Id-1, PAI-1), inflammation (VCAM-1, ICAM-1), oxidative stress (NQO1, HO-1) and heart remodeling (PDGFβ, COL1A1, β-MHC). In conclusion, the results of this study confirmed that sEng, even combined with a high-fat diet inducing hypercholesterolemia administered for six months, does not affect the structure of the heart with respect to hypertrophy, fibrosis, inflammation and oxidative stress. Interestingly, pSmad2/3/p-eNOS signaling was reduced in both the heart in this study and the aorta in the previous study, suggesting a possible alteration of NO metabolism caused by six months exposure to high sEng levels and HFD. Thus, we might conclude that sEng combined with a high-fat diet might be related to the alteration of NO production due to altered pSmad2/3/p-eNOS signaling in the heart and aorta.
Collapse
Affiliation(s)
- Barbora Vitverova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Iveta Najmanova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Matej Vicen
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Katarina Tripska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ivone Cristina Igreja Sa
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Radek Hyspler
- Centrum for Research and Development, University Hospital, Hradec Kralove, Czech Republic
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, and the Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
- * E-mail:
| |
Collapse
|
22
|
Gawrys J, Gajecki D, Szahidewicz-Krupska E, Doroszko A. Intraplatelet L-Arginine-Nitric Oxide Metabolic Pathway: From Discovery to Clinical Implications in Prevention and Treatment of Cardiovascular Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1015908. [PMID: 32215167 PMCID: PMC7073508 DOI: 10.1155/2020/1015908] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/12/2020] [Indexed: 12/31/2022]
Abstract
Despite the development of new drugs and other therapeutic strategies, cardiovascular disease (CVD) remains still the major cause of morbidity and mortality in the world population. A lot of research, performed mostly in the last three decades, revealed an important correlation between "classical" demographic and biochemical risk factors for CVD, (i.e., hypercholesterolemia, hyperhomocysteinemia, smoking, renal failure, aging, diabetes, and hypertension) with endothelial dysfunction associated directly with the nitric oxide deficiency. The discovery of nitric oxide and its recognition as an endothelial-derived relaxing factor was a breakthrough in understanding the pathophysiology and development of cardiovascular system disorders. The nitric oxide synthesis pathway and its regulation and association with cardiovascular risk factors were a common subject for research during the last decades. As nitric oxide synthase, especially its endothelial isoform, which plays a crucial role in the regulation of NO bioavailability, inhibiting its function results in the increase in the cardiovascular risk pattern. Among agents altering the production of nitric oxide, asymmetric dimethylarginine-the competitive inhibitor of NOS-appears to be the most important. In this review paper, we summarize the role of L-arginine-nitric oxide pathway in cardiovascular disorders with the focus on intraplatelet metabolism.
Collapse
Affiliation(s)
- Jakub Gawrys
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Poland
| | - Damian Gajecki
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Poland
| | - Ewa Szahidewicz-Krupska
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Poland
| | - Adrian Doroszko
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Poland
| |
Collapse
|
23
|
Kirollos S, Skilton M, Patel S, Arnott C. A Systematic Review of Vascular Structure and Function in Pre-eclampsia: Non-invasive Assessment and Mechanistic Links. Front Cardiovasc Med 2019. [PMID: 31803759 DOI: 10.3389/fcvm.2019.00166, 10.3389/fmed.2019.00166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Hypertensive disorders of pregnancy, such as pre-eclampsia, are known to be independently associated with the development of premature cardiovascular disease (CVD) in women. In pre-eclampsia, the placenta secretes excess anti-angiogenic factors into the maternal circulation, leading to widespread endothelial damage, and inflammation. This endothelial damage is evidenced to persist beyond the acute illness. However, whether it is permanent and responsible for the elevated rates of premature CVD seen in this at-risk group remains unclear. A systematic review of the available literature with respect to vascular structure and function prior to, during and after a pregnancy complicated by pre-eclampsia was performed. Studies non-invasively assessing vascular structure using carotid intima-media thickness (CIMT), retinal microvasculature caliber, CT coronary angiogram, or coronary calcium scores were included. Vascular function was assessed using brachial flow-mediated dilation (FMD), pulse wave analysis (PWA), and peripheral arterial tonometry (PAT). In total 59 articles were included (13 CIMT, 5 CTCA/Ca score, five retinal microvasculature, 27 FMD, 7 PAT, and 14 PWV/PWA), consisting of prospective and retrospective cohort, and case-control studies. Change in vascular structure was evidenced with significant increases in CIMT by 73-180 μm greater than that of non-affected women. This is tempered by other studies reporting resolution of structural changes postpartum, highlighting the need for further research. Accelerated coronary calcification and plaque deposition was identified, with greater rates of increased calcium scores and subclinical coronary artery disease shown by CTCA in women with a history of pre-eclampsia at 30 years postpartum. Impaired endothelial function was consistently reported prior to, during and immediately after pregnancy as evidenced by differences in FMD of 1.7-12.2% less than non-affected women, an increase in PWV by 13.2-26%, and reduced retinal microvascular caliber and arterial elasticity indices. The evidence was less conclusive for the persistence of long-term endothelial dysfunction. Understanding the underlying mechanistic links between pre-eclampsia and CVD is a key step to identifying targeted therapies aimed at "repairing the endothelium" and attenuating risk. This review has highlighted the need for a greater understanding of vascular structure and function following pre-eclampsia through high quality studies with large sample sizes, particularly in the longer postpartum period when clinical CVD disease starts to manifest.
Collapse
Affiliation(s)
- Shady Kirollos
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Michael Skilton
- Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, Faculty of Medicine and Health University of Sydney, Sydney, NSW, Australia
| | - Sanjay Patel
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Department of Coronary Diseases, The Heart Research Institute, Sydney, NSW, Australia
| | - Clare Arnott
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Department of Coronary Diseases, The Heart Research Institute, Sydney, NSW, Australia.,Department of Cardiology, The George Institute for Global Health, Sydney, NSW, Australia
| |
Collapse
|
24
|
Kirollos S, Skilton M, Patel S, Arnott C. A Systematic Review of Vascular Structure and Function in Pre-eclampsia: Non-invasive Assessment and Mechanistic Links. Front Cardiovasc Med 2019; 6:166. [PMID: 31803759 PMCID: PMC6873347 DOI: 10.3389/fcvm.2019.00166] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 10/30/2019] [Indexed: 12/14/2022] Open
Abstract
Hypertensive disorders of pregnancy, such as pre-eclampsia, are known to be independently associated with the development of premature cardiovascular disease (CVD) in women. In pre-eclampsia, the placenta secretes excess anti-angiogenic factors into the maternal circulation, leading to widespread endothelial damage, and inflammation. This endothelial damage is evidenced to persist beyond the acute illness. However, whether it is permanent and responsible for the elevated rates of premature CVD seen in this at-risk group remains unclear. A systematic review of the available literature with respect to vascular structure and function prior to, during and after a pregnancy complicated by pre-eclampsia was performed. Studies non-invasively assessing vascular structure using carotid intima-media thickness (CIMT), retinal microvasculature caliber, CT coronary angiogram, or coronary calcium scores were included. Vascular function was assessed using brachial flow-mediated dilation (FMD), pulse wave analysis (PWA), and peripheral arterial tonometry (PAT). In total 59 articles were included (13 CIMT, 5 CTCA/Ca score, five retinal microvasculature, 27 FMD, 7 PAT, and 14 PWV/PWA), consisting of prospective and retrospective cohort, and case-control studies. Change in vascular structure was evidenced with significant increases in CIMT by 73–180 μm greater than that of non-affected women. This is tempered by other studies reporting resolution of structural changes postpartum, highlighting the need for further research. Accelerated coronary calcification and plaque deposition was identified, with greater rates of increased calcium scores and subclinical coronary artery disease shown by CTCA in women with a history of pre-eclampsia at 30 years postpartum. Impaired endothelial function was consistently reported prior to, during and immediately after pregnancy as evidenced by differences in FMD of 1.7–12.2% less than non-affected women, an increase in PWV by 13.2–26%, and reduced retinal microvascular caliber and arterial elasticity indices. The evidence was less conclusive for the persistence of long-term endothelial dysfunction. Understanding the underlying mechanistic links between pre-eclampsia and CVD is a key step to identifying targeted therapies aimed at “repairing the endothelium” and attenuating risk. This review has highlighted the need for a greater understanding of vascular structure and function following pre-eclampsia through high quality studies with large sample sizes, particularly in the longer postpartum period when clinical CVD disease starts to manifest.
Collapse
Affiliation(s)
- Shady Kirollos
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Michael Skilton
- Boden Collaboration for Obesity, Nutrition, Exercise, and Eating Disorders, Faculty of Medicine and Health University of Sydney, Sydney, NSW, Australia
| | - Sanjay Patel
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Department of Coronary Diseases, The Heart Research Institute, Sydney, NSW, Australia
| | - Clare Arnott
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Department of Coronary Diseases, The Heart Research Institute, Sydney, NSW, Australia.,Department of Cardiology, The George Institute for Global Health, Sydney, NSW, Australia
| |
Collapse
|
25
|
Zhang L, Li X, Zhou C, You Z, Zhang J, Cao G. The diagnosis values of serum STAT4 and sEng in preeclampsia. J Clin Lab Anal 2019; 34:e23073. [PMID: 31628681 PMCID: PMC7031581 DOI: 10.1002/jcla.23073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/12/2019] [Accepted: 09/27/2019] [Indexed: 11/10/2022] Open
Abstract
Objective To detect the levels of signal transducer and activator of transcription 4 (STAT4) and soluble endoglin (sEng) in preeclampsia patients and analyze the diagnostic values of STAT4 and sEng in preeclampsia. Methods Fifty‐four pregnant women with preeclampsia from October 2017 to June 2018 were included in this study. Twenty‐eight matched healthy pregnant women were set as the control group. The general clinical characteristics were measured. Serum STAT4 and sEng were detected by ELISA. Correlation between STAT4 and sEng, and their diagnostic value in preeclampsia were analyzed. Results Compared with control, the prothrombin time in preeclampsia was significantly lower, while the mean arterial pressure, 24‐hour urine protein, serum creatinine, fibrinogen, and ALT were significantly higher. The circulating levels of STAT4 and sEng were significantly increased in the preeclampsia. The serum levels of STAT4 and sEng in preeclampsia were positively correlated. For the diagnosis of preeclampsia by the serum STAT4, AUC is 0.902, and the sensitivity and specificity are 0.893 and 0.929. By the serum sEng, AUC is 0.873, and the sensitivity and specificity are 0.816 and 0.905. Conclusion The serum levels of STAT4 and sEng were significantly increased in preeclampsia with disease severity status, which have promise as diagnostic markers in preeclampsia.
Collapse
Affiliation(s)
- Luyan Zhang
- Department of Laboratory Medicine, Ningbo Mingzhou Hospital, Ningbo, China
| | - Xuechun Li
- Department of Laboratory Medicine, Ningbo Mingzhou Hospital, Ningbo, China
| | - Chengcheng Zhou
- Department of Laboratory Medicine, Ningbo Mingzhou Hospital, Ningbo, China
| | - Zhengming You
- Department of Laboratory Medicine, Ningbo Mingzhou Hospital, Ningbo, China
| | - Jianwei Zhang
- Department of Laboratory Medicine, Ningbo Mingzhou Hospital, Ningbo, China
| | - Guomei Cao
- Department of Laboratory Medicine, Ningbo Mingzhou Hospital, Ningbo, China
| |
Collapse
|
26
|
Jouvray M, Launay D, Dubucquoi S, Sobanski V, Podevin C, Lambert M, Morell-Dubois S, Maillard H, Hatron PY, Hachulla E, Giovannelli J. Whole-Body Distribution and Clinical Association of Telangiectases in Systemic Sclerosis. JAMA Dermatol 2019; 154:796-805. [PMID: 29799952 DOI: 10.1001/jamadermatol.2018.0916] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Importance In systemic sclerosis (SSc), to date, no study has precisely described the total number and fine distribution of telangiectases (TAs), their clinical association with the disease, and the biological mechanisms causing their development. Objectives To describe the whole-body distribution of TAs and assess the association between the whole-body TA number and the characteristics of patients with SSc. Design, Setting, and Participants A single-center, cross-sectional study was conducted between July 11, 2016, and March 15, 2017, at the National Referral Centre for Rare Systemic and Autoimmune Diseases in France. A population-based sample of 106 adults who fulfilled the 2013 American College of Rheumatology/European League Against Rheumatism criteria for SSc were included; 8 patients who had previously received laser treatment for TAs were excluded. Main Outcomes and Measures The number of TAs on the whole body (total and those >5 mm) and TA distribution in different areas were recorded. The association with clinical and biological data was studied using univariate and multivariate linear regression. Results A total of 106 patients (83 [78.3%] women) were enrolled, including 12 with precapillary pulmonary hypertension (PH). Mean (SD) age was 60.6 (13.5) years. Telangiectasia distribution was 37.2% on the face, 33.2% on the upper limbs, including 26.4% on the hands, 28.1% on the trunk, including 17.1% for the upper part of the trunk, and 1.5% on the lower limbs. In analysis using the multivariate linear regression model, the whole-body TA number was independently associated with male sex (percentage change, 144.4%; 95% CI, 7.5% to 455.9%; P = .03), PH (162.8%; 95% CI, 5.6% to 553.8%; P = .04), history of pulmonary embolism (336.4%; 95% CI, 39.0% to 1270.1%; P = .01), glomerular filtration rate (-1.6%; 95% CI, -3.2% to -0.1% per 1-mL/min/1.73 m2 increase; P = .04), and soluble endoglin level (28.2%; 95% CI, 1.2% to 62.5% per 1-ng/mL increase; P = .04). Receiver operating characteristic analyses assessing the ability of TAs to identify the presence of PH revealed that the area under the curve was significant for the TA number on the whole body (0.77; 95% CI, 0.57 to 0.88), on the hands and face (0.81; 95% CI, 0.57 to 0.91), and on the hands (95% CI, 0.77; 95% CI, 0.57 to 0.89). Conclusions and Relevance In the patients in this study with SSc, TAs were predominantly located on the face, hands, and the upper part of the trunk. Telangiectases appeared to be associated with vasculopathy features of SSc, particularly with PH and soluble endoglin levels.
Collapse
Affiliation(s)
- Mathieu Jouvray
- University Lille, Lille Inflammation Research International Center, Lille, France.,Inserm, Lille, France.,CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - David Launay
- University Lille, Lille Inflammation Research International Center, Lille, France.,Inserm, Lille, France.,CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - Sylvain Dubucquoi
- University Lille, Lille Inflammation Research International Center, Lille, France.,Inserm, Lille, France.,Institut d'Immunologie, CHU Lille, Institut d'Immunologie, Lille, France
| | - Vincent Sobanski
- University Lille, Lille Inflammation Research International Center, Lille, France.,Inserm, Lille, France.,CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - Céline Podevin
- University Lille, Lille Inflammation Research International Center, Lille, France.,Inserm, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - Marc Lambert
- University Lille, Lille Inflammation Research International Center, Lille, France.,Inserm, Lille, France.,CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - Sandrine Morell-Dubois
- CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - Hélène Maillard
- CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - Pierre-Yves Hatron
- CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - Eric Hachulla
- University Lille, Lille Inflammation Research International Center, Lille, France.,Inserm, Lille, France.,CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| | - Jonathan Giovannelli
- University Lille, Lille Inflammation Research International Center, Lille, France.,Inserm, Lille, France.,CHU Lille, Département De Médecine Interne et Immunologie Clinique, Lille, France.,Centre de Référence des Maladies Autoimmunes et Systémiques Rares du Nord et Nord-Ouest de France, Lille, France
| |
Collapse
|
27
|
Abstract
Endoglin (ENG) is a coreceptor of the transforming growth factor-β (TGFβ) family signaling complex, which is highly expressed on endothelial cells and plays a key role in angiogenesis. Its extracellular domain can be cleaved and released into the circulation as soluble ENG (sENG). High circulating levels of sENG contribute to the pathogenesis of preeclampsia (PE). Circulating bone morphogenetic protein 9 (BMP9), a vascular quiescence and endothelial-protective factor, binds sENG with high affinity, but how sENG participates in BMP9 signaling complexes is not fully resolved. sENG was thought to be a ligand trap for BMP9, preventing type II receptor binding and BMP9 signaling. Here we show that, despite cell-surface ENG being a dimer linked by disulfide bonds, sENG purified from human placenta and plasma from PE patients is primarily in a monomeric form. Incubating monomeric sENG with the circulating form of BMP9 (prodomain-bound form) in solution leads to the release of the prodomain and formation of a sENG:BMP9 complex. Furthermore, we demonstrate that binding of sENG to BMP9 does not inhibit BMP9 signaling. Indeed, the sENG:BMP9 complex signals with comparable potency and specificity to BMP9 on human primary endothelial cells. The full signaling activity of the sENG:BMP9 complex required transmembrane ENG. This study confirms that rather than being an inhibitory ligand trap, increased circulating sENG might preferentially direct BMP9 signaling via cell-surface ENG at the endothelium. This is important for understanding the role of sENG in the pathobiology of PE and other cardiovascular diseases.
Collapse
|
28
|
Li Q, Lin F, Ke D, Cheng Q, Gui Y, Zhou Y, Wu Y, Wang Y, Zhu P. Combination of Endoglin and ASCVD Risk Assessment Improves Carotid Subclinical Atherosclerosis Recognition. J Atheroscler Thromb 2019; 27:331-341. [PMID: 31406054 PMCID: PMC7192815 DOI: 10.5551/jat.50898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aim: Our study investigated the association between soluble endoglin and carotid subclinical atherosclerosis. Methods: We used endoglin as an adjunct to atherosclerotic cardiovascular disease (ASCVD) risk, in recognition of carotid clinical atherosclerosis, in order to explore a new model to refine risk assessment. Out of 3,452 participants, 978 subjects with detected soluble endoglin were enrolled in a cross-sectional investigation in Fujian Province were enrolled. Soluble endoglin concentration in serum samples was evaluated using an enzyme-linked immunosorbent assay method. Carotid ultrasonography was used to detect intima-media thickness and carotid plaque. Results: The mean 10-year ASCVD risk by the new Pooled Cohort Equations accounted for 10.04% (± 12.35). The mean soluble endoglin level was 15.35 ng/ml (± 6.64). Multivariable regression demonstrated that age, systolic blood pressure, diastolic blood pressure, total cholesterol, high density lipoprotein cholesterol, and serum uric acid were independent determinants of soluble endoglin. Adding tests of ASCVD and endoglin together, in parallel, will increase the sensitivity and decrease specificity in recognizing carotid subclinical atherosclerosis. Evaluating the added value of endoglin to the ASCVD risk model showed significantly improved discrimination with analysis of C-statistics, continuous net reclassification index and integrated discrimination index. Both ASCVD risk and soluble endoglin showed positively linear correlation with carotid intima-media thickness (cIMT) (β = 0.006, P < 0.001; β = 0.485, P < 0.001). Even with adjustment for other factors, the relationship between log-transformed soluble endoglin with cIMT was still significant (β = 0.369, P < 0.001). Conclusions: The combination of ASCVD risk and endoglin levels increases carotid atherosclerosis recognition.
Collapse
Affiliation(s)
- Qiaowei Li
- Shengli Clinical Medical College of Fujian Medical University, Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian Institute of Clinical Geriatrics, Fujian Provincial Center for Geriatrics
| | - Fan Lin
- Shengli Clinical Medical College of Fujian Medical University, Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian Institute of Clinical Geriatrics, Fujian Provincial Center for Geriatrics
| | - Douli Ke
- Shengli Clinical Medical College of Fujian Medical University, Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian Institute of Clinical Geriatrics, Fujian Provincial Center for Geriatrics
| | - Qiong Cheng
- Department of Neurology, Fujian Provincial Hospital, Fujian Provincial Center for Geriatrics, Shengli Clinical Medical College of Fujian Medical University
| | - Yongzhi Gui
- Shengli Clinical Medical College of Fujian Medical University, Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian Institute of Clinical Geriatrics, Fujian Provincial Center for Geriatrics
| | - Yuyan Zhou
- Shengli Clinical Medical College of Fujian Medical University, Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian Institute of Clinical Geriatrics, Fujian Provincial Center for Geriatrics
| | - Yicheng Wu
- Shengli Clinical Medical College of Fujian Medical University, Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian Institute of Clinical Geriatrics, Fujian Provincial Center for Geriatrics
| | - Yinzhou Wang
- Department of Neurology, Fujian Provincial Hospital, Fujian Provincial Center for Geriatrics, Shengli Clinical Medical College of Fujian Medical University
| | - Pengli Zhu
- Shengli Clinical Medical College of Fujian Medical University, Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian Institute of Clinical Geriatrics, Fujian Provincial Center for Geriatrics
| |
Collapse
|
29
|
Dolezelova E, Sa ICI, Prasnicka A, Hroch M, Hyspler R, Ticha A, Lastuvkova H, Cermanova J, Pericacho M, Visek J, Lasticova M, Micuda S, Nachtigal P. High soluble endoglin levels regulate cholesterol homeostasis and bile acids turnover in the liver of transgenic mice. Life Sci 2019; 232:116643. [PMID: 31299237 DOI: 10.1016/j.lfs.2019.116643] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/01/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023]
Abstract
AIMS Increased plasma soluble endoglin concentrations (sEng) are frequently detected in metabolic disorders accompanied with hypercholesterolemia in serum, but effect of sEng on the cholesterol biochemistry is unknown. Cholesterol and bile acids (BA) are important products of liver metabolism with numerous functions within the organism. Turnover of these substances requires precise regulation due to potential toxicities during their cumulation. In this study, we hypothesized that high sEng levels affect cholesterol homeostasis and BA turnover in mice liver. MAIN METHODS Nine-month-old transgenic male mice overexpressing human sEng and wild-type mice underwent plasma, bile, stool, and organ samples analysis by analytical, qRT-PCT and Western blot methods. KEY FINDINGS sEng mice demonstrated decreased plasma total and LDL cholesterol concentrations due to upregulation of hepatic Sr-b1 and Ldlr receptors, increased liver cholesterol content, and increased Abcg8-mediated cholesterol efflux into bile. sEng also increased conversion of cholesterol into bile acids (BA) via upregulation of Cyp7a1 and increased Mdr1 expression. Plasma concentrations of BA were increased in sEng mice due to their enhanced reabsorption via ileum. Increased hepatic disposition of BA led to their increased biliary excretion coupled with choleretic activity. SIGNIFICANCE For the first time, we have shown that high sEng plasma levels affect cholesterol and BA homeostasis on the basis of complex liver and intestinal effects. The significance of these findings for pathophysiology of diseases associated with increased sEng concentrations remains to be elucidated in prospective studies.
Collapse
Affiliation(s)
- Eva Dolezelova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Ivone Cristina Igreja Sa
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Alena Prasnicka
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Milos Hroch
- Department of Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Czech Republic
| | - Radomir Hyspler
- Centrum for Research and Development, University Hospital, Hradec Kralove, Czech Republic
| | - Alena Ticha
- Centrum for Research and Development, University Hospital, Hradec Kralove, Czech Republic
| | - Hana Lastuvkova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Czech Republic
| | - Jolana Cermanova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Czech Republic
| | - Miguel Pericacho
- Biomedical Research Institute of Salamanca and Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain
| | - Jakub Visek
- 3rd Department of Internal Medicine, Metabolism and Gerontology, University Hospital, Hradec Kralove, Czech Republic
| | - Martina Lasticova
- 3rd Department of Internal Medicine, Metabolism and Gerontology, University Hospital, Hradec Kralove, Czech Republic
| | - Stanislav Micuda
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Czech Republic.
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic.
| |
Collapse
|
30
|
Buda V, Andor M, Baibata DE, Cozlac R, Radu G, Coricovac D, Danciu C, Ledeti I, Cheveresan A, Nica C, Tuduce P, Tomescu MC. Decreased sEng plasma levels in hypertensive patients with endothelial dysfunction under chronic treatment with Perindopril. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1915-1925. [PMID: 31239642 PMCID: PMC6556213 DOI: 10.2147/dddt.s186378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 03/15/2019] [Indexed: 12/25/2022]
Abstract
Purpose: Endoglin is a transmembrane glycoprotein which plays an important role in maintaining cardiovascular homeostasis. One of its forms, soluble endoglin (sEng), a molecule with antiangiogenic properties, has been found overexpressed in patients suffering from hypercholesterolemia, diabetes mellitus and hypertension, and is proposed as a marker of endothelial damage. Accordingly, we aimed to quantify the efficacy of various antihypertensive regimens on sEng levels, in hypertensive patients with endothelial dysfunction. Patients and methods: 323 patients were enrolled, and there were 99 patients with normal blood pressure values, 106 hypertensive patients under chronic treatment with different types of antihypertensive molecules (beta blockers, calcium channel blockers, and diuretics) in monotherapy, and 118 hypertensive patients under chronic treatment with perindopril. sEng plasma levels were quantified and were correlated with classical methods of assessing the endothelial damage. Results: Patients under chronic treatment with perindopril had lower sEng plasma levels compared with the other group of hypertensive patients under different regimens of antihypertensive treatment (sEng: 4.73±1.39 versus 5.63±2.33, p<0.01). Conclusion: Decreased sEng plasma levels were found in patients under chronic treatment with perindopril, when compared with other antihypertensive regimens of treatment (beta blockers, calcium channel blockers, and/or diuretics).
Collapse
Affiliation(s)
- Valentina Buda
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Minodora Andor
- Department of Medical Semiotics, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Dana Emilia Baibata
- Department of Cardiology VI, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania.,Cardiovascular Diseases Institute, Timisoara, Romania
| | - Ramona Cozlac
- Department of Cardiology VI, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania.,Cardiovascular Diseases Institute, Timisoara, Romania
| | - Gabriela Radu
- Department of Medical Semiotics, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Dorina Coricovac
- Department of Toxicology, Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Corina Danciu
- Department of Pharmacognosy, Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ionut Ledeti
- Department of Physical Chemistry, Faculty of Pharmacy, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Adelina Cheveresan
- Department of Pharmacology, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Cristian Nica
- Department of Surgery, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| | - Paul Tuduce
- Department of Balneophysiotherapy, Rheumatology and Rehabilitation, Faculty of Medicine, Vasile Goldis Western University, Arad, Romania
| | - Mirela Cleopatra Tomescu
- Department of Medical Semiotics, Faculty of Medicine, "Victor Babeş" University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
31
|
Vicen M, Vitverova B, Havelek R, Blazickova K, Machacek M, Rathouska J, Najmanová I, Dolezelova E, Prasnicka A, Sternak M, Bernabeu C, Nachtigal P. Regulation and role of endoglin in cholesterol-induced endothelial and vascular dysfunction in vivo and in vitro. FASEB J 2019; 33:6099-6114. [PMID: 30753095 DOI: 10.1096/fj.201802245r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Our objective was to investigate the effect of cholesterol [hypercholesterolemia and 7-ketocholesterol (7K)] on endoglin (Eng) expression and regulation with respect to endothelial or vascular dysfunction in vivo and in vitro. In vivo experiments were performed in 2-mo-old atherosclerosis-prone apolipoprotein E-deficient/LDL receptor-deficient (ApoE-/-/LDLR-/-) female mice and their wild-type C57BL/6J littermates. In in vitro experiments, human aortic endothelial cells (HAECs) were treated with 7K. ApoE-/-/LDLR-/- mice developed hypercholesterolemia accompanied by increased circulating levels of P-selectin and Eng and a disruption of NO metabolism. Functional analysis of the aorta demonstrated impaired vascular reactivity, and Western blot analysis revealed down-regulation of membrane Eng/Smad2/3/eNOS signaling in ApoE-/-/LDLR-/- mice. 7K increased Eng expression via Krüppel-like factor 6 (KLF6), liver X nuclear receptor, and NF-κB in HAECs. 7K-induced Eng expression was prevented by the treatment with 2-hydroxypropyl-β-cyclodextrin; 8-{[5-chloro-2-(4-methylpiperazin-1-yl) pyridine-4-carbonyl] amino}-1-(4-fluorophenyl)-4, 5-dihydrobenzo[g]indazole-3-carboxamide; or by KLF6 silencing. 7K induced increased adhesion and transmigration of monocytic human leukemia promonocytic cell line cells and was prevented by Eng silencing. We concluded that hypercholesterolemia altered Eng expression and signaling, followed by endothelial or vascular dysfunction before formation of atherosclerotic lesions in ApoE-/-/LDLR-/- mice. By contrast, 7K increased Eng expression and induced inflammation in HAECs, which was followed by an increased adhesion and transmigration of monocytes via endothelium, which was prevented by Eng inhibition. Thus, we propose a relevant role for Eng in endothelial or vascular dysfunction or inflammation when exposed to cholesterol.-Vicen, M., Vitverova, B., Havelek, R., Blazickova, K., Machacek, M., Rathouska, J., Najmanová, I., Dolezelova, E., Prasnicka, A., Sternak, M., Bernabeu, C., Nachtigal, P. Regulation and role of endoglin in cholesterol-induced endothelial and vascular dysfunction in vivo and in vitro.
Collapse
Affiliation(s)
- Matej Vicen
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Barbora Vitverova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Radim Havelek
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Katerina Blazickova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Miloslav Machacek
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jana Rathouska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Iveta Najmanová
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Eva Dolezelova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Alena Prasnicka
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Magdalena Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego, Krakow, Poland
| | - Carmelo Bernabeu
- Center for Biological Research, Spanish National Research Council (CSIC), Madrid, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
32
|
Doghish AS, Bassyouni AA, Mahfouz MH, Abd El-Aziz HG, Zakaria RY. Plasma endoglin in Type2 diabetic patients with nephropathy. Diabetes Metab Syndr 2019; 13:764-768. [PMID: 30641803 DOI: 10.1016/j.dsx.2018.11.058] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/30/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Diabetic nephropathy may be a common complication of diabetes mellitus. Endoglin is glycoprotein located on cell surfaces of endothelial cells and is part of the transforming growth factor beta (TGF- β) receptor. Endoglin expression is enhanced in endothelial cells during injury and inflammation. The aim of this study was to estimate the plasma level of soluble endoglin (sEng) in type 2 diabetic patients (with and without nephropathy). Also to explore its availability as marker for disease progression. METHODS In this study, sixty eight patients with type 2 diabetes mellitus (T2DM) were included; the patients were sub-grouped to normoalbuminuria without nephropathy and moderately increased albuminuria (microalbuminuria) with nephropathy groups with 13 individuals as control group. Plasma soluble endoglin level was determined using ELISA technique. Fasting plasma glucose (FPG), glycated haemoglobin (HbA1c), lipid profile, and creatinine were determined using colorimetric assay, whereas glomerular filtration rate (GFR) was calculated. RESULTS The plasma level of sEng of both normoalbuminuria group 1 and microalbuminuria group 2 were significantly higher when compared to control group. While, the plasma level of sEng in microalbuminuria group 2 was nonsignificant lower when compared to normoalbuminuria group 1. Also, there was a significant positive association between plasma level of sEng and HbA1c, HDL-C and urinary albumin concentration in normoalbuminuria group. CONCLUSION Plasma level of soluble Endoglin is markedly increase prior to alteration in endothelial function, and increases to lesser extent with the developing of diabetic nephropathy which indicated disease progression and development of vascular abnormalities.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, 13465, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Badr University in Cairo, Cairo, Egypt.
| | - Atef A Bassyouni
- National Institute of Diabetes and Endocrinology (NIDE), Cairo, Egypt
| | - Mohamed H Mahfouz
- National Institute of Diabetes and Endocrinology (NIDE), Cairo, Egypt
| | - Heba G Abd El-Aziz
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, 13465, Cairo, Egypt
| | - Rania Y Zakaria
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, 13465, Cairo, Egypt
| |
Collapse
|
33
|
Gallardo-Vara E, Tual-Chalot S, Botella LM, Arthur HM, Bernabeu C. Soluble endoglin regulates expression of angiogenesis-related proteins and induction of arteriovenous malformations in a mouse model of hereditary hemorrhagic telangiectasia. Dis Model Mech 2018; 11:dmm.034397. [PMID: 30108051 PMCID: PMC6176985 DOI: 10.1242/dmm.034397] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/29/2018] [Indexed: 12/16/2022] Open
Abstract
Endoglin is a transmembrane glycoprotein expressed in vascular endothelium that plays a key role in angiogenesis. Mutations in the endoglin gene (ENG) cause hereditary hemorrhagic telangiectasia type 1 (HHT1), characterized by arteriovenous malformations (AVMs) in different organs. These vascular lesions derive from abnormal processes of angiogenesis, whereby aberrant vascular remodeling leads to focal loss of capillaries. Current treatments for HHT1 include antiangiogenic therapies. Interestingly, a circulating form of endoglin (also known as soluble endoglin, sEng), proteolytically released from the membrane-bound protein and displaying antiangiogenic activity, has been described in several endothelial-related pathological conditions. Using human and mouse endothelial cells, we find that sEng downregulates several pro-angiogenic and pro-migratory proteins involved in angiogenesis. However, this effect is much reduced in endothelial cells that lack endogenous transmembrane endoglin, suggesting that the antiangiogenic activity of sEng is dependent on the presence of endogenous transmembrane endoglin protein. In fact, sEng partially restores the phenotype of endoglin-silenced endothelial cells to that of normal endothelial cells. Moreover, using an established neonatal retinal model of HHT1 with depleted endoglin in the vascular endothelium, sEng treatment decreases the number of AVMs and has a normalizing effect on the vascular phenotype with respect to vessel branching, vascular density and migration of the vascular plexus towards the retinal periphery. Taken together, these data show that circulating sEng can influence vascular development and AVMs by modulating angiogenesis, and that its effect on endothelial cells depends on the expression of endogenous endoglin. This article has an associated First Person interview with the first author of the paper. Summary: Soluble endoglin regulates vascular development and arteriovenous malformations by modulating angiogenesis, and its effect on endothelial cells depends on expression of endogenous membrane-bound endoglin.
Collapse
Affiliation(s)
- Eunate Gallardo-Vara
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain
| | - Simon Tual-Chalot
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| | - Luisa M Botella
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain
| | - Helen M Arthur
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Preeclampsia (PE) is a hypertensive disorder exclusive for pregnancy. It affects women all over the world and poses a great threat to life, both for mother and child. No definitive treatment exists and placenta delivery comprises the only known cure for PE. One of the most severe complications observed in preeclamptic women is the occurrence of cardiovascular diseases (CVDs) later in life. RECENT FINDINGS Both PE and CVDs share some of their pathogenic pathways and gene variations. Thus far, a number of publications have examined those relationships; however, almost all of them focus only on common risk factors. The precise pathomechanism and genetic basis of PE and its associated cardiovascular complications remain unknown. Therefore, the aim of this review is to unify and clarify the current state of knowledge and provide direction for future studies, especially those regarding the genetic aspect.
Collapse
Affiliation(s)
- Michalina Lisowska
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, Łódź, Poland.
| | - Tadeusz Pietrucha
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, Łódź, Poland
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, Łódź, Poland
| |
Collapse
|
35
|
Badmus OO, Michael OS, Rabiu S, Olatunji LA. Gestational glucocorticoid exposure disrupts glucose homeostasis that is accompanied by increased endoglin and DPP-4 activity instead of GSK-3 in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:66-75. [PMID: 29677638 DOI: 10.1016/j.etap.2018.04.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 05/27/2023]
Abstract
Gestational glucocorticoid (GC) treatment has been associated with cardiometabolic disorder (CMD) in offspring's in later life. Elevated dipeptidyl peptidase-4 (DPP-4) activity, endoglin and glycogen synthase kinase-3 (GSK-3) has also been implicated in the development of insulin resistance (IR) and/or vascular inflammation. We aimed to investigate the impact of GC exposure on glucose metabolism and the circulating levels of inflammatory biomarkers, DPP-4 activity and GSK-3 in pregnant rats. Pregnant Wistar rats received either vehicle or dexamethasone (DEX; 0.2 mg/kg; po) between gestational days 14 and 19. Gestational GC exposure resulted in impaired glucose homeostasis that is accompanied with elevated circulating levels of inflammatory biomarkers (endoglin, uric acid, and platelet/lymphocyte ratio), oxidative stress (malondialdehyde), blood viscosity, reduced NO level and increased DPP-4 activity. However, these effects were associated with atherogenic dyslipidemia and reduced GSK-3.We conclude that plasma endoglin, a marker of vascular inflammation, and plasma DPP-4 activity are increased in pregnant rats treated with GC during late gestation. Therefore, glucose deregulation associated with gestational GC exposure is through endoglin-/DPP-4-dependent but GSK-3-independent pathway.
Collapse
Affiliation(s)
- Olufunto O Badmus
- Department of Physiology & Hope Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria; Department of Public Health, Kwara State University, Malete, Nigeria
| | - Olugbenga S Michael
- Department of Physiology & Hope Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria; Department of Physiology, Cardiometabolic Research Unit, College of Health and Medical sciences, Bowen University, Iwo, Nigeria
| | - Saheed Rabiu
- Department of Physiology & Hope Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A Olatunji
- Department of Physiology & Hope Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.
| |
Collapse
|
36
|
Decreased Count and Dysfunction of Circulating EPCs in Postmenopausal Hypercholesterolemic Females via Reducing NO Production. Stem Cells Int 2018; 2018:2543847. [PMID: 29760721 PMCID: PMC5924981 DOI: 10.1155/2018/2543847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/14/2018] [Indexed: 12/27/2022] Open
Abstract
Endothelial progenitor cells (EPCs) contribute to the endogenous endothelial repair program during hypercholesterolemia. EPC count and migratory and proliferative capacities remain unchanged in the premenopausal female with hypercholesterolemia. However, the changes of count and activity of circulating EPCs in the hypercholesterolemic postmenopausal females are unknown. Here, we find that the migratory and proliferative capacities of circulating EPCs were decreased in patients with hypercholesterolemia versus normocholesterolemia. No significant differences were found between postmenopausal females and age-matched males. NO production showed positive correlation with the activity and count of circulating EPCs in patients with hypercholesterolemia. Flow-mediated dilatation (FMD) is directly interrelated with EPC counts and function. Our findings reveal that decreased EPC count and endothelial dysfunction lead to less NO production in hypercholesterolemic postmenopausal females. Maintaining the EPC numbers and activity might be emerging as a potential therapeutic strategy to reduce the risk of cardiovascular injury in elder women.
Collapse
|
37
|
Wang K, Lu C, Liu Y, Tao Y. In vitro
effects of sEng and TGF-β on human umbilical vein endothelial cells and trophoblasts. J Obstet Gynaecol Res 2018; 44:1023-1030. [PMID: 29673026 DOI: 10.1111/jog.13643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/20/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Ketao Wang
- Suzhou Hospital Affiliated to Nanjing Medical University; Suzhou Jiangsu Province China
| | - Caihua Lu
- Suzhou Hospital Affiliated to Nanjing Medical University; Suzhou Jiangsu Province China
| | - Yanpo Liu
- Department of Speacial Diseases; Branch Hospital of Dezhou Municiple Hospital; Dezhou China
| | - Ye Tao
- Suzhou Science and Technology Town Hospital; Suzhou Jiangsu Province China
| |
Collapse
|
38
|
Soluble endoglin and hypercholesterolemia aggravate endothelial and vessel wall dysfunction in mouse aorta. Atherosclerosis 2018; 271:15-25. [DOI: 10.1016/j.atherosclerosis.2018.02.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 11/19/2022]
|
39
|
Goumans MJ, Zwijsen A, Ten Dijke P, Bailly S. Bone Morphogenetic Proteins in Vascular Homeostasis and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031989. [PMID: 28348038 DOI: 10.1101/cshperspect.a031989] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo-angiogenesis as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - An Zwijsen
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, 3000 Leuven, Belgium
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Mécale (INSERM), U1036, 38000 Grenoble, France.,Laboratoire Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France.,University of Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
40
|
Li H, Yao J, Chang X, Wu J, Duan T, Wang K. LIFR increases the release of soluble endoglin via the upregulation of MMP14 expression in preeclampsia. Reproduction 2018; 155:297-306. [PMID: 29363569 DOI: 10.1530/rep-17-0732] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022]
Abstract
Preeclampsia (PE) is a pregnancy-specific disorder that is the main cause of maternal and perinatal morbidity and mortality worldwide. Inadequate trophoblastic invasion and endothelial dysfunction in the placenta are considered the foundation of the pathogenesis of preeclampsia in which soluble endoglin (sENG) plays an antiangiogenic role in the development of PE. The leukemia inhibitory factor receptor (LIFR) has been widely studied and is highly involved in arterial injury in vivo and in the migration of cancer cells in vitro Here, we tested the hypothesis that LIFR may be correlated with preeclampsia through its regulation of the release of sENG. Our data showed that LIFR protein, the expression of which significantly decreased with the progression of pregnancy, was located in the syncytiotrophoblast and cytotrophoblast. The LIFR protein level was increased in pregnancies with preeclampsia compared with normotensive full-term pregnancies. After the overexpression of LIFR in HTR8/SVneo cells, the release of sENG as well as the migration and invasion were significantly enhanced. Moreover, we also observed that LIFR induced the expression of matrix metalloproteinase14 (MMP14) and that the knockdown or inhibition of MMP14 decreased the release of sENG, as well as increased the LIFR-induced migration and invasion of HTR8/SVneo cells. These studies demonstrated that LIFR promoted the release of sENG through MMP14 in vitro, which indicates that LIFR may be involved in the development of preeclampsia.
Collapse
Affiliation(s)
- Hua Li
- Clinical and Translational Research CenterShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Julei Yao
- Clinical and Translational Research CenterShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xinwen Chang
- Clinical and Translational Research CenterShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jinting Wu
- Clinical and Translational Research CenterShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Tao Duan
- Clinical and Translational Research CenterShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China .,Department of ObstetricsShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Kai Wang
- Clinical and Translational Research CenterShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
41
|
Chronic Venous Insufficiency: Transforming Growth Factor-β Isoforms and Soluble Endoglin Concentration in Different States of Wound Healing. Int J Mol Sci 2017; 18:ijms18102206. [PMID: 29065449 PMCID: PMC5666886 DOI: 10.3390/ijms18102206] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 01/01/2023] Open
Abstract
Venous leg ulcer (VLU) is a huge healthcare problem with poorly understood pathophysiology. Transforming growth factor-β (TGF-β) and endoglin (Eng), are inflammatory and wound healing mediators. Eng, co-receptor for TGF-β type-II receptors, may be cleaved forming soluble Eng (sEng), antagonizing TGF-β signaling, a crucial process in vascular pathologies. We evaluated the accumulation in wound fluid (WF) of TGF-β isoforms and sEng in healing stages, showing the effects of sulodexide treatments, a glycosaminoglycan with clinical efficacy in VLU healing. Patients with inflammatory (Infl) and granulating (Gran) VLU were recruited. WFs and THP-1 monocytes exposed to Infl and Gran WF (treated/untreated with sulodexide) were analyzed for TGF-β isoforms and sEng by multiplex immunoassay. In both Infl and Gran WF, TGF-β1 and β2 were similar; TGF-β3 was significantly increased in Infl compared to Gran WFs (p = 0.033). sEng was significantly elevated in Gran compared to Infl WFs (p = 0.002). In THP-1 monocytes there was a significant increase in sEng after co-treatment of WF and sulodexide. The increase in TGF-β3 found in Infl WF highlights its negative effect on wound healing, while the increased levels of sEng in Gran WF affects the leukocyte adhesion/transmigration through the endothelium, reducing the inflammatory response and favoring the wound healing. Glycosaminoglycan sulodexide potentiates the effects of sEng release from monocyte, representing an important therapeutic option for wound healing.
Collapse
|
42
|
Chen L, Chen Z, Ge M, Tang O, Cheng Y, Zhou H, Shen Y, Qin F. Monocytic cell junction proteins serve important roles in atherosclerosis via the endoglin pathway. Mol Med Rep 2017; 16:6750-6756. [PMID: 28901429 PMCID: PMC5865831 DOI: 10.3892/mmr.2017.7444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 06/15/2017] [Indexed: 01/22/2023] Open
Abstract
The formation of atherosclerosis is recognized to be caused by multiple factors including pathogenesis in monocytes during inflammation. The current study provided evidence that monocytic junctions were significantly altered in patients with atherosclerosis, which suggested an association between cell junctions and atherosclerosis. Claudin‑1, occludin‑1 and ZO‑1 were significantly enhanced in atherosclerosis, indicating that the tight junction pathway was activated during the pathogenesis of atherosclerosis. In addition, the gene expression of 5 connexin members involved in the gap junction pathway were quantified, indicating that connexin 43 and 46 were significantly up‑regulated in atherosclerosis. Furthermore, inflammatory factors including endoglin and SMAD were observed, suggesting that immune regulative factors were down‑regulated in this pathway. Silicon‑based analysis additionally identified that connexins and tight junctions were altered in association with monocytic inflammation regulations, endoglin pathway. The results imply that reduced expression of the immune regulation pathway in monocytes is correlated with the generation of gap junctions and tight junctions which serve important roles in atherosclerosis.
Collapse
Affiliation(s)
- Lina Chen
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Zhongliang Chen
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Menghua Ge
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Oushan Tang
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Yinhong Cheng
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Haoliang Zhou
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Yu Shen
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Fengming Qin
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
43
|
Bláha V, Bláha M, Lánská M, Solichová D, Kujovská Krčmová L, Havel E, Vyroubal P, Zadák Z, Žák P, Sobotka L. Lipoprotein apheresis in the treatment of dyslipidaemia - the Czech Republic experience. Physiol Res 2017; 66:S91-S100. [PMID: 28379034 DOI: 10.33549/physiolres.933584] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In 1984, we started using therapeutic plasmapheresis (plasma exchange) as a method of extracorporeal lipoprotein elimination for the treatment of hypercholesterolemic patients. We evaluated the results of long-term therapy in 14 patients, 8 men and 6 women. The average age was 55.6+/-13.2 (range 28-70), median 59.5 years. 14 patients were diagnosed with familial hypercholesterolemia (FH): 5 homozygous, 9 heterozygous. Ten patients in the group were treated using immunoadsorption lipoprotein apheresis and 4 using hemorheopheresis. Immunoapheretic interventions decreased LDL-cholesterol (82+/-1 %), ApoB (73+/-13 %) and even Lp(a) by 82+/-19 %, respectively. Selected non-invasive methods are important for long-term and repeated follow-up. Carotid intima-media thickness showed improvement or stagnation in 75 % of the patients. Biomarkers of endothelial dysfunction such as endoglin (in the control group: 3.85+/-1.25 microg/l, in lipoprotein apheresis-treated hypercholesterolemic individuals 5.74+/-1.47 microg/l), CD40 ligand (before lipoprotein apheresis: 6498+/-2529 ng/l, after lipoprotein apheresis: 4057+/-2560 ng/l) and neopterin (before lipoprotein apheresis: 5.7+/-1.1 nmol/l, after lipoprotein apheresis: 5.5+/-1.3 nmol/l) related to the course of atherosclerosis, but did not reflect the actual activity of the disease nor facilitate the prediction or planning of therapy. Hemorheopheresis may improve blood flow in microcirculation in familial hypercholesterolemia and also in some other microcirculation disorders via significantly decreased activity of thrombomodulin (p<0.0001), tissue factor (p<0.0001), aggregation of thrombocytes (p<0.0001) and plasma and whole blood viscosity (p<0.0001). In conclusion, lipoprotein apheresis and hemorheopheresis substantially lowered LDL-cholesterol in severe hypercholesterolemia. Our experience with long-term therapy also shows good tolerance and a small number of complications (6.26 % non-serious clinical complications).
Collapse
Affiliation(s)
- V Bláha
- Third Department of Internal Medicine, Metabolism and Gerontology, University Hospital Hradec Králové and Charles University Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
High soluble endoglin levels do not induce changes in structural parameters of mouse heart. Heart Vessels 2017; 32:1013-1024. [DOI: 10.1007/s00380-017-0976-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/31/2017] [Indexed: 10/19/2022]
|
45
|
Varejckova M, Gallardo-Vara E, Vicen M, Vitverova B, Fikrova P, Dolezelova E, Rathouska J, Prasnicka A, Blazickova K, Micuda S, Bernabeu C, Nemeckova I, Nachtigal P. Soluble endoglin modulates the pro-inflammatory mediators NF-κB and IL-6 in cultured human endothelial cells. Life Sci 2017; 175:52-60. [DOI: 10.1016/j.lfs.2017.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 01/01/2023]
|
46
|
Soh MC, Nelson-Piercy C. Biomarkers for Adverse Pregnancy Outcomes in Rheumatic Diseases. Rheum Dis Clin North Am 2017; 43:201-214. [PMID: 28390563 DOI: 10.1016/j.rdc.2016.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Pregnancy is a delicate balance of angiogenic factors. Adverse pregnancy outcomes in the form of placental insufficiency occur when antiangiogenic factors predominate, which manifests as maternal-placental syndrome (MPS). Women with rheumatic disease are at increased risk of MPS. Endothelial damage from circulating antiangiogenic factors and other inflammatory molecules in combination with preexisting maternal vascular risk factors is the likely underlying pathophysiological process for MPS. It is likely that these changes persist, and additional "insults" from ongoing inflammation, medications, and disease damage contribute to the development of accelerated cardiovascular disease seen in young women with rheumatic disease.
Collapse
Affiliation(s)
- May Ching Soh
- Silver Star High-Risk Pregnancy Unit, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Headley Way, Oxford OX3 9DU, UK; de Sweit Obstetric Medicine Department, Queen Charlotte's & Chelsea Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London W12 0HS, UK; Women's Health Academic Centre, King's College London, St Thomas' Hospital, 10th Floor, North Wing, Westminster Bridge Road, London SE1 7EH, UK.
| | - Catherine Nelson-Piercy
- de Sweit Obstetric Medicine Department, Queen Charlotte's & Chelsea Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London W12 0HS, UK; Women's Health Academic Centre, King's College London, St Thomas' Hospital, 10th Floor, North Wing, Westminster Bridge Road, London SE1 7EH, UK
| |
Collapse
|
47
|
Plasma Soluble Endoglin Levels Are Inversely Associated With the Severity of Coronary Atherosclerosis—Brief Report. Arterioscler Thromb Vasc Biol 2017; 37:49-52. [DOI: 10.1161/atvbaha.116.308494] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 10/06/2016] [Indexed: 11/16/2022]
Abstract
Objective—
Transforming growth factor-β inhibits migration and proliferation of endothelial and smooth muscle cells. Endoglin is a transmembrane receptor for transforming growth factor-β1 and transforming growth factor-β3. Endoglin is released into blood as a soluble form (soluble endoglin [sEng]), but plasma sEng levels in patients with coronary artery disease (CAD) have not been elucidated.
Approach and Results—
We measured plasma sEng levels in 244 patients undergoing coronary angiography. The severity of coronary atherosclerosis was evaluated as the numbers of >50% stenotic vessels and segments. CAD was found in 147 patients, of whom 55 had 1-vessel, 42 had 2-vessel, and 50 had 3-vessel disease. Compared with 97 patients without CAD, 147 with CAD had lower sEng levels (median 4.04 versus 4.37 ng/mL;
P
<0.005). A stepwise decrease in sEng levels was found based on the number of stenotic vessels: 4.37 in CAD(−), 4.23 in 1-vessel, 4.13 in 2-vessel, and 3.74 ng/mL in 3-vessel disease (
P
<0.005). sEng levels inversely correlated with the number of stenotic segments (
r
=−0.25;
P
<0.001). In multivariate analysis, sEng was an independent factor for 3-vessel disease and CAD. Odds ratios for CAD and 3-vessel disease were 0.97 (95% confidence interval, 0.95–0.99;
P
<0.02) and 0.96 (95% confidence interval, 0.93–0.99;
P
<0.01) for a 0.1 ng/mL increase in sEng levels, respectively.
Conclusions—
Plasma sEng levels were low in patients with CAD, especially 3-vessel disease, and were inversely associated with the severity of coronary atherosclerosis.
Collapse
|
48
|
Çelik N, Cinaz P, Bideci A, Derinkuyu B, Emeksiz HC, Döğer E, Damar Ç, Yüce Ö, Çamurdan O. Endoglin and obestatin levels, cardiometabolic risk factors and subclinical atherosclerosis in children aged 10-18 years. J Pediatr Endocrinol Metab 2016; 29:1173-1180. [PMID: 27682709 DOI: 10.1515/jpem-2016-0024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/09/2016] [Indexed: 01/22/2023]
Abstract
BACKGROUND The aim of this study was to investigate the early signs of atherosclerosis and to evaluate serum endoglin and obestatin levels as predictors of subclinical atherosclerosis in obese children. METHODS A total of 95 children (60 obese and 35 controls) aged 10-18 years were included in the study. Their endoglin and obestatin levels and biochemical parameters were measured. The carotid intima media thickness (cIMT) and brachial artery flow-mediated dilatation (FMD) responses were evaluated. RESULTS The cIMT values were higher (p < 0.001) and FMD responses were lower (p = 0.003) in the obese group than in the control group. A logistic regression multivariate analysis revealed that cIMT was independently associated with the body mass index (BMI) Z-score (β = 0.323, p = 0.003) and low density lipoprotein (LDL) (β = 0.29, p = 0.008), while FMD % was independently associated with waist circumference (β = -0.36, p = 0.002). The obese and control groups were similar in endoglin (p = 0.67) and obestatin levels (p = 0.70). The endoglin level was inversely correlated with the cholesterol and LDL levels (r = -0.23, p = 0.032; rho = -0.25, p = 0.019). CONCLUSIONS The cIMT and brachial artery FMD response in obese children are significantly different compared to healthy controls. Circulating endoglin and obestatin levels are not predictive markers for subclinical atherosclerosis in obese children aged 10-18 years old.
Collapse
|