1
|
Kalies K, Knöpp K, Wurmbrand L, Korte L, Dutzmann J, Pilowski C, Koch S, Sedding D. Isolation of circulating endothelial cells provides tool to determine endothelial cell senescence in blood samples. Sci Rep 2024; 14:4271. [PMID: 38383692 PMCID: PMC10882010 DOI: 10.1038/s41598-024-54455-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/13/2024] [Indexed: 02/23/2024] Open
Abstract
Circulating endothelial cells (CEC) are arising as biomarkers for vascular diseases. However, whether they can be utilized as markers of endothelial cell (EC) senescence in vivo remains unknown. Here, we present a protocol to isolate circulating endothelial cells for a characterization of their senescent signature. Further, we characterize different models of EC senescence induction in vitro and show similar patterns of senescence being upregulated in CECs of aged patients as compared to young volunteers. Replication-(ageing), etoposide-(DNA damage) and angiotensin II-(ROS) induced senescence models showed the expected cell morphology and proliferation-reduction effects. Expression of senescence-associated secretory phenotype markers was specifically upregulated in replication-induced EC senescence. All models showed reduced telomere lengths and induction of the INK4a/ARF locus. Additional p14ARF-p21 pathway activation was observed in replication- and etoposide-induced EC senescence. Next, we established a combined magnetic activated- and fluorescence activated cell sorting (MACS-FACS) based protocol for CEC isolation. Interestingly, CECs isolated from aged volunteers showed similar senescence marker patterns as replication- and etoposide-induced senescence models. Here, we provide first proof of senescence in human blood derived circulating endothelial cells. These results hint towards an exciting future of using CECs as mirror cells for in vivo endothelial cell senescence, of particular interest in the context of endothelial dysfunction and cardiovascular diseases.
Collapse
Affiliation(s)
- Katrin Kalies
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany.
| | - Kai Knöpp
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Leonie Wurmbrand
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Laura Korte
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Straße 1, 30625, Hannover, Germany
| | - Jochen Dutzmann
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Claudia Pilowski
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Susanne Koch
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Daniel Sedding
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| |
Collapse
|
2
|
Bakhashab S, O’Neill J, Barber R, Arden C, Weaver JU. Upregulation of Anti-Angiogenic miR-106b-3p Correlates Negatively with IGF-1 and Vascular Health Parameters in a Model of Subclinical Cardiovascular Disease: Study with Metformin Therapy. Biomedicines 2024; 12:171. [PMID: 38255276 PMCID: PMC10813602 DOI: 10.3390/biomedicines12010171] [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: 12/24/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Well-controlled type 1 diabetes mellitus (T1DM) is regarded as a model of subclinical cardiovascular disease (CVD), characterized by inflammation and adverse vascular health. However, the underlying mechanisms are not fully understood. We investigated insulin-like growth factor-1 (IGF-1) and IGF-binding protein-3 (IGFBP-3) levels, their correlation to miR-106b-3p expression in a subclinical CVD model, and the cardioprotective effect of metformin. A total of 20 controls and 29 well-controlled T1DM subjects were studied. Plasma IGF-1, IGFBP-3 levels, and miR-106b-3p expression in colony-forming unit-Hills were analyzed and compared with vascular markers. miR-106b-3p was upregulated in T1DM (p < 0.05) and negatively correlated with pro-angiogenic markers CD34+/100-lymphocytes (p < 0.05) and IGF-1 (p < 0.05). IGF-1 was downregulated in T1DM (p < 0.01), which was associated with increased inflammatory markers TNF-α, CRP, and IL-10 and reduced CD34+/100-lymphocytes. IGFBP-3 had no significant results. Metformin had no effect on IGF-1 but significantly reduced miR-106b-3p (p < 0.0001). An Ingenuity Pathway analysis predicted miR-106b-3p to inhibit PDGFA, PIK3CG, GDNF, and ADAMTS13, which activated CVD. Metformin was predicted to be cardioprotective by inhibiting miR-106b-3p. In conclusion: Subclinical CVD is characterized by a cardio-adverse profile of low IGF-1 and upregulated miR-106b-3p. We demonstrated that the cardioprotective effect of metformin may be via downregulation of upregulated miR-106b-3p and its effect on downstream targets.
Collapse
Affiliation(s)
- Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, P.O. Box 80218, Jeddah 21589, Saudi Arabia;
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| | - Josie O’Neill
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Rosie Barber
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Catherine Arden
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Jolanta U. Weaver
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Department of Diabetes, Queen Elizabeth Hospital, Newcastle upon Tyne NE9 6SH, UK
- Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
3
|
Mitsis A, Avraamides P, Lakoumentas J, Kyriakou M, Sokratous S, Karmioti G, Drakomathioulakis M, Theodoropoulos KC, Nasoufidou A, Evangeliou A, Vassilikos V, Fragakis N, Ziakas A, Tzikas S, Kassimis G. Role of inflammation following an acute myocardial infarction: design of INFINITY. Biomark Med 2023; 17:971-981. [PMID: 38235565 DOI: 10.2217/bmm-2023-0491] [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] [Indexed: 01/19/2024] Open
Abstract
After a myocardial infarction, the inflammatory response is connected to major adverse outcomes such as ischemia-reperfusion injury, adverse cardiac remodeling, infarct size and poor prognosis. INFlammatIoN amI sTudY (INFINITY) is a multicenter, prospective, observational, cohort study designed to investigate the prognostic role of the cytokines IL-6, IL-10, IL-18 and IL-17 and the adipokines leptin, apelin and chemerin in patients with acute coronary syndrome. The study will test if these inflammatory biomarkers reflect different clinical manifestations of coronary artery disease and have a prognostic role in a 6-month follow-up period. This study represents an opportunity to investigate further the prognostic role of a selected combination of proinflammatory and anti-inflammatory biomarkers in the prognosis and risk stratification of acute coronary syndrome patients.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, Nicosia, 2029, Cyprus
| | | | - John Lakoumentas
- Department of Medical Physics, School of Medicine, University of Patras, Patras, 26504, Greece
| | - Michaela Kyriakou
- Cardiology Department, Nicosia General Hospital, Nicosia, 2029, Cyprus
| | | | - Georgia Karmioti
- Cardiology Department, Nicosia General Hospital, Nicosia, 2029, Cyprus
| | | | - Konstantinos C Theodoropoulos
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, 54636, Greece
| | - Athina Nasoufidou
- Second Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - Alexandros Evangeliou
- Third Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - Vassilios Vassilikos
- Third Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - Antonios Ziakas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, 54636, Greece
| | - Stergios Tzikas
- Third Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - George Kassimis
- Second Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| |
Collapse
|
4
|
Fang Y, Chen L, Imoukhuede PI. Toward Blood-Based Precision Medicine: Identifying Age-Sex-Specific Vascular Biomarker Quantities on Circulating Vascular Cells. Cell Mol Bioeng 2023; 16:189-204. [PMID: 37456786 PMCID: PMC10338416 DOI: 10.1007/s12195-023-00771-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Abnormal angiogenesis is central to vascular disease and cancer, and noninvasive biomarkers of vascular origin are needed to evaluate patients and therapies. Vascular endothelial growth factor receptors (VEGFRs) are often dysregulated in these diseases, making them promising biomarkers, but the need for an invasive biopsy has limited biomarker research on VEGFRs. Here, we pioneer a blood biopsy approach to quantify VEGFR plasma membrane localization on two circulating vascular proxies: circulating endothelial cells (cECs) and circulating progenitor cells (cPCs). Methods Using quantitative flow cytometry, we examined VEGFR expression on cECs and cPCs in four age-sex groups: peri/premenopausal females (aged < 50 years), menopausal/postmenopausal females (≥ 50 years), and younger and older males with the same age cut-off (50 years). Results cECs in peri/premenopausal females consisted of two VEGFR populations: VEGFR-low (~ 55% of population: population medians ~ 3000 VEGFR1 and 3000 VEGFR2/cell) and VEGFR-high (~ 45%: 138,000 VEGFR1 and 39,000-236,000 VEGFR2/cell), while the menopausal/postmenopausal group only possessed the VEGFR-low cEC population; and 27% of cECs in males exhibited high plasma membrane VEGFR expression (206,000 VEGFR1 and 155,000 VEGFR2/cell). The absence of VEGFR-high cEC subpopulations in menopausal/postmenopausal females suggests that their high-VEGFR cECs are associated with menstruation and could be noninvasive proxies for studying the intersection of age-sex in angiogenesis. VEGFR1 plasma membrane localization in cPCs was detected only in menopausal/postmenopausal females, suggesting a menopause-specific regenerative mechanism. Conclusions Overall, our quantitative, noninvasive approach targeting cECs and cPCs has provided the first insights into how sex and age influence VEGFR plasma membrane localization in vascular cells. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00771-1.
Collapse
Affiliation(s)
- Yingye Fang
- Department of Bioengineering, University of Washington, Seattle, WA USA
| | - Ling Chen
- Division of Biostatistics, Washington University in St. Louis School of Medicine, St. Louis, MO USA
| | - P. I. Imoukhuede
- Department of Bioengineering, University of Washington, Seattle, WA USA
| |
Collapse
|
5
|
Cavalcante S, Teixeira M, Gouveia M, Duarte A, Ferreira M, Simões MI, Conceição M, Costa M, Ribeiro IP, Gonçalves AC, Oliveira J, Ribeiro F. Reaktion der endothelialen Progenitorzellen auf ein Multikomponenten-Trainingsprogramm bei Erwachsenen mit kardiovaskulären Risikofaktoren. GERMAN JOURNAL OF EXERCISE AND SPORT RESEARCH 2023. [DOI: 10.1007/s12662-023-00882-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
6
|
de Bakker M, Kraan J, Akkerhuis KM, Oemrawsingh R, Asselbergs FW, Hoefer I, Kardys I, Boersma E. Longitudinal profile of circulating endothelial cells in post-acute coronary syndrome patients. Biomarkers 2023; 28:152-159. [PMID: 36617894 DOI: 10.1080/1354750x.2022.2162966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
IntroductionPatients who have experienced an acute coronary syndrome (ACS) are at risk of a recurrent event, but their level of risk varies. Because of their close temporal relationship with vascular injury, longitudinal measurements of circulating endothelial cells (CECs) carry potential to improve individual risk assessment.MethodsWe conducted an explorative nested case-control study within our multicenter, prospective, observational biomarker study (BIOMArCS) of 844 ACS patients. Following an index ACS, high-frequency blood sampling was performed during 1-year follow-up. CECs were identified using flow cytometric analyses in 15 cases with recurrent event, and 30 matched controls.ResultsCases and controls had a median (25th-75thpercentile) age of 64.1 (58.1-75.1) years and 80% were men. During the months preceding the endpoint, the mean (95%CI) CEC concentration in cases was persistently higher than in controls (12.8 [8.2-20.0] versus 10.0 [7.0-14.4] cells/ml), although this difference was non-significant (P = 0.339). In controls, the mean cell concentration was significantly (P = 0.030) lower in post 30-day samples compared to samples collected within one day after index ACS: 10.1 (7.5-13.6) versus 17.0 (10.8-26.6) cells/ml. Similar results were observed for CEC subsets co-expressing CD133 and CD309 (VEGFR-2) or CD106 (VCAM-1).ConclusionDespite their close relation to vascular damage, no increase in cell concentrations were found prior to the occurrence of a secondary adverse cardiac event.
Collapse
Affiliation(s)
- Marie de Bakker
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jaco Kraan
- Department of Medical Oncology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rohit Oemrawsingh
- Department of Cardiology, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Folkert W Asselbergs
- Amsterdam University Medical Centers, Department of Cardiology, University of Amsterdam, Amsterdam, The Netherlands.,Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Imo Hoefer
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
7
|
Xier Z, Zhu YX, Tang SW, Kong C, Aili D, Huojia G, Peng H. Plasma VWF: Ag levels predict long-term clinical outcomes in patients with acute myocardial infarction. Front Cardiovasc Med 2023; 9:1013815. [PMID: 36684571 PMCID: PMC9845945 DOI: 10.3389/fcvm.2022.1013815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 12/02/2022] [Indexed: 01/05/2023] Open
Abstract
Background A vital role in coronary artery disease is played by Von Willebrand factor (VWF), which serves as a bridge between platelets and the subendothelial matrix after vessel damage. The purpose of the study was to assess the validity of plasma VWF antigen (VWF: Ag) levels as a predictor of clinical outcomes after acute myocardial infarction (AMI). Methods Three hundred and seventy-four patients were studied following coronary angiography, including 209 patients suffering from acute myocardial infarction and 165 healthy participants. Coronary angiography was followed by measurement of plasma VWF: Ag levels. Over a 2-year follow-up period, major adverse cardiopulmonary and cerebrovascular events (MACEs) were the primary endpoint. All-cause mortality was investigated as a secondary endpoint. Results When compared to controls, patients with AMI had mean plasma VWF: Ag levels that were ~1.63 times higher (0.860 ± 0.309 vs. 0.529 ± 0.258 IU/ml; P < 0.001). The plasma VWF: Ag levels were substantially higher in patients who experienced MACEs after myocardial infarction vs. those without MACEs (1.088 ± 0.253 vs. 0.731 ± 0.252 IU/ml; P < 0.001). For predicting long-term MACEs using the optimal cut-off value (0.7884 IU/ml) of VWF: Ag, ROC curve area for VWF: Ag was 0.847, with a sensitivity of 87.2% and a specificity of 66.3% (95%CI: 0.792-0.902; P = 0.001). Two-year follow-up revealed a strong link between higher plasma VWF: Ag levels and long-term MACEs. At the 2-year follow-up, multivariate regression analysis revealed an independent relationship between plasma VWF: Ag levels and MACEs (HR = 6.004, 95%CI: 2.987-12.070). Conclusion We found evidence that plasma VWF: Ag levels were independent risk factors for AMI. Meanwhile, higher plasma VWF: Ag levels are associated with long-term MACEs in people with AMI.
Collapse
|
8
|
Effect of Low High-Density Lipoprotein Level on Endothelial Activation and Prothrombotic Processes in Coronary Artery Disease-A Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148637. [PMID: 35886486 PMCID: PMC9316205 DOI: 10.3390/ijerph19148637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022]
Abstract
High-density lipoproteins (HDL) play an important role in the prevention of atherosclerosis. The aim of the study was to assess the relationship between serum HDL-C concentration and proinflammatory/prothrombic activation in coronary artery disease (CAD) patients. The study group included 27 acute myocardial infarction (AMI) patients and 30 stable angina pectoris (SA) patients. The control group consisted of 23 people without cardiac symptoms. In the AMI and SA groups, a lower HDL-C and a higher LDL-C/HDL-C index were observed. The SA patients had lower total cholesterol, LDL-C, sE-selectin ligand, as well as higher triglycerides and CD40 concentration in comparison with both the control and AMI groups. A higher von Willebrand Factor and intercellular adhesion molecule-1 were found in both study groups. Low HDL-C concentration in the CAD patients may intensify pro-inflammatory endothelial activation and prothrombotic processes. A low concentration of HDL-C and a high value of the LDL-C/HDL-C index seem to be better indices of atherogenic processes than the LDL-C concentration alone.
Collapse
|
9
|
Heinisch PP, Bello C, Emmert MY, Carrel T, Dreßen M, Hörer J, Winkler B, Luedi MM. Endothelial Progenitor Cells as Biomarkers of Cardiovascular Pathologies: A Narrative Review. Cells 2022; 11:cells11101678. [PMID: 35626716 PMCID: PMC9139418 DOI: 10.3390/cells11101678] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 01/25/2023] Open
Abstract
Endothelial progenitor cells (EPC) may influence the integrity and stability of the vascular endothelium. The association of an altered total EPC number and function with cardiovascular diseases (CVD) and risk factors (CVF) was discussed; however, their role and applicability as biomarkers for clinical purposes have not yet been defined. Endothelial dysfunction is one of the key mechanisms in CVD. The assessment of endothelial dysfunction in vivo remains a major challenge, especially for a clinical evaluation of the need for therapeutic interventions or for primary prevention of CVD. One of the main challenges is the heterogeneity of this particular cell population. Endothelial cells (EC) can become senescent, and the majority of circulating endothelial cells (CEC) show evidence of apoptosis or necrosis. There are a few viable CECs that have properties similar to those of an endothelial progenitor cell. To use EPC levels as a biomarker for vascular function and cumulative cardiovascular risk, a correct definition of their phenotype, as well as an update on the clinical application and practicability of current isolation methods, are an urgent priority.
Collapse
Affiliation(s)
- Paul Philipp Heinisch
- Department of Congenital and Pediatric Heart Surgery, German Heart Center Munich, School of Medicine, Technical University of Munich, 80636 Munich, Germany;
- Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilians-Universität, 80636 Munich, Germany
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (C.B.); (M.M.L.)
- Correspondence:
| | - Corina Bello
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (C.B.); (M.M.L.)
| | - Maximilian Y. Emmert
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany;
- Institute of Regenerative Medicine (IREM), University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Thierry Carrel
- Department of Cardiac Surgery, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Martina Dreßen
- Department of Cardiovascular Surgery, Institute Insure, German Heart Center Munich, School of Medicine & Health, Technical University of Munich, Lazarettstrasse 36, 80636 Munich, Germany;
| | - Jürgen Hörer
- Department of Congenital and Pediatric Heart Surgery, German Heart Center Munich, School of Medicine, Technical University of Munich, 80636 Munich, Germany;
- Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilians-Universität, 80636 Munich, Germany
| | - Bernhard Winkler
- Department of Cardiovascular Surgery, Hospital Hietzing, 1130 Vienna, Austria;
| | - Markus M. Luedi
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (C.B.); (M.M.L.)
| |
Collapse
|
10
|
Cavalcante S, Teixeira M, Duarte A, Ferreira M, Simões MI, Conceição M, Costa M, Ribeiro IP, Gonçalves AC, Oliveira J, Ribeiro F. Endothelial Progenitor Cell Response to Acute Multicomponent Exercise Sessions with Different Durations. BIOLOGY 2022; 11:biology11040572. [PMID: 35453771 PMCID: PMC9025950 DOI: 10.3390/biology11040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 12/04/2022]
Abstract
It is widely accepted that exercise training has beneficial effects on vascular health. Although a dose-dependent relation has been suggested, little is known about the effects of different exercise durations on endothelial markers. This study aimed to assess the effect of single exercise sessions with different durations in the circulating levels of endothelial progenitor cells (EPCs) and endothelial cells (CECs) among adults with cardiovascular risk factors. Ten participants performed two multicomponent exercise sessions, one week apart, lasting 30 and 45 min (main exercise phase). Before and after each exercise session, blood samples were collected to quantify EPCs and CECs by flow cytometry. The change in EPCs was significantly different between sessions by 3.0% (95% CI: 1.3 to 4.7), being increased by 1.8 ± 1.7% (p = 0.009) in the 30 min session vs. −1.2 ± 2.0% (p > 0.05) in the 45 min session. No significant change was observed in CECs [−2.0%, 95%CI: (−4.1 to 0.2)] between the sessions. In conclusion, a multicomponent exercise session of 30 min promotes an acute increase in the circulating levels of EPCs without increasing endothelial damage (measured by the levels of CECs) among adults with cardiovascular risk factors.
Collapse
Affiliation(s)
- Suiane Cavalcante
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, 4099-002 Porto, Portugal; (S.C.); (J.O.)
| | - Manuel Teixeira
- Institute of Biomedicine—iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Ana Duarte
- Unidade Cuidados na Comunidade Cubo Mágico da Saúde, ACES Baixo Vouga, 3800-120 Aveiro, Portugal; (A.D.); (M.F.); (M.I.S.); (M.C.)
| | - Miriam Ferreira
- Unidade Cuidados na Comunidade Cubo Mágico da Saúde, ACES Baixo Vouga, 3800-120 Aveiro, Portugal; (A.D.); (M.F.); (M.I.S.); (M.C.)
| | - Maria I. Simões
- Unidade Cuidados na Comunidade Cubo Mágico da Saúde, ACES Baixo Vouga, 3800-120 Aveiro, Portugal; (A.D.); (M.F.); (M.I.S.); (M.C.)
| | - Maria Conceição
- Unidade Cuidados na Comunidade Cubo Mágico da Saúde, ACES Baixo Vouga, 3800-120 Aveiro, Portugal; (A.D.); (M.F.); (M.I.S.); (M.C.)
| | - Mariana Costa
- Câmara Municipal de Oliveira do Bairro—Projeto Não Fique Parado, 3800-120 Aveiro, Portugal;
| | - Ilda P. Ribeiro
- Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine (FMUC), University of Coimbra, 3004-531 Coimbra, Portugal;
- Institute for Clinical and Biomedical Research (iCBR), Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Institute for Clinical and Biomedical Research (iCBR)—Group of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine (FMUC), Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-531 Coimbra, Portugal;
- Laboratory of Oncobiology and Hematology, University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - José Oliveira
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, 4099-002 Porto, Portugal; (S.C.); (J.O.)
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4099-002 Porto, Portugal
| | - Fernando Ribeiro
- Institute of Biomedicine—iBiMED, School of Health Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence:
| |
Collapse
|
11
|
Mitsis A, Kadoglou NPE, Lambadiari V, Alexiou S, Theodoropoulos KC, Avraamides P, Kassimis G. Prognostic role of inflammatory cytokines and novel adipokines in acute myocardial infarction: An updated and comprehensive review. Cytokine 2022; 153:155848. [PMID: 35301174 DOI: 10.1016/j.cyto.2022.155848] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 12/19/2022]
Abstract
Acute myocardial infarction (AMI) is one of the major causes of morbidity and mortality worldwide. The inflammation response during and after AMI is common and seems to play a key role in the peri-AMI period, related with ischaemia-reperfusion injury, adverse cardiac remodelling, infarct size and poor prognosis. In this article, we provide an updated and comprehensive overview of the most important cytokines and adipokines involved in the complex pathophysiology mechanisms in AMI, summarizing their prognostic role post-AMI. Data so far support that elevated levels of the major proinflammatory cytokines TNFα, IL-6 and IL-1 and the adipokines adiponectin, visfatin and resistin, are linked to high mortality and morbidity. In contrary, there is evidence that anti-inflammatory cytokines and adipokines as IL-10, omentin-1 and ghrelin can suppress the AMI-induced inflammatory response and are correlated with better prognosis. Mixed data make unclear the role of the novel adipokines leptin and apelin. After all, imbalance of pro-inflammatory and anti-inflammatory cytokines may result in worst AMI prognosis. The incorporation of these inflammation biomarkers in established prognostic models could further improve their prognostic power improving overall the management of AMI patients.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, Cyprus.
| | | | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Institute and Diabetes Centre, Athens University Medical School, Attikon University General Hospital, Athens, Greece
| | - Sophia Alexiou
- Second Cardiology Department, "Hippokration" Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - George Kassimis
- Second Cardiology Department, "Hippokration" Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
12
|
Lopes J, Teixeira M, Cavalcante S, Gouveia M, Duarte A, Ferreira M, Simões MI, Conceição M, Ribeiro IP, Gonçalves AC, Schmidt C, de Jesus BB, Almeida R, Viamonte S, Santos M, Ribeiro F. Reduced Levels of Circulating Endothelial Cells and Endothelial Progenitor Cells in Patients with Heart Failure with Reduced Ejection Fraction. Arch Med Res 2022; 53:289-295. [DOI: 10.1016/j.arcmed.2022.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/03/2022] [Accepted: 02/02/2022] [Indexed: 11/26/2022]
|
13
|
Inflammatory cardiovascular risk markers and silent myocardial ischemia in type 2 diabetic patients. VOJNOSANIT PREGL 2022. [DOI: 10.2298/vsp201012010m] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background/Aim. A special feature of coronary heart disease (CHD) in patients with type 2 diabetes mellitus (T2DM) is that it is often a symptomatic and occurs as a consequence of cardiovascular autonomic neuropathy. Dysregulation of the autonomic nervous system is associated with elevated values of inflammatory markers such as high-sensitivity C-reactive protein (hs-CRP) and interleukin- 6 (IL-6), which accelerate atherosclerosis and the occurrence of cardiovascular complications in patients with T2DM. The aim of the study was to evaluate the importance of determining inflammatory cardiovascular risk markers IL-6 and hs-CRP in screening for the presence of CHD in asymptomatic patients with T2DM. Methods. The study included 169 patients with T2DM without any symptoms and signs of CHD. Ergometric testing proved or ruled out the presence of silent CHD. The levels of hs- CRP and IL-6 were determined by ELISA. Results. IL-6 values were significantly higher in patients with a positive ergometric test (6.83 ? 1.99 pg/mL) compared to patients with a negative ergometric test (3.04 ? 1.39 pg/mL) (p < 0.001). We also found that hs-CRP values in patients with a positive ergometric test were significantly higher compared to patients with a negative ergometric test (6.37 ? 2.25 vs 1.67 ? 1.41 mg/L; p < 0.001). Combinations of IL-6 and hs-CRP with age, HbA1c values, and duration of diabetes, presented through three binary logistic regression models, are significant predictors of silent CHD proven by ergometric testing, ie, with their increase, the probability of a positive ergometric test also increases (p < 0.01). The sensitivity of the associated finding of elevated IL-6 and hs-CRP values in the detection of silent CHD by ergometric testing was 90%, and the specificity was 86%. Conclusion. Hs-CRP and IL-6 are significant predictors of silent CHD, and their determination could be recommended for improving cardiovascular risk stratification in asymptomatic patients with T2DM.
Collapse
|
14
|
Sharda AV, Bogue T, Barr A, Mendez LM, Flaumenhaft R, Zwicker JI. Circulating Protein Disulfide Isomerase Is Associated with Increased Risk of Thrombosis in JAK2-Mutated Myeloproliferative Neoplasms. Clin Cancer Res 2021; 27:5708-5717. [PMID: 34400417 DOI: 10.1158/1078-0432.ccr-21-1140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/07/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Thromboembolic events (TE) are the most common complications of myeloproliferative neoplasms (MPN). Clinical parameters, including patient age and mutation status, are used to risk-stratify patients with MPN, but a true biomarker of TE risk is lacking. Protein disulfide isomerase (PDI), an endoplasmic reticulum protein vital for protein folding, also possesses essential extracellular functions, including regulation of thrombus formation. Pharmacologic PDI inhibition prevents thrombus formation, but whether pathologic increases in PDI increase TE risk remains unknown. EXPERIMENTAL DESIGN We evaluated the association of plasma PDI levels and risk of TE in a cohort of patients with MPN with established diagnosis of polycythemia vera (PV) or essential thrombocythemia (ET), compared with healthy controls. Plasma PDI was measured at enrollment and subjects followed prospectively for development of TE. RESULTS A subset of patients, primarily those with JAK2-mutated MPN, had significantly elevated plasma PDI levels as compared with controls. Plasma PDI was functionally active. There was no association between PDI levels and clinical parameters typically used to risk-stratify patients with MPN. The risk of TE was 8-fold greater in those with PDI levels above 2.5 ng/mL. Circulating endothelial cells from JAK2-mutated MPN patients, but not platelets, demonstrated augmented PDI release, suggesting endothelial activation as a source of increased plasma PDI in MPN. CONCLUSIONS The observed association between plasma PDI levels and increased risk of TE in patients with JAK2-mutated MPN has both prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Anish V Sharda
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.,Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Thomas Bogue
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Alexandra Barr
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Lourdes M Mendez
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Jeffrey I Zwicker
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts. .,Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Wang X, Fang F, Ni Y, Yu H, Ma J, Deng L, Li C, Shen Y, Liu X. The Combined Contribution of Vascular Endothelial Cell Migration and Adhesion to Stent Re-endothelialization. Front Cell Dev Biol 2021; 9:641382. [PMID: 33748131 PMCID: PMC7969796 DOI: 10.3389/fcell.2021.641382] [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: 12/14/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Coronary stent placement inevitably causes mechanical damage to the endothelium, leading to endothelial denudation and in-stent restenosis (ISR). Re-endothelialization depends mainly on the migration of vascular endothelial cells (VECs) adjacent to the damaged intima, as well as the mobilization and adhesion of circulating VECs. To evaluate the combined contribution of VEC migration and adhesion to re-endothelialization under flow and the influence of stent, in vitro models were constructed to simulate various endothelial denudation scales (2 mm/5 mm/10 mm) and stent deployment depths (flat/groove/bulge). Our results showed that (1) in 2 mm flat/groove/bulge models, both VEC migration and adhesion combined completed the percentage of endothelial recovery about 27, 16, and 12%, and migration accounted for about 21, 15, and 7%, respectively. It was suggested that the flat and groove models were in favor of VEC migration. (2) With the augmentation of the injury scales (5 and 10 mm), the contribution of circulating VEC adhesion on endothelial repair increased. Taken together, endothelial restoration mainly depended on the migration of adjacent VECs when the injury scale was 2 mm. The adhered cells contributed to re-endothelialization in an injury scale-dependent way. This study is helpful to provide new enlightenment for surface modification of cardiovascular implants.
Collapse
Affiliation(s)
- Xiaoli Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Fei Fang
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Yinghao Ni
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Hongchi Yu
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Jia Ma
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Li Deng
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Chunli Li
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Yang Shen
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Xiaoheng Liu
- West China School of Basic Medical Sciences and Forensic Medicine, Institute of Biomedical Engineering, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Di Martino ML, Frau A, Losa F, Muggianu E, Mura MN, Rotta G, Scotti L, Marongiu F. Role of circulating endothelial cells in assessing the severity of systemic sclerosis and predicting its clinical worsening. Sci Rep 2021; 11:2681. [PMID: 33514797 PMCID: PMC7846576 DOI: 10.1038/s41598-020-80604-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 12/09/2020] [Indexed: 11/09/2022] Open
Abstract
Endothelial damage and fibro-proliferative vasculopathy of small vessels are pathological hallmarks of systemic sclerosis (SSc). The consequence is the detachment of resident elements that become circulating endothelial cells (CECs). The aim of our study was to evaluate the potential of CECs as biomarker in SSc. We enrolled 50 patients with limited cutaneous (lcSSc) and diffuse cutaneous (dcSSc) subset of SSc, who underwent clinical evaluation to establish the organ involvement. CECs were measured by flow-cytometry utilizing a polychromatic panel. An evident difference was observed in CEC counts comparing controls to SSc patients (median 10.5 vs. 152 cells/ml, p < 0.0001) and for the first time, between the two subsets of disease (median lcSSc 132 vs. dcSSc 716 CEC/ml, p < 0.0001). A significant correlation was established between CECs and some SSc clinical parameters, such as digital ulcers, skin and pulmonary involvement, presence of Scl-70 antibodies, nailfold videocapillaroscopy patterns and EUSTAR activity index. After 12 months, CECs correlated with clinical worsening of patients, showing that a number higher than 414 CEC/ml is a strong negative prognostic factor (RR 5.70). Our results indicate that CECs are a direct indicator of systemic vascular damage. Therefore, they can be used as a reliable marker of disease severity.
Collapse
Affiliation(s)
- Maria Luisa Di Martino
- Department of Medical Sciences and Public Health and Unit of Internal Medicine, University of Cagliari, SS554-km 4,500, 09042, Monserrato, Cagliari, Italy.
| | - Alessandra Frau
- Department of Medical Sciences and Public Health and Unit of Internal Medicine, University of Cagliari, SS554-km 4,500, 09042, Monserrato, Cagliari, Italy
| | - Francesca Losa
- Department of Medical Sciences and Public Health and Unit of Internal Medicine, University of Cagliari, SS554-km 4,500, 09042, Monserrato, Cagliari, Italy
| | - Emma Muggianu
- Department of Medical Sciences and Public Health and Unit of Internal Medicine, University of Cagliari, SS554-km 4,500, 09042, Monserrato, Cagliari, Italy
| | - Mario Nicola Mura
- Department of Medical Sciences and Public Health and Unit of Internal Medicine, University of Cagliari, SS554-km 4,500, 09042, Monserrato, Cagliari, Italy
| | | | - Lorenza Scotti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Francesco Marongiu
- Department of Medical Sciences and Public Health and Unit of Internal Medicine, University of Cagliari, SS554-km 4,500, 09042, Monserrato, Cagliari, Italy
| |
Collapse
|
17
|
Möller C, Schutte AE, Smith W, Botha-Le Roux S. Von Willebrand factor, its cleaving protease (ADAMTS13), and inflammation in young adults: The African-PREDICT study. Cytokine 2020; 136:155265. [PMID: 32927287 DOI: 10.1016/j.cyto.2020.155265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/30/2020] [Accepted: 08/21/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND The role of inflammation in the early development of vascular dysfunction remains complex. Interleukin-6 (IL-6) and C-reactive protein (CRP) can cause an acute imbalance in the von Willebrand factor (vWF)-ADAMTS13 interaction, indicating a possible link between markers of haemostasis and low-grade inflammation. To better understand these inter-relationships in the early phases of disease development, we investigated whether vWF and ADAMTS13 associate with the pro-inflammatory markers, IL-6 and CRP in healthy young adults. We considered the role of blood types, sex and race on these relationships. METHODS In healthy black and white men and women (n = 1113; 24 ± 5 years; no previous diagnosis or medication use for chronic diseases) we analysed von Willebrand factor antigen (vWFag), ADAMTS13, IL-6 and CRP, and grouped blood types as non-O (A, B and AB) and O. Covariates included socioeconomic status, age, estimated glomerular filtration rate, 24-hour systolic blood pressure, waist circumference, glucose, total cholesterol, platelet count, γ-glutamyl transferase and total energy expenditure. RESULTS In the total group, vWFag was highest in the third tertile of both IL-6 and CRP (p ≤ 0.014), while ADAMTS13 was lowest in the third compared to the first IL-6 tertile (p = 0.006). In multivariate regression, vWFag associated positively with IL-6 (Adj R2 = 0.169; β = 0.123; p = 0.001) and CRP (Adj R2 = 0.163; β=0.094; p = 0.019) in the total group, in the O blood group (all p ≤ 0.051) and white men (all p ≤ 0.035). ADAMTS13 associated negatively with IL-6 (Adj R2 = 0.053; β = -0.154; p = 0.015) and CRP (Adj R2 = 0.055; β = -0.177; p = 0.009), only in the O blood group. CONCLUSIONS Markers of haemostasis associated independently with low-grade inflammation in the O type blood group and white men. An interplay between the haemostatic and inflammatory systems may already exist in young healthy adults and is dependent on blood groups, sex and race. This extends our understanding on the role of inflammation in the early development of vascular dysfunction prior to cardiovascular compromise.
Collapse
Affiliation(s)
- Christine Möller
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa; Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa; Faculty of Medicine, University of New South Wales, The George Institute for Global Health, Sydney, Australia
| | - Wayne Smith
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa; Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Shani Botha-Le Roux
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa; Medical Research Council: Research Unit for Hypertension and Cardiovascular Disease, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
18
|
Circulating Endothelial Cells From Septic Shock Patients Convert to Fibroblasts Are Associated With the Resuscitation Fluid Dose and Are Biomarkers for Survival Prediction. Crit Care Med 2020; 47:942-950. [PMID: 30998606 DOI: 10.1097/ccm.0000000000003778] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To determine whether circulating endothelial cells from septic shock patients and from nonseptic shock patients are transformed in activated fibroblast by changing the expression level of endothelial and fibrotic proteins, whether the level of the protein expression change is associated with the amount of administered resuscitation fluid, and whether this circulating endothelial cell protein expression change is a biomarker to predict sepsis survival. DESIGN Prospective study. SETTING Medical-surgical ICUs in a tertiary care hospital. PATIENTS Forty-three patients admitted in ICU and 22 healthy volunteers. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Circulating mature endothelial cells and circulating endothelial progenitor cells from septic shock and nonseptic shock patients showed evidence of endothelial fibrosis by changing the endothelial protein expression pattern. The endothelial proteins were downregulated, whereas fibroblast-specific markers were increased. The magnitude of the expression change in endothelial and fibrotic proteins was higher in the septic shock nonsurvivors patients but not in nonseptic shock. Interestingly, the decrease in the endothelial protein expression was correlated with the administered resuscitation fluid better than the Acute Physiology and Chronic Health Evaluation II and Sequential Organ Failure Assessment scores in the septic shock nonsurvivors patients but not in nonseptic shock. Notably, the significant difference between endothelial and fibrotic protein expression indicated a nonsurvival outcome in septic shock but not in nonseptic shock patients. Remarkably, area under the receiver operating characteristic curve analysis showed that endothelial protein expression levels predicted the survival outcome better than the Acute Physiology and Chronic Health Evaluation II and Sequential Organ Failure Assessment scores in septic shock but not in nonseptic shock patients. CONCLUSIONS Circulating endothelial cells from septic shock patients are acutely converted into fibroblasts. Endothelial and fibrotic protein expression level are associated with resuscitation fluid administration magnitude and can be used as biomarkers for an early survival diagnosis of sepsis.
Collapse
|
19
|
Effect of Acute Myocardial Infarction on a Disintegrin and Metalloprotease with Thrombospondin Motif 13 and Von Willebrand Factor and Their Relationship with Markers of Inflammation. Int J Vasc Med 2020; 2020:4981092. [PMID: 32095288 PMCID: PMC7036134 DOI: 10.1155/2020/4981092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/14/2022] Open
Abstract
Objectives Coagulation mechanisms and fibrinolytic assembly are important components role players of acute myocardial infarction (AMI) progression. Our study objective was to see the serial variations in the levels of Von Willebrand factor (VWF) and A Disintegrin and Metalloprotease with ThromboSpondin motif (ADAMTS13) over the course of AMI and to determine their relationship with the cardiovascular risk markers and the patient's clinical characteristics. Methods This project was done at the departments of Emergency Medicine, Physiology and Cardiac sciences of King Saud University Medical City. We studied ADAMTS13, VWF, fibrinogen, and CRP levels in 80 patients with AMI when patients were admitted; post AMI by 3-4 days and at follow-up of 3 months. We compared them with a control group consisting of 36 subjects. Results AMI had significantly lower levels of ADAMTS13 at AMI and after 3-4 days; at follow-up the difference in levels was nonsignificant, when compared with controls. Similarly, VWF levels were significantly higher in AMI and remained high even at follow-up compared to control subjects. VWF/ADAMTS13 ratio was also significantly higher at AMI and 3-4 days while at follow-up difference was nonsignificant compared to control subjects. Regression analysis between hsCRP and ADAMTS13 showed an inverse relationship (r = 0.376, p < 0.01), while correlation with VWF was significantly positive (r = 0.376, p < 0.01), while correlation with VWF was significantly positive (. Conclusions Increased levels of VWF and reduced levels of ADAMTS13 activity may contribute to the pathogenesis of acute myocardial infarction and might prove to be important mediators of AMI progression.
Collapse
|
20
|
Shavadia JS, Granger CB, Alemayehu W, Westerhout CM, Povsic TJ, Brener SJ, van Diepen S, Defilippi C, Armstrong PW. High-throughput targeted proteomics discovery approach and spontaneous reperfusion in ST-segment elevation myocardial infarction. Am Heart J 2020; 220:137-144. [PMID: 31812755 DOI: 10.1016/j.ahj.2019.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 09/20/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Although spontaneous reperfusion (SR) prior to primary percutaneous coronary intervention (pPCI) is associated with improved outcomes, its pathophysiology remains unclear. The objective of the study was to explore associations between SR in ST-segment elevation myocardial infarction (STEMI) using a multimarker cardiovascular proteins strategy METHODS: We evaluated STEMI patients from the Assessment of Pexelizumab in Acute Myocardial Infarction trial treated with pPCI within 6 hours from symptom onset. SR was core laboratory-defined as pre-PCI Thrombolysis in Myocardial Infarction flow 2 or 3. Ninety-one cardiovascular disease-related serum biomarkers drawn prior to PCI were analyzed using a high-throughput "targeted discovery" panel. Expression levels for individual biomarkers were compared between patients with/without SR. A hierarchical clustering method of biomarkers identified clusters of biomarkers that differentiated the 2 groups. Associations between individual biomarkers and clusters with SR were further evaluated by multivariable logistic regression. RESULTS Of 683 patients studied, 290 had spontaneous reperfusion; those with compared to without SR were more likely noninferior STEMI and had lower clinical acuity and lower baseline levels of troponin and creatine kinase. SR was associated with a lower occurrence of 90-day composite of death, heart failure, or cardiogenic shock. Fifty-two of 91 individual biomarkers were significantly univariably associated with SR. Forty-five remained significant with adjustment for false discovery rate. Using cluster analysis, 26 biomarkers clusters were identified, explaining 72% of total covariance, and 13 biomarker clusters were significantly associated with SR after multivariable adjustment. SR was associated with higher mean expression levels of proteins in all 13 clusters. The cluster most strongly associated with SR consisted of novel proteins across various distinct, yet interlinked, pathobiological processes (kallikrein-6, matrix extracellular phosphoglycoprotein, matrix mettaloproteinaise-3, and elafin). CONCLUSIONS Spontaneous reperfusion prior to pPCI in STEMI was associated with a lower risk of adverse clinical events. These exploratory data from a targeted discovery proteomics platform identifies novel proteins across diverse, yet complementary, pathobiological axes that show promise in providing mechanistic insights into spontaneous reperfusion in STEMI. CONDENSED ABSTRACT Spontaneous reperfusion has been established with improved STEMI outcomes, yet its pathobiology is unclear and appears to involve diverse physiological processes. Using a 91-biomarker high-throughput proteomics platform, we studied 683 STEMI patients in the APEX AMI trial (290 had core laboratory-adjudicated pre-PCI TIMI 2/3 flow) and identified 52 proteins that univariably associate with spontaneous reperfusion. Cluster analysis identified 26 biomarker clusters (explaining 72% of total variance), 13 of which, after multivariable adjustment, were significantly associated with spontaneous reperfusion. Four proteins (kallikrein-6, matrix extracellular phosphoglycoprotein, matrix mettaloproteinaise-3, and elafin) across diverse, yet complementary, pathways appear to be associated most strongly with spontaneous reperfusion.
Collapse
Affiliation(s)
- Jay S Shavadia
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Duke Clinical Research Institute, Durham, NC, USA.
| | | | | | | | | | - Sorin J Brener
- Department of Medicine, Cardiac Catheterization Laboratory, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY, USA
| | - Sean van Diepen
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | - Paul W Armstrong
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
21
|
Gu J, Zhu H, Zhu D, Li M, Xiao M, Yan D, Shen S. VWF, CXCL8 and IL6 might be potential druggable genes for acute coronary syndrome (ACS). Comput Biol Chem 2019; 83:107125. [DOI: 10.1016/j.compbiolchem.2019.107125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 01/31/2023]
|
22
|
von Känel R, Princip M, Schmid JP, Barth J, Znoj H, Schnyder U, Meister-Langraf RE. Association of sleep problems with neuroendocrine hormones and coagulation factors in patients with acute myocardial infarction. BMC Cardiovasc Disord 2018; 18:213. [PMID: 30463526 PMCID: PMC6249741 DOI: 10.1186/s12872-018-0947-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/01/2018] [Indexed: 12/14/2022] Open
Abstract
Background Obstructive sleep apnea (OSA) and insomnia are frequent sleep problems that are associated with poor prognosis in patients with coronary heart disease. The mechanisms linking poor sleep with an increased cardiovascular risk are incompletely understood. We examined whether a high risk of OSA as well as insomnia symptoms are associated with neuroendocrine hormones and coagulation factors in patients admitted with acute myocardial infarction. Methods We assessed 190 patients (mean age 60 years, 83% men) in terms of OSA risk (STOP screening tool for the assessment of high vs. low OSA risk) and severity of insomnia symptoms (Jenkins Sleep Scale for the assessment of subjective sleep difficulties) within 48 h of an acute coronary intervention. Circulating concentrations of epinephrine, norepinephrine, cortisol, fibrinogen, D-dimer, and von Willebrand factor were measured the next morning. The association of OSA risk and insomnia symptoms with neuroendocrine hormones and coagulation factors was computed using multivariate models adjusting for demographic factors, health behaviors, somatic and psychiatric comorbidities, cardiac disease-related variables, and OSA risk in the model for insomnia symptoms, respectively, for insomnia symptoms in the model for OSA risk. Results High OSA risk was identified in 41% of patients and clinically relevant insomnia symptoms were reported by 27% of patients. Compared to those with low OSA risk, patients with high OSA risk had lower levels of epinephrine (p = 0.015), norepinephrine (p = 0.049) and cortisol (p = 0.001). More severe insomnia symptoms were associated with higher levels of fibrinogen (p = 0.037), driven by difficulties initiating sleep, and with lower levels of norepinephrine (p = 0.024), driven by difficulties maintaining sleep. Conclusions In patients with acute myocardial infarction, sleep problems are associated with neuroendocrine hormones and coagulation activity. The pattern of these relationships is not uniform for patients with a high risk of OSA and those with insomnia symptoms, and whether they contribute to adverse cardiovascular outcomes needs to be established. Trial registration ClinicalTrials.gov NCT01781247.
Collapse
Affiliation(s)
- Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, Culmannstrasse 8, CH-8091, Zurich, Switzerland.
| | - Mary Princip
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, Culmannstrasse 8, CH-8091, Zurich, Switzerland
| | - Jean-Paul Schmid
- Department of Cardiology, Clinic Barmelweid, Barmelweid, Switzerland
| | - Jürgen Barth
- Complementary and Integrative Medicine, University of Zurich, Zurich, Switzerland
| | - Hansjörg Znoj
- Department of Clinical Psychology and Psychotherapy, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
23
|
Diego-Nieto A, Vidriales MB, Alonso-Orcajo N, Moreno-Samos JC, Martin-Herrero F, Carbonell R, Cid B, Cruz-Gonzalez I, Martin-Moreiras JC, Cuellas C, Pascual C, Lopez-Benito M, Sanchez PL, Fernandez-Vazquez F, de Prado AP. No Differences in Levels of Circulating Progenitor Endothelial Cells or Circulating Endothelial Cells Among Patients Treated With Ticagrelor Compared With Clopidogrel During Non- ST -Segment-Elevation Myocardial Infarction. J Am Heart Assoc 2018; 7:e009444. [PMID: 30371302 PMCID: PMC6404906 DOI: 10.1161/jaha.118.009444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Ticagrelor use during acute coronary syndromes demonstrated a decrease in all‐cause mortality in the PLATO (Platelet Inhibition and Patient Outcomes) trial. This effect has been attributed to a non–platelet‐derived improvement in endothelial function. The aim of this study was to determine differences in the number of endothelial progenitor cells and/or circulating endothelial cells found in peripheral blood in patients treated with either ticagrelor or clopidogrel during non–ST‐segment–elevation myocardial infarction. Methods and Results In this multicenter, randomized study (NCT02244710), patients were considered for inclusion after non–ST‐segment–elevation myocardial infarction whenever they were P2Y12‐inhibitor naïve. Ticagrelor and clopidogrel were allocated at a 1:1 ratio. Blood samples for determining endothelial progenitor cells and circulating endothelial cells were extracted before the antiplatelet loading dose, 48 hours after presentation of index symptoms, and 1 month after the event. A multichannel cytometer was used for optimal cell characterization. A total of 96 patients fulfilled the inclusion criteria. Circulating endothelial cell levels corrected by white blood cells were as follows at baseline, 48 hours, and 1 month: 44 (28–64), 50 (33–63), and 38 (23–62) cells/mL, respectively, for clopidogrel and 38 (29–60), 45 (32–85), and 35 (24–71) cells/mL, respectively, for ticagrelor (P=0.6). Endothelial progenitor cell levels were 29 (15–47), 27 (15–33), and 18 (10–25) cells/mL, respectively, for clopidogrel and 20 (11–33), 22 (12–32), and 18 (11–29) cells/mL, respectively, for ticagrelor (P=0.9). No differences in intraindividual changes were found. Conclusions Patients treated with ticagrelor during non–ST‐segment–elevation myocardial infarction, in comparison to clopidogrel, showed similar levels of endothelial progenitor cells and circulating endothelial cells. These data suggest that the endothelial protective effect mediated by ticagrelor is not related to bone marrow physiology modulation. Clinical Trial Registration URL: https://www.clinicaltrials.gov. Unique identifier: NCT02244710.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Belen Cid
- 4 Department of Cardiology City of Universitu Hospital of Santiago de Compostela
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhang B, Li XL, Zhao CR, Pan CL, Zhang Z. Interleukin-6 as a Predictor of the Risk of Cardiovascular Disease: A Meta-Analysis of Prospective Epidemiological Studies. Immunol Invest 2018; 47:689-699. [PMID: 29873573 DOI: 10.1080/08820139.2018.1480034] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Bo Zhang
- Department of Cardiovascular Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiao-Ling Li
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Cun-Rui Zhao
- Department of Cardiovascular Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chen-Liang Pan
- Department of Cardiovascular Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zheng Zhang
- Department of Cardiovascular Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
25
|
Lanuti P, Simeone P, Rotta G, Almici C, Avvisati G, Azzaro R, Bologna G, Budillon A, Di Cerbo M, Di Gennaro E, Di Martino ML, Diodato A, Doretto P, Ercolino E, Falda A, Gregorj C, Leone A, Losa F, Malara N, Marini M, Mastroroberto P, Mollace V, Morelli M, Muggianu E, Musolino G, Neva A, Pierdomenico L, Pinna S, Piovani G, Roca MS, Russo D, Scotti L, Tirindelli MC, Trunzo V, Venturella R, Vitagliano C, Zullo F, Marchisio M, Miscia S. A standardized flow cytometry network study for the assessment of circulating endothelial cell physiological ranges. Sci Rep 2018; 8:5823. [PMID: 29643468 PMCID: PMC5895616 DOI: 10.1038/s41598-018-24234-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
Circulating endothelial cells (CEC) represent a restricted peripheral blood (PB) cell subpopulation with high potential diagnostic value in many endothelium-involving diseases. However, whereas the interest in CEC studies has grown, the standardization level of their detection has not. Here, we undertook the task to align CEC phenotypes and counts, by standardizing a novel flow cytometry approach, within a network of six laboratories. CEC were identified as alive/nucleated/CD45negative/CD34bright/CD146positive events and enumerated in 269 healthy PB samples. Standardization was demonstrated by the achievement of low inter-laboratory Coefficients of Variation (CVL), calculated on the basis of Median Fluorescence Intensity values of the most stable antigens that allowed CEC identification and count (CVL of CD34bright on CEC ~ 30%; CVL of CD45 on Lymphocytes ~ 20%). By aggregating data acquired from all sites, CEC numbers in the healthy population were captured (medianfemale = 9.31 CEC/mL; medianmale = 11.55 CEC/mL). CEC count biological variability and method specificity were finally assessed. Results, obtained on a large population of donors, demonstrate that the established procedure might be adopted as standardized method for CEC analysis in clinical and in research settings, providing a CEC physiological baseline range, useful as starting point for their clinical monitoring in endothelial dysfunctions.
Collapse
Affiliation(s)
- Paola Lanuti
- Department of Medicine and Aging Sciences, University "G.d'Annunzio", Chieti-Pescara, Italy
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, University "G.d'Annunzio", Chieti-Pescara, Italy
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy
| | | | - Camillo Almici
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, Spedali Civili of Brescia, Brescia, Italy
| | - Giuseppe Avvisati
- Hematology, Stem Cell Transplantation, Transfusion Medicine and Cellular Therapy, Department of Medicine, Campus Bio-Medico University Hospital, Roma, Italy
| | - Rosa Azzaro
- Transfusion Service, Department of Hematology-Oncology and Stem Cell Transplantation Unit, Napoli, Italy
| | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, University "G.d'Annunzio", Chieti-Pescara, Italy
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Department of Research, Istituto Nazionale Tumori- IRCCS G. Pascale, Napoli, Italy
| | - Melania Di Cerbo
- Hematology, Stem Cell Transplantation, Transfusion Medicine and Cellular Therapy, Department of Medicine, Campus Bio-Medico University Hospital, Roma, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Department of Research, Istituto Nazionale Tumori- IRCCS G. Pascale, Napoli, Italy
| | - Maria Luisa Di Martino
- Unit of Internal Medicine, Allergy and Clinical Immunology, Department of Medical Sciences "M. Aresu", University of Cagliari Monserrato, Cagliari, Italy
| | - Annamaria Diodato
- Transfusion Service, Department of Hematology-Oncology and Stem Cell Transplantation Unit, Napoli, Italy
| | - Paolo Doretto
- Clinical Pathology Laboratory, Department of Laboratory Medicine, AAS5, Pordenone Hospital, Pordenone, Italy
| | - Eva Ercolino
- Department of Medicine and Aging Sciences, University "G.d'Annunzio", Chieti-Pescara, Italy
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy
| | - Alessandra Falda
- Clinical Pathology Laboratory, Department of Laboratory Medicine, AAS5, Pordenone Hospital, Pordenone, Italy
| | - Chiara Gregorj
- Hematology, Stem Cell Transplantation, Transfusion Medicine and Cellular Therapy, Department of Medicine, Campus Bio-Medico University Hospital, Roma, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit, Department of Research, Istituto Nazionale Tumori- IRCCS G. Pascale, Napoli, Italy
| | - Francesca Losa
- Unit of Internal Medicine, Allergy and Clinical Immunology, Department of Medical Sciences "M. Aresu", University of Cagliari Monserrato, Cagliari, Italy
| | - Natalia Malara
- Department of Health Science University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Mirella Marini
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, Spedali Civili of Brescia, Brescia, Italy
| | - Pasquale Mastroroberto
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Vincenzo Mollace
- Department of Health Science University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Michele Morelli
- Department of Obstetrics and Gynecology, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Emma Muggianu
- Unit of Internal Medicine, Allergy and Clinical Immunology, Department of Medical Sciences "M. Aresu", University of Cagliari Monserrato, Cagliari, Italy
| | - Giuseppe Musolino
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Arabella Neva
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, Spedali Civili of Brescia, Brescia, Italy
| | - Laura Pierdomenico
- Department of Medicine and Aging Sciences, University "G.d'Annunzio", Chieti-Pescara, Italy
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy
| | - Silvia Pinna
- Unit of Internal Medicine, Allergy and Clinical Immunology, Department of Medical Sciences "M. Aresu", University of Cagliari Monserrato, Cagliari, Italy
| | - Giovanna Piovani
- Department Molecular Medicine and Translational, University of Brescia, Brescia, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Department of Research, Istituto Nazionale Tumori- IRCCS G. Pascale, Napoli, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, Brescia, Italy
| | - Lorenza Scotti
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milano, Italy
| | - Maria Cristina Tirindelli
- Hematology, Stem Cell Transplantation, Transfusion Medicine and Cellular Therapy, Department of Medicine, Campus Bio-Medico University Hospital, Roma, Italy
| | - Valentina Trunzo
- Department of Health Science University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Roberta Venturella
- Department of Obstetrics and Gynecology, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit, Department of Research, Istituto Nazionale Tumori- IRCCS G. Pascale, Napoli, Italy
| | - Fulvio Zullo
- Department of Obstetrics and Gynecology, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Marco Marchisio
- Department of Medicine and Aging Sciences, University "G.d'Annunzio", Chieti-Pescara, Italy.
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy.
| | - Sebastiano Miscia
- Department of Medicine and Aging Sciences, University "G.d'Annunzio", Chieti-Pescara, Italy
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy
| |
Collapse
|
26
|
|
27
|
Enumeration of circulating endothelial cell frequency as a diagnostic marker in aortic valve surgery - a flow cytometric approach. J Cardiothorac Surg 2017; 12:68. [PMID: 28793899 PMCID: PMC5551027 DOI: 10.1186/s13019-017-0631-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/02/2017] [Indexed: 12/02/2022] Open
Abstract
Background The frequency of circulating endothelial cells (CEC) in patients’ peripheral blood can be assessed as a direct marker of endothelial damage. However, conventional enumeration methods are extremely challenging. We developed a novel, automated approach to determine CEC frequencies and tested this method on two groups of patients undergoing conventional (CAVR) versus trans-catheter aortic valve implantation (TAVI). Methods CEC frequencies were assessed by a flow cytometric approach, including automated pre-enrichment of CD34 positive blood cell subpopulation and isotype controls. The efficacy and reproducibility of the CEC enumeration method was validated by spiking blood samples of healthy control donors with defined numbers of endothelial cells. Results CEC frequencies were significantly higher in the TAVI group before (9.8 ± 4.1 vs. 5.5 ± 2.2, p = 0.019) and 1 h after surgery (13.4 ± 5.1 vs. 8.2 ± 4.1, p = 0.030) corresponding to higher Euroscore, STS score in higher risk patients from the TAVI group. Five days after surgery, CEC frequencies became significantly higher in the more invasive CAVR group (39.0 ± 13.0 vs. 14.3 ± 4.4, p < 0.001) compared to minimally invasive TAVI approach. Conclusions The new flow cytometric approach might be a robust and reliable method for CEC enumeration. Initial results show that CEC frequency is a valid clinical marker for the assessment of pre-operative risk, invasiveness of surgical procedure and clinical outcome. Further studies are necessary to validate the practical clinical usefulness and the potential superiority compared to conventional markers.
Collapse
|
28
|
Dunbar RL, Movva R, Bloedon LT, Duffy D, Norris RB, Navab M, Fogelman AM, Rader DJ. Oral Apolipoprotein A-I Mimetic D-4F Lowers HDL-Inflammatory Index in High-Risk Patients: A First-in-Human Multiple-Dose, Randomized Controlled Trial. Clin Transl Sci 2017; 10:455-469. [PMID: 28795506 PMCID: PMC5673907 DOI: 10.1111/cts.12487] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 06/13/2017] [Indexed: 12/26/2022] Open
Abstract
A single dose of the apolipoprotein (apo)A-I mimetic peptide D-4F rendered high-density lipoprotein (HDL) less inflammatory, motivating the first multiple-dose study. We aimed to assess safety/tolerability, pharmacokinetics, and pharmacodynamics of daily, orally administered D-4F. High-risk coronary heart disease (CHD) subjects added double-blinded placebo or D-4F to statin for 13 days, randomly assigned 1:3 to ascending cohorts of 100, 300, then 500 mg (n = 62; 46 men/16 women). D-4F was safe and well-tolerated. Mean ± SD plasma D-4F area under the curve (AUC, 0-8h) was 6.9 ± 5.7 ng/mL*h (100 mg), 22.7 ± 19.6 ng/mL*h (300 mg), and 104.0 ± 60.9 ng/mL*h (500 mg) among men, higher among women. Whereas placebo dropped HDL inflammatory index (HII) 28% 8 h postdose (range, 1.25-0.86), 300-500 mg D-4F effectively halved HII: 1.35-0.57 and 1.22-0.63, respectively (P < 0.03 vs. placebo). Oral D-4F peptide dose predicted HII suppression, whereas plasma D-4F exposure was dissociated, suggesting plasma penetration is unnecessary. In conclusion, oral D-4F dosing rendered HDL less inflammatory, affirming oral D-4F as a potential therapy to improve HDL function.
Collapse
Affiliation(s)
- Richard L Dunbar
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rajesh Movva
- Eastern Maine Medical Center, Bangor, Maine, USA
| | | | - Danielle Duffy
- Department of Medicine, Division of Cardiology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert B Norris
- Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mohamad Navab
- University of California, Los Angeles, California, USA
| | | | - Daniel J Rader
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
|
30
|
van Diepen S, Alemayehu WG, Zheng Y, Theroux P, Newby LK, Mahaffey KW, Granger CB, Armstrong PW. Temporal changes in biomarkers and their relationships to reperfusion and to clinical outcomes among patients with ST segment elevation myocardial infarction. J Thromb Thrombolysis 2016; 42:376-85. [DOI: 10.1007/s11239-016-1390-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
31
|
Ryder JR, O'Connell MJ, Rudser KD, Fox CK, Solovey AN, Hebbel RP, Kelly AS. Reproducibility of circulating endothelial cell enumeration and activation in children and adolescents. Biomark Med 2016; 10:463-71. [PMID: 27071934 DOI: 10.2217/bmm-2015-0051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION We examined the reproducibility of circulating endothelial cells (CEC) enumeration and activation among youth. MATERIALS AND METHODS CECs from 151 youth were measured at baseline and 1 week follow-up. Enumeration of CEC in fresh whole blood was determined by direct assessment of buffy coat smears (CD146+ nucleated cells) and activated CEC (%VCAM-1 expression) was determined after immunomagnetic enrichment and co-staining of nuclei, plus positivity for P1H12 and VCAM-1. RESULTS No statistically significant difference in CEC enumeration (1.2 ± 2.5 vs 1.3 ± 2.2 CEC/milliliter of whole blood, p = 0.745) or activated CEC (57.1 ± 24.4 vs 58.0 ± 21.3 %VCAM-1, p = 0.592) between baseline and 1 week follow-up. CONCLUSION On a cohort basis, CEC enumeration and activation are reproducible in youth. Relatively high individual biological variability may limit its clinical utility.
Collapse
Affiliation(s)
- Justin R Ryder
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Michael J O'Connell
- Division of Biostatistics, School of Public Health, Minneapolis, MN 55455, USA
| | - Kyle D Rudser
- Division of Biostatistics, School of Public Health, Minneapolis, MN 55455, USA
| | - Claudia K Fox
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Anna N Solovey
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Robert P Hebbel
- Vascular Biology Center, Division of Hematology, Oncology & Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Aaron S Kelly
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
32
|
Obeid J, Nguyen T, Cellucci T, Larché MJ, Timmons BW. Effects of acute exercise on circulating endothelial and progenitor cells in children and adolescents with juvenile idiopathic arthritis and healthy controls: a pilot study. Pediatr Rheumatol Online J 2015; 13:41. [PMID: 26458943 PMCID: PMC4604015 DOI: 10.1186/s12969-015-0038-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/30/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Youth with juvenile idiopathic arthritis (JIA) may be at risk of poor cardiovascular health. Circulating endothelial progenitor cells (EPCs) and circulating endothelial cells (CECs) are markers of cardiovascular repair and damage, respectively, and respond to exercise. The objectives of this study were to compare resting levels of EPCs and CECs in JIA and controls, and to assess the effects of distinct types of exercise on EPCs and CECs in JIA and controls. METHODS Seven youth with JIA and six controls completed 3 visits. First, aerobic fitness was assessed. Participants then performed either moderate intensity, continuous exercise (MICE) or high intensity, intermittent exercise (HIIE) on separate days. Blood samples were collected at the beginning (REST), mid-point (MID) and end of exercise (POST) for determination of EPCs (CD31(+)CD34(bright)CD45(dim)CD133(+)) and CECs (CD31(bright)CD34(+)CD45(-)CD133(-)) by flow cytometry. Between group differences in EPCs and CECs were examined using two-way ANOVA, followed by Tukey's HSD post hoc, where appropriate. Statistical significance set at p ≤ 0.05. RESULTS Both EPCs and CECs were similar between groups at REST (p = 0.18-0.94). During MICE, EPCs remained unchanged in JIA (p = 0.95) but increased significantly at POST in controls (REST: 0.91 ± 0.55 × 10(6) cells/L vs. POST: 1.53 ± 0.36 × 10(6) cells/L, p = 0.04). Compared with controls, lower levels of EPCs were observed in JIA at MID (0.48 ± 0.50 × 10(6) cells/L vs. 1.10 ± 0.39 × 10(6) cells/L, p = 0.01) and POST (0.38 ± 0.34 × 10(6) cells/L vs. 1.53 ± 0.36 × 10(6) cells/L, p < 0.001) during MICE. No changes were detected in CECs with MICE in JIA and controls (p = 0.69). Neither EPCs nor CECs were modified with HIIE (p = 0.28-0.69). CONCLUSION Youth with JIA demonstrated a blunted EPC response to MICE when compared with controls. Future work should examine factors that may increase or normalize EPC mobilization in JIA.
Collapse
Affiliation(s)
- Joyce Obeid
- Child Health and Exercise Medicine Program, McMaster University, HSC 3N27G, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada. .,Department of Pediatrics, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1, ON, Canada.
| | - Thanh Nguyen
- Child Health and Exercise Medicine Program, McMaster University, HSC 3N27G, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada. .,Department of Pediatrics, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1, ON, Canada.
| | - Tania Cellucci
- Department of Pediatrics, Division of Rheumatology, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1, ON, Canada. .,Department of Pediatrics, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1, ON, Canada.
| | - Maggie J. Larché
- Departments of Medicine and Pediatrics, Division of Rheumatology, McMaster University, Hamilton, ON Canada ,Department of Medicine, Charlton Medical Centre, 25 Charlton Ave E, Suite 702, Hamilton, L8N1Y2 ON Canada
| | - Brian W. Timmons
- Child Health and Exercise Medicine Program, McMaster University, HSC 3N27G, 1280 Main Street West, Hamilton, L8S 4K1 ON Canada ,Department of Pediatrics, McMaster University, 1280 Main St W, Health Sciences Centre, Hamilton, L8S 4K1 ON Canada
| |
Collapse
|
33
|
Tuzcu ZB, Asicioglu E, Sunbul M, Ozben B, Arikan H, Koc M. Circulating endothelial cell number and markers of endothelial dysfunction in previously preeclamptic women. Am J Obstet Gynecol 2015; 213:533.e1-7. [PMID: 26116870 DOI: 10.1016/j.ajog.2015.06.043] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/03/2015] [Accepted: 06/16/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Patients with preeclampsia (PE) have endothelial dysfunction and an increased future risk of cardiovascular (CV) mortality. The number of circulating endothelial cells (CECs) is markedly increased in conditions associated with a high degree of endothelial cell activation/injury including PE. We hypothesized that the number of CECs continues to be increased in women with a history of PE, reflecting ongoing endothelial cell activation/injury. STUDY DESIGN CECs, flow-mediated vasodilation, levels of adhesion molecules and soluble vascular endothelial growth factor receptor-1 (sVEGFR1), and urine albumin/creatinine ratio were determined in 21 healthy women with ongoing normal pregnancy, 24 healthy currently nonpregnant women with a history of normal pregnancy, a total of 17 women with currently active mild (n = 11) or severe (n = 6) PE without hemolysis, elevated liver enzymes, and low platelet count (HELLP) syndrome, and 16 currently nonpregnant women with a history of mild (n = 10) or severe (n = 6) PE. RESULTS Blood samples from women with active preeclampsia had higher CECs (9.9 ± 7.9 cells/mL) than healthy pregnant women (3.0 ± 4.1 cells/mL; P < .001), healthy nonpregnant women with a history of normal pregnancy (3.4 ± 4.0 cells/mL; P < .001), or women with a history of preeclampsia (2.4 ± 2.0 cells/mL; P < .001). The number of CECs were similar between women with a history of preeclampsia and healthy nonpregnant women with a history of normal pregnancy. Patients with active preeclampsia had significantly higher soluble vascular cell adhesion molecule-1, soluble E-selectin, sVEGFR1, and urinary albumin/creatinine ratio than healthy pregnant women. However, soluble vascular cell adhesion molecule-1, soluble E-selectin, urinary albumin/creatinine ratio were similar in women with a history of preeclampsia and healthy nonpregnant women with a history of normal pregnancy. However, women with a history of preeclampsia had higher sVEGFR1 levels than women with a history of normal pregnancy (P < .05). CONCLUSION Markers of endothelial activation, dysfunction, and damage were increased in patients with PE. After the delivery, this activation status is similar to the age-matched nonpregnant women with a history of normal pregnancy. However, sVEGFR-1 levels remain higher in women with a history of preeclampsia compared with women without a history of preeclampsia.
Collapse
|
34
|
Circulating endothelial cells in coronary artery disease and acute coronary syndrome. Trends Cardiovasc Med 2015; 25:578-87. [DOI: 10.1016/j.tcm.2015.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/28/2015] [Accepted: 01/28/2015] [Indexed: 01/27/2023]
|
35
|
Korjian S, Daaboul Y, Halaby R, Goldhaber SZ, Cohen AT, Singh K, Susheela AT, Harrington RA, Hull RD, Hernandez AF, Gibson CM. Extended-Duration Thromboprophylaxis Among Acute Medically Ill Patients. J Cardiovasc Pharmacol Ther 2015; 21:227-32. [DOI: 10.1177/1074248415601894] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/02/2015] [Indexed: 01/08/2023]
Abstract
Acute medical illnesses are associated with a prolonged elevation in inflammatory markers that predisposes patients to thrombosis beyond the duration of their hospital stay. In parallel, both observational and randomized data have demonstrated a rate of postdischarge venous thromboembolic events that often exceeds that observed in the hospital setting. Despite this significant residual risk of venous thromboembolic events following discharge among acute medically ill patients, no therapeutic strategies have been recommended to address this unmet need. Available randomized trials have demonstrated the efficacy of extending the duration of thromboprophylaxis with available anticoagulants; however, the efficacy is offset, at least in part, by an increase in bleeding events. Identification of the optimal therapeutic strategies, treatment duration, and risk assessment tools that reconcile both efficacy and safety of extended-duration thromboprophylaxis among acute medically ill patients is an area of ongoing investigation.
Collapse
Affiliation(s)
- Serge Korjian
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yazan Daaboul
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Rim Halaby
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Samuel Z. Goldhaber
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander T. Cohen
- Department of Haematology, Guys and St Thomas’ NHS Trust, London, United Kingdom
| | - Kiran Singh
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ammu T. Susheela
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Robert A. Harrington
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Russell D. Hull
- Department of Medicine, Foothills Hospital, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Adrian F. Hernandez
- Duke Clinical Research Institute and Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - C. Michael Gibson
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Cuadrado-Godia E, Regueiro A, Núñez J, Díaz-Ricard M, Novella S, Oliveras A, Valverde MA, Marrugat J, Ois A, Giralt-Steinhauer E, Sanchís J, Escolar G, Hermenegildo C, Heras M, Roquer J. Endothelial Progenitor Cells Predict Cardiovascular Events after Atherothrombotic Stroke and Acute Myocardial Infarction. A PROCELL Substudy. PLoS One 2015; 10:e0132415. [PMID: 26332322 PMCID: PMC4557832 DOI: 10.1371/journal.pone.0132415] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/13/2015] [Indexed: 12/21/2022] Open
Abstract
Introduction The aim of this study was to determine prognostic factors for the risk of new vascular events during the first 6 months after acute myocardial infarction (AMI) or atherothrombotic stroke (AS). We were interested in the prognostic role of endothelial progenitor cells (EPC) and circulating endothelial cells (CEC) Methods Between February 2009 and July 2012, 100 AMI and 50 AS patients were consecutively studied in three Spanish centres. Patients with previously documented coronary artery disease or ischemic strokes were excluded. Samples were collected within 24h of onset of symptoms. EPC and CEC were studied using flow cytometry and categorized by quartiles. Patients were followed for up to 6 months. NVE was defined as new acute coronary syndrome, transient ischemic attack (TIA), stroke, or any hospitalization or death from cardiovascular causes. The variables included in the analysis included: vascular risk factors, carotid intima-media thickness (IMT), atherosclerotic burden and basal EPC and CEC count. Multivariate survival analysis was performed using Cox regression analysis. Results During follow-up, 19 patients (12.66%) had a new vascular event (5 strokes; 3 TIAs; 4 AMI; 6 hospitalizations; 1 death). Vascular events were associated with age (P = 0.039), carotid IMT≥0.9 (P = 0.044), and EPC count (P = 0.041) in the univariate analysis. Multivariate Cox regression analysis showed an independent association with EPC in the lowest quartile (HR: 10.33, 95%CI (1.22–87.34), P = 0.032] and IMT≥0.9 [HR: 4.12, 95%CI (1.21–13.95), P = 0.023]. Conclusions Basal EPC and IMT≥0.9 can predict future vascular events in patients with AMI and AS, but CEC count does not affect cardiovascular risk.
Collapse
Affiliation(s)
- Elisa Cuadrado-Godia
- Department of Neurology, Neurovascular Research Group, IMIM-Hospital del Mar (Institut Hospital del Mar d'Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
- * E-mail:
| | - Ander Regueiro
- Cardiology Department, Thorax Institute, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Julio Núñez
- Cardiology Department, Hospital Clínico Universitario, Valencia.School of Medicine.Universitat de València, Valencia, Spain
| | - Maribel Díaz-Ricard
- Hemotherapy-Hemostasis Department, Biomedical Diagnostics Center, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Susana Novella
- Valencia INCLIVA Biomedical Research Institute, Hospital Clínico, Valencia; Department of Physiology, Universitat de València, València, Spain
| | - Anna Oliveras
- Nephrology Department, Hospital del Mar. Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel A. Valverde
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jaume Marrugat
- Epidemiology and Cardiovascular Genetics Group. IMIM, Barcelona, Spain
| | - Angel Ois
- Department of Neurology, Neurovascular Research Group, IMIM-Hospital del Mar (Institut Hospital del Mar d'Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Eva Giralt-Steinhauer
- Department of Neurology, Neurovascular Research Group, IMIM-Hospital del Mar (Institut Hospital del Mar d'Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Juan Sanchís
- Cardiology Department, Hospital Clínico Universitario, Valencia.School of Medicine.Universitat de València, Valencia, Spain
| | - Ginès Escolar
- Hemotherapy-Hemostasis Department, Biomedical Diagnostics Center, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Carlos Hermenegildo
- Valencia INCLIVA Biomedical Research Institute, Hospital Clínico, Valencia; Department of Physiology, Universitat de València, València, Spain
| | - Magda Heras
- Cardiology Department, Thorax Institute, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Jaume Roquer
- Department of Neurology, Neurovascular Research Group, IMIM-Hospital del Mar (Institut Hospital del Mar d'Investigacions Mèdiques), Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
37
|
Free hemoglobin increases von Willebrand factor-mediated platelet adhesion in vitro: implications for circulatory devices. Blood 2015; 126:2338-41. [PMID: 26307534 DOI: 10.1182/blood-2015-05-648030] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/16/2015] [Indexed: 12/18/2022] Open
Abstract
Intravascular hemolysis occurs in patients on extracorporeal membrane oxygenation. High levels of free acellular adult hemoglobin (free HbA) are associated with clotting in this mechanical device that can result in thrombotic complications. Adsorption of fibrinogen onto the surface of biomaterial correlates with platelet adhesion, which is mediated by von Willebrand factor (VWF). Because free Hb interacts with VWF, we studied the effect of hemoglobin (Hb) on platelet adhesion to fibrin(ogen) under conditions of different hydrodynamic forces. This effect was investigated using purified human HbA and fibrinogen, extracellular matrix, collagen, or purified plasma VWF as surface-coated substrates to examine flow-dependent platelet adhesion. Antibodies and VWF-deficient plasma were also used. Free Hb (≥50 mg/dL) effectively augmented platelet adhesion, and microthrombi formation on fibrin(ogen), extracellular matrix, and collagen at high shear stress. The effect of free Hb was effectively blocked by anti-glycoprotein Ibα (GPIbα) antibodies or depletion of VWF. Unexpectedly, free Hb also promoted firm platelet adhesion and stable microthrombi on VWF. Lastly, we determined that Hb interacts directly with the A1 domain. This study is the first to demonstrate that extracellular Hb directly affects the GPIbα-VWF interaction in thrombosis, and describes another mechanism by which hemolysis is connected to thrombotic events.
Collapse
|
38
|
Lanuti P, Rotta G, Almici C, Avvisati G, Budillon A, Doretto P, Malara N, Marini M, Neva A, Simeone P, Di Gennaro E, Leone A, Falda A, Tozzoli R, Gregorj C, Di Cerbo M, Trunzo V, Mollace V, Marchisio M, Miscia S. Endothelial progenitor cells, defined by the simultaneous surface expression of VEGFR2 and CD133, are not detectable in healthy peripheral and cord blood. Cytometry A 2015; 89:259-70. [DOI: 10.1002/cyto.a.22730] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/18/2015] [Accepted: 07/16/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Paola Lanuti
- Department of Medicine and Aging Science; School of Medicine and Health Science, University “G. d'Annunzio” of Chieti-Pescara; Chieti 66013 Italy
- Center for Ageing Sciences (Ce.S.I.), “Università G. d'Annunzio” Foundation; Chieti 66013 Italy
| | | | - Camillo Almici
- Department of Transfusion Medicine; Laboratory for Stem Cells Manipulation and Cryopreservation, AO Spedali Civili di Brescia; Brescia 25123 Italy
| | - Giuseppe Avvisati
- Department of Hematology; Stem Cell Transplantation, Transfusion Medicine and Cellular Therapy, Campus Bio-Medico University Hospital; Rome 00128 Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS; Naples 80131 Italy
| | - Paolo Doretto
- Department of Laboratory Medicine; Clinical Pathology Laboratory, “S. Maria Degli Angeli” Hospital; Pordenone 33170 Italy
| | - Natalia Malara
- Department of Health Science; Interregional Research Center for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro; Catanzaro 88100 Italy
- Department of Experimental and Clinical Medicine; BioNEM Lab, University “Magna Graecia” of Catanzaro, Catanzaro 88100; Italy
| | - Mirella Marini
- Department of Transfusion Medicine; Laboratory for Stem Cells Manipulation and Cryopreservation, AO Spedali Civili di Brescia; Brescia 25123 Italy
| | - Arabella Neva
- Department of Transfusion Medicine; Laboratory for Stem Cells Manipulation and Cryopreservation, AO Spedali Civili di Brescia; Brescia 25123 Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Science; School of Medicine and Health Science, University “G. d'Annunzio” of Chieti-Pescara; Chieti 66013 Italy
- Center for Ageing Sciences (Ce.S.I.), “Università G. d'Annunzio” Foundation; Chieti 66013 Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS; Naples 80131 Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale-IRCCS; Naples 80131 Italy
| | - Alessandra Falda
- Department of Laboratory Medicine; Clinical Pathology Laboratory, “S. Maria Degli Angeli” Hospital; Pordenone 33170 Italy
| | - Renato Tozzoli
- Department of Laboratory Medicine; Clinical Pathology Laboratory, “S. Maria Degli Angeli” Hospital; Pordenone 33170 Italy
| | - Chiara Gregorj
- Department of Hematology; Stem Cell Transplantation, Transfusion Medicine and Cellular Therapy, Campus Bio-Medico University Hospital; Rome 00128 Italy
| | - Melania Di Cerbo
- Department of Hematology; Stem Cell Transplantation, Transfusion Medicine and Cellular Therapy, Campus Bio-Medico University Hospital; Rome 00128 Italy
| | - Valentina Trunzo
- Department of Health Science; Interregional Research Center for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro; Catanzaro 88100 Italy
| | - Vincenzo Mollace
- Department of Health Science; Interregional Research Center for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro; Catanzaro 88100 Italy
| | - Marco Marchisio
- Department of Medicine and Aging Science; School of Medicine and Health Science, University “G. d'Annunzio” of Chieti-Pescara; Chieti 66013 Italy
- Center for Ageing Sciences (Ce.S.I.), “Università G. d'Annunzio” Foundation; Chieti 66013 Italy
| | - Sebastiano Miscia
- Department of Medicine and Aging Science; School of Medicine and Health Science, University “G. d'Annunzio” of Chieti-Pescara; Chieti 66013 Italy
- Center for Ageing Sciences (Ce.S.I.), “Università G. d'Annunzio” Foundation; Chieti 66013 Italy
| |
Collapse
|
39
|
Obeid J, Nguyen T, Walker RG, Gillis LJ, Timmons BW. Circulating endothelial cells in children: role of fitness, activity, and adiposity. Med Sci Sports Exerc 2015; 46:1974-80. [PMID: 24561817 DOI: 10.1249/mss.0000000000000313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Circulating endothelial cells (CEC) are thought to be useful biomarkers of endothelial dysfunction and overall cardiovascular health. The extent to which CEC are influenced by fitness, physical activity, and adiposity in youth remains unknown, as they have seldom been examined in the pediatric population. This study assessed resting levels of CEC in boys and girls of different chronological and biological age and explored the relationship between these cells and aerobic fitness, physical activity, and adiposity. METHODS Seventy-five children (39 males; median [interquartile range], age = 14.4 [5.8]) completed two study visits. During the first visit, basic anthropometric data were collected and biological age was calculated. Peak mechanical power (Wpeak) was determined using the McMaster All-Out Continuous cycling test. Participants then wore an accelerometer over a 7-d period to assess habitual levels of moderate-to-vigorous physical activity. During visit 2, percent body fat (%BF) was assessed by dual-energy x-ray absorptiometry. A fasted blood sample was also collected from which concentrations of CEC, identified as CD31CD34CD45CD133, were quantified by flow cytometry. RESULTS No differences were seen in CEC by sex, chronological age, or biological age. The median (interquartile range) CEC concentration was 32.3 × 10 (63.0 × 10), representing 1.3% (2.7%) of collected peripheral blood mononuclear cells. CEC concentration was associated with Wpeak normalized to lean body mass (r = 0.36, P < 0.01) and time spent in moderate-to-vigorous physical activity (r = -0.27, P = 0.02). No relationship was observed between CEC and %BF. CONCLUSION Healthy children demonstrate relatively low concentrations of CECs. Because CEC represent a population of mature endothelial cells shed from the intima after irreversible damage, they may be more reflective of recent physical activity levels rather than fitness or level of adiposity.
Collapse
Affiliation(s)
- Joyce Obeid
- Child Health & Exercise Medicine Program, Department of Pediatrics, McMaster University, Hamilton, ON, CANADA
| | | | | | | | | |
Collapse
|
40
|
Kachamakova-Trojanowska N, Bukowska-Strakova K, Zukowska M, Dulak J, Jozkowicz A. The real face of endothelial progenitor cells - Circulating angiogenic cells as endothelial prognostic marker? Pharmacol Rep 2015; 67:793-802. [PMID: 26321283 DOI: 10.1016/j.pharep.2015.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 02/08/2023]
Abstract
Endothelial progenitor cells (EPCs) have been extensively studied for almost 19 years now and were considered as a potential marker for endothelial regeneration ability. On the other hand, circulating endothelial cells (CEC) were studied as biomarker for endothelial injury. Yet, in the literature, there is also huge incoherency in regards to terminology and protocols used. This results in misleading conclusions on the role of so called "EPCs", especially in the clinical field. The discrepancies are mainly due to strong phenotypic overlap between EPCs and circulating angiogenic cells (CAC), therefore changes in "EPC" terminology have been suggested. Other factors leading to inconsistent results are varied definitions of the studied populations and the lack of universal data reporting, which could strongly affect data interpretation. The current review is focused on controversies concerning the use of "EPCs"/CAC and CEC as putative endothelial diagnostic markers.
Collapse
Affiliation(s)
- Neli Kachamakova-Trojanowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Monika Zukowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
41
|
|
42
|
Pourcyrous M, Basuroy S, Tcheranova D, Arheart KL, Elabiad MT, Leffler CW, Parfenova H. Brain-derived circulating endothelial cells in peripheral blood of newborn infants with seizures: a potential biomarker for cerebrovascular injury. Physiol Rep 2015; 3:3/3/e12345. [PMID: 25804265 PMCID: PMC4393173 DOI: 10.14814/phy2.12345] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neonatal seizures have been associated with cerebrovascular endothelial injury and neurological disabilities. In a piglet model, the long-term loss of endothelial regulation of cerebral blood flow coincides with the surge of brain-derived circulating endothelial cells (BCECs) in blood. We hypothesized that BCECs could serve as a noninvasive biomarker of cerebrovascular injury in neonates with seizures. In a prospective pilot feasibility study, we enrolled newborn infants with confirmed diagnoses of perinatal asphyxia and intraventricular hemorrhage (IVH); both are commonly associated with seizures. Infants without clinical evidence of cerebrovascular injuries were representative of the control group. BCECs were detected in the CD45-negative fraction of peripheral blood mononuclear cells by coexpression of CD31 (common endothelial antigen) and GLUT1 (blood-brain barrier antigen) via automated flow cytometry method. In Infants with asphyxia (n = 12) and those with IVH grade III/IV (n = 5), the BCEC levels were 9.9 ± 0.9% and 19.0 ± 2.0%, respectively. These levels were significantly higher than the control group (n = 27), 0.9 ± 0.2%, P < 0.001. BCECs in infants with cerebrovascular insults with documented clinical seizures (n = 10; 16.8 ± 1.3%) were significantly higher than infants with cerebrovascular insults with subclinical or no seizures (n = 7; 9.5 ± 1.2%); P < 0.001. BCEC levels decreased with seizure control. BCECs levels were elevated in infants with seizures caused by severe IVH and perinatal asphyxia. We suggest that monitoring BCEC levels in peripheral blood can potentially offer a biological marker that reflects cerebrovascular insult and recovery. Further studies with a larger number of patients are required to support these findings.
Collapse
Affiliation(s)
- Massroor Pourcyrous
- Department of Pediatrics, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Neuroscience Institute, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Obstetrics and Gynecology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Shyamali Basuroy
- Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Dilyara Tcheranova
- Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Kristopher L Arheart
- Division of Biostatistics and Pediatrics, Department of Public Health Sciences, Miller School of Medicine University of Miami, Coral Gables, Florida
| | - Mohamad T Elabiad
- Department of Pediatrics, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Charles W Leffler
- Department of Pediatrics, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Neuroscience Institute, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| | - Helena Parfenova
- Department of Physiology, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee Department of Neuroscience Institute, The University of Tennessee Health Science Center (UTHSC), Memphis, Tennessee
| |
Collapse
|
43
|
Investigation of Proposed Mechanisms of Chemotherapy-Induced Venous Thromboembolism. Clin Appl Thromb Hemost 2015; 21:420-7. [DOI: 10.1177/1076029615575071] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Venous thromboembolism (VTE) during chemotherapy is common, with 7% mortality in metastatic breast cancer (MBC). In a prospective cohort study of patients with breast cancer, we investigated whether vascular endothelial cell activation (VECA), and whether apoptosis, is the cause of chemotherapy-induced VTE. Methods: Serum markers of VECA, E-selectin (E-sel), vascular cell adhesion molecule 1 (VCAM-1) and d-dimer (fibrin degradation and hypercoagulability marker) were measured prechemotherapy and at 1, 4, and 8 days following chemotherapy. Clinical deep vein thrombosis (DVT) or pulmonary embolism and occult DVT detected by duplex ultrasound imaging were recorded as VTE-positive (VTE+). In patients with MBC, hypercoagulable response to chemotherapy was compared between patients with and without cancer progression. Development of VTE and cancer progression was assessed 3 months following starting chemotherapy. Results: Of the 134 patients, 10 (7.5%) developed VTE (6 [17%] of 36 MBC receiving palliation, 0 of 11 receiving neoadjuvant to downsize tumor, and 4 [5%] of 87 early breast cancer receiving adjuvant chemotherapy, P = .06). Levels of E-sel and VCAM-1 decreased in response to chemotherapy ( P < .001) in both VTE+ and patients not developing VTE (VTE−). However, decrease in VECA markers was similar in VTE+ and VTE− patients, implying this is not the cause of VTE. In patients with MBC following chemotherapy, d-dimer (geometric mean) increased by 36% in the 21 patients with MBC responding to chemotherapy but steadily decreased by 11% in the 15 who progressed (day 4, P < .01), implying patients with tumor response (apoptosis) had an early hypercoagulable response. Conclusions: During chemotherapy for breast cancer, VECA is induced; however, this is not the primary mechanism for VTE. Chemotherapy-induced apoptosis may enhance hypercoagulability and initiate VTE.
Collapse
|
44
|
Moussa MD, Santonocito C, Fagnoul D, Donadello K, Pradier O, Gaussem P, De Backer D, Vincent JL. Evaluation of endothelial damage in sepsis-related ARDS using circulating endothelial cells. Intensive Care Med 2015; 41:231-8. [PMID: 25510299 DOI: 10.1007/s00134-014-3589-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/27/2014] [Indexed: 02/07/2023]
Abstract
PURPOSE Endothelial cell activation and dysfunction are involved in the pathophysiology of ARDS. Circulating endothelial cells (CECs) may be a useful marker of endothelial dysfunction and damage but have been poorly studied in ARDS. We hypothesized that the CEC count may be elevated in patients with sepsis-related ARDS compared to those with sepsis without ARDS. METHODS ARDS was defined according to the Berlin consensus definition. The study population included 17 patients with moderate or severe ARDS, 9 with mild ARDS, 13 with sepsis and no ARDS, 13 non-septic patients, and 12 healthy volunteers. Demographic, hemodynamic, and prognostic variables, including PaO(2)/FiO(2) ratio, 28-day survival, blood lactate, APACHE II, and SOFA score, were recorded. CECs were counted in arterial blood samples using the reference CD146 antibody-based immunomagnetic isolation and UEA1-FITC staining method. Measurements were performed 12-24 h after diagnosis of ARDS and repeated daily for 3 days. RESULTS The median day-1 CEC count was significantly higher in patients with moderate or severe ARDS than in mild ARDS or septic-control patients [27.2 (18.3-49.4) vs. 17.4 (11-24.5) cells/ml (p < 0.034), and 18.4 (9.1-31) cells/ml (p < 0.035), respectively]. All septic patients (with or without ARDS) had higher day-1 CEC counts than the non-septic patients [19.6 (14.2-30.6) vs. 10.8 (5.7-13.2) cells/ml, p = 0.002]. CONCLUSION The day-1 CEC count was significantly higher in ARDS patients than in other critically ill patients, and in moderate or severe ARDS patients compared to those with milder disease, making it a potentially useful marker of ARDS severity.
Collapse
Affiliation(s)
- Mouhamed Djahoum Moussa
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles (ULB), 808 Route de Lennik, 1070, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Ding J, Chen Q, Zhuang X, Feng Z, Xu L, Chen F. Low paraoxonase 1 arylesterase activity and high von Willebrand factor levels are associated with severe coronary atherosclerosis in patients with non-diabetic stable coronary artery disease. Med Sci Monit 2014; 20:2421-9. [PMID: 25420483 PMCID: PMC4254670 DOI: 10.12659/msm.890911] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background Paraoxonase 1 (PON1) activity and von Willebrand factor (VWF) release are associated with lesion initiation in atherosclerosis. Diabetes can complicate coronary artery disease (CAD) due to the production of advanced glycation end products. This study evaluated PON1 activity and VWF levels in non-post-acute coronary syndrome, stable CAD (SCAD) patients without diabetes. Material/Methods Non-diabetic SCAD patients and patients experiencing acute stress periods were selected (n=130). Forty-seven cases with normal coronary angiography and 50 healthy individuals served as controls. The non-diabetic SCAD group was then stratified into single-vessel lesions, multiple-vessel lesions, and mild or severe luminal stenosis according to the number and the degree of luminal stenoses. Serum PON1 paraoxonase and arylesterase activities, and plasma VWF levels were measured, as well as serum total cholesterol, total triglycerides, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and apolipoprotein A1. PON1 arylesterase activity was detected with an ordinary chemistry system using a novel phenylacetate derivative. Results Both PON1 paraoxonase and arylesterase were lower in the non-diabetic SCAD group, but VWF levels were higher (versus controls, all P<0.001). PON1 paraoxonase activity (OR=0.991), PON1 arylesterase activity (OR=0.981), and VWF (OR 2.854) influenced SCAD in multiple logistic regression. Decreased PON1 arylesterase activity and increased VWF levels were associated with severe atherosclerosis in non-diabetic SCAD patients. We also observed a slight negative correlation between VWF and PON1 paraoxonase/arylesterase. Conclusions PON1 and VWF are detectable markers that may predict the severity of stenoses, ideally facilitating a non-diabetic SCAD diagnosis before the sudden onset of life-threatening symptoms.
Collapse
Affiliation(s)
- Jieying Ding
- Department of Clinical Laboratories, Ninth People's Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai, China (mainland)
| | - Qizhi Chen
- Department of Cardiology, Ninth People's Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai, China (mainland)
| | - Xing Zhuang
- Department of Clinical Laboratories, Ninth People's Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai, China (mainland)
| | - Zhilei Feng
- Department of Clinical Laboratories, Ninth People's Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai, China (mainland)
| | - Lili Xu
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China (mainland)
| | - Fuxiang Chen
- Department of Clinical Laboratories, Ninth People's Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
46
|
Radecke CE, Warrick AE, Singh GD, Rogers JH, Simon SI, Armstrong EJ. Coronary artery endothelial cells and microparticles increase expression of VCAM-1 in myocardial infarction. Thromb Haemost 2014; 113:605-16. [PMID: 25413339 DOI: 10.1160/th14-02-0151] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 10/10/2014] [Indexed: 11/05/2022]
Abstract
Coronary artery disease (CAD) is characterised by progressive atherosclerotic plaque leading to flow-limiting stenosis, while myocardial infarction (MI) occurs due to plaque rupture or erosion with abrupt coronary artery occlusion. Multiple inflammatory pathways influence plaque stability, but direct assessment of endothelial inflammation at the site of coronary artery stenosis has largely been limited to pathology samples or animal models of atherosclerosis. We describe a technique for isolating and characterising endothelial cells (ECs) and EC microparticles (EMPs) derived directly from the site of coronary artery plaque during balloon angioplasty and percutaneous coronary intervention. Coronary artery endothelial cells (CAECs) were identified using imaging flow cytometry (IFC), and individual CAEC and EMP expression of the pro-atherogenic adhesion molecule vascular cell adhesion molecule-1 (VCAM-1) was assessed immediately following angioplasty. Patients with MI registered 73 % higher VCAM-1 expression on their CAECs and 79 % higher expression on EMPs compared to patients with stable CAD. In contrast, VCAM-1 expression was absent on ECs in the peripheral circulation from these same subjects. VCAM-1 density was significantly higher on CAECs and EMPs among patients with MI and positively correlated with markers of myocardial infarct size. We conclude that increased VCAM-1 expression on EC and formation of EMP at the site of coronary plaque is positively correlated with the extent of vascular inflammation in patients with myocardial infarction.
Collapse
Affiliation(s)
| | | | | | | | - Scott I Simon
- Scott I. Simon, Department of Biomedical Engineering, University of California at Davis, 451 East Health Sciences Drive, Davis, CA 95616, USA, Tel.: +1 530 752 0299, E-mail
| | | |
Collapse
|
47
|
Circulating endothelial cells and chronic kidney disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:364738. [PMID: 24949439 PMCID: PMC4052150 DOI: 10.1155/2014/364738] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/30/2014] [Accepted: 05/03/2014] [Indexed: 12/31/2022]
Abstract
Endothelial dysfunction may play a crucial role in initiation of the pathogenesis of vascular disease and atherosclerosis. The identification and quantification of circulating endothelial cells (CEC) have been developed as a novel marker of endothelial function. We describe, in great detail, mechanisms of endothelial dysfunction and CEC detachment. We also review the relationship between numbers of CEC and disease severity and response to treatment. In addition, we describe the possible clinical use of CEC in chronic kidney disease (CKD) and kidney transplantation. In summary, CEC have been developed as a novel approach to assess the endothelial damage. Measurement of the CEC level would provide an important diagnostic and prognostic value on the endothelium status and the long-term outcome of vascular dysfunction.
Collapse
|
48
|
Association between circulating endothelial cells and carotid atherosclerosis in patients receiving maintenance hemodialysis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:753759. [PMID: 24772436 PMCID: PMC3977501 DOI: 10.1155/2014/753759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 02/08/2014] [Accepted: 02/16/2014] [Indexed: 02/01/2023]
Abstract
Accelerated atherosclerosis is the major cause of mortality in maintenance hemodialysis (MHD) patients, and endothelial injury associated with MHD might contribute strongly to pathogenesis. The current study was designed to explore possible associations between circulating endothelial cells (CECs) and intima-media thickness of common carotid artery (CCA-IMT) as an indicator of carotid atherosclerosis. Sixty-two MHD patients and 26 age- and sex-matched healthy volunteers were recruited. The number of CECs was determined in peripheral blood using multiparametric flow cytometry. CCA-IMT and presence of plaques in the common carotid arteries were assessed with ultrasound. Laboratory tests results and the demographics were recorded. The finding indicated that numbers of CECs were higher in patients before hemodialysis (predialysis) compared with numbers in controls (P = 0.045). CCA-IMT was also significantly higher in patients than in controls (P < 0.01). A positive relationship was observed between predialysis CECs numbers and CCA-IMT (P < 0.01) in MHD patients. In multiple linear regression analysis, the relationship between the predialysis CECs level and CCA-IMT remained the same even if adjusting for confounding effects. Accordingly, the investigation indicates that the CECs level is positively associated with CCA-IMT in our hemodialysis patients. CECs might be an important marker to the severity of carotid atherosclerosis in MHD patients.
Collapse
|
49
|
Sonneveld MAH, de Maat MPM, Leebeek FWG. Von Willebrand factor and ADAMTS13 in arterial thrombosis: a systematic review and meta-analysis. Blood Rev 2014; 28:167-78. [PMID: 24825749 DOI: 10.1016/j.blre.2014.04.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/14/2014] [Indexed: 01/08/2023]
Abstract
Von Willebrand Factor (VWF) plays an important role in hemostasis by mediating platelet adhesion and aggregation. Ultralarge VWF multimers are cleaved by ADAMTS13 in smaller, less procoagulant forms. An association between high VWF levels and cardiovascular disease has frequently been reported, and more recently also an association has been observed between low ADAMTS13 levels and arterial thrombosis. We reviewed the current literature and performed meta-analyses on the relationship between both VWF and ADAMTS13 with arterial thrombosis. Most studies showed an association between high VWF levels and arterial thrombosis. It remains unclear whether ADAMTS13 is a causal independent risk factor because the association between low ADAMTS13 and arterial thrombosis is so far only shown in case-control studies. Prospective studies are awaited. A causal role for ADAMTS13 is supported by mice studies of cerebral infarction where the infusion of recombinant human ADAMTS13 reduced the infarct size.
Collapse
Affiliation(s)
| | - Moniek P M de Maat
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
50
|
Freestone B, Krishnamoorthy S, Lip GYH. Assessment of endothelial dysfunction. Expert Rev Cardiovasc Ther 2014; 8:557-71. [DOI: 10.1586/erc.09.184] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|