1
|
Gold DA, Maisuradze N, Mullinax BJ, Sawan MA, Barker M, Hebbo E, Shekiladze N, Kindya B, A Jaber W, Sandesara PB, Quyyumi AA, Nicholson WJ. Future Directions of Chronic Total Occlusion Management: Identifying the Right Patient for Intervention With a Focus on Biomarkers. Catheter Cardiovasc Interv 2025; 105:1462-1471. [PMID: 40047319 DOI: 10.1002/ccd.31466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 05/08/2025]
Abstract
The management of a chronic total occlusion (CTO) of a coronary artery has been a conundrum in interventional cardiology, as revascularization has not been proven to provide a mortality benefit. However, there are subgroups of patients with a CTO that have high levels of ischemia on myocardial perfusion imaging and high circulating levels of high sensitivity troponin-I (hsTn-I) and N terminal pro-brain natriuretic peptide (NT pro-BNP) that are at a particularly high-risk for adverse cardiovascular events. These high-risk subgroups of patients with a CTO may have not been well represented in prior clinical trials, and may gain a mortality benefit from revascularization of the CTO. Conversely, patients with low levels of ischemia and these biomarkers are at lower risk and may not gain a mortality benefit from revascularization of their CTO. It is important for future randomized controlled trials to investigate the efficacy of CTO PCI in patients with elevated biomarkers and high ischemic burden on myocardial perfusion imaging to determine if patients at high-risk gain a mortality benefit from revascularization.
Collapse
Affiliation(s)
- Daniel A Gold
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Nodari Maisuradze
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Billy Joe Mullinax
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Mariem A Sawan
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Madeleine Barker
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Elsa Hebbo
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Nikoloz Shekiladze
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Bryan Kindya
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Wissam A Jaber
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Pratik B Sandesara
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - William J Nicholson
- Emory Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
2
|
Caruso P, Maiorino MI, Longo M, Maio A, Scappaticcio L, Di Martino N, Carbone C, Barrasso M, Caputo M, Gicchino M, Bellastella G, Giugliano D, Esposito K. Liraglutide improves peripheral perfusion and markers of angiogenesis and inflammation in people with type 2 diabetes and peripheral artery disease: An 18-month follow-up of a randomized clinical trial. Diabetes Obes Metab 2025. [PMID: 40276845 DOI: 10.1111/dom.16419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
AIMS In a six-month randomized clinical trial, improved peripheral perfusion has been shown with liraglutide, associated with favourable vascular effects in people with type 2 diabetes and peripheral artery disease (PAD). We aimed to evaluate the durability of these benefits and to elucidate some mechanisms underlying liraglutide's effect over an 18-month follow-up. METHODS STARDUST was a randomized clinical trial which compared liraglutide up to 1.8 mg/day with tailored therapeutic prescriptions to manage cardiovascular risk factors in 55 participants with type 2 diabetes and PAD. We report data of people who have reached the 18-month follow-up for the primary outcome (transcutaneous oxygen pressure, TcPO2) and also for additional secondary outcomes (markers of inflammation, angiogenesis and kidney function), as well as glycemic and metabolic parameters. TcPO2 was assessed with transcutaneous oximetry. Circulating levels of angiogenic progenitor cells and serum inflammation markers were evaluated by flow cytometry and enzyme-linked immunosorbent assay, respectively. RESULTS Compared with the control group, significant differences favouring the liraglutide group were observed at 18 months for TcPO2 [estimated treated difference (95% CI), 10.9 mmHg (7.6 to 14.1 mmHg), p < 0.001]. At 18 months of follow-up, participants in the liraglutide group, as compared with those in the control group, had a significant reduction in urine albumin to creatinine ratio (estimated difference, -103.9 mg/g Cr, 95%CI, -170.8 to -37.1, p = 0.003), C-reactive protein (-0.5 mg/dL, 95%CI, -0.8 to -0.2, p = 0.002), as well as interleukin-6 (-32.6 pg/mL, 95%CI, -54.6 to -10.5, p = 0.004). Compared with the control group, participants in the liraglutide group showed significantly higher concentrations of circulating progenitor cells and endothelial progenitor cells at both 6 and 18 months, for CD34+, CD133+, KDR+, CD34+/KDR+ and CD34+/CD133+/KDR+. Liraglutide was also associated with a higher increase in vascular endothelial growth factor A at 18 months (70.1 pg/mL, 95%CI, 44.7 to 95.4, p < 0.001). CONCLUSIONS In people with type 2 diabetes and PAD, liraglutide increased peripheral perfusion, with amelioration of markers of angiogenesis and inflammation over an 18-month follow-up.
Collapse
Affiliation(s)
- Paola Caruso
- Division of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Ida Maiorino
- Division of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Miriam Longo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- PhD Program of Translational Medicine, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonietta Maio
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lorenzo Scappaticcio
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Nicole Di Martino
- PhD Program of Translational Medicine, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Carla Carbone
- Division of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mariluce Barrasso
- Division of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
- PhD Program of Translational Medicine, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mariangela Caputo
- Division of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maurizio Gicchino
- Division of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Bellastella
- Division of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Dario Giugliano
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Katherine Esposito
- Division of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- PhD Program of Translational Medicine, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
3
|
Caradonna E, Abate F, Schiano E, Paparella F, Ferrara F, Vanoli E, Difruscolo R, Goffredo VM, Amato B, Setacci C, Setacci F, Novellino E. Trimethylamine-N-Oxide (TMAO) as a Rising-Star Metabolite: Implications for Human Health. Metabolites 2025; 15:220. [PMID: 40278349 PMCID: PMC12029716 DOI: 10.3390/metabo15040220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
The intestinal microbiota, hosting trillions of microorganisms that inhabit the gastrointestinal tract, functions as a symbiotic organism that plays a crucial role in regulating health by producing biologically active molecules that can enter systemic circulation. Among them, trimethylamine-N-oxide (TMAO), an organic compound derived from dietary sources and microbial metabolism, has emerged as a critical biomarker linking diet, the gut microbiota, and the host metabolism to various pathological conditions. This comprehensive review highlights TMAO's biosynthesis, physiological functions, and clinical significance, focusing on its mechanistic contributions to cardiovascular and neurodegenerative diseases. Notably, TMAO-mediated pathways include endothelial dysfunction, inflammation via NLRP3 inflammasome activation, and cholesterol metabolism disruption, which collectively accelerate atherosclerosis and disease progression. Nonetheless, this work underscores the innovative potential of targeting TMAO through dietary, nutraceutical, and microbiota-modulating strategies to mitigate its pathological effects, marking a transformative approach in the prevention and management of TMAO-related disorders.
Collapse
Affiliation(s)
- Eugenio Caradonna
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy (F.F.)
| | - Federico Abate
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy;
| | - Elisabetta Schiano
- Inventia Biotech-Healthcare Food Research Center S.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy;
| | - Francesca Paparella
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy (F.F.)
| | - Fulvio Ferrara
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy (F.F.)
| | - Emilio Vanoli
- School of Nursing, Cardiovascular Diseases, University of Pavia, 27100 Pavia, Italy;
| | | | - Vito Maria Goffredo
- Department of Interdisciplinary Medicine, Università Degli Studi di Bari, 70124 Bari, Italy;
| | - Bruno Amato
- Department of Public Health, Università Degli Studi di Napoli Federico II, 80138 Naples, Italy;
| | - Carlo Setacci
- Vascular and Endovascuar Surgery Unit, “Le Scotte” Hospital of Siena, Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy;
| | - Francesco Setacci
- Vascular Surgery Unit, Università degli Studi di Enna “Kore”, 94100 Enna, Italy;
| | - Ettore Novellino
- Department of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| |
Collapse
|
4
|
Gendron N, Planquette B, Roche A, Chocron R, Helley D, Philippe A, Morange PE, Gaussem P, Sanchez O, Smadja DM. Circulating CD34 + Cells: A New Biomarker of Residual Pulmonary Vascular Obstruction after Pulmonary Embolism. Stem Cell Rev Rep 2025:10.1007/s12015-025-10865-0. [PMID: 40085375 DOI: 10.1007/s12015-025-10865-0] [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] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Pulmonary embolism (PE) is a life-threatening condition with long-term complications, including residual pulmonary vascular obstruction (RPVO). RPVO is associated with an increased risk of venous thromboembolism recurrence, chronic symptoms, and reduced quality of life. We hypothesize that an endothelial activation and vascular injury play a central role in the pathophysiology of RPVO. This prospective monocentric study investigates the potential of circulating biomarkers, including CD34⁺ cells, circulating endothelial cells (CECs), and platelet-derived growth factor BB (PDGF-BB), as indicators of vascular sequelae and predictors of RPVO. We included 56 patients with a first episode of PE. Biomarker levels were measured at PE diagnosis and six months later, coinciding with RPVO assessment using ventilation-perfusion lung scans. This defined groups of patients with (RPVO ≥ 10%) and without (RPVO < 10%) perfusion defects. Associations between biomarker levels, presence of perfusion defects, and clinical parameters were analyzed. At PE diagnosis, CEC and PDGF-BB levels were significantly elevated in patients compared to healthy controls, while CD34⁺ levels showed no difference. At the six-month follow-up, patients with perfusion defects exhibited significantly lower CD34⁺ cell levels compared to those without (median 1440 cells/mL vs. 2960 cells/mL). No significant differences in CEC or PDGF-BB levels were observed at follow-up. In conclusion, low CD34⁺ cell levels at RPVO assessment suggest a decreased regenerative potential contributing to thrombus persistence. CD34⁺ cells may serve as biomarkers for perfusion defects and warrant further study for their potential role in guiding clinical management of PE complications.
Collapse
Affiliation(s)
- Nicolas Gendron
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
| | - Benjamin Planquette
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - Anne Roche
- INSERM UMR-S 999 « Pulmonary Hypertension: Pathophysiology and Novel Therapies », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Faculté de Médecine, HPPIT, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Richard Chocron
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- Emergency Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - Dominique Helley
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Aurélien Philippe
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Pierre-Emmanuel Morange
- F-CRIN INNOVTE, Saint-Étienne, France
- Laboratory of Hematology, La Timone Hospital, Marseille, France
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Pascale Gaussem
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Olivier Sanchez
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - David M Smadja
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France.
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France.
- F-CRIN INNOVTE, Saint-Étienne, France.
| |
Collapse
|
5
|
Liu Y, Li P, Yang Y. Advancements in utilizing CD34 + stem cells for repairing diabetic vascular damage. Biochem Biophys Res Commun 2025; 750:151411. [PMID: 39889623 DOI: 10.1016/j.bbrc.2025.151411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Diabetes-related vascular damage is a frequent complication of diabetes that results in structural and functional impairment of blood vessels. This damage significantly heightens the risk of cardiovascular events. CD34+ stem cells have shown great potential in the treatment of diabetes-related vascular damage due to their differentiation and vascular repair capabilities. This article provides a review of the research hotspots on the role and mechanisms of CD34+ stem cells in the repair of diabetes-related vascular damage, including changes in cell quantity and function during diabetes, as well as the latest research on activating, protecting, or repairing these cells to prevent or treat vascular damage. The article also summarizes the impact of diabetes on the mobilization and function of CD34+ stem cells, emphasizing how diabetes negatively affects their ability to promote angiogenesis. These deficits can result in various complications, including issues with small blood vessels, coronary heart disease, foot problems, and retinal complications. On the clinical side, the article highlights the positive effects of CD34+ stem cell therapy in improving vascular function and tissue repair in diabetic patients, while also mentioning the inconsistencies in results between diabetes models and clinical studies, which necessitate further research to optimize treatment strategies. It emphasizes the importance of enhancing the mobilization, homing, and repair capabilities of CD34+ stem cells, as well as combining them with other treatment methods, to develop more effective strategies for treating diabetes-related vascular damage.
Collapse
Affiliation(s)
- Yiting Liu
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| |
Collapse
|
6
|
Heydari K, Johnson C, Diane Cooper I, Hersi K, Tanba C, Sun J, Solomon MA, Elinoff JM. Flow Cytometric Determination of Circulating Progenitor Cells in Patients With Pulmonary Arterial Hypertension: A Systematic Review. Pulm Circ 2025; 15:e70065. [PMID: 40125448 PMCID: PMC11925724 DOI: 10.1002/pul2.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by progressive narrowing and obliteration of distal, pre-capillary pulmonary vessels. Yet, noninvasive biomarkers that reflect this disease-defining process are lacking. A systematic review of PAH studies that measured circulating progenitor cells (CPCs) or circulating endothelial cells (CECs) in PAH by flow cytometry was performed to understand how future studies, leveraging state-of-the-art single-cell analyses, can advance the field. The study was conducted in accordance with the Preferred Reporting Items for Systematic Reviews. Of the 2422 studies identified, 20 met inclusion criteria. Nineteen studies measured CPCs by flow cytometry, only one study examined CECs. A total of 647 PAH patients were included across all 19 CPC studies. Marker schemes chosen to define CPCs, and the methods of flow cytometry used, varied significantly across studies. Meta-analysis of a subgroup of CPC studies (n = 8) similarly identified a significant amount of heterogeneity even amongst studies using the same marker scheme. In conclusion, a systematic review of CPC studies in PAH patients reveals the limitations of the current literature. Future studies should include contemporary risk assessments, disease duration, reporting of comorbid conditions, and serial sampling over time. Furthermore, methods that incorporate best practices for detecting rare cell populations by flow cytometry are essential and should be reported in sufficient detail in future publications. With the emergence of single-cell technologies, future studies of circulating progenitor and endothelial cells in PAH remain relevant and may incorporate several insights from the current review to build upon the existing literature.
Collapse
Affiliation(s)
- Kimia Heydari
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
| | - Carrie Johnson
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
| | | | - Kadija Hersi
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Carl Tanba
- Department of Pulmonary and Critical Care MedicineTufts Medical CenterBostonMarylandUSA
| | - Junfeng Sun
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
| | - Michael A. Solomon
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Jason M. Elinoff
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
7
|
Groner JA, Nicholson L, Bauer JA, Huang H, Lindstrom M, Root E. Exposure to air pollution and cardiovascular risk in young children - a pilot project. Pediatr Res 2024; 96:1718-1723. [PMID: 38982164 DOI: 10.1038/s41390-024-03377-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE To examine relationships between traffic-related air pollution (TRAP) and markers of pre-clinical cardiovascular risk in young children. STUDY DESIGN We studied a cohort of healthy children ages 2-5 recruited from pediatric primary care sites (n = 122). We obtained child weight, height, blood pressure and hair nicotine levels. A blood sample was obtained for biomarkers of systemic inflammation, oxidation, and prevalence of circulating endothelial progenitor cells. This manuscript represents a secondary analysis. TRAP exposure (particulate levels, nitrogen dioxide, nitrogen oxides, and proximity to major roadways) was assessed using national air pollution data based on child's census tract of residence. RESULTS TRAP exposure had significant positive associations with prevalence of two of the three EPC subtypes (CD34 + /CD133 + /CD45- and CD133 + /CD45-) in unadjusted correlations. In a linear regression model, adjusting for sex, age, race, ethnicity, body mass index, parental education, child insurance, and secondhand smoke exposure, one EPC subtype (CD133 + /CD45-) had a positive significant correlation to every TRAP measure. No significant relationships between air pollution and measures of inflammation and oxidation was found. CONCLUSION Our findings of the upregulation of EPCs may signal a response to early vascular damage during early childhood due to air pollution exposure. IMPACT Traffic-related air pollution (TRAP) - known cardiovascular risk factor during adulthood Current pilot study in very young children shows upregulation of cells which protect the endothelial lining of blood vessels (endothelial progenitor cells, EPCs) Upregulation of EPCs aligns with other cardiovascular risks during childhood (obesity, prematurity, type 1 diabetes) Demonstrated with TRAP exposure lower than EPA threshold Response to air pollution may be protective of cardiovascular damage during early childhood.
Collapse
Affiliation(s)
- Judith A Groner
- AAP Julius B. Richmond Center of Excellence, Itasca, IL, USA.
- Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Lisa Nicholson
- Statistical Consulting, Nationwide Children's Hospital, Columbus, OH, USA
| | - John Anthony Bauer
- AAP Julius B. Richmond Center of Excellence, Itasca, IL, USA
- Kentucky Children's Hospital, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
- University of Kentucky Center for Appalachian Research in Environmental Sciences, Lexington, USA
| | - Hong Huang
- Kentucky Children's Hospital, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
- University of Kentucky Center for Appalachian Research in Environmental Sciences, Lexington, USA
| | - Megan Lindstrom
- Department of Geography, The Ohio State University, Columbus, USA
| | - Elisabeth Root
- Department of Geography and Division of Epidemiology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
8
|
Bock PM, Monteiro RB, Maraschin CK, Alegretti AP, Farias MG, Spagnol F, Lopez PLDC, Santos LP, Helal L, Moraes RS, Umpierre D, Schaan BD. Circulating endothelial progenitor cells and inflammatory markers in type 1 diabetes after an acute session of aerobic exercise. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230499. [PMID: 39876965 PMCID: PMC11771752 DOI: 10.20945/2359-4292-2023-0499] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 09/04/2024] [Indexed: 01/31/2025]
Abstract
Objective To determine circulating endothelial progenitor cells (EPC) counts and levels of inflammatory markers in individuals with and without type 1 diabetes mellitus (T1DM) in response to an intense aerobic exercise session. Subjects and methods In total, 15 adult men with T1DM and 15 healthy individuals underwent a 30-minute aerobic exercise session on a cycle ergometer at 60% of the peak heart rate. The EPC count (CD45dim/CD34+/KDR+), tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6) levels were measured before and 60 minutes after the session. Results We found no difference within or between groups regarding EPC counts before and after the aerobic exercise: healthy individuals, 0.02% change (95% confidence interval [CI] -0.04%-0.08%); individuals with T1DM, 0.00% (95%CI -0.01%-0.01%). We also found no difference in TNF-α levels before and after exercise in healthy individuals (210.2, interquartile range [IQR] 142.1-401.2 pg/mL and 191.3, IQR 136.4-350.5 pg/mL, respectively) and in patients with T1DM (463.8, IQR 201.4-4306.0 pg/mL and 482.7, IQR 143.8-4304.3 pg/mL, respectively). Similarly, no difference in IL-6 levels was observed before and after exercise in healthy individuals (148.2, IQR 147.5-148.6 pg/mL and 148.2, IQR 147.7-148.6 pg/mL, respectively) and individuals with T1DM (147.2, IQR 145.9-147.7 pg/mL and 147.2, IQR 146.8-147.8 pg/mL, respectively). Conclusions Patients with T1DM and healthy controls had comparable EPC responses to aerobic exercise, most likely due to the absence of a chronic inflammatory state.
Collapse
Affiliation(s)
- Patrícia Martins Bock
- Instituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em SaúdeHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilInstituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em Saúde (IATS) – CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio GrandeRio GrandeRSBrasilUniversidade Federal do Rio Grande, Rio Grande, RS, Brasil
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Raíssa Borges Monteiro
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Clara Krummenauer Maraschin
- Universidade Federal do Rio Grande do SulFaculdade de MedicinaDepartamento de Clínica MédicaPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
| | - Ana Paula Alegretti
- Serviço de Diagnóstico LaboratorialHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilServiço de Diagnóstico Laboratorial (SDLab), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Mariela Granero Farias
- Serviço de Diagnóstico LaboratorialHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilServiço de Diagnóstico Laboratorial (SDLab), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Fabiane Spagnol
- Serviço de Diagnóstico LaboratorialHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilServiço de Diagnóstico Laboratorial (SDLab), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Patricia Luciana da Costa Lopez
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Lucas Porto Santos
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| | - Lucas Helal
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| | - Ruy Silveira Moraes
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| | - Daniel Umpierre
- Instituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em SaúdeHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilInstituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em Saúde (IATS) – CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| | - Beatriz D. Schaan
- Instituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em SaúdeHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilInstituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em Saúde (IATS) – CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do SulFaculdade de MedicinaDepartamento de Clínica MédicaPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| |
Collapse
|
9
|
Salybekov AA, Tashov K, Sheng Y, Salybekova A, Shinozaki Y, Asahara T, Kobayashi S. Cardioimmunology in Health and Diseases: Impairment of the Cardio-Spleno-Bone Marrow Axis Following Myocardial Infarction in Diabetes Mellitus. Int J Mol Sci 2024; 25:11833. [PMID: 39519382 PMCID: PMC11546687 DOI: 10.3390/ijms252111833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
A comprehensive understanding of the cardio-spleen-bone marrow immune cell axis is essential for elucidating the alterations occurring during the pathogenesis of diabetes mellitus (DM). This study investigates the dynamics of immune cell kinetics in DM after myocardial infarction (MI) over time. MI was induced in diabetic and healthy control groups using C57BL/N6 mice, with sacrifices occurring at days 1, 3, 7, and 28 post-MI to collect heart, peripheral blood (PB), spleen, and bone marrow (BM) samples. Cell suspensions from each organ were isolated and analyzed via flow cytometry. Additionally, the endothelial progenitor cell-colony-forming assay (EPC-CFA) was performed using mononuclear cells derived from BM, PB, and the spleen. The results indicated that, despite normal production in BM and the spleen, CD45+ cells were lower in the PB of DM mice at days 1 to 3. Further analysis revealed a reduction in total and pro-inflammatory neutrophils (N1s) in PB at days 1 to 3 and in the spleen at days 3 to 7 in DM mice, suggesting that DM-induced alterations in splenic neutrophils fail to meet the demand in PB and ischemic tissues. Infiltrating macrophages (total, M1, M2) were reduced at day 3 in the DM-ischemic heart, with total and M1 (days 1-3) and M2 (days 3-7) macrophages being significantly decreased in DM-PB compared to controls, indicating impaired macrophage recruitment and polarization in DM. Myeloid dendritic cells (mDCs) in the heart were higher from days 1 to 7, which corresponded with the enhanced recruitment of CD8+ cells from days 1 to 28 in the DM-infarcted myocardium. Total CD4+ cells decreased in DM-PB at days 1 to 3, suggesting a delayed adaptive immune response to MI. B cells were reduced in PB at days 1 to 3, in myocardium at day 3, and in the spleen at day 7, indicating compromised mobilization from BM. EPC-CFA results showed a marked decrease in definitive EPC colonies in the spleen and BM from days 1 to 28 in DM mice compared to controls in vitro, highlighting that DM severely impairs EPC colony-forming activity by limiting the differentiation of EPCs from primitive to definitive forms. Taking together, this study underscores significant disruptions in the cardio-spleen-bone marrow immune cell axis following MI in DM, revealing delayed innate and adaptive immune responses along with impaired EPC differentiation.
Collapse
Affiliation(s)
- Amankeldi A. Salybekov
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan;
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Kanat Tashov
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Yin Sheng
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Ainur Salybekova
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Yoshiko Shinozaki
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara 259-1193, Japan
| | - Takayuki Asahara
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara 259-1193, Japan; (K.T.); (Y.S.)
| | - Shuzo Kobayashi
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan;
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
| |
Collapse
|
10
|
von Känel R. Unraveling the connection: Tracing the link between neurobiological and peripheral physiological responses to acute stress, and long-term cardiovascular risk. J Psychosom Res 2024; 186:111431. [PMID: 37414618 DOI: 10.1016/j.jpsychores.2023.111431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023]
Affiliation(s)
- Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
11
|
Tiller C, Reindl M, Lechner I, Reinstadler SJ. Circulating progenitor cells: A promising biomarker for coronary collateral formation in CTO? Int J Cardiol 2024; 411:132267. [PMID: 38885793 DOI: 10.1016/j.ijcard.2024.132267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Affiliation(s)
- Christina Tiller
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Martin Reindl
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Ivan Lechner
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Sebastian J Reinstadler
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
12
|
Gold DA, Sandesara PB, Kindya B, Gold ME, Jain V, Vatsa N, Desai SR, Yadalam A, Razavi A, Elhage Hassan M, Ko YA, Liu C, Alkhoder A, Rahbar A, Hossain MS, Waller EK, Jaber WA, Nicholson WJ, Quyyumi AA. Circulating Progenitor Cells and Coronary Collaterals in Chronic Total Occlusion. Int J Cardiol 2024; 407:132104. [PMID: 38677332 PMCID: PMC11559591 DOI: 10.1016/j.ijcard.2024.132104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND The role of circulating progenitor cells (CPC) in collateral formation that occurs in the presence of chronic total occlusions (CTO) of a coronary artery is not well established. In stable patients with a CTO, we investigated whether CPC levels are associated with (a) collateral development and (b) ischemic burden, as measured by circulating high sensitivity troponin-I (hsTn-I) levels. METHODS CPCs were enumerated by flow cytometry as CD45med+ blood mononuclear cells expressing CD34 and both CD34 and CD133 epitopes. The association between CPC counts and both Rentrop collateral grade (0, 1, 2, or 3) and hsTn-I levels were evaluated using multivariate regression analysis, after adjusting for demographic and clinical characteristics. RESULTS In 89 patients (age 65.5, 72% male, 27% Black), a higher CPC count was positively associated with a higher Rentrop collateral grade; [CD34+ adjusted odds ratio (OR) 1.49 95% confidence interval (CI) (0.95, 2.34) P = 0.082] and [CD34+/CD133+ OR 1.57 95% CI (1.05, 2.36) P = 0.028]. Every doubling of CPC counts was also associated with lower hsTn-I levels [CD34+ β -0.35 95% CI (-0.49, -0.15) P = 0.002] and [CD34+/CD133+ β -0.27 95% CI (-0.43, -0.08) P = 0.009] after adjustment. CONCLUSION Individuals with higher CPC counts have greater collateral development and lower ischemic burden in the presence of a CTO.
Collapse
Affiliation(s)
- Daniel A Gold
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Pratik B Sandesara
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Bryan Kindya
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Matthew E Gold
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Vardhmaan Jain
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Nishant Vatsa
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Shivang R Desai
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Adithya Yadalam
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Alexander Razavi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Malika Elhage Hassan
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Chang Liu
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ayman Alkhoder
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Alireza Rahbar
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Mohammad S Hossain
- Division of Hematology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Edmund K Waller
- Division of Hematology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Wissam A Jaber
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - William J Nicholson
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
13
|
Tkacz M, Zgutka K, Tomasiak P, Tarnowski M. Responses of Endothelial Progenitor Cells to Chronic and Acute Physical Activity in Healthy Individuals. Int J Mol Sci 2024; 25:6085. [PMID: 38892272 PMCID: PMC11173310 DOI: 10.3390/ijms25116085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Endothelial progenitor cells (EPCs) are circulating cells of various origins that possess the capacity for renewing and regenerating the endothelial lining of blood vessels. During physical activity, in response to factors such as hypoxia, changes in osmotic pressure, and mechanical forces, endothelial cells undergo intense physiological stress that results in endothelial damage. Circulating EPCs participate in blood vessel repair and vascular healing mainly through paracrine signalling. Furthermore, physical activity may play an important role in mobilising this important cell population. In this narrative review, we summarise the current knowledge on the biology of EPCs, including their characteristics, assessment, and mobilisation in response to both chronic and acute physical activity in healthy individuals.
Collapse
Affiliation(s)
- Marta Tkacz
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Zolnierska 48, 70-210 Szczecin, Poland
| | - Katarzyna Zgutka
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Zolnierska 48, 70-210 Szczecin, Poland
| | - Patrycja Tomasiak
- Institute of Physical Culture Sciences, University of Szczecin, 70-453 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Zolnierska 48, 70-210 Szczecin, Poland
- Institute of Physical Culture Sciences, University of Szczecin, 70-453 Szczecin, Poland
| |
Collapse
|
14
|
Kwon JY, Maeng YS. Human Cord Blood Endothelial Progenitor Cells and Pregnancy Complications (Preeclampsia, Gestational Diabetes Mellitus, and Fetal Growth Restriction). Int J Mol Sci 2024; 25:4444. [PMID: 38674031 PMCID: PMC11050478 DOI: 10.3390/ijms25084444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Hemangioblasts give rise to endothelial progenitor cells (EPCs), which also express the cell surface markers CD133 and c-kit. They may differentiate into the outgrowth endothelial cells (OECs) that control neovascularization in the developing embryo. According to numerous studies, reduced levels of EPCs in circulation have been linked to human cardiovascular disorders. Furthermore, preeclampsia and senescence have been linked to levels of EPCs produced from cord blood. Uncertainties surround how preeclampsia affects the way EPCs function. It is reasonable to speculate that preeclampsia may have an impact on the function of fetal EPCs during the in utero period; however, the present literature suggests that maternal vasculopathies, including preeclampsia, damage fetal circulation. Additionally, the differentiation potential and general activity of EPCs may serve as an indicator of the health of the fetal vascular system as they promote neovascularization and repair during pregnancy. Thus, the purpose of this review is to compare-through the assessment of their quantity, differentiation potency, angiogenic activity, and senescence-the angiogenic function of fetal EPCs obtained from cord blood for normal and pregnancy problems (preeclampsia, gestational diabetes mellitus, and fetal growth restriction). This will shed light on the relationship between the angiogenic function of fetal EPCs and pregnancy complications, which could have an effect on the management of long-term health issues like metabolic and cardiovascular disorders in offspring with abnormal vasculature development.
Collapse
Affiliation(s)
- Ja-Young Kwon
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University Health System, Seoul 03722, Republic of Korea;
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Yong-Sun Maeng
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University Health System, Seoul 03722, Republic of Korea;
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
15
|
Wang Y, Huang J, Ang TFA, Zhu Y, Tao Q, Mez J, Alosco M, Denis GV, Belkina A, Gurnani A, Ross M, Gong B, Han J, Lunetta KL, Stein TD, Au R, Farrer LA, Zhang X, Qiu WQ. The association between circulating CD34+CD133+ endothelial progenitor cells and reduced risk of Alzheimer's disease in the Framingham Heart Study. EXPLORATION OF MEDICINE 2024; 5:193-214. [PMID: 38854406 PMCID: PMC11160969 DOI: 10.37349/emed.2024.00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/22/2024] [Indexed: 06/11/2024] Open
Abstract
Aim Endothelial dysfunction has been associated with both cerebrovascular pathology and Alzheimer's disease (AD). However, the connection between circulating endothelial cells and the risk of AD remains uncertain. The objective was to leverage data from the Framingham Heart Study to investigate various circulating endothelial subtypes and their potential correlations with the risk of AD. Methods The study conducted data analyses using Cox proportional hazard regression and linear regression methods. Additionally, genome-wide association study (GWAS) was carried out to further explore the data. Results Among the eleven distinct circulating endothelial subtypes, only circulating endothelial progenitor cells (EPCs) expressing CD34+CD133+ were found to be negatively and dose-dependently associated with reduced AD risk. This association persisted even after adjusting for age, sex, years of education, apolipoprotein E (APOE) ε4 status, and various vascular diseases. Particularly noteworthy was the significant association observed in individuals with hypertension and cerebral microbleeds. Consistently, positive associations were identified between CD34+CD133+ EPCs and specific brain regions, such as higher proportions of circulating CD34+CD133+ cells correlating with increased volumes of white matter and the hippocampus. Additionally, a GWAS study unveiled that CD34+CD133+ cells influenced AD risk specifically in individuals with homozygous genotypes for variants in two stem cell-related genes: kirre like nephrin family adhesion molecule 3 (KIRREL3, rs580382 CC and rs4144611 TT) and exocyst complex component 6B (EXOC6B, rs61619102 CC). Conclusions The findings suggest that circulating CD34+CD133+ EPCs possess a protective effect and may offer a new therapeutic avenue for AD, especially in individuals with vascular pathology and those carrying specific genotypes of KIRREL3 and EXOC6B genes.
Collapse
Affiliation(s)
- Yixuan Wang
- Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jinghan Huang
- Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ting Fang Alvin Ang
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Yibo Zhu
- Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Qiushan Tao
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jesse Mez
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Michael Alosco
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Gerald V. Denis
- Hematology & Medical Oncology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Anna Belkina
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Ashita Gurnani
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Mark Ross
- School of Energy, Geosciences, Infrastructure and Society, Institute of Life and Earth Sciences, Heriot-Watt University, EH14 4AS Edinburgh, UK
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jingyan Han
- Vascular Biology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Kathryn L. Lunetta
- Departments of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Thor D. Stein
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, Boston, MA 02132, USA
| | - Rhoda Au
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Departments of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
| | - Lindsay A. Farrer
- Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Departments of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- Departments of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Xiaoling Zhang
- Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Departments of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Wei Qiao Qiu
- Alzheimer’s Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
16
|
Mizuno T, Hoshino T, Ishizuka K, Toi S, Takahashi S, Wako S, Arai S, Kitagawa K. Association of circulating CD34+ cells level and prognosis after ischemic stroke. Int J Stroke 2024; 19:460-469. [PMID: 37978860 DOI: 10.1177/17474930231217192] [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: 11/19/2023]
Abstract
BACKGROUND CD34 is a transmembrane phosphoglycoprotein and a marker of hematopoietic and nonhematopoietic stem/progenitor cells. In experimental studies, CD34+ cells are rich sources of endothelial progenitor cells and can promote neovascularization and endothelial repair. The potential role of CD34+ cells in stroke patients remains unclear. AIMS We aimed to assess the prognostic effect of circulating CD34+ cell levels on the risk of vascular events and functional prognosis in stroke patients. PATIENTS AND METHODS In this prospective observational study, patients with ischemic stroke were consecutively enrolled within 1 week of onset and followed up for 1 year. Patients were divided into three groups according to tertiles of the level of circulating CD34+ cells (Tertile 1, <0.51/µL; Tertile 2, 0.51-0.96/µL; and Tertile 3, >0.96/µL). The primary outcome was a composite of major adverse cardiovascular events (MACEs), including nonfatal stroke, nonfatal acute coronary syndrome, major peripheral artery disease, and vascular death. The secondary outcomes included the modified Rankin scale (mRS) scores. RESULTS A total of 524 patients (mean age, 71.3 years; male, 60.1%) were included. High CD34+ cell levels were associated with younger age (p < 0.001) and low National Institutes of Health Stroke Scale scores at admission (p = 0.010). No significant differences were found in the risk of MACEs among the three groups (annual rates: 15.0%, 13.4%, and 12.6% in Tertiles 1, 2, and 3, respectively; log-rank p = 0.70). However, there were significant differences in the mRS scores at 3 months (median (interquartile range); 2 (1-4), 1 (1-3), and 1 (0-2) in Tertiles 1, 2, and 3, respectively; p = 0.010) and 1 year (3 (1-4), 2 (1-4), and 1 (0-3); p < 0.001) among these groups. After multivariable adjustments, a higher CD34+ cell level was independently associated with good functional outcomes (mRS score of 0-2) at 3 months (adjusted odds ratio (OR), 1.43; 95% confidence interval (CI), 1.01-2.05) and 1 year (adjusted OR, 1.53; 95% CI, 1.09-2.16). CONCLUSION Although no correlations were found between circulating CD34+ cell levels and vascular event risk, elevated CD34+ cell levels were associated with favorable functional recovery in stroke patients. DATA ACCESS STATEMENT Data supporting the findings of this study are available from the corresponding author on reasonable request. CLINICAL TRIAL REGISTRATION The TWMU Stroke Registry is registered at https://upload.umin.ac.jp as UMIN000031913.
Collapse
Affiliation(s)
- Takafumi Mizuno
- Department of Neurology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Takao Hoshino
- Department of Neurology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Kentaro Ishizuka
- Department of Neurology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Sono Toi
- Department of Neurology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Shuntaro Takahashi
- Department of Neurology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Sho Wako
- Department of Neurology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Satoko Arai
- Department of Neurology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| |
Collapse
|
17
|
Almuwaqqat Z, Wittbrodt M, Moazzami K, Garcia M, Lima B, Martini A, Sullivan S, Nye JA, Pearce BD, Shah AJ, Waller EK, Vaccarino V, Bremner JD, Quyyumi AA. Acute psychological stress-induced progenitor cell mobilization and cardiovascular events. J Psychosom Res 2024; 178:111412. [PMID: 38281471 PMCID: PMC10823179 DOI: 10.1016/j.jpsychores.2023.111412] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 05/29/2023] [Accepted: 06/12/2023] [Indexed: 01/30/2024]
Abstract
OBJECTIVE Certain brain activation responses to psychological stress are associated with worse outcomes in CVD patients. We hypothesized that elevated acute psychological stress, manifesting as greater activity within neural centers for emotional regulation, mobilizes CPC from the bone marrow to the peripheral blood and predicts future cardiovascular events. METHODS In 427 patients with stable CAD undergoing a laboratory-based mental stress (MS) test, CPCs were enumerated using flow cytometry as CD34-expressing mononuclear cells (CD34+) before and 45 min after stress. Changes in brain regional blood flow with MS were measured using high resolution-positron emission tomography (HR-PET). Association between the change in CPC with MS and the risk of cardiovascular death or myocardial infarction (MI) during a 5-year follow-up period was analyzed. RESULTS MS increased CPC counts by a mean of 150 [630] cells/mL (15%), P < 0.001. Greater limbic lobe activity, indicative of activation of emotion-regulating centers, was associated with greater CPC mobilization (P < 0.005). Using Fine and Gray models after adjustment for demographioc, clinical risk factors and medications use, greater CPC mobilization was associated with a higher adjusted risk of adverse events; a rise of 1000 cells/mL was associated with a 50% higher risk of cardiovascular death/MI [hazards ratio, 1.5, 95% confidence interval, 1.1-2.2). CONCLUSION Greater limbic lobe activity, brain areas involved in emotional regulation, is associated with MS-induced CPC mobilization. This mobilization isindependently associated with cardiovascular events. These findings provide novel insights into mechanisms through which psychological stressors modulate cardiovascular risk.
Collapse
Affiliation(s)
- Zakaria Almuwaqqat
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Matthew Wittbrodt
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA
| | - Kasra Moazzami
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Mariana Garcia
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Bruno Lima
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Afif Martini
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Samaah Sullivan
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center-Houston, Houston, Texas
| | - Jonathon A Nye
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Bradley D Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Amit J Shah
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Viola Vaccarino
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - J Douglas Bremner
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA; Atlanta VA Medical Center, Decatur, GA, USA; Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
18
|
Krishnaraj A, Bakbak E, Teoh H, Pan Y, Firoz IN, Pandey AK, Terenzi DC, Verma R, Bari B, Bakbak AI, Kunjummar SP, Yanagawa B, Connelly KA, Mazer CD, Rotstein OD, Quan A, Bhatt DL, McGuire DK, Hess DA, Verma S. Vascular Regenerative Cell Deficiencies in South Asian Adults. J Am Coll Cardiol 2024; 83:755-769. [PMID: 38355246 DOI: 10.1016/j.jacc.2023.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 10/26/2023] [Accepted: 12/06/2023] [Indexed: 02/16/2024]
Abstract
BACKGROUND South Asian individuals shoulder a disproportionate burden of cardiometabolic diseases. OBJECTIVES The purpose of this study was to determine if vascular regenerative cell content varies significantly between South Asian and White European people. METHODS Between January 2022 and January 2023, 60 South Asian and 60 White European adults with either documented cardiovascular disease or established diabetes with ≥1 other cardiovascular risk factor were prospectively enrolled. Vascular regenerative cell content in venous blood was enumerated using a flow cytometry assay that is based on high aldehyde dehydrogenase (ALDHhi) activity and cell surface marker phenotyping. The primary outcome was the difference in frequency of circulating ALDHhi progenitor cells, monocytes, and granulocytes between the 2 groups. RESULTS Compared with White European participants, those of South Asian ethnicity were younger (69 ± 10 years vs 66 ± 9 years; P < 0.05), had lower weight (88 ± 19 kg vs 75 ± 13 kg; P < 0.001), and exhibited a greater prevalence of type 2 diabetes (62% vs 92%). South Asian individuals had markedly lower circulating frequencies of pro-angiogenic ALDHhiSSClowCD133+ progenitor cells (P < 0.001) and ALDHhiSSCmidCD14+CD163+ monocytes with vessel-reparative capacity (P < 0.001), as well as proportionally more ALDHhi progenitor cells with high reactive oxygen species content (P < 0.05). After correction for sex, age, body mass index, and glycated hemoglobin, South Asian ethnicity was independently associated with lower ALDHhiSSClowCD133+ cell count. CONCLUSIONS South Asian people with cardiometabolic disease had less vascular regenerative and reparative cells suggesting compromised vessel repair capabilities that may contribute to the excess vascular risk in this population. (The Role of South Asian vs European Origins on Circulating Regenerative Cell Exhaustion [ORIGINS-RCE]; NCT05253521).
Collapse
Affiliation(s)
- Aishwarya Krishnaraj
- Division of Cardiac Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Ehab Bakbak
- Division of Cardiac Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Hwee Teoh
- Division of Cardiac Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Division of Endocrinology and Metabolism, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada
| | - Yi Pan
- Division of Cardiac Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada
| | - Irene N Firoz
- Division of Cardiac Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Arjun K Pandey
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Raj Verma
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Basel Bari
- Markham Health+ Plex, Markham, Ontario, Canada
| | | | | | - Bobby Yanagawa
- Division of Cardiac Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Division of Cardiology, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - C David Mazer
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Department of Anesthesia, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ori D Rotstein
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Division of General Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada
| | - Adrian Quan
- Division of Cardiac Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai Health System, New York, New York, USA
| | - Darren K McGuire
- Division of Cardiology, University of Texas Southwestern Medical Center and Parkland Health and Hospital System, Dallas, Texas, USA
| | - David A Hess
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Physiology and Pharmacology, Western University, London, Ontario, Canada; Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, Canada.
| | - Subodh Verma
- Division of Cardiac Surgery, St Michael's Hospital of Unity Health Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
19
|
Fadini GP, Marassi M. Vitamin D and Cardiovascular Risk: Stemming the Tide of Poor Outcomes in Coronary Heart Disease. JACC. ADVANCES 2024; 3:100803. [PMID: 38939406 PMCID: PMC11198725 DOI: 10.1016/j.jacadv.2023.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | | |
Collapse
|
20
|
Bonora BM, Morieri ML, Marassi M, Cappellari R, Avogaro A, Fadini GP. Improved prediction of long-term kidney outcomes in people with type 2 diabetes by levels of circulating haematopoietic stem/progenitor cells. Diabetologia 2023; 66:2346-2355. [PMID: 37712954 PMCID: PMC10627906 DOI: 10.1007/s00125-023-06002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/25/2023] [Indexed: 09/16/2023]
Abstract
AIM/HYPOTHESIS We examined whether prediction of long-term kidney outcomes in individuals with type 2 diabetes can be improved by measuring circulating levels of haematopoietic stem/progenitor cells (HSPCs), which are reduced in diabetes and are associated with cardiovascular risk. METHODS We included individuals with type 2 diabetes who had a baseline determination of circulating HSPCs in 2004-2019 at the diabetes centre of the University Hospital of Padua and divided them into two groups based on their median value per ml of blood. We collected updated data on eGFR and albuminuria up to December 2022. The primary endpoint was a composite of new-onset macroalbuminuria, sustained ≥40% eGFR decline, end-stage kidney disease or death from any cause. The analyses were adjusted for known predictors of kidney disease in the population with diabetes. RESULTS We analysed 342 participants (67.8% men) with a mean age of 65.6 years. Those with low HSPC counts (n=171) were significantly older and had a greater prevalence of hypertension, heart failure and nephropathy (45.0% vs 33.9%; p=0.036), as evidenced by lower eGFR and higher albuminuria at baseline. During a median follow-up of 6.7 years, participants with high vs low HSPC counts had lower rates of the composite kidney outcome (adjusted HR 0.69 [95% CI 0.49, 0.97]), slower decline in eGFR and a similar increase in albuminuria. Adding the HSPC information to the risk score of the CKD Prognosis Consortium significantly improved discrimination of individuals with future adverse kidney outcomes. CONCLUSIONS/INTERPRETATION HSPC levels predict worsening of kidney function and improve the identification of individuals with type 2 diabetes and adverse kidney outcomes over and beyond a clinical risk score.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | | | | | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padua, Italy.
- Veneto Institute of Molecular Medicine, Padua, Italy.
| |
Collapse
|
21
|
Kwon H, Jung YJ, Lee Y, Son GH, Kim HO, Maeng YS, Kwon JY. Impaired Angiogenic Function of Fetal Endothelial Progenitor Cells via PCDH10 in Gestational Diabetes Mellitus. Int J Mol Sci 2023; 24:16082. [PMID: 38003275 PMCID: PMC10671254 DOI: 10.3390/ijms242216082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Maternal hyperglycemia, induced by gestational diabetes mellitus (GDM), has detrimental effects on fetal vascular development, ultimately increasing the risk of cardiovascular diseases in offspring. The potential underlying mechanisms through which these complications occur are due to functional impairment and epigenetic changes in fetal endothelial progenitor cells (EPCs), which remain less defined. We confirm that intrauterine hyperglycemia leads to the impaired angiogenic function of fetal EPCs, as observed through functional assays of outgrowth endothelial cells (OECs) derived from fetal EPCs of GDM pregnancies (GDM-EPCs). Notably, PCDH10 expression is increased in OECs derived from GDM-EPCs, which is associated with the inhibition of angiogenic function in fetal EPCs. Additionally, increased PCDH10 expression is correlated with the hypomethylation of the PCDH10 promoter. Our findings demonstrate that in utero exposure to GDM can induce angiogenic dysfunction in fetal EPCs through altered gene expression and epigenetic changes, consequently increasing the susceptibility to cardiovascular diseases in the offspring of GDM mothers.
Collapse
Affiliation(s)
- Hayan Kwon
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (H.K.); (Y.J.J.); (Y.L.)
| | - Yun Ji Jung
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (H.K.); (Y.J.J.); (Y.L.)
| | - Yeji Lee
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (H.K.); (Y.J.J.); (Y.L.)
| | - Ga-Hyun Son
- Department of Obstetrics and Gynecology, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea;
| | - Hyun Ok Kim
- Korea Cell-Based Artificial Blood Project, Regenerative Medicine Acceleration Foundation, Seoul 04512, Republic of Korea;
| | - Yong-Sun Maeng
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (H.K.); (Y.J.J.); (Y.L.)
| | - Ja-Young Kwon
- Department of Obstetrics and Gynecology, Institute of Women’s Life Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (H.K.); (Y.J.J.); (Y.L.)
| |
Collapse
|
22
|
Ye G, Chen X, Zhou Y, Zhou J, Song Y, Yang X, Yang L. Prognostic Value of Endothelial Progenitor Cells in Acute Myocardial Infarction Patients. Mediators Inflamm 2023; 2023:4450772. [PMID: 37899988 PMCID: PMC10613116 DOI: 10.1155/2023/4450772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 10/31/2023] Open
Abstract
Objective To determine prognostic role of endothelial progenitor cells (EPCs) in intensive care patients with acute myocardial infarction (AMI). Materials and Methods From December 2018 to July 2021, a total of 91 eligible patients with AMI were consecutively examined in a single intensive care unit (ICU) in China. Patients with a history of acute coronary artery disease were excluded from the study. Samples were collected within 24 hr of onset of symptoms. EPCs, defined as coexpression of CD34+/CD133+ cells or CD133+/CD34+/KDR+, were studied using flow cytometry and categorized by quartiles. Based on the 28-days mortality outcome, the patients were further divided into two groups: death and survival. The study incorporated various variables, including cardiovascular risk factors such as body mass index, hypertension, diabetes, hypercholesterolemia, atherosclerotic burden, and medication history, as well as clinical characteristics such as APACHEⅡscore, central venous-arterial carbon dioxide difference (GAP), homocysteine, creatinine, C-reactive protein, HbAlc, and cardiac index. Cox regression analysis was employed to conduct a multivariate analysis. Results A total of 91 patients with AMI who were admitted to the ICU were deemed eligible for inclusion in the study. Among these patients, 23 (25.3%) died from various causes during the follow-up period. The counts of EPCs were found to be significantly higher in the survival group compared to the death group (P < 0.05). In the univariate analysis, it was observed that the 28-days mortality rate was associated with the several factors, including the APACHEⅡscore (P=0.00), vasoactive inotropic score (P=0.03), GAP (P=0.00), HCY (P=0.00), creatinine (P=0.00), C-reactive protein (P=0.00), HbAlc (P=0.00), CI (P=0.01), quartiles of CD34+/CD133+ cells (P=0.00), and quartiles of CD34+/CD133+/KDR+ cells (P=0.00). CD34+/CD133+/KDR+ cells retained statistical significance in Cox regression models even after controlling for clinical variables (HR: 6.258 × 10-10 and P=0.001). Nevertheless, no significant correlation was observed between CD34+/CD133+ cells and all-cause mortality. Conclusions The decreased EPCs levels, especially for CD34+/CD133+/KDR+ cells subsets, were an independent risk factor for 28-days mortality in AMI patients.
Collapse
Affiliation(s)
- Gongjie Ye
- Department of Intensive Care Unit, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315040, China
| | - Xiaodan Chen
- Department of Clinical Laboratory, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China
| | - Yinchao Zhou
- Ningbo University, Ningbo 315211, Zhejiang, China
| | - Jianqing Zhou
- Internal Medicine-Cardiovascular Department, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China
| | - Yongfei Song
- Ningbo Institute for Medicine and Biomedical Engineering Combined Innovation, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315000, Zhejiang, China
| | - Xiaoyong Yang
- Department of Rehabilitation, Zhenhai Longsai Hospital, Ningbo 315000, Zhejiang, China
| | - Lei Yang
- Department of Intensive Care Unit, Zhenhai Longsai Hospital, 6 Gulou West Road, Chengguan, Zhaobaoshan Street, Zhenhai District, Ningbo 315299, Zhejiang, China
| |
Collapse
|
23
|
Ahmadi AR, Atiee G, Chapman B, Reynolds L, Sun J, Cameron AM, Wesson RN, Burdick JF, Sun Z. A phase I, first-in-human study to evaluate the safety and tolerability, pharmacokinetics, and pharmacodynamics of MRG-001 in healthy subjects. Cell Rep Med 2023; 4:101169. [PMID: 37633275 PMCID: PMC10518600 DOI: 10.1016/j.xcrm.2023.101169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/13/2023] [Accepted: 08/01/2023] [Indexed: 08/28/2023]
Abstract
Preclinical studies demonstrate that pharmacological mobilization and recruitment of endogenous bone marrow stem cells and immunoregulatory cells by a fixed-dose drug combination (MRG-001) improves wound healing, promotes tissue regeneration, and prevents allograft rejection. In this phase I, first-in-human study, three cohorts receive subcutaneous MRG-001 or placebo, every other day for 5 days. The primary outcome is safety and tolerability of MRG-001. Fourteen subjects received MRG-001 and seven received a placebo. MRG-001 is safe over the selected dose range. There are no clinically significant laboratory changes. The intermediate dose group demonstrates the most significant white blood cell, stem cell, and immunoregulatory cell mobilization. PBMC RNA sequencing and gene set enrichment analysis reveal 31 down-regulated pathways in the intermediate MRG-001 dose group compared with no changes in the placebo group. MRG-001 is safe across all dose ranges. MRG-001 may be a clinically useful therapy for immunoregulation and tissue regeneration (ClinicalTrials.gov: NCT04646603).
Collapse
Affiliation(s)
| | | | | | | | - John Sun
- MedRegen LLC, Baltimore, MD, USA
| | - Andrew M Cameron
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Russell N Wesson
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | | | - Zhaoli Sun
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
24
|
Marassi M, Fadini GP. The cardio-renal-metabolic connection: a review of the evidence. Cardiovasc Diabetol 2023; 22:195. [PMID: 37525273 PMCID: PMC10391899 DOI: 10.1186/s12933-023-01937-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023] Open
Abstract
Type 2 diabetes (T2D), cardiovascular disease (CVD) and chronic kidney disease (CKD), are recognized among the most disruptive public health issues of the current century. A large body of evidence from epidemiological and clinical research supports the existence of a strong interconnection between these conditions, such that the unifying term cardio-metabolic-renal (CMR) disease has been defined. This coexistence has remarkable epidemiological, pathophysiologic, and prognostic implications. The mechanisms of hyperglycemia-induced damage to the cardio-renal system are well validated, as are those that tie cardiac and renal disease together. Yet, it remains controversial how and to what extent CVD and CKD can promote metabolic dysregulation. The aim of this review is to recapitulate the epidemiology of the CMR connections; to discuss the well-established, as well as the putative and emerging mechanisms implicated in the interplay among these three entities; and to provide a pathophysiological background for an integrated therapeutic intervention aiming at interrupting this vicious crosstalks.
Collapse
Affiliation(s)
- Marella Marassi
- Department of Medicine, Division of Metabolic Diseases, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, Division of Metabolic Diseases, University of Padova, Via Giustiniani 2, 35128, Padua, Italy.
- Veneto Institute of Molecular Medicine, 35129, Padua, Italy.
| |
Collapse
|
25
|
Yanai H, Adachi H, Hakoshima M, Katsuyama H. Significance of Endothelial Dysfunction Amelioration for Sodium-Glucose Cotransporter 2 Inhibitor-Induced Improvements in Heart Failure and Chronic Kidney Disease in Diabetic Patients. Metabolites 2023; 13:736. [PMID: 37367894 DOI: 10.3390/metabo13060736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Beyond lowering plasma glucose levels, sodium-glucose cotransporter 2 inhibitors (SGLT2is) significantly reduce hospitalization for heart failure (HF) and retard the progression of chronic kidney disease (CKD) in patients with type 2 diabetes. Endothelial dysfunction is not only involved in the development and progression of cardiovascular disease (CVD), but is also associated with the progression of CKD. In patients with type 2 diabetes, hyperglycemia, insulin resistance, hyperinsulinemia and dyslipidemia induce the development of endothelial dysfunction. SGLT2is have been shown to improve endothelial dysfunction, as assessed by flow-mediated vasodilation, in individuals at high risk of CVD. Along with an improvement in endothelial dysfunction, SGLT2is have been shown to improve oxidative stress, inflammation, mitochondrial dysfunction, glucotoxicity, such as the advanced signaling of glycation end products, and nitric oxide bioavailability. The improvements in endothelial dysfunction and such endothelium-derived factors may play an important role in preventing the development of coronary artery disease, coronary microvascular dysfunction and diabetic cardiomyopathy, which cause HF, and play a role in retarding CKD. The suppression of the development of HF and the progression of CKD achieved by SGLT2is might have been largely induced by their capacity to improve vascular endothelial function.
Collapse
Affiliation(s)
- Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Hiroki Adachi
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Mariko Hakoshima
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Hisayuki Katsuyama
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| |
Collapse
|
26
|
Nardone V, Belfiore MP, De Chiara M, De Marco G, Patanè V, Balestrucci G, Buono M, Salvarezza M, Di Guida G, D'Angiolella D, Grassi R, D'Onofrio I, Cimmino G, Della Corte CM, Gambardella A, Morgillo F, Ciardiello F, Reginelli A, Cappabianca S. CARdioimaging in Lung Cancer PatiEnts Undergoing Radical RadioTherapy: CARE-RT Trial. Diagnostics (Basel) 2023; 13:1717. [PMID: 37238201 DOI: 10.3390/diagnostics13101717] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) is a common, steady growing lung tumour that is often discovered when a surgical approach is forbidden. For locally advanced inoperable NSCLC, the clinical approach consists of a combination of chemotherapy and radiotherapy, eventually followed by adjuvant immunotherapy, a treatment that is useful but may cause several mild and severe adverse effect. Chest radiotherapy, specifically, may affect the heart and coronary artery, impairing heart function and causing pathologic changes in myocardial tissues. The aim of this study is to evaluate the damage coming from these therapies with the aid of cardiac imaging. METHODS This is a single-centre, prospective clinical trial. Patients with NSCLC who are enrolled will undergo computed tomography (CT) and magnetic resonance imaging (MRI) before chemotherapy 3 months, 6 months, and 9-12 months after the treatment. We expect to enrol 30 patients in 2 years. CONCLUSIONS Our clinical trial will be an opportunity not only to highlight the timing and the radiation dose needed for pathological cardiac tissue changes to happen but will also provide useful data to set new follow-up schedules and strategies, keeping in mind that, more often than not, patients affected by NSCLC may present other heart- and lung-related pathological conditions.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Maria Paola Belfiore
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Marco De Chiara
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Giuseppina De Marco
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Vittorio Patanè
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Giovanni Balestrucci
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Mauro Buono
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Maria Salvarezza
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Gaetano Di Guida
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Domenico D'Angiolella
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Roberta Grassi
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Ida D'Onofrio
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
- Radiotherapy Unit, Ospedale del Mare, ASL Napoli 1 Centro, 80138 Naples, Italy
| | - Giovanni Cimmino
- Department of Translational Medical Science, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | | | - Antonio Gambardella
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Floriana Morgillo
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
27
|
Carulli E, Pompilio G, Vinci MC. Human Hematopoietic Stem/Progenitor Cells in Type One Diabetes Mellitus Treatment: Is There an Ideal Candidate? Cells 2023; 12:cells12071054. [PMID: 37048127 PMCID: PMC10093723 DOI: 10.3390/cells12071054] [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: 02/21/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a highly prevalent autoimmune disease causing the destruction of pancreatic islet β-cells. The resulting insulin production deficiency leads to a lifelong need for insulin re-placement therapy, systemic complications, and reduced life quality and expectancy. Cell therapy has been extensively attempted to restore insulin independence (IID), and autologous nonmyeloablative hematopoietic stem cell transplantation (AHST) has appeared to give the most promising results, but with a highly variable quote of patients achieving IID across the studies. We performed a comprehensive review of the trials involving stem cells, and in particular AHST, for the treatment of T1DM. We then pooled the patients enrolled in the different trials and looked for the patient characteristics that could be associated with the achievement of IID. We found a significantly higher probability of achieving IID in older patients (OR 1.17, 95%CI 1.06–1.33, p = 0.002) and a significantly lower probability in patients with a history of ketoacidosis (OR 0.23, 95%CI 0.06–0.78, p = 0.023). This suggests that there could be a population of patients more likely to benefit from AHST, but further data would be required to depict the profile of the ideal candidate.
Collapse
Affiliation(s)
- Ermes Carulli
- Doctoral Programme in Translational Medicine, Università di Milano, 20122 Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università di Milano, 20122 Milan, Italy
- National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
- Correspondence:
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (G.P.); (M.C.V.)
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università di Milano, 20122 Milan, Italy
| | - Maria Cristina Vinci
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (G.P.); (M.C.V.)
| |
Collapse
|
28
|
Dhindsa DS, Desai SR, Jin Q, Sandesara PB, Mehta A, Liu C, Tahhan AS, Nayak A, Ejaz K, Hooda A, Moazzami K, Islam SJ, Rogers SC, Almuwaqqat Z, Mokhtari A, Hesaroieh I, Ko YA, Sperling LS, Waller EK, Quyyumi AA. Circulating progenitor cells and outcomes in patients with coronary artery disease. Int J Cardiol 2023; 373:7-16. [PMID: 36460208 PMCID: PMC9840693 DOI: 10.1016/j.ijcard.2022.11.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 11/10/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Low quantities of circulating progenitor cells (CPCs), specifically CD34+ populations, reflect impairment of intrinsic regenerative capacity. This study investigates the relationship between subsets of CPCs and adverse outcomes. METHODS 1366 individuals undergoing angiography for evaluation of coronary artery disease (CAD) were enrolled into the Emory Cardiovascular Biobank. Flow cytometry identified CPCs as CD45med blood mononuclear cells expressing the CD34 epitope, with further enumeration of hematopoietic CPCs as CD133+/CXCR4+ cells and endothelial CPCs as vascular endothelial growth factor receptor-2 (VEGFR2+) cells. Adjusted Cox or Fine and Gray's sub-distribution hazard regression models analyzed the relationship between CPCs and 1) all-cause death and 2) a composite of cardiovascular death and non-fatal myocardial infarction (MI). RESULTS Over a median 3.1-year follow-up period (IQR 1.3-4.9), there were 221 (16.6%) all-cause deaths and 172 (12.9%) cardiovascular deaths/MIs. Hematopoietic CPCs were highly correlated, and the CD34+/CXCR4+ subset was the best independent predictor. Lower counts (≤median) of CD34+/CXCR4+ and CD34+/VEGFR2+ cells independently predicted all-cause mortality (HR 1.46 [95% CI 1.06-2.01], p = 0.02 and 1.59 [95% CI 1.15-2.18], p = 0.004) and cardiovascular death/MI (HR 1.50 [95% CI 1.04-2.17], p = 0.03 and 1.47 [95% CI 1.01-2.03], p = 0.04). A combination of low CD34+/CXCR4+ and CD34+/VEGFR2+ CPCs predicted all-cause death (HR 2.1, 95% CI 1.4-3.0; p = 0.0002) and cardiovascular death/MI (HR 2.0, 95% CI 1.3-3.2; p = 0.002) compared to those with both lineages above the cut-offs. CONCLUSIONS Lower levels of hematopoietic and endothelial CPCs indicate diminished endogenous regenerative capacity and independently correlate with greater mortality and cardiovascular risk in patients with CAD.
Collapse
Affiliation(s)
- Devinder S Dhindsa
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Shivang R Desai
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Qingchun Jin
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Pratik B Sandesara
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Anurag Mehta
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Chang Liu
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ayman S Tahhan
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Aditi Nayak
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Kiran Ejaz
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ananya Hooda
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Kasra Moazzami
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Shabatun J Islam
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Steven C Rogers
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Zakaria Almuwaqqat
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ali Mokhtari
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Iraj Hesaroieh
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Laurence S Sperling
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
29
|
Yanai H, Adachi H, Hakoshima M, Katsuyama H. Glucagon-Like Peptide 1 Receptor Agonists Versus Sodium-Glucose Cotransporter 2 Inhibitors for Atherosclerotic Cardiovascular Disease in Patients With Type 2 Diabetes. Cardiol Res 2023; 14:12-21. [PMID: 36896226 PMCID: PMC9990545 DOI: 10.14740/cr1459] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/11/2023] [Indexed: 02/27/2023] Open
Abstract
Beyond improving hemoglobin A1c (HbA1c) in adults with type 2 diabetes, glucagon-like peptide 1 receptor agonists (GLP-1RA) have been approved for reducing risk of major adverse cardiovascular events (MACE) with established cardiovascular disease (CVD) or multiple CV risk factors. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) also reduced the risk for the primary composite CV outcome in patients with type 2 diabetes at high risk for CV events. In the American Diabetes Association (ADA) and European Association of Study in Diabetes (EASD) consensus report 2022, there is the description "In people with established atherosclerotic CVD (ASCVD) or with a high risk for ASCVD, GLP-1RA were prioritized over SGLT2i"; however, the evidence supporting such statement is limited. Therefore, we studied the superiority of GLP-1RA over SGLT2i for prevention of ASCVD from various viewpoints. We could not find a meaningful difference in the risk reduction in three-point MACE (3P-MACE), mortality due to any cause, mortality due to CV cause and nonfatal myocardial infarction between GLP-1RA and SGLT2i trials. The risk of nonfatal stroke decreased in all five GLP-1RA trials; however, two of three SGLT2i trials showed an increase in risk of nonfatal stroke. The risk of hospitalization for heart failure (HHF) decreased in all three SGLT2i trials, and one GLP-1RA trial showed an increase in risk of HHF. The risk reduction of HHF in SGLT2i trials was greater than that in GLP-1RA trials. These findings were consistent with current systematic reviews and meta-analyses. The risk reduction of 3P-MACE was significantly and negatively correlated with changes in HbA1c (R = -0.861, P = 0.006) and body weight (R = -0.895, P = 0.003) in GLP-1RA and SGLT2i trials. The studies using SGLT2i failed to reduce carotid intima media thickness (cIMT), the surrogate marker for atherosclerosis; however, several studies using GLP-1RA successfully reduced cIMT in patients with type 2 diabetes. Compared with SGLT2i, GLP-1RA had a higher probability of decreasing serum triglyceride. GLP-1RA have multiple vascular biological anti-atherogenic properties.
Collapse
Affiliation(s)
- Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, Chiba, Japan
| | - Hiroki Adachi
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, Chiba, Japan
| | - Mariko Hakoshima
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, Chiba, Japan
| | - Hisayuki Katsuyama
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, Chiba, Japan
| |
Collapse
|
30
|
Wang Y, Huang J, Ang TFA, Zhu Y, Tao Q, Mez J, Alosco M, Denis GV, Belkina A, Gurnani A, Ross M, Gong B, Han J, Lunetta KL, Stein TD, Au R, Farrer LA, Zhang X, Qiu WQ. Circulating Endothelial Progenitor Cells Reduce the Risk of Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.16.23284571. [PMID: 36711847 PMCID: PMC9882408 DOI: 10.1101/2023.01.16.23284571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Cerebrovascular damage coexists with Alzheimer's disease (AD) pathology and increases AD risk. However, it is unclear whether endothelial progenitor cells reduce AD risk via cerebrovascular repair. By using the Framingham Heart Study (FHS) offspring cohort, which includes data on different progenitor cells, the incidence of AD dementia, peripheral and cerebrovascular pathologies, and genetic data (n = 1,566), we found that elevated numbers of circulating endothelial progenitor cells with CD34+CD133+ co-expressions had a dose-dependent association with decreased AD risk (HR = 0.67, 95% CI: 0.46-0.96, p = 0.03) after adjusting for age, sex, years of education, and APOE ε4. With stratification, this relationship was only significant among those individuals who had vascular pathologies, especially hypertension (HTN) and cerebral microbleeds (CMB), but not among those individuals who had neither peripheral nor central vascular pathologies. We applied a genome-wide association study (GWAS) and found that the number of CD34+CD133+ cells impacted AD risk depending on the homozygous genotypes of two genes: KIRREL3 rs580382 CC carriers (HR = 0.31, 95% CI: 0.17-0.57, p<0.001), KIRREL3 rs4144611 TT carriers (HR = 0.29, 95% CI: 0.15-0.57, p<0.001), and EXOC6B rs61619102 CC carriers (HR = 0.49, 95% CI: 0.31-0.75, p<0.001) after adjusting for confounders. In contrast, the relationship did not exist in their counterpart genotypes, e.g. KIRREL3 TT/CT or GG/GT carriers and EXOC6B GG/GC carriers. Our findings suggest that circulating CD34+CD133+ endothelial progenitor cells can be therapeutic in reducing AD risk in the presence of cerebrovascular pathology, especially in KIRREL3 and EXOC6B genotype carriers.
Collapse
|
31
|
Dassie F, Albiero M, Bettini S, Cappellari R, Milan G, Ciciliot S, Naggert JK, Avogaro A, Vettor R, Maffei P, Fadini GP. Hematopoietic Stem Cells and Metabolic Deterioration in Alström Syndrome, a Rare Genetic Model of the Metabolic Syndrome. Endocrinology 2023; 164:7005410. [PMID: 36702623 DOI: 10.1210/endocr/bqad011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/28/2023]
Abstract
Alström syndrome (AS) is a rare genetic disease caused by ALMS1 mutations, characterized by short stature, and vision and hearing loss. Patients with AS develop the metabolic syndrome, long-term organ complications, and die prematurely. We explored the association between AS and a shortage of hematopoietic stem/progenitor cells (HSPCs), which is linked to metabolic diseases and predicts diabetic complications. We included patients with AS at a national referral center. We measured HSPCs with flow cytometry at baseline and follow-up. We followed patients up to January 2022 for metabolic worsening and end-organ damage. We evaluated HSPC levels and mobilization as well as bone marrow histology in a murine model of AS. In 23 patients with AS, we found significantly lower circulating HSPCs than in healthy blood donors (-40%; P = .002) and age/sex-matched patients (-25%; P = .022). Longitudinally, HSPCs significantly declined by a further 20% in patients with AS over a median of 36 months (interquartile range 30-44). Patients with AS who displayed metabolic deterioration over 5.3 years had lower levels of HSPCs, both at baseline and at last observation, than those who did not deteriorate. Alms1-mutated mice were obese and insulin resistant and displayed significantly reduced circulating HSPCs, despite no overt hematological abnormality. Contrary to what was observed in diabetic mice, HSPC mobilization and bone marrow structure were unaffected. We found depletion of HSPCs in patients with AS, which was recapitulated in Alms1-mutated mice. Larger and longer studies will be needed to establish HSPCs shortage as a driver of metabolic deterioration leading to end-organ damage in AS.
Collapse
Affiliation(s)
- Francesca Dassie
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, 35128 Padova, Italy
- Veneto Institute of Molecular Medicine, 35128 Padova, Italy
| | - Silvia Bettini
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | | | - Gabriella Milan
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | | | | | - Angelo Avogaro
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Roberto Vettor
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Pietro Maffei
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, 35128 Padova, Italy
- Veneto Institute of Molecular Medicine, 35128 Padova, Italy
| |
Collapse
|
32
|
The Long Telling Story of "Endothelial Progenitor Cells": Where Are We at Now? Cells 2022; 12:cells12010112. [PMID: 36611906 PMCID: PMC9819021 DOI: 10.3390/cells12010112] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Endothelial progenitor cells (EPCs): The name embodies years of research and clinical expectations, but where are we now? Do these cells really represent the El Dorado of regenerative medicine? Here, past and recent literature about this eclectic, still unknown and therefore fascinating cell population will be discussed. This review will take the reader through a temporal journey that, from the first discovery, will pass through years of research devoted to attempts at their definition and understanding their biology in health and disease, ending with the most recent evidence about their pathobiological role in cardiovascular disease and their recent applications in regenerative medicine.
Collapse
|
33
|
Baumbach A, Cui YX, Evans RN, Culliford L, Johnson T, Rogers CA, Reeves BC, Bucciarelli-Ducci C, Harris J, Hamilton M, Madeddu P. A cohort study of circulating progenitor cells after ST-segment elevation and non-ST segment elevation myocardial infarction in non-diabetic and diabetic patients. Front Cardiovasc Med 2022; 9:1011140. [DOI: 10.3389/fcvm.2022.1011140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022] Open
Abstract
BackgroundMyocardial infarction induces elevation of progenitor cells in the circulation, a reparative response inhibited by type-2 diabetes.ObjectivesDetermine if myocardial infarct severity and diabetes interactively influence the migratory activity of CD34+/CXCR4+ progenitor cells and if the migratory test predicts cardiac outcomes.Materials and methodsA longitudinal study was conducted on patients with or without diabetes with a STEMI or NSTEMI. CD34+/CXCR4+ cells were measured in the peripheral blood using flow cytometry, and migratory activity was tested in vitro on cells isolated from samples collected on days 0 and 4 post-infarct. Cardiac function was assessed at three months using cardiac MRI.ResultsOf 1,149 patients screened, 71 (6.3%) were eligible and consented. Fifty had STEMI (16 with diabetes) and 21 NSTEMI (8 with diabetes). The proportion of CD34+/CXCR4+ cells within blood mononuclear cells was 1.96 times higher after STEMI compared with NSTEMI (GMR = 1.96, 95% CI 0.87, 4.37) and 1.55 times higher in patients with diabetes compared to patients without diabetes (GMR = 1.55, 95% CI 0.77, 3.13). In the latter, STEMI was associated with a 2.42-times higher proportion of migrated CD34 + /CXCR4 + cells compared with NSTEMI (GMR = 2.42, 95% CI 0.66, 8.81). In patients with diabetes, the association was the opposite, with a 55% reduction in the proportion of migrated CD34+/CXCR4+ cells. No statistically significant associations were observed between the frequency in peripheral blood or in vitro migration capacity of CD34+/CXCR4+ cells and MRI outcomes.ConclusionWe document the interaction between infarct and diabetes on the migratory activity of CD34+/CXCR4+ cells. The test did not predict functional outcomes in the studied cohort.
Collapse
|
34
|
Xie D, Li Y, Xu M, Zhao X, Chen M. Effects of dulaglutide on endothelial progenitor cells and arterial elasticity in patients with type 2 diabetes mellitus. Cardiovasc Diabetol 2022; 21:200. [PMID: 36199064 PMCID: PMC9533545 DOI: 10.1186/s12933-022-01634-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Randomised controlled trial showed that dulaglutide can reduce the risk of atherosclerotic cardiovascular disease (ASCVD) in patients with type 2 diabetes mellitus (T2DM), but the underlying mechanisms remain unclear. This study aimed to investigate the effect of dulaglutide on the number and function of endothelial progenitor cells (EPCs) in the peripheral blood of patients with T2DM and its role in improving arterial elasticity, so as to determine potential mechanisms of preventive effect of dulaglutide on ASCVD. Methods Sixty patients with T2DM were treated with 1000 mg/day of metformin and randomly divided into two groups for 12 weeks: metformin monotherapy group (MET group, n = 30), and metformin combined with dulaglutide group (MET-DUL group, n = 30). Before and after treatment, the number of CD34+CD133+KDR+ EPCs and the brachial–ankle pulse wave velocity (baPWV) of the participants were measured, and EPC proliferation, adhesion, migration, and tubule formation were assessed in vitro. Results There were no significant differences in the number and function of EPCs and baPWV changes in MET group (P > 0.05). In MET-DUL group, nitric oxide (NO) levels and the number of EPCs increased after treatment (P < 0.05), while the levels of C-reactive protein (CRP), interleukin-6 (IL-6), tumour necrosis factor-α (TNF-α), advanced glycation end products (AGEs), and baPWV decreased (P < 0.05). EPC proliferation, adhesion, migration, and tubule formation abilities were significantly enhanced (P < 0.05). Correlation analysis showed that in MET-DUL group, the changes in CRP, IL-6, TNF-α, and AGEs were negatively correlated with the number of EPCs and their proliferation and migration abilities (P < 0.05). Body weight, NO, CRP, and IL-6 levels were independent factors affecting the number of EPCs (P < 0.05). The changes in number of EPCs, proliferation and migration abilities of EPCs, and NO and IL-6 levels were independent influencing factors of baPWV changes (P < 0.05). Conclusion Dulaglutide can increase the number and function of EPCs in peripheral blood and improve arterial elasticity in patients with T2DM; it is accompanied by weight loss, inflammation reduction, and high NO levels. Dulaglutide regulation of EPCs may be a mechanism of cardiovascular protection.
Collapse
Affiliation(s)
- Dandan Xie
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei, 230032, Anhui, People's Republic of China
| | - Yutong Li
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei, 230032, Anhui, People's Republic of China
| | - Murong Xu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei, 230032, Anhui, People's Republic of China
| | - Xiaotong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei, 230032, Anhui, People's Republic of China
| | - Mingwei Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Shushan District, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
35
|
Lin A, Pehrson M, Sarno G, Fraser A, Rich-Edwards JW, Gonҫalves I, Pihlsgård M, Timpka S. Coronary Artery Restenosis in Women by History of Preeclampsia. J Am Heart Assoc 2022; 11:e026287. [PMID: 36073639 DOI: 10.1161/jaha.122.026287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background A history of preeclampsia is associated with increased risk of coronary artery disease and experimental evidence suggests that a history of preeclampsia also increases the risk of restenosis. However, the extent to which a history of preeclampsia is associated with risk of restenosis after percutaneous coronary intervention in women is unknown. Methods and Results We included 6065 parous women aged ≤65 years with first percutaneous coronary intervention on 9452 segments 2006 to 2017, linking nationwide data on percutaneous coronary intervention and delivery history in Sweden. Main outcomes were clinical restenosis and target lesion revascularization within 2 years. We accounted for segment-, procedure-, and patient-related potential predictors of restenosis in proportional hazards regression models. Restenosis occurred in 345 segments (3.7%) and target lesion revascularization was performed on 383 patients (6.3%). A history of preeclampsia was neither significantly associated with risk of restenosis (predictor-accounted hazard ratio [HR], 0.71 [95% CI, 0.41-1.23]) nor target lesion revascularization (0.74 [95% CI, 0.51-1.07]) compared with a normotensive pregnancy history. When term and preterm preeclampsia were investigated separately, segments in women with a history of term preeclampsia had a lower risk of restenosis (predictor-accounted HR, 0.45 [95% CI, 0.21-0.94]). A history of preeclampsia was not significantly associated with death by any cause within 2 years of the index procedure (predictor-accounted HR 1.06, [95% CI, 0.62-1.80]). Conclusions A history of preeclampsia was not associated with increased risk of restenosis but instead some evidence pointed to a decreased risk. To facilitate future studies and allow for replication, concomitant collection of data on pregnancy complication history and percutaneous coronary intervention outcomes in women is warranted.
Collapse
Affiliation(s)
- Annie Lin
- Perinatal and Cardiovascular Epidemiology Lund University Diabetes Centre, Clinical Sciences Malmö, Lund University Malmö Sweden
| | - Moa Pehrson
- Perinatal and Cardiovascular Epidemiology Lund University Diabetes Centre, Clinical Sciences Malmö, Lund University Malmö Sweden
| | - Giovanna Sarno
- Department of Medical Sciences Cardiology and Uppsala Clinical Research Center, Uppsala University Uppsala Sweden
| | - Abigail Fraser
- Population Health Science, Bristol Medical School University of Bristol Bristol United Kingdom
| | - Janet W Rich-Edwards
- Division of Women's Health Department of Medicine, Brigham and Women's Hospital and Harvard Medical School Boston MA
| | - Isabel Gonҫalves
- Department of Cardiology and Cardiovascular Research Translational Studies Clinical Sciences Malmö, Lund University Malmö Sweden
| | - Mats Pihlsgård
- Perinatal and Cardiovascular Epidemiology Lund University Diabetes Centre, Clinical Sciences Malmö, Lund University Malmö Sweden
| | - Simon Timpka
- Perinatal and Cardiovascular Epidemiology Lund University Diabetes Centre, Clinical Sciences Malmö, Lund University Malmö Sweden.,Department of Obstetrics and Gynecology Skåne University Hospital Malmö Sweden
| |
Collapse
|
36
|
Maio A, Maiorino MI, Longo M, Scappaticcio L, Pernice V, Cirillo P, Caruso P, Paglionico VA, Bellastella G, Esposito K. Change in Circulating Levels of Endothelial Progenitor Cells and Sexual Function in Women With Type 1 Diabetes. J Clin Endocrinol Metab 2022; 107:e3910-e3918. [PMID: 35583559 PMCID: PMC9387708 DOI: 10.1210/clinem/dgac316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Endothelial progenitor cells (EPCs), which are involved in the mechanisms of vascular repair and sexual function, are decreased in diabetic women compared with general population. OBJECTIVE This work aimed to investigate the circulating levels of EPCs and the change in sexual function during the menstrual cycle in women with type 1 diabetes (T1DM) compared with healthy women. METHODS This case-control observational study was conducted at the Unit of Endocrinology and Metabolic Diseases at University Hospital "Luigi Vanvitelli'' of Naples. Participants included 36 women with T1DM and 64 age-matched healthy controls. EPCs were quantified by flow cytometry and sexual function was assessed using the Female Sexual Function Index (FSFI) and the Female Sexual Distress Scale. All assessments were made at the follicular, ovulatory, and luteal phases of the same menstrual cycle. Main outcome measures included differences in EPCs levels and sexual function between patients and controls. RESULTS Compared with controls, women with T1DM showed significantly lower levels of both CD34 + (P < .001) and CD34 + CD133 + cells (P < .001) in the ovulatory phase, and CD34 + KDR + cells both in the ovulatory phase and in the luteal phase (P < .001 for both). Diabetic women showed significantly lower total FSFI scores and higher FSDS score than control women in all phases of the menstrual cycle. FSFI total score was predicted by both CD34 + CD133 + and CD34 + KDR + cells in the follicular phase, CD34 + and CD34 + KDR + CD133 + cells in the ovulatory phase, and CD34 + KDR + and CD34 + KDR + CD133 + cells in the luteal phase. CONCLUSION Women with T1DM show lower levels of EPCs during the menstrual cycle compared with controls. EPCs count predicts sexual function in this selected population.
Collapse
Affiliation(s)
| | - Maria Ida Maiorino
- Correspondence: Maria Ida Maiorino, MD, PhD, Unit of Endocrinology and Metabolic Diseases, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza L. Miraglia 2, 80138 Naples, Italy.
| | - Miriam Longo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Unit of Endocrinology and Metabolic Diseases, University Hospital Luigi Vanvitelli, 80138 Naples, Italy
| | - Lorenzo Scappaticcio
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Vlenia Pernice
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Paolo Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Paola Caruso
- Unit of Endocrinology and Metabolic Diseases, University Hospital Luigi Vanvitelli, 80138 Naples, Italy
| | - Vanda Amoresano Paglionico
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Unit of Endocrinology and Metabolic Diseases, University Hospital Luigi Vanvitelli, 80138 Naples, Italy
| | - Giuseppe Bellastella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Unit of Endocrinology and Metabolic Diseases, University Hospital Luigi Vanvitelli, 80138 Naples, Italy
| | - Katherine Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Unit of Endocrinology and Metabolic Diseases, University Hospital Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
37
|
Bonora BM, Cappellari R, Grasso M, Mazzucato M, D'Anna M, Avogaro A, Fadini GP. Glycaemic Control Achieves Sustained Increases of Circulating Endothelial Progenitor Cells in Patients Hospitalized for Decompensated Diabetes: An Observational Study. Diabetes Ther 2022; 13:1327-1337. [PMID: 35676613 PMCID: PMC9240124 DOI: 10.1007/s13300-022-01273-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/12/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND AND AIM Diabetes reduces the levels of circulating endothelial progenitor cells (EPCs), which contribute to vascular homeostasis. In turn, low EPCs levels predict progression of chronic complications. Several studies have shown that hyperglycaemia exerts detrimental effects on EPCs. Improvement in glucose control with glucose-lowering medications is associated with an increase of EPCs, but only after a long time of good glycaemic control. In the present study, we examined the effect of a rapid glycaemic amelioration on EPC levels in subjects hospitalized for decompensated diabetes. METHODS We used flow cytometry to quantify EPCs (CD34+/CD133+KDR+) in patients hospitalized for/with decompensated diabetes at admission, at discharge, and 2 months after the discharge. During hospitalization, all patients received intensive insulin therapy. RESULTS Thirty-nine patients with type 1 or type 2 diabetes were enrolled. Average (± SEM) fasting glucose decreased from 409.2 ± 25.9 mg/dl at admission to 190.4 ± 12.0 mg/dl at discharge and to 169.0 ± 10.3 at 2 months (both p < 0.001). EPCs (per million blood cells) significantly increased from hospital admission (13.1 ± 1.4) to discharge (16.4 ± 1.1; p = 0.022) and remained stable after 2 months (15.5 ± 1.7; p = 0.023 versus baseline). EPCs increased significantly more in participants with newly-diagnosed diabetes than in those with pre-existing diabetes. The increase in EPCs was significant in type 1 but not in type 2 diabetes and in those without chronic complications. CONCLUSION In individuals hospitalized for decompensated diabetes, insulin therapy rapidly increases EPC levels for up to 2 months. EPC defect, reflecting impaired vascular repair capacity, may be reversible in the early diabetes stages.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- Venetian Institute of Molecular Medicine, 35128, Padua, Italy
| | | | - Marco Grasso
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Marta Mazzucato
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Marianna D'Anna
- Venetian Institute of Molecular Medicine, 35128, Padua, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy.
- Venetian Institute of Molecular Medicine, 35128, Padua, Italy.
| |
Collapse
|
38
|
Yiliyasi Y, Wusainahong K, An L, Liu Q, Fan Z, Fan J. Co-Culture of Bone Marrow Mesenchymal Stem Cells Regulates the Fibrotic Response of Endometrial Stromal Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study explored the protective effects and mechanisms of bone marrow mesenchymal stem cells (BMSCs) on the fibrotic response of endometrial stromal cells. The endometrial cells were isolated from intrauterine adhesions (IUA) patients and assigned into control group, TGF-β1
group, BMSC group which was co-cultured with BMSCs in presence of TGF-β1; Exo group (treated with BMSCs-originated exosomes); Exo-inhibitor group (treated with exosome-specific inhibitors) followed by analysis of α-SMA and Vimentin level, cell viability and expression
of TGFBR2, TGFβ-1, Fibronectin (FN), α-SMA, Collagen 1α1 (Col1a1), Smad2/3 and p-Smad2/3. After TGF-β1 treatment, cells exhibited higher expression of p-Smad2/3, TGFBR2, FN, TGF-SMA and COL1A1 along with reduced cell proliferation. However,
BMSCs-originated exosomes or co-culture with BMSCs reversed these changes which could be inverted by exosome-specific inhibitors. In conclusion, BMSCs-originated exosomes and BMSCs exerted an anti-fibrosis effect, which was possibly through regulation of TGFβ1/Smad2/Smad3 signalling
pathway in endometrial stromal cells.
Collapse
Affiliation(s)
- Yilinuer Yiliyasi
- Department of Obstetrics and Gynecology, Beijing Changping District Hospital of Traditional Chinese Medicine, Beijing, 102200, China
| | - Kunduozi Wusainahong
- Department of Gynaecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, 830001, China
| | - Liping An
- Department of Obstetrics and Gynecology, Beijing Changping District Hospital of Traditional Chinese Medicine, Beijing, 102200, China
| | - Qinghua Liu
- Department of Obstetrics and Gynecology, Beijing Changping District Hospital of Traditional Chinese Medicine, Beijing, 102200, China
| | - ZeLing Fan
- Department of Obstetrics and Gynecology, Beijing Changping District Hospital of Traditional Chinese Medicine, Beijing, 102200, China
| | - JunHua Fan
- Department of Obstetrics and Gynecology, Beijing Changping District Hospital of Traditional Chinese Medicine, Beijing, 102200, China
| |
Collapse
|
39
|
Fadini GP, Albiero M. Impaired haematopoietic stem / progenitor cell traffic and multi-organ damage in diabetes. Stem Cells 2022; 40:716-723. [PMID: 35552468 PMCID: PMC9406601 DOI: 10.1093/stmcls/sxac035] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022]
Abstract
During antenatal development, hematopoietic stem/progenitor cells (HSPCs) arise from a specialized endothelium and migrate from the extraembryonic mesoderm to the fetal liver before establishing hematopoiesis in the bone marrow (BM). It is still debated whether, in adulthood, HSPCs display such ontologic overlap with vascular cells and capacity for endothelial differentiation. Yet, adult HSPCs retain a prominent migratory activity and traffic in the bloodstream to secondary lymphoid organs and all peripheral tissues, before eventually returning to the BM. While patrolling parenchymatous organs, HSPCs locate close to the vasculature, where they establish local hematopoietic islands and contribute to tissue homeostasis by paracrine signals. Solid evidence shows that diabetes mellitus jeopardizes the traffic of HSPCs from BM to the circulation and peripheral tissues, a condition called “mobilopathy.” A reduction in the levels of circulating HSPCs is the most immediate and apparent consequence, which has been consistently observed in human diabetes, and is strongly associated with future risk for multi-organ damage, including micro- and macro-angiopathy. But the shortage of HSPCs in the blood is only the visible tip of the iceberg. Abnormal HSPC traffic results from a complex interplay among metabolism, innate immunity, and hematopoiesis. Notably, mobilopathy is mechanistically connected with diabetes-induced myelopoiesis. Impaired traffic of HSPCs and enhanced generation of pro-inflammatory cells synergize for tissue damage and impair the resolution of inflammation. We herein summarize the current evidence that diabetes affects HSPC traffic, which are the causes and consequences of such alteration, and how it contributes to the overall disease burden.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
40
|
Sforza A, Vigorelli V, Rurali E, Perrucci GL, Gambini E, Arici M, Metallo A, Rinaldi R, Fiorina P, Barbuti A, Raucci A, Sacco E, Rocchetti M, Pompilio G, Genovese S, Vinci MC. Liraglutide preserves CD34+ stem cells from dysfunction Induced by high glucose exposure. Cardiovasc Diabetol 2022; 21:51. [PMID: 35397526 PMCID: PMC8994898 DOI: 10.1186/s12933-022-01486-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
Background Glucagon like peptide-1 receptor agonists (GLP-1RAs) have shown to reduce mortality and cardiovascular events in patients with type 2 diabetes mellitus (T2DM). Since the impairment in number and function of vasculotrophic circulating CD34+ hematopoietic stem progenitor cells (HSPCs) in T2D has been reported to increase cardiovascular (CV) risk, we hypothesized that one of the mechanisms whereby GLP-1 RAs exert CV protective effects may be related to the ability to improve CD34+ HSPC function. Methods In cord blood (CB)-derived CD34+ HSPC, the expression of GLP-1 receptor (GLP-1R) mRNA, receptor protein and intracellular signaling was evaluated by RT-qPCR and Western Blot respectively. CD34+ HSPCs were exposed to high glucose (HG) condition and GLP-1RA liraglutide (LIRA) was added before as well as after functional impairment. Proliferation, CXCR4/SDF-1α axis activity and intracellular ROS production of CD34+ HSPC were evaluated. Results CD34+ HSPCs express GLP-1R at transcriptional and protein level. LIRA treatment prevented and rescued HSPC proliferation, CXCR4/SDF-1α axis activity and metabolic imbalance from HG-induced impairment. LIRA stimulation promoted intracellular cAMP accumulation as well as ERK1/2 and AKT signaling activation. The selective GLP-1R antagonist exendin (9–39) abrogated LIRA-dependent ERK1/2 and AKT phosphorylation along with the related protective effects. Conclusion We provided the first evidence that CD34+ HSPC express GLP-1R and that LIRA can favorably impact on cell dysfunction due to HG exposure. These findings open new perspectives on the favorable CV effects of GLP-1 RAs in T2DM patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01486-9.
Collapse
|
41
|
Albiero M, D'Anna M, Bonora BM, Zuccolotto G, Rosato A, Giorgio M, Iori E, Avogaro A, Fadini GP. Hematopoietic and Nonhematopoietic p66Shc Differentially Regulates Stem Cell Traffic and Vascular Response to Ischemia in Diabetes. Antioxid Redox Signal 2022; 36:593-607. [PMID: 34538132 DOI: 10.1089/ars.2021.0097] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aims: Peripheral artery disease (PAD) is a severe complication of diabetes, characterized by defective traffic of hematopoietic stem/progenitor cells (HSPCs). We examined the hematopoietic versus nonhematopoietic role of p66Shc in regulating HSPC traffic and blood flow recovery after ischemia in diabetic mice. Results: Using streptozotocin-induced diabetes, chimeric mice with green fluorescent protein (GFP)+ bone marrow (BM), and the hind limb ischemia model, we found that the physiologic mobilization and homing of HSPCs were abolished by diabetes, along with impaired vascular recovery. Hematopoietic deletion of p66Shc, obtained by transplanting p66Shc-/- BM cells into wild-type (Wt) recipients, but not nonhematopoietic deletion, constrained hyperglycemia-induced myelopoiesis, rescued postischemic HSPC mobilization, and improved blood flow recovery in diabetic mice. In Wt diabetic mice transplanted with BM cells from GFP+p66Shc-/- mice, the amount of HSPCs homed to ischemic muscles was greater than in mice transplanted with GFP+p66Shc+/+ cells, with recruited cells displaying higher expression of adhesion molecules and Vegf. In 40 patients with diabetes, p66Shc gene expression in mononuclear cells was correlated with myelopoiesis and elevated in the presence of PAD. In 13 patients with diabetes and PAD, p66Shc expression in HSPC-mobilized peripheral blood cells was inversely correlated with VEGF expression. Innovation: For the first time, we dissect the role of hematopoietic versus nonhematopoietic p66Shc in regulating HSPC traffic and ischemic responses. Conclusion: Hematopoietic deletion of p66Shc was sufficient to rescue HSPC mobilization and homing in diabetes after ischemia and improved blood flow recovery. Inhibiting p66Shc in blood cells may be a novel strategy to counter PAD in diabetes. Antioxid. Redox Signal. 36, 593-607. Clinical Trial No.: NCT02790957.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marianna D'Anna
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | - Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gaia Zuccolotto
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
| | - Antonio Rosato
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy.,Veneto Institute of Oncology - IOV IRCCS, Padua, Italy
| | - Marco Giorgio
- European Institute of Oncology (IEO), Milan, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy.,Veneto Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
42
|
Bonora BM, Fogar P, Zuin J, Falaguasta D, Cappellari R, Cattelan A, Marinello S, Ferrari A, Avogaro A, Plebani M, Basso D, Fadini GP. Hyperglycemia, Reduced Hematopoietic Stem Cells, and Outcome of COVID-19. Diabetes 2022; 71:788-794. [PMID: 35061020 DOI: 10.2337/db21-0965] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/15/2022] [Indexed: 12/15/2022]
Abstract
Admission hyperglycemia has emerged worldwide as a predictor of poor coronavirus disease 2019 (COVID-19) outcome. Hyperglycemia leads to a defect in circulating hematopoietic stem/progenitor cells (HSPCs), which, in turn, predicts diabetic complications. Here, we explored whether reduced HSPCs mediated at least part of the prognostic effect of hyperglycemia on COVID-19 outcome. We found that patients with COVID-19 (n = 100) hospitalized in a nonintensive setting displayed dramatically (50-60%) reduced levels of HSPCs measured by flow cytometry as CD34+, CD34+CD45dim, or CD34+CD133+ cells, compared with control subjects (n = 595). This finding was highly significant (all P < 10-10) after multivariable adjustment, or manual 1:1 patient match, or propensity score matching. Admission hyperglycemia (≥7.0 mmol/L) was present in 45% of patients, was associated with a significant further ∼30% HSPCs reduction, and predicted a 2.6-fold increased risk of the primary outcome of adverse COVID-19 course (admittance to the intensive care unit or death). Low HSPCs were also associated with advanced age, higher peak C-reactive protein, and neutrophil-to-lymphocyte ratio. Independently from confounders, 1 SD lower CD34+ HSPCs was associated with a more than threefold higher risk of adverse outcome. Upon formal analysis, reduction of HSPCs was a significant mediator of the admission hyperglycemia on COVID-19 outcome, being responsible for 28% of its prognostic effect.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Paola Fogar
- Department of Medicine, University of Padova, Padua, Italy
| | - Jenny Zuin
- Department of Medicine, University of Padova, Padua, Italy
| | | | | | | | | | - Anna Ferrari
- Department of Medicine, University of Padova, Padua, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, Padua, Italy
| | - Mario Plebani
- Department of Medicine, University of Padova, Padua, Italy
| | - Daniela Basso
- Department of Medicine, University of Padova, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
43
|
Taylor GS, Shaw A, Scragg JH, Smith K, Campbell MD, McDonald TJ, Shaw JA, Ross MD, West DJ. Type 1 Diabetes Patients With Different Residual Beta-Cell Function but Similar Age, HBA1c, and Cardiorespiratory Fitness Have Differing Exercise-Induced Angiogenic Cell Mobilisation. Front Endocrinol (Lausanne) 2022; 13:797438. [PMID: 35222269 PMCID: PMC8874313 DOI: 10.3389/fendo.2022.797438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Many individuals with type 1 diabetes retain residual beta-cell function. Sustained endogenous insulin and C-peptide secretion is associated with reduced diabetes related complications, but underlying mechanisms remain unclear. Lower circulating numbers of endothelial and hematopoietic progenitor cells (EPCs and HPCs), and the inability to increase the count of these cells in response to exercise, are also associated with increased diabetes complications and cardiovascular disease. It is unknown whether residual beta-cell function influences HPCs and EPCs. Thus, this study examined the influence of residual beta-cell function in type 1 diabetes upon exercise-induced changes in haematopoietic (HPCs) and endothelial progenitor cells (EPCs). METHODS Participants with undetectable stimulated C-peptide (n=11; Cpepund), 10 high C-peptide (Cpephigh; >200 pmol/L), and 11 non-diabetes controls took part in this observational exercise study, completing 45 minutes of intensive walking at 60% V˙O2peak . Clinically significant HPCs (CD34+) and EPCs (CD34+VEGFR2+) phenotypes for predicting future adverse cardiovascular outcomes, and subsequent cell surface expression of chemokine receptor 4 (CXCR4) and 7 (CXCR7), were enumerated at rest and immediately post-exercise by flow cytometry. RESULTS Exercise increased HPCs and EPCs phenotypes similarly in the Cpephigh and control groups (+34-121% across phenotypes, p<0.04); but Cpepund group did not significantly increase from rest, even after controlling for diabetes duration. Strikingly, the post-exercise Cpepund counts were still lower than Cpephigh at rest. CONCLUSIONS Residual beta-cell function is associated with an intact exercise-induced HPCs and EPCs mobilisation. As key characteristics (age, fitness, HbA1c) were similar between groups, the mechanisms underpinning the absent mobilisation within those with negative C-peptide, and the vascular implications, require further investigation.
Collapse
Affiliation(s)
- Guy S. Taylor
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- *Correspondence: Daniel J. West, ; Guy S. Taylor,
| | - Andy Shaw
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jadine H. Scragg
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Kieran Smith
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Matthew D. Campbell
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, United Kingdom
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Timothy J. McDonald
- National Institute for Health Research Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
- Academic Department of Blood Sciences, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - James A. Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Newcastle Centre for Diabetes Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Mark D. Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Daniel J. West
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- *Correspondence: Daniel J. West, ; Guy S. Taylor,
| |
Collapse
|
44
|
Wang Y, Yao M, Wang J, Liu H, Zhang X, Zhao L, Hu X, Guan H, Lyu Z. Effects of Antidiabetic Drugs on Endothelial Function in Patients With Type 2 Diabetes Mellitus: A Bayesian Network Meta-Analysis. Front Endocrinol (Lausanne) 2022; 13:818537. [PMID: 35370959 PMCID: PMC8969579 DOI: 10.3389/fendo.2022.818537] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/14/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The changes of endothelial function in type 2 diabetes mellitus (T2DM) patients are closely associated with the development of cardiovascular disease (CVD). However, it is still unclear whether commonly used antidiabetic drugs can improve endothelial function. Flow-mediated dilation (FMD) is a noninvasive tool for evaluating endothelial function, which typically examines changes in the brachial artery diameter in response to ischemia using ultrasound. We performed a network meta-analysis (NMA) to explore the associations between changes in endothelial function and antidiabetic drugs by evaluating FMD in T2DM patients. METHODS We systematically searched several electronic databases for randomized controlled trials (RCTs) published from inception until January 25, 2022 with no language restriction. The primary outcome was FMD change in all studies, and we performed subgroup analysis in T2DM patients without CVD. NMA was performed to calculate the mean differences (MDs) with 95% confidence intervals (CIs). RESULTS From the 1,987 candidate articles identified in the initial search, 30 RCTs were eventually included in the analysis. In all studies, glucagon-like peptide-1 receptor (GLP-1R) agonists [MD = 3.70 (1.39-5.97)], TZD [MD = 1.96 (0.006-3.89)] produced improvement of FMD change compared to lifestyle intervention. GLP-1R agonists [MD = 3.33 (1.36-5.34) and MD = 3.30 (1.21-5.43)] showed significantly greater improvements in FMD change in pairwise comparisons with sulfonylureas and placebo. SGLT-2i also showed efficacy compared to sulfonylureas (MD = 1.89, 95% CI, 0.10, 3.75). In studies of T2DM patients without CVD, GLP-1R agonists [MD = 3.53 (1.24-5.76)], and TZD [MD = 2.30 (0.27-3.24)] produced improvements in FMD change compared to lifestyle treatment. GLP-1R agonists [MD = 3.25 (1.13-5.40), and MD = 3.85 (1.68-6.13)] showed significantly greater improvements in pairwise comparisons with sulfonylureas, and placebo. CONCLUSION In T2DM patients, both GLP-1R agonists, SGLT-2i and TZD have favorable effects to improve endothelial function in T2DM patients. In T2DM patients without CVD, GLP-1R agonists had a greater effect to improve endothelial function than sulfonylureas. These suggested that GLP-1R agonists are associated with significantly improved endothelial function in T2DM patients.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mingyan Yao
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jincheng Wang
- Department of Radiology, Peking University Cancer Hospital, Beijing, China
| | - Hongzhou Liu
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xuelian Zhang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ling Zhao
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaodong Hu
- Department of Endocrinology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Haixia Guan
- Department of Endocrinology Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Haixia Guan, ; Zhaohui Lyu,
| | - Zhaohui Lyu
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Haixia Guan, ; Zhaohui Lyu,
| |
Collapse
|
45
|
A Meta-Analysis of Randomized Controlled Trials on Therapeutic Efficacy and Safety of Autologous Cell Therapy for Atherosclerosis Obliterans. J Vasc Surg 2021; 75:1440-1449.e5. [PMID: 34788653 DOI: 10.1016/j.jvs.2021.10.051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Atherosclerosis obliterans (ASO) is a chronic occlusive arterial disease and the most common type of peripheral arterial disease. Current treatment options like medication and vascularization have limited effects for "no-option" patients, and stem cell therapy is considered a viable option although its application and efficacy have not been standardized. The objective of this review was to assess the safety and efficacy of autologous stem cell therapy in patients with ASO. METHODS We performed a literature search of published RCTs for ASO patients receiving stem cell therapy without a revascularization option. PubMed, Embase, and the Cochrane Library were searched. This study was conducted by a pair of authors independently and audited by a third author. Data were synthesized with a random-effect model. RESULTS 630 patients in 12 RCTs were included. The results showed that cell therapy significantly improved total amputation (RR: 0.64, p = 0.004, 95% CI: [0.47, 0.87]), major amputation (RR: 0.69, p = 0.02, 95% CI: [0.50, 0.94]), ankle-brachial index (ABI) (MD = 0.08, p = 0.004, 95% CI: [0.02, 0.13]), transcutaneous oxygen tension (TcO2) (MD = 11.52, p = 0.004, 95% CI: [3.60, 19.43]) and rest pain score (MD = -0.64, p = 0.007, 95% CI: [-1.10, -0.17]) compared to placebo or standard care. However, current studies showed cell therapy was not superior to placebo or standard care in all-cause death (RR: 0.75, p = 0.34, 95% CI: [0.41, 1.36]) and ulcer size (MD = -8.85, p = 0.39, CI: [-29.05,11.36]). LIMITATION The number of trials included was limited. Moreover, most trials were designed for "no-option" patients and thus the results should be applied with caution to other PAD patients. CONCLUSION ASO patients can benefit from autologous cell therapy in limb salvage, limb blood perfusion, and rest pain alleviation.
Collapse
|
46
|
Wu W, Zhang J, Shao L, Huang H, Meng Q, Shen Z, Teng X. Evaluation of Circulating Endothelial Progenitor Cells in Abdominal Aortic Aneurysms after Endovascular Aneurysm Repair. Int J Stem Cells 2021; 15:136-143. [PMID: 34711694 PMCID: PMC9148833 DOI: 10.15283/ijsc21027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/11/2021] [Accepted: 08/02/2021] [Indexed: 11/10/2022] Open
Abstract
Background and Objectives Circulating endothelial progenitor cells (EPCs) participate in vascular repair and predict cardiovascular outcomes. The aim of this study was to investigate the correlation between EPCs and abdominal aortic aneurysms (AAAs). Methods and Results Patients (age 67±9.41 years) suffering from AAAs (aortic diameters 58.09±11.24 mm) were prospectively enrolled in this study. All patients received endovascular aneurysm repair (EVAR). Blood samples were taken preoperatively and 14 days after surgery from patients with aortic aneurysms. Samples were also obtained from age-matched control subjects. Circulating EPCs were defined as those cells that were double positive for CD34 and CD309. Rat models of AAA formation were generated by the peri-adventitial elastase application of either saline solution (control; n=10), or porcine pancreatic elastase (PPE; n=14). The aortas were analyzed using an ultrasonic video system and immunohistochemistry. The levels of CD34+/CD309+ cells in the peripheral blood mononuclear cell populations were measured by flow cytometry. The baseline numbers of circulating EPCs (CD34+/CD309+) in the peripheral blood were significantly smaller in AAA patients compared with control subjects. The number of EPCs doubled by the 14th day after EVAR. A total of 78.57% of rats in the PPE group (11/14) formed AAAs (dilation ratio >150%). The numbers of EPCs from defined AAA rats were significantly decreased compared with the control group. Conclusions EPC levels may be useful for monitoring abdominal aorta aneurysms and rise after EVAR in patients with aortic aneurysms, and might contribute to the rapid endothelialization of vessels.
Collapse
Affiliation(s)
- Weihua Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China.,Center of Clinical Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinlong Zhang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Haoyue Huang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Qingyou Meng
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Xiaomei Teng
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
47
|
Camps-Renom P, Jiménez-Xarrié E, Soler M, Puig N, Aguilera-Simón A, Marín R, Prats-Sánchez L, Delgado-Mederos R, Martínez-Domeño A, Guisado-Alonso D, Guasch-Jiménez M, Martí-Fàbregas J. Endothelial Progenitor Cells Count after Acute Ischemic Stroke Predicts Functional Outcome in Patients with Carotid Atherosclerosis. J Stroke Cerebrovasc Dis 2021; 30:106144. [PMID: 34649037 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES Circulating Endothelial Progenitor Cells (EPCs) predict cardiovascular outcomes in patients with coronary disease. However, the predictive value of EPCs after ischemic stroke is not well established. We aimed to study the prognostic role of EPCs in patients with acute ischemic stroke and carotid atherosclerosis, focusing on post-stroke functional outcome and stroke recurrences. MATERIALS AND METHODS We studied consecutive adult patients with an acute (<7 days) anterior circulation ischemic stroke and carotid atherosclerosis. Cardioembolic strokes were excluded. We measured circulating EPCs by flow cytometry (CD34+/CD133+/KDR+) at inclusion (7±1 days after stroke) and at one year of follow-up. At three months and at one year we registered the modified Rankin Scale score, stroke recurrences and coronary syndromes during the follow-up. RESULTS We studied 80 patients with a mean age of 74.3±10.4 years. We divided the population in tertiles according to the EPCs count. At three months we observed a favorable outcome in 25/36 (69.4%) patients in the lowest, 19/22 (86.4%) in the medium and 21/22 (95.5%) in the highest tercile (p=0.037). In the multivariable analysis a higher EPCs count was associated with favorable functional outcome after adjusting for age and baseline NIHSS score (OR=3.61, 95%CI 1.34-9.76; p=0.011). This association persisted at one year of follow-up. We did not find association between counts of EPCs and stroke recurrence. CONCLUSIONS In patients with acute ischemic stroke and carotid atherosclerosis, a higher count of EPCs was associated with favorable functional outcome in the mid and long-term follow-up. Counts of EPCs did not predict stroke recurrences.
Collapse
Affiliation(s)
- Pol Camps-Renom
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain..
| | - Elena Jiménez-Xarrié
- Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Marta Soler
- Facility of Cytometry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona, Spain
| | - Núria Puig
- Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Ana Aguilera-Simón
- Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Rebeca Marín
- Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Luis Prats-Sánchez
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Raquel Delgado-Mederos
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Alejandro Martínez-Domeño
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Daniel Guisado-Alonso
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Marina Guasch-Jiménez
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Joan Martí-Fàbregas
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| |
Collapse
|
48
|
Guyonnet L, Detriché G, Gendron N, Philippe A, Latremouille C, Soret L, Capel A, Peronino C, Jansen P, Ivak P, Carpentier A, Mirault T, Netuka I, Guerin CL, Smadja DM. Elevated Circulating Stem Cells Level is Observed One Month After Implantation of Carmat Bioprosthetic Total Artificial Heart. Stem Cell Rev Rep 2021; 17:2332-2337. [PMID: 34622384 DOI: 10.1007/s12015-021-10270-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2021] [Indexed: 11/26/2022]
Abstract
The Aeson® total artificial heart (A-TAH) has been developed as a total heart replacement for patients at risk of death from biventricular failure. We previously described endothelialization of the hybrid membrane inside A-TAH probably at the origin of acquired hemocompatibility. We aimed to quantify vasculogenic stem cells in peripheral blood of patients with long-term A-TAH implantation. Four male adult patients were included in this study. Peripheral blood mononuclear cells were collected before A-TAH implantation (T0) and after implantation at one month (T1), between two and five months (T2), and then between six and twelve months (T3). Supervised analysis of flow cytometry data confirmed the presence of the previously identified Lin-CD133+CD45- and Lin-CD34+ with different CD45 level intensities. Lin-CD133+CD45-, Lin-CD34+CD45- and Lin-CD34+CD45+ were not modulated after A-TAH implantation. However, we demonstrated a significant mobilization of Lin-CD34+CD45dim (p = 0.01) one month after A-TAH implantation regardless of the expression of CD133 or c-Kit. We then visualized data for the resulting clusters on a uniform manifold approximation and projection (UMAP) plot showing all single cells of the live Lin- and CD34+ events selected from down sampled files concatenated at T0 and T1. The three clusters upregulated at T1 are CD45dim clusters, confirming our results. In conclusion, using a flow cytometry approach, we demonstrated in A-TAH-transplanted patients a significant mobilization of Lin-CD34+CD45dim in peripheral blood one month after A-TAH implantation. Using a flow cytometry approach, we demonstrated in A-TAH transplanted patients a significant mobilization of Lin-CD34+CD45dim in peripheral blood one month after A-TAH implantation. This cell population could be at the origin of newly formed endothelial cells on top of hybrid membrane in Carmat bioprosthetic total artificial heart.
Collapse
Affiliation(s)
- Léa Guyonnet
- Institut Curie, Cytometry Platform, 75006, Paris, France
| | - Grégoire Detriché
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, 75006, Paris, France
- Vascular Medicine Department and Georges Pompidou European Hospital, AP-HP, 75015, Paris, France
| | - Nicolas Gendron
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, 75006, Paris, France
- Hematology and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Georges Pompidou European Hospital, 75015, Paris, France
| | - Aurélien Philippe
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, 75006, Paris, France
- Hematology and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Georges Pompidou European Hospital, 75015, Paris, France
| | | | - Lou Soret
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, 75006, Paris, France
- Hematology and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Georges Pompidou European Hospital, 75015, Paris, France
| | | | - Christophe Peronino
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, 75006, Paris, France
- Hematology and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Georges Pompidou European Hospital, 75015, Paris, France
- Carmat SA, Vélizy-Villacoublay, France
| | | | - Peter Ivak
- Department of Cardiovascular Surgery, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Alain Carpentier
- Cardiac Surgery Department and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Georges Pompidou European Hospital, Université de Paris, 75015, Paris, France
| | - Tristan Mirault
- Vascular Medicine Department and Georges Pompidou European Hospital, AP-HP, 75015, Paris, France
- PARCC, INSERM, Université de Paris, 75015, Paris, France
| | - Ivan Netuka
- Department of Cardiovascular Surgery, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Coralie L Guerin
- Institut Curie, Cytometry Platform, 75006, Paris, France
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, 75006, Paris, France
| | - David M Smadja
- Innovative Therapies in Haemostasis, INSERM, Université de Paris, 75006, Paris, France.
- Hematology and Biosurgical Research Lab (Carpentier Foundation), AP-HP, Georges Pompidou European Hospital, 75015, Paris, France.
| |
Collapse
|
49
|
Kröpfl JM. Circulating progenitor cells as predictor of mortality in cardiovascular disease: Could physical activity change the global outcome? Atherosclerosis 2021; 333:83-84. [PMID: 34391572 DOI: 10.1016/j.atherosclerosis.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Julia M Kröpfl
- University of Basel, Department of Sport, Exercise and Health, Division of Sports and Exercise Medicine, Birsstrasse 320 B, CH-4052, Basel, Switzerland.
| |
Collapse
|
50
|
Mause SF, Ritzel E, Deck A, Vogt F, Liehn EA. Endothelial Progenitor Cells Modulate the Phenotype of Smooth Muscle Cells and Increase Their Neointimal Accumulation Following Vascular Injury. Thromb Haemost 2021; 122:456-469. [PMID: 34214997 DOI: 10.1055/s-0041-1731663] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Smooth muscle cells (SMCs) are the main driver of neointima formation and restenosis following vascular injury. In animal models, endothelial progenitor cells (EPCs) accelerate endothelial regeneration and reduce neointima formation after arterial injury; however, EPC-capture stents do not reduce target vessel failure compared with conventional stents. Here we examined the influence of EPCs on features of SMCs pivotal for their impact on injury-induced neointima formation including proliferation, migration, and phenotype switch. METHODS AND RESULTS EPCs, their conditioned medium, and EPC-derived microparticles induced proliferation of SMCs while limiting their apoptosis. In transwell membrane experiments and scratch assays, EPCs stimulated migration of SMCs and accelerated their recovery from scratch-induced injury. Treatment of SMCs with an EPC-derived conditioned medium or microparticles triggered transformation of SMCs toward a synthetic phenotype. However, co-cultivation of EPCs and SMCs enabling direct cell-cell contacts preserved their original phenotype and protected from the transformative effect of SMC cholesterol loading. Adhesion of EPCs to SMCs was stimulated by SMC injury and reduced by blocking CXCR2 and CCR5. Interaction of EPCs with SMCs modulated their secretory products and synergistically increased the release of selected chemokines. Following carotid wire injury in athymic mice, injection of EPCs resulted not only in reduced neointima formation but also in altered cellular composition of the neointima with augmented accumulation of SMCs. CONCLUSION EPCs stimulate proliferation and migration of SMCs and increase their neointimal accumulation following vascular injury. Furthermore, EPCs context-dependently modify the SMC phenotype with protection from the transformative effect of cholesterol when a direct cell-cell contact is established.
Collapse
Affiliation(s)
- Sebastian F Mause
- Department of Internal Medicine I, Cardiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Elisabeth Ritzel
- Department of Otorhinolaryngology Head and Neck Surgery, Klinikum Stuttgart, Stuttgart, Germany.,Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Annika Deck
- Department of Internal Medicine I, Cardiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Felix Vogt
- Department of Internal Medicine I, Cardiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Elisa A Liehn
- Department of Internal Medicine I, Cardiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany.,Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| |
Collapse
|