1
|
Fadini GP. Do epigenetic echoes of gestational diabetes craft a transgenerational path to cardio-metabolic disease? Eur Heart J 2024; 45:5186-5188. [PMID: 39523019 DOI: 10.1093/eurheartj/ehae719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
2
|
Wang Z, Li R, Yang G, Wang Y. Cancer stem cell biomarkers and related signalling pathways. J Drug Target 2024; 32:33-44. [PMID: 38095181 DOI: 10.1080/1061186x.2023.2295222] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/10/2023] [Indexed: 12/20/2023]
Abstract
Cancer stem cells (CSCs) represent a distinct subset of neoplastic cells characterised by their heightened capacity for tumorigenesis. These cells are implicated in the facilitation of cancer metastasis, recurrence, and resistance to conventional therapeutic interventions. Extensive scientific research has been devoted to the identification of biomarkers and the elucidation of molecular mechanisms in order to improve targeted therapeutic approaches. Accurate identification of cancer stem cells based on biomarkers can provide a theoretical basis for drug combinations of malignant tumours. Targeted biomarker-based therapies also offer a silver lining for patients with advanced malignancies. This review aims comprehensively to consolidate the latest findings on CSCs biomarkers, targeted agents as well as biomarkers associated signalling pathways in well-established cancer types, thereby contributing to improved prognostic outcomes.
Collapse
Affiliation(s)
- Zhe Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Department of Infectious Disease, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Rui Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Guilin Yang
- Department of Infectious Disease, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yijin Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
3
|
Vaccarino V, Bremner JD. Stress and cardiovascular disease: an update. Nat Rev Cardiol 2024; 21:603-616. [PMID: 38698183 DOI: 10.1038/s41569-024-01024-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/08/2024] [Indexed: 05/05/2024]
Abstract
Psychological stress is generally accepted to be associated with an increased risk of cardiovascular disease (CVD), but results have varied in terms of how stress is measured and the strength of the association. Additionally, the mechanisms and potential causal links have remained speculative despite decades of research. The physiological responses to stress are well characterized, but their contribution to the development and progression of CVD has received little attention in empirical studies. Evidence suggests that physiological responses to stress have a fundamental role in the risk of CVD and that haemodynamic, vascular and immune perturbations triggered by stress are especially implicated. Stress response physiology is regulated by the corticolimbic regions of the brain, which have outputs to the autonomic nervous system. Variation in these regulatory pathways might explain interindividual differences in vulnerability to stress. Dynamic perturbations in autonomic, immune and vascular functions are probably also implicated as CVD risk mechanisms of chronic, recurring and cumulative stressful exposures, but more data are needed from prospective studies and from assessments in real-life situations. Psychological assessment remains insufficiently recognized in clinical care and prevention. Although stress-reduction interventions might mitigate perceived stress levels and potentially reduce cardiovascular risk, more data from randomized trials are needed.
Collapse
Affiliation(s)
- Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
| | - J Douglas Bremner
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Department of Radiology and Diagnostic Imaging, Emory University School of Medicine, Atlanta, GA, USA
- Veterans Administration Medical Center, Decatur, GA, USA
| |
Collapse
|
4
|
Tan C, Chen J, Tu T, Chen L, Zou J. Lycopene inhibits pyroptosis of endothelial progenitor cells induced by ox-LDL through the AMPK/mTOR/NLRP3 pathway. Open Med (Wars) 2024; 19:20240973. [PMID: 38919547 PMCID: PMC11197008 DOI: 10.1515/med-2024-0973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/16/2024] [Accepted: 05/03/2024] [Indexed: 06/27/2024] Open
Abstract
The malfunction of endothelial progenitor cells (EPCs) due to ox-LDL is a risk contributor for arteriosclerotic disease. Meanwhile, lycopene possesses anti-inflammatory and antioxidative qualities. This investigation aimed to determine if lycopene can protect EPCs from ox-LDL-induced damage and to elucidate the underlying mechanism. The effects of lycopene on the survival, migration, and tube-forming capacity of EPCs were determined via in vitro assays. Expression of proteins related to pyroptosis and cellular proteins related to AMPK/mTOR/NLRP3 signaling was determined by western blot/flow cytometry. Our results demonstrated that lycopene treatment significantly enhanced proliferation, tube formation, and migration of EPCs stimulated by ox-LDL. Additionally, lycopene was found to suppress pyroptosis in ox-LDL-induced EPCs through the activation of AMPK, which led to the inhibition of mTOR phosphorylation and subsequent downregulation of the downstream NLRP3 inflammasome. In summary, our study suggests that lycopene mitigates ox-LDL-induced dysfunction in EPCs and inhibits pyroptosis via AMPK/mTOR/NLRP3 signaling. Our study suggests that lycopene may act as promising therapies for preventing atherosclerosis.
Collapse
Affiliation(s)
- Chujun Tan
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Junqiu Chen
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, 518057, China
| | - Tengcan Tu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Lifang Chen
- Department of Cardiology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 528200, China
| | - Jun Zou
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528200, China
| |
Collapse
|
5
|
Liou YJ, Chen MH, Hsu JW, Huang KL, Huang PH, Bai YM. Dysfunction of circulating endothelial progenitor cells in major depressive disorder. Acta Neuropsychiatr 2024; 36:153-161. [PMID: 38178721 DOI: 10.1017/neu.2023.49] [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] [Indexed: 01/06/2024]
Abstract
OBJECTIVES Despite mounting evidence demonstrates circulating endothelial progenitor cells (cEPCs) quantitative changes in depression, no study has investigated cEPC functions in major depressive disorder (MDD). We investigated the role of cEPC adhesive and apoptotic functions in MDD. METHODS We recruited 68 patients with MDD and 56 healthy controls (HCs). The depression symptoms, anxiety, psychosomatic symptoms, subjective cognitive dysfunction, quality of life, and functional disability were evaluated using the Hamilton Depression Rating Scale and Montgomery-Åsberg Depression Rating Scale, Hamilton Anxiety Rating Scale, Depression and Somatic Symptoms Scale (DSSS), Perceived Deficits Questionnaire-Depression, 12-Item Short Form Health Survey (SF-12), and Sheehan Disability Scale (SDS), respectively. Working memory and executive function were assessed using a 2-back task and Wisconsin Card Sorting Test (WCST). Inflammatory marker (soluble interleukin-6 receptor, C-reactive protein, and tumor necrosis factor-α receptor-1), cEPC adhesive, and apoptotic levels were measured using in vitro assays. RESULTS The MDD patients showed significantly lower cEPC adhesive levels than the HCs, and this difference in adhesive function remained statistically significant even after adjusting for inflammatory marker levels. The cEPC adhesion levels were in inverse correlations with commission and omission errors in 2-back task, the percent perseverative response and percent perseverative errors in WCST, and the DSSS and SDS scores, but in positive correlations with SF-12 physical and mental component scores. cEPC apoptotic levels did not differ significantly between the groups. CONCLUSION The findings indicate that cEPC adhesive function is diminished in MDD and impacts various aspects of cognitive and psychosocial functions associated with the disorder.
Collapse
Affiliation(s)
- Ying-Jay Liou
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ju-Wei Hsu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kai-Lin Huang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
6
|
Bonora BM, Marassi M, Fogar P, Zuin J, Cappellari R, Marinello S, Ferrari A, Cattelan A, Avogaro A, Basso D, Fadini GP. Circulating haematopoietic stem cells and long-term outcomes of COVID-19. Eur J Clin Invest 2024; 54:e14150. [PMID: 38088242 DOI: 10.1111/eci.14150] [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: 10/01/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND AIMS An acute depletion of circulating haematopoietic stem/progenitor cells (HSPCs) occurs during COVID-19, especially among patients with a poorer disease course. We herein examined whether HSPCs levels at hospital admission for COVID-19 predict 1-year mortality and the long-COVID syndrome. MATERIALS AND METHODS Patients hospitalized for COVID-19 in an infectious disease ward were consecutively enrolled. Circulating HSPC levels were assessed by flow cytometry as cells expressing CD34 and/or CD133. Follow-up was performed for 12 months after hospitalization through the review of electronic medical records and demographic local registers. RESULTS The study included 100 patients, 36 of whom reported symptoms of long-COVID and 20 died during follow-up. The reduction of 1-SD of HSPCs was associated with a 3- to 5-fold increase in the risk of 1-year mortality. Age, admission hyperglycaemia, C-reactive protein peak, liver enzymes, the need of high-flow oxygen and/or invasive ventilation were predictors of mortality at univariate analysis. Among pre-existing comorbidities, coronary heart disease and chronic kidney disease, but not diabetes, were associated with 1-year mortality. In multivariate analyses, HSPCs remained significantly associated with 1-year mortality independently of confounders. The development of pneumonia an in-hospital treatment with glucocorticoids and convalescent plasma were associated with long-COVID symptoms at follow-up. HSPCs, diabetes and other comorbidities were not predictors of long-COVID. CONCLUSIONS In a cohort of patients hospitalized for COVID-19, lower HSPC levels at the time of admission were independent predictors of 1-year mortality. However, COVID-19 severity, but not HSPC level, was significantly associated with the development of long-COVID symptoms.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University Hospital of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Marella Marassi
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Paola Fogar
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Jenny Zuin
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | | | - Serena Marinello
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Anna Ferrari
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Daniela Basso
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University Hospital of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
7
|
Krauz K, Kempiński M, Jańczak P, Momot K, Zarębiński M, Poprawa I, Wojciechowska M. The Role of Epicardial Adipose Tissue in Acute Coronary Syndromes, Post-Infarct Remodeling and Cardiac Regeneration. Int J Mol Sci 2024; 25:3583. [PMID: 38612394 PMCID: PMC11011833 DOI: 10.3390/ijms25073583] [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: 02/15/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Epicardial adipose tissue (EAT) is a fat deposit surrounding the heart and located under the visceral layer of the pericardium. Due to its unique features, the contribution of EAT to the pathogenesis of cardiovascular and metabolic disorders is extensively studied. Especially, EAT can be associated with the onset and development of coronary artery disease, myocardial infarction and post-infarct heart failure which all are significant problems for public health. In this article, we focus on the mechanisms of how EAT impacts acute coronary syndromes. Particular emphasis was placed on the role of inflammation and adipokines secreted by EAT. Moreover, we present how EAT affects the remodeling of the heart following myocardial infarction. We further review the role of EAT as a source of stem cells for cardiac regeneration. In addition, we describe the imaging assessment of EAT, its prognostic value, and its correlation with the clinical characteristics of patients.
Collapse
Affiliation(s)
- Kamil Krauz
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (K.K.); (M.K.); (P.J.); (K.M.)
| | - Marcel Kempiński
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (K.K.); (M.K.); (P.J.); (K.M.)
| | - Paweł Jańczak
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (K.K.); (M.K.); (P.J.); (K.M.)
| | - Karol Momot
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (K.K.); (M.K.); (P.J.); (K.M.)
| | - Maciej Zarębiński
- Department of Invasive Cardiology, Independent Public Specialist Western Hospital John Paul II, Lazarski University, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland; (M.Z.); (I.P.)
| | - Izabela Poprawa
- Department of Invasive Cardiology, Independent Public Specialist Western Hospital John Paul II, Lazarski University, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland; (M.Z.); (I.P.)
| | - Małgorzata Wojciechowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (K.K.); (M.K.); (P.J.); (K.M.)
| |
Collapse
|
8
|
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
|
9
|
Desai SR, Ko YA, Liu C, Hafeez Z, Park J, Faaborg-Andersen C, Alvi Z, Alras Z, Alkhoder AA, Martini A, Varughese A, Ejaz K, Cheung B, Wang M, Gold DA, Gold ME, Jain V, Vatsa N, Islam SJ, Almuwaqqat Z, Dhindsa DS, Mehta A, Kim JH, Wilson P, Waller EK, Vaccarino V, Quyyumi AA. Vitamin D Deficiency, Inflammation, and Diminished Endogenous Regenerative Capacity in Coronary Heart Disease. JACC. ADVANCES 2024; 3:100804. [PMID: 38939377 PMCID: PMC11198268 DOI: 10.1016/j.jacadv.2023.100804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/13/2023] [Accepted: 11/10/2023] [Indexed: 06/29/2024]
Abstract
Background Vitamin D deficiency (VDD) is associated with coronary heart disease (CHD) and poor outcomes, but supplementation does not improve prognosis. VDD has been implicated in and may promote greater risk through inflammation and impaired progenitor cell function. Objectives The authors examined VDD, high-sensitivity C-reactive protein (hsCRP), circulating progenitor cell (CPC) counts, and outcomes in patients with CHD. They hypothesized that the higher risk with VDD is mediated by inflammation and impaired regenerative capacity. Methods A total of 5,452 individuals with CHD in the Emory Cardiovascular Biobank had measurement of 25-hydroxyvitamin D, subsets of whom had hsCRP measurements and CPCs estimated as CD34-expressing mononuclear cell counts. Findings were validated in an independent cohort. 25-hydroxyvitamin D <20 ng/mL was considered VDD. Cox and Fine-Gray models determined associations between marker levels and: 1) all-cause mortality; 2) cardiovascular mortality; and 3) major adverse cardiovascular events, a composite of adverse CHD outcomes. Results VDD (43.6% of individuals) was associated with higher adjusted cardiovascular mortality (HR: 1.57, 95% CI: 1.09-2.28). There were significant interactions between VDD and hsCRP and CPC counts in predicting cardiovascular mortality. Individuals with both VDD and elevated hsCRP had the greatest risk (HR: 2.82, 95% CI: 2.16-3.67). Only individuals with both VDD and low CPC counts were at high risk (HR: 2.25, 95% CI: 1.46-3.46). These findings were reproduced in the validation cohort. Conclusions VDD predicts adverse outcomes in CHD. Those with VDD, inflammation and/or diminished regenerative capacity are at a significantly greater risk of cardiovascular mortality. Whether targeted supplementation in these high-risk groups improves risk warrants further study.
Collapse
Affiliation(s)
- Shivang R. Desai
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yi-An Ko
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Chang Liu
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Zaki Hafeez
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Christian Faaborg-Andersen
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Zain Alvi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zahran Alras
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ayman A. Alkhoder
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Afif Martini
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anil Varughese
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kiran Ejaz
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Brian Cheung
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maggie Wang
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Daniel A. Gold
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Matthew E. Gold
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vardhmaan Jain
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nishant Vatsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shabatun J. Islam
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zakaria Almuwaqqat
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Devinder S. Dhindsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anurag Mehta
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- VCU Health Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Jonathan H. Kim
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Peter Wilson
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Edmund K. Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Viola Vaccarino
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Arshed A. Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
10
|
Altabas V, Marinković Radošević J, Špoljarec L, Uremović S, Bulum T. The Impact of Modern Anti-Diabetic Treatment on Endothelial Progenitor Cells. Biomedicines 2023; 11:3051. [PMID: 38002051 PMCID: PMC10669792 DOI: 10.3390/biomedicines11113051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Diabetes is one of the leading chronic diseases globally with a significant impact on mortality. This condition is associated with chronic microvascular and macrovascular complications caused by vascular damage. Recently, endothelial progenitor cells (EPCs) raised interest due to their regenerative properties. EPCs are mononuclear cells that are derived from different tissues. Circulating EPCs contribute to regenerating the vessel's intima and restoring vascular function. The ability of EPCs to repair vascular damage depends on their number and functionality. Diabetic patients have a decreased circulating EPC count and impaired EPC function. This may at least partially explain the increased risk of diabetic complications, including the increased cardiovascular risk in these patients. Recent studies have confirmed that many currently available drugs with proven cardiovascular benefits have beneficial effects on EPC count and function. Among these drugs are also medications used to treat different types of diabetes. This manuscript aims to critically review currently available evidence about the ways anti-diabetic treatment affects EPC biology and to provide a broader context considering cardiovascular complications. The therapies that will be discussed include lifestyle adjustments, metformin, sulphonylureas, gut glucosidase inhibitors, thiazolidinediones, dipeptidyl peptidase 4 inhibitors, glucagon-like peptide 1 receptor analogs, sodium-glucose transporter 2 inhibitors, and insulin.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Jelena Marinković Radošević
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
| | - Lucija Špoljarec
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
| | | | - Tomislav Bulum
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
- Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, 10000 Zagreb, Croatia
| |
Collapse
|
11
|
Bakbak E, Krishnaraj A, Park B, Verma S, Hess DA. Vascular regenerative cells in cardiometabolic disease. Curr Opin Cardiol 2023; 38:546-551. [PMID: 37668181 DOI: 10.1097/hco.0000000000001089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
PURPOSE OF REVIEW This review will provide an overview of the recent literature linking the pathophysiology of cardiometabolic disease with the depletion and dysfunction of circulating vascular regenerative (VR) cell content. Moreover, we provide rationale for the use of VR cells as a biomarker for cardiovascular risk and the use of pharmacological agents to improve VR cell content. RECENT FINDINGS Recent studies demonstrate the potential of VR cells as a biomarker of cardiovascular risk and as a therapeutic target. Notably, lipid-lowering agents, antihyperglycemic therapies such as sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonists, as well as exercise and weight loss, have all been found to improve VR cell content, providing mechanistic evidence supporting a role in mitigating adverse cardiovascular outcomes in people with cardiometabolic-based disease. SUMMARY The importance of VR cells as a biomarker in assessing cardiovascular risk is becoming increasingly apparent. This review highlights recent literature supporting the accurate use of VR cell characterization to monitor the capacity for vessel repair and novel strategies to improve vessel health. Future research is required to validate and optimize these emerging approaches.
Collapse
Affiliation(s)
- Ehab Bakbak
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital
- Department of Pharmacology and Toxicology
| | - Aishwarya Krishnaraj
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital
- Department of Pharmacology and Toxicology
| | - Brady Park
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital
- Department of Pharmacology and Toxicology
| | - Subodh Verma
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital
- Department of Pharmacology and Toxicology
- Department of Surgery, University of Toronto, Toronto
| | - David A Hess
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital
- Department of Pharmacology and Toxicology
- Department of Physiology and Pharmacology, Western University, London
- Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| |
Collapse
|
12
|
Pelliccia F, Zimarino M, Niccoli G, Morrone D, De Luca G, Miraldi F, De Caterina R. In-stent restenosis after percutaneous coronary intervention: emerging knowledge on biological pathways. EUROPEAN HEART JOURNAL OPEN 2023; 3:oead083. [PMID: 37808526 PMCID: PMC10558044 DOI: 10.1093/ehjopen/oead083] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/24/2023] [Accepted: 08/15/2023] [Indexed: 10/10/2023]
Abstract
Percutaneous coronary intervention (PCI) has evolved significantly over the past four decades. Since its inception, in-stent restenosis (ISR)-the progressive reduction in vessel lumen diameter after PCI-has emerged as the main complication of the procedure. Although the incidence of ISR has reduced from 30% at 6 months with bare-metal stents to 7% at 4 years with drug-eluting stents (DESs), its occurrence is relevant in absolute terms because of the dimensions of the population treated with PCI. The aim of this review is to summarize the emerging understanding of the biological pathways that underlie ISR. In-stent restenosis is associated with several factors, including patient-related, genetic, anatomic, stent, lesion, and procedural characteristics. Regardless of associated factors, there are common pathophysiological pathways involving molecular phenomena triggered by the mechanical trauma caused by PCI. Such biological pathways are responses to the denudation of the intima during balloon angioplasty and involve inflammation, hypersensitivity reactions, and stem cell mobilization particularly of endothelial progenitor cells (EPCs). The results of these processes are either vessel wall healing or neointimal hyperplasia and/or neo-atherosclerosis. Unravelling the key molecular and signal pathways involved in ISR is crucial to identify appropriate therapeutic strategies aimed at abolishing the 'Achille's heel' of PCI. In this regard, we discuss novel approaches to prevent DES restenosis. Indeed, available evidence suggests that EPC-capturing stents promote rapid stent re-endothelization, which, in turn, has the potential to decrease the risk of stent thrombosis and allow the use of a shorter-duration dual antiplatelet therapy.
Collapse
Affiliation(s)
- Francesco Pelliccia
- Department of Cardiovascular Sciences, University Sapienza, Viale del Policlinico 155, 00161 Rome, Italy
| | - Marco Zimarino
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University, Viale Abruzzo, 332, 66100 Chieti, Italy
- Department of Cardiology, “SS. Annunziata Hospital”, ASL 2 Abruzzo, Via dei Vestini, 66100 Chieti, Italy
| | - Giampaolo Niccoli
- Department of Cardiology, University of Parma, Piazzale S. Francesco, 3, 43121 Parma, Italy
| | - Doralisa Morrone
- Department of Surgical, Medical and Molecular Pathology and of Critical Sciences, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy
| | - Giuseppe De Luca
- Division of Cardiology, AOU “Policlinico G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy
- Division of Cardiology, IRCCS Hospital Galeazzi-Sant'Ambrogio, Via Cristina Belgioioso 173, 20157 Milan, Italy
| | - Fabio Miraldi
- Department of Cardiovascular Sciences, University Sapienza, Viale del Policlinico 155, 00161 Rome, Italy
| | - Raffaele De Caterina
- Department of Surgical, Medical and Molecular Pathology and of Critical Sciences, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy
| |
Collapse
|
13
|
Forman DE, Kuchel GA, Newman JC, Kirkland JL, Volpi E, Taffet GE, Barzilai N, Pandey A, Kitzman DW, Libby P, Ferrucci L. Impact of Geroscience on Therapeutic Strategies for Older Adults With Cardiovascular Disease: JACC Scientific Statement. J Am Coll Cardiol 2023; 82:631-647. [PMID: 37389519 PMCID: PMC10414756 DOI: 10.1016/j.jacc.2023.05.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023]
Abstract
Geroscience posits that cardiovascular disease (CVD) and other chronic diseases result from progressive erosion of the effectiveness of homeostatic mechanisms that oppose age-related accumulation of molecular damage. This hypothetical common root to chronic diseases explains why patients with CVD are often affected by multimorbidity and frailty and why older age negatively affects CVD prognosis and treatment response. Gerotherapeutics enhance resilience mechanisms that counter age-related molecular damage to prevent chronic diseases, frailty, and disability, thereby extending healthspan. Here, we describe the main resilience mechanisms of mammalian aging, with a focus on how they can affect CVD pathophysiology. We next present novel gerotherapeutic approaches, some of which are already used in management of CVD, and explore their potential to transform care and management of CVD. The geroscience paradigm is gaining traction broadly in medical specialties, with potential to mitigate premature aging, reduce health care disparities, and improve population healthspan.
Collapse
Affiliation(s)
- Daniel E Forman
- Department of Medicine (Geriatrics and Cardiology) University of Pittsburgh, Pittsburgh, Pennsylvania, USA; GRECC, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA.
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - John C Newman
- Buck Institute for Research on Aging, Novato California, USA; Division of Geriatrics, University of California San Francisco, San Francisco, California, USA
| | - James L Kirkland
- Division of General Internal Medicine, Department of Medicine and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Elena Volpi
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas, USA
| | - George E Taffet
- Department of Medicine (Geriatrics and Cardiovascular Sciences), Baylor College of Medicine, Houston, Texas, USA
| | - Nir Barzilai
- Einstein Institute for Aging Research, Bronx, New York, USA; Einstein-NSC and Glenn Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Dalane W Kitzman
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Libby
- Cardiovascular Medicine and Geriatrics, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | |
Collapse
|
14
|
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: 7] [Impact Index Per Article: 7.0] [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
|
15
|
Yang Z, Wang T, Dong Y, Liu L, Xue X, Wu J, Hao L, Yuan J, Cui J, Qiao S, Yang W. Analysis of Risk Factors for Major Adverse Cardiovascular Events in Patients with Coronary Stent Restenosis after Revascularization. Rev Cardiovasc Med 2023; 24:146. [PMID: 39076758 PMCID: PMC11273033 DOI: 10.31083/j.rcm2405146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 07/31/2024] Open
Abstract
Background To investigate the risk factors for myocardial infarction, recurrent in-stent restenosis (ISR) and target vessel revascularization (TVR) in patients with coronary ISR within 4 years after revascularization. Methods A total of 1884 patients who were hospitalized at Fuwai Hospital for ISR and successfully treated with coronary intervention between January 2017 and December 2018 were included to determine whether there were myocardial infarction, recurrent ISR, TVR and other major adverse cardiovascular events (MACEs) within 4 years after intervention. The patients were divided into the MACE group (215 patients) and the non-MACE group (1669 patients). The clinical data of patients in the two groups were compared, and the risk factors for postoperative MACEs in the ISR patients were obtained by multivariate logistic regression analysis. The receiver operating characteristic (ROC) curve was used to determine the optimal prediction threshold for postoperative MACEs in ISR patients. The difference in survival curves between the two groups was compared using Kaplan‒Meier survival analysis. Results The albumin (43.42 ± 4.77 vs. 44.17 ± 4.46, p = 0.021), direct bilirubin (2.5 (2, 3.5) vs. 2.8 (2.07, 3.73), p = 0.036) and free triiodothyronine (FT3) (2.85 ± 0.43 vs. 2.92 ± 0.42, p = 0.019) levels in the MACE group were significantly lower than those in the non-MACE group, and there was a significant negative correlation between albumin and FT3 and MACEs. The results of univariate and multivariate logistic regression analyses revealed that FT3 was an independent predictor of postoperative MACEs in ISR patients (Odds Ratio (OR) = 0.626, 95% CI: 0.429-0.913, p = 0.015). The ROC curve analysis determined that an FT3 value of 2.785 pmol/L was the optimal prediction threshold. According to the threshold, ISR patients were divided into the FT3 < 2.785 group and the FT3 ≥ 2.785 group. The Kaplan‒Meier analysis revealed that the postoperative recurrence rate of MACEs of the FT3 < 2.785 group was substantially greater than that of the FT3 ≥ 2.785 group (Hazard Ratio (HR) = 0.76, 95% CI: 0.58-0.994, p = 0.044). Conclusions FT3 can be used as an independent predictor of postoperative myocardial infarction, recurrent ISR and TVR in ISR patients. When FT3 is < 2.785 pmol/L, the incidence of postoperative myocardial infarction, recurrent ISR and TVR in ISR patients increases significantly.
Collapse
Affiliation(s)
- Zhuoxuan Yang
- Department of Cardiology, Yuncheng Central Hospital of Shanxi Province, 044000 Yuncheng, Shanxi, China
| | - Tianjie Wang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Ying Dong
- Department of Graduate, Changzhi Medical College, 046012 Changzhi, Shanxi, China
| | - Long Liu
- Department of Cardiology, Yuncheng Central Hospital of Shanxi Province, 044000 Yuncheng, Shanxi, China
| | - Xuan Xue
- Department of Cardiology, Yuncheng Central Hospital of Shanxi Province, 044000 Yuncheng, Shanxi, China
| | - Jine Wu
- Department of Cardiology, Yuncheng Central Hospital of Shanxi Province, 044000 Yuncheng, Shanxi, China
| | - Liuyi Hao
- Department of Cardiology, Yuncheng Central Hospital of Shanxi Province, 044000 Yuncheng, Shanxi, China
| | - Jiansong Yuan
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Jingang Cui
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Shubin Qiao
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Weixian Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| |
Collapse
|
16
|
Li N, Xu X, Qi Z, Gao C, Zhao P, Yang J, Damirin A. Lpar1-mediated Effects in Endothelial Progenitor Cells Are Crucial for Lung Repair in Acute Respiratory Distress Syndrome/Acute Lung Injury. Am J Respir Cell Mol Biol 2023; 68:161-175. [PMID: 36287629 DOI: 10.1165/rcmb.2021-0331oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Acute respiratory distress syndrome/acute lung injury (ARDS/ALI) involves acute respiratory failure characterized by vascular endothelial and lung alveolar epithelial injury. Endothelial progenitor cells (EPCs) can mediate vasculogenesis. However, the limitations of EPCs, such as low survival and differentiation, are believed to inhibit the effectiveness of autologous cell therapies. This study demonstrated that lysophosphatidic acid (LPA), a bioactive small molecule without immunogenicity, is involved in the survival and antiapoptotic effects in human umbilical cord mesenchymal stem cells. This study aimed to explore whether LPA improves the survival of EPCs, enhancing the cellular therapeutic efficacy in ARDS, and these results will expand the application of LPA in stem cells and regenerative medicine. LPA promoted the colony formation, proliferation, and migration of EPCs and upregulated the expression of vascular endothelial-derived growth factor (VEGF) in EPCs. LPA pretreatment of transplanted EPCs improved the therapeutic effect by increasing EPC numbers in the rat lungs. LPA enhanced EPC proliferation and migration through Lpar1 coupled to Gi/o and Gq/11, respectively. Activation of extracellular signal-related kinase 1/2, or ERK1/2, was related to LPA-induced EPC proliferation but not migration. LPA/Lpar1-mediated Gi/o protein was also shown to be involved in promoting VEGF expression and inhibiting IL-1α expression in EPCs. Low LPA concentrations are present after lung injury; thus, the restoration of LPA may promote endothelial cell homeostasis and lung repair in ARDS. Inhalation of LPA significantly promoted the homing of endogenous EPCs to the lung and reduced lung injury in both rats with LPS-induced ALI and Streptococcus pneumoniae-infected mice. Taken together, these data indicated that LPA/Lpar1-mediated effects in EPCs are involved in maintaining endothelial cell homeostasis and lung tissue repair under physiological conditions.
Collapse
Affiliation(s)
- Narengerile Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; and.,The Third Affiliated Hospital, Inner Mongolia Medical University, Baotou, Inner Mongolia, China
| | - Xiyuan Xu
- The Third Affiliated Hospital, Inner Mongolia Medical University, Baotou, Inner Mongolia, China
| | - Zhimin Qi
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Chanchan Gao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Pengfei Zhao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Jingping Yang
- The Third Affiliated Hospital, Inner Mongolia Medical University, Baotou, Inner Mongolia, China
| | - Alatangaole Damirin
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| |
Collapse
|
17
|
Nian W, Fu C. Exosomes in Myocardial Infarction: Therapeutic Potential and Clinical Application. J Cardiovasc Transl Res 2023; 16:87-96. [PMID: 35672604 DOI: 10.1007/s12265-022-10284-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
Myocardial infarction (MI) remains the leading fatal disease in the world, and with subsequent adverse ventricular remodeling often leading to the development of heart failure, finding new ways to improve the prognosis of MI is important. Exosomes are extracellular vesicles of 30-150 nm secreted by various cells in the body. It is now well recognized that exosomes play an important role in MI, and exosomes may become a new approach to post-MI treatment. It is valuable to study how exosomes are involved in post-MI progression and how exosomes can be modified to improve their effectiveness. In this review, we focus on summarizing the therapeutic potential of exosomes for MI and the current status of clinical applications to provide evidence for the formal use of exosomes in the clinic.
Collapse
Affiliation(s)
| | - Cong Fu
- Department of Cardiology, Yi Ji Shan Hospital Affiliated to Wan Nan Medical College, 92# West Zhe Shan Road, Wuhu, Anhui, China. .,Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wan Nan Medical College, Wuhu, China.
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Gori T. Restenosis after Coronary Stent Implantation: Cellular Mechanisms and Potential of Endothelial Progenitor Cells (A Short Guide for the Interventional Cardiologist). Cells 2022; 11:cells11132094. [PMID: 35805178 PMCID: PMC9265311 DOI: 10.3390/cells11132094] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 02/05/2023] Open
Abstract
Coronary stents are among the most common therapies worldwide. Despite significant improvements in the biocompatibility of these devices throughout the last decades, they are prone, in as many as 10–20% of cases, to short- or long-term failure. In-stent restenosis is a multifactorial process with a complex and incompletely understood pathophysiology in which inflammatory reactions are of central importance. This review provides a short overview for the clinician on the cellular types responsible for restenosis with a focus on the role of endothelial progenitor cells. The mechanisms of restenosis are described, along with the cell-based attempts made to prevent it. While the focus of this review is principally clinical, experimental evidence provides some insight into the potential implications for prevention and therapy of coronary stent restenosis.
Collapse
Affiliation(s)
- Tommaso Gori
- German Center for Cardiac and Vascular Research (DZHK) Standort Rhein-Main, Department of Cardiology, University Medical Center Mainz, 55131 Mainz, Germany
| |
Collapse
|
21
|
Ruzanova V, Proskurina A, Efremov Y, Kirikovich S, Ritter G, Levites E, Dolgova E, Potter E, Babaeva O, Sidorov S, Taranov O, Ostanin A, Chernykh E, Bogachev S. Chronometric Administration of Cyclophosphamide and a Double-Stranded DNA-Mix at Interstrand Crosslinks Repair Timing, Called "Karanahan" Therapy, Is Highly Efficient in a Weakly Immunogenic Lewis Carcinoma Model. Pathol Oncol Res 2022; 28:1610180. [PMID: 35693632 PMCID: PMC9185167 DOI: 10.3389/pore.2022.1610180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/27/2022] [Indexed: 12/12/2022]
Abstract
Background and Aims: A new technology based on the chronometric administration of cyclophosphamide and complex composite double-stranded DNA-based compound, which is scheduled in strict dependence on interstrand crosslinks repair timing, and named “Karanahan”, has been developed. Being applied, this technology results in the eradication of tumor-initiating stem cells and full-scale apoptosis of committed tumor cells. In the present study, the efficacy of this novel approach has been estimated in the model of Lewis carcinoma. Methods: To determine the basic indicative parameters for the approach, the duration of DNA repair in tumor cells, as well as their distribution along the cell cycle, have been assessed. Injections were done into one or both tumors in femoral region of the engrafted mice in accordance with the developed regimen. Four series of experiments were carried out at different periods of time. The content of poorly differentiated CD34+/TAMRA+ cells in the bone marrow and peripheral blood has been determined. Immunostaining followed by the flow cytometry was used to analyze the subpopulations of immune cells. Results: The high antitumor efficacy of the new technology against the developed experimental Lewis carcinoma was shown. It was found that the therapy efficacy depended on the number of tumor growth sites, seasonal and annual peculiarities. In some experiments, a long-term remission has been reached in 70% of animals with a single tumor and in 60% with two tumors. In mice with two developed grafts, mobilization capabilities of both poorly differentiated hematopoietic cells of the host and tumor stem-like cells decrease significantly. Being applied, this new technology was shown to activate a specific immune response. There is an increase in the number of NK cell populations in the blood, tumor, and spleen, killer T cells and T helper cells in the tumor and spleen, CD11b+Ly-6C+ and CD11b+Ly-6G+ cells in the tumor. A population of mature dendritic cells is found in the tumor. Conclusion: The performed experiments indicate the efficacy of the Karanahan approach against incurable Lewis carcinoma. Thus, the discussed therapy is a new approach for treating experimental neoplasms, which has a potential as a personalized anti-tumor therapeutic approach in humans.
Collapse
Affiliation(s)
- Vera Ruzanova
- Laboratory of Induced Cellular Processes, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk National Research State University, Novosibirsk, Russia
| | - Anastasia Proskurina
- Laboratory of Induced Cellular Processes, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Yaroslav Efremov
- Department of Natural Sciences, Novosibirsk National Research State University, Novosibirsk, Russia.,Common Use Center for Microscopic Analysis of Biological Objects SB RAS, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Svetlana Kirikovich
- Laboratory of Induced Cellular Processes, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Genrikh Ritter
- Laboratory of Induced Cellular Processes, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenii Levites
- Laboratory of Induced Cellular Processes, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenia Dolgova
- Laboratory of Induced Cellular Processes, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Ekaterina Potter
- Laboratory of Induced Cellular Processes, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oksana Babaeva
- Oncology Department, Municipal Hospital No. 1, Novosibirsk, Russia
| | - Sergey Sidorov
- Department of Natural Sciences, Novosibirsk National Research State University, Novosibirsk, Russia.,Oncology Department, Municipal Hospital No. 1, Novosibirsk, Russia
| | - Oleg Taranov
- Laboratory of Microscopic Research, State Research Center of Virology and Biotechnology "Vector", Koltsovo, Russia
| | - Alexandr Ostanin
- Laboratory of Cellular Immunotherapy, Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena Chernykh
- Laboratory of Cellular Immunotherapy, Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Sergey Bogachev
- Laboratory of Induced Cellular Processes, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
22
|
Deng H, Gong Y, Chen Y, Zhang G, Chen H, Cheng T, Jin L, Wang Y. Porphyromonas gingivalis lipopolysaccharide affects the angiogenic function of endothelial progenitor cells via Akt/FoxO1 signaling. J Periodontal Res 2022; 57:859-868. [PMID: 35694806 DOI: 10.1111/jre.13024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/20/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
AIMS Endothelial progenitor cells (EPCs) function as the angiogenic switch of many physiological and pathological conditions. We aimed to investigate the effects of Porphyromonas gingivalis lipopolysaccharide on the angiogenic capacity of EPCs and delineate the underlying mechanisms. MATERIALS AND METHODS EPCs were isolated from human umbilical blood. CCK-8 assay was undertaken to analyze the cell viability. The migration and tube formation capacity were assessed by wound healing and tube formation, respectively. The protein expression of Akt/p-Akt, endothelial nitric oxide synthase (eNOS)/p-eNOS, and Forkhead box O1 (FoxO1)/p-FoxO1 was determined by Western blot. The intracellular localization of FoxO1 was evaluated by immunofluorescent staining. RESULTS P. gingivalis LPS at 10 μg/ml significantly increased the viability (10.9 ± 2.9%), migration (16.3 ± 3.1%), and tube formation (38.6 ± 5.5%) of EPCs, along with increased phosphorylation of Akt, eNOS, and FoxO1. Mechanistically, Akt inhibition by specific inhibitor wortmannin and FoxO1 forced expression by adenovirus transfection in EPCs markedly attenuated the P. gingivalis LPS-induced eNOS activation, tube formation, and migration. Moreover, P. gingivalis LPS-induced phosphorylation and nuclear exclusion of FoxO1 were blunted by Akt inhibition. CONCLUSIONS The present study suggests that P. gingivalis LPS could affect the angiogenic function of EPCs through the Akt/FoxO1 signaling. The current findings may shed light on the clinical association of periodontitis with aberrant angiogenesis seen in atherosclerotic plaque rupture.
Collapse
Affiliation(s)
- Hui Deng
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yixuan Gong
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yuan Chen
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Guigui Zhang
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Hui Chen
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Tianfan Cheng
- Division of Periodontology and Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Lijian Jin
- Division of Periodontology and Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Pelliccia F, Zimarino M, De Luca G, Viceconte N, Tanzilli G, De Caterina R. Endothelial Progenitor Cells in Coronary Artery Disease: From Bench to Bedside. Stem Cells Transl Med 2022; 11:451-460. [PMID: 35365823 PMCID: PMC9154346 DOI: 10.1093/stcltm/szac010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 02/04/2022] [Indexed: 11/14/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are a heterogeneous group of cells present in peripheral blood at various stages of endothelial differentiation. EPCs have been extensively investigated in patients with coronary artery disease (CAD), with controversial findings both on their role in atherosclerosis progression and in the process of neointimal growth after a percutaneous coronary intervention (PCI). Despite nearly 2 decades of experimental and clinical investigations, however, the significance of EPCs in clinical practice remains unclear and poorly understood. This review provides an update on the role of EPCs in the most common clinical scenarios that are experienced by cardiologists managing patients with CAD. We here summarize the main findings on the association of EPCs with cardiovascular risk factors, coronary atherosclerosis, and myocardial ischemia. We then discuss the potential effects of EPCs in post-PCI in-stent restenosis, as well as most recent findings with EPC-coated stents. Based on the mounting evidence of the relationship between levels of EPCs and several different adverse cardiovascular events, EPCs are emerging as novel predictive biomarkers of long-term outcomes in patients with CAD.
Collapse
Affiliation(s)
| | - Marco Zimarino
- Institute of Cardiology, “G. d’Annunzio” University, Chieti, Italy
- Cath Lab, SS. Annunziata Hospital, Chieti, Italy
| | - Giuseppe De Luca
- Division of Cardiology, Azienda Ospedaliero-Universitaria Maggiore della Carità, Università del Piemonte Orientale, Novara, Italy
| | - Nicola Viceconte
- Department of Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Gaetano Tanzilli
- Department of Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | |
Collapse
|
24
|
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: 6.0] [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
|
25
|
Li S, Zheng Y, Huang Y, He W, Liu X, Zhu W. Association of body mass index and prognosis in patients with HFpEF: A dose-response meta-analysis. Int J Cardiol 2022; 361:40-46. [PMID: 35568057 DOI: 10.1016/j.ijcard.2022.05.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Although agreements regarding the negative effects of obesity on the development of heart failure with preserved ejection fraction (HFpEF) have been reached, the relationship between body mass index (BMI) and adverse outcomes in HFpEF patients are still debatable. Therefore, we conducted the dose-response meta-analysis to investigate this relationship. METHODS We searched the PubMed and Embase databases up to February 2022 for studies that evaluated the association between BMI and prognostic outcomes in patients with HFpEF. A cubic spline random-effects model was used to fit the potential dose-response curve. The effect estimates were expressed as adjusted hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS A total of 11 studies involving 69,273 patients with HFpEF were included. The summary HR for all-cause mortality was 0.90 (95% CI, 0.84-0.95) per 5 units increase in BMI, but the association was U-shaped (Pnonlinear < 0.01) with the nadir of risk at a BMI of 32-34 kg/m2. The summary HR for HF hospitalization was 1.12 (95% CI, 1.05-1.19) with a significant positive linear association (Pnonlinear = 0.54). CONCLUSIONS For patients with HFpEF, there was a positive linear association of BMI with HF hospitalization, while a U-shaped relationship between BMI and all-cause mortality was observed with the lowest event rate at a BMI of 32-34 kg/m2.
Collapse
Affiliation(s)
- Siyuan Li
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yuxiang Zheng
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yuwen Huang
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Wenfeng He
- Department of Medical Geneticsthe, Jiangxi Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510030, China.
| | - Wengen Zhu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
26
|
Wang Y, Kong Y, Zhao HY, Zhang YY, Wang YZ, Xu LP, Zhang XH, Liu KY, Huang XJ. Prophylactic NAC promoted hematopoietic reconstitution by improving endothelial cells after haploidentical HSCT: a phase 3, open-label randomized trial. BMC Med 2022; 20:140. [PMID: 35473809 PMCID: PMC9044574 DOI: 10.1186/s12916-022-02338-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/14/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Poor graft function (PGF) or prolonged isolated thrombocytopenia (PT), which are characterized by pancytopenia or thrombocytopenia, have become serious complications after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Our previous single-arm trial suggests that N-acetyl-L-cysteine (NAC) prophylaxis reduced PGF or PT after allo-HSCT. Therefore, an open-label, randomized, phase 3 trial was performed to investigate the efficacy and tolerability of NAC prophylaxis to reduce PGF or PT after allo-HSCT. METHODS A phase 3, open-label randomized trial was performed. Based on the percentage of CD34+VEGFR2 (CD309)+ endothelial cells (ECs) in bone marrow (BM) detected by flow cytometry at 14 days before conditioning, patients aged 15 to 60 years with acute leukemia undergoing haploidentical HSCT were categorized as low-risk (EC ≥ 0.1%) or high-risk (EC < 0.1%); patients at high risk were randomly assigned (2:1) to receive NAC prophylaxis or nonprophylaxis. The primary endpoint was PGF and PT incidence at +60 days post-HSCT. RESULTS Between April 18, 2019, and June 24, 2021, 120 patients with BM EC <0.1% were randomly assigned for NAC (group A, N = 80) or nonprophylaxis (group B, N = 40), and 105 patients with EC≥0.1% (group C) were also analyzed. The +60 days incidence of PGF and PT was 7.5% (95% CI, 1.7 to 13.3%) and 22.5% (95% CI, 9.1 to 35.9%) in group A and group B (hazard ratio, 0.317; 95% CI, 0.113 to 0.890; P = 0.021) and 11.4% (95% CI, 5.2 to 17.6%) in group C (hazard ratio, 0.643; 95% CI, 0.242 to 1.715; P = 0.373). Consistently, NAC prophylaxis gradually improved BM ECs and CD34+ cells in group A, whereas reduced their reactive oxygen species (ROS) levels post-HSCT. Within 60 days post-HSCT, the most common grade 3 to 5 adverse events for the NAC and control groups were infections (19/80 [24%] vs. 10/40 [25%]) and gastrointestinal adverse events (16/80 [20%] vs. 7/40 [18%]). There were no treatment-related deaths. CONCLUSIONS N-Acetyl-L-cysteine prophylaxis can prevent the occurrence of poor hematopoietic function and is well tolerated in haploidentical HSCT. It may offer a potential pathogenesis-oriented therapeutic approach for patients with poor hematopoietic function. TRIAL REGISTRATION This trial was registered at ClinicalTrials.gov as #NCT03967665.
Collapse
Affiliation(s)
- Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuan Kong
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Hong-Yan Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuan-Yuan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Ya-Zhe Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
27
|
Bonora BM, Palano MT, Testa G, Fadini GP, Sangalli E, Madotto F, Persico G, Casciaro F, Vono R, Colpani O, Scavello F, Cappellari R, Abete P, Orlando P, Carnelli F, Berardi AG, De Servi S, Raucci A, Giorgio M, Madeddu P, Spinetti G. Hematopoietic progenitor cell liabilities and alarmins S100A8/A9-related inflammaging associate with frailty and predict poor cardiovascular outcomes in older adults. Aging Cell 2022; 21:e13545. [PMID: 35166014 PMCID: PMC8920446 DOI: 10.1111/acel.13545] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/13/2021] [Accepted: 12/19/2021] [Indexed: 12/18/2022] Open
Abstract
Frailty affects the physical, cognitive, and social domains exposing older adults to an increased risk of cardiovascular disease and death. The mechanisms linking frailty and cardiovascular outcomes are mostly unknown. Here, we studied the association of abundance (flow cytometry) and gene expression profile (RNAseq) of stem/progenitor cells (HSPCs) and molecular markers of inflammaging (ELISA) with the cardiorespiratory phenotype and prospective adverse events of individuals classified according to levels of frailty. Two cohorts of older adults were enrolled in the study. In a cohort of pre-frail 35 individuals (average age: 75 years), a physical frailty score above the median identified subjects with initial alterations in cardiorespiratory function. RNA sequencing revealed S100A8/A9 upregulation in HSPCs from the bone marrow (>10-fold) and peripheral blood (>200-fold) of individuals with greater physical frailty. Moreover higher frailty was associated with increased alarmins S100A8/A9 and inflammatory cytokines in peripheral blood. We then studied a cohort of 104 more frail individuals (average age: 81 years) with multidomain health deficits. Reduced levels of circulating HSPCs and increased S100A8/A9 concentrations were independently associated with the frailty index. Remarkably, low HSPCs and high S100A8/A9 simultaneously predicted major adverse cardiovascular events at 1-year follow-up after adjustment for age and frailty index. In conclusion, inflammaging characterized by alarmin and pro-inflammatory cytokines in pre-frail individuals is mirrored by the pauperization of HSPCs in frail older people with comorbidities. S100A8/A9 is upregulated within HSPCs, identifying a phenotype that associates with poor cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Gianluca Testa
- Department of Medicine and Health Sciences “Vincenzo Tiberio” University of Molise Campobasso Italy
| | | | | | | | | | | | | | | | - Francesco Scavello
- Unit of Experimental Cardio‐Oncology and Cardiovascular Aging Centro Cardiologico Monzino‐IRCCS Milan Italy
| | | | - Pasquale Abete
- Department of Translational Medical Sciences University of Naples Federico II Naples Italy
| | | | | | | | | | - Angela Raucci
- Unit of Experimental Cardio‐Oncology and Cardiovascular Aging Centro Cardiologico Monzino‐IRCCS Milan Italy
| | - Marco Giorgio
- European Institute of Oncology (IEO) Milan Italy
- Department of Biomedical Sciences University of Padova Padua Italy
| | - Paolo Madeddu
- Experimental Cardiovascular Medicine Bristol Medical School: Translational Health Sciences University of Bristol Bristol UK
| | | |
Collapse
|
28
|
Lyotropic Liquid Crystals: A Biocompatible and Safe Material for Local Cardiac Application. Pharmaceutics 2022; 14:pharmaceutics14020452. [PMID: 35214184 PMCID: PMC8879243 DOI: 10.3390/pharmaceutics14020452] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/03/2022] [Accepted: 02/17/2022] [Indexed: 02/01/2023] Open
Abstract
The regeneration of cardiac tissue is a multidisciplinary research field aiming to improve the health condition of the post-heart attack patient. Indeed, myocardial tissue has a poor ability to self-regenerate after severe damage. The scientific efforts focused on the research of a biomaterial able to adapt to heart tissue, thus guaranteeing the in situ release of active substances or growth promoters. Many types of hydrogels were proposed for this purpose, showing several limitations. The aim of this study was to suggest a new usage for glyceryl monooleate-based lyotropic liquid crystals (LLCs) as a biocompatible and inert material for a myocardial application. The main advantages of LLCs are mainly related to their easy in situ injection as lamellar phase and their instant in situ transition in the cubic phase. In vivo studies proved the biocompatibility and the inertia of LLCs after their application on the myocardial tissue of mice. In detail, the cardiac activity was monitored through 28 days, and no significant alterations were recorded in the heart anatomy and functionality. Moreover, gross anatomy showed the ability of LLCs to be bio-degraded in a suitable time frame. Overall, these results permitted us to suppose a potential use of LLCs as materials for cardiac drug delivery.
Collapse
|
29
|
Wu H, Norton V, Cui K, Zhu B, Bhattacharjee S, Lu YW, Wang B, Shan D, Wong S, Dong Y, Chan SL, Cowan D, Xu J, Bielenberg DR, Zhou C, Chen H. Diabetes and Its Cardiovascular Complications: Comprehensive Network and Systematic Analyses. Front Cardiovasc Med 2022; 9:841928. [PMID: 35252405 PMCID: PMC8891533 DOI: 10.3389/fcvm.2022.841928] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus is a worldwide health problem that usually comes with severe complications. There is no cure for diabetes yet and the threat of these complications is what keeps researchers investigating mechanisms and treatments for diabetes mellitus. Due to advancements in genomics, epigenomics, proteomics, and single-cell multiomics research, considerable progress has been made toward understanding the mechanisms of diabetes mellitus. In addition, investigation of the association between diabetes and other physiological systems revealed potentially novel pathways and targets involved in the initiation and progress of diabetes. This review focuses on current advancements in studying the mechanisms of diabetes by using genomic, epigenomic, proteomic, and single-cell multiomic analysis methods. It will also focus on recent findings pertaining to the relationship between diabetes and other biological processes, and new findings on the contribution of diabetes to several pathological conditions.
Collapse
Affiliation(s)
- Hao Wu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Vikram Norton
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Kui Cui
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Bo Zhu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Sudarshan Bhattacharjee
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Yao Wei Lu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Beibei Wang
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Dan Shan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Scott Wong
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Yunzhou Dong
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Siu-Lung Chan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Douglas Cowan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Diane R. Bielenberg
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Hong Chen
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
30
|
Vitale E, Rossin D, Perveen S, Miletto I, Lo Iacono M, Rastaldo R, Giachino C. Silica Nanoparticle Internalization Improves Chemotactic Behaviour of Human Mesenchymal Stem Cells Acting on the SDF1α/CXCR4 Axis. Biomedicines 2022; 10:biomedicines10020336. [PMID: 35203545 PMCID: PMC8961775 DOI: 10.3390/biomedicines10020336] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Human mesenchymal stem cell (hMSC)-based therapy is an emerging resource in regenerative medicine. Despite the innate ability of hMSCs to migrate to sites of injury, homing of infused hMSCs to the target tissue is inefficient. It was shown that silica nanoparticles (SiO2-NPs), previously developed to track the stem cells after transplantation, accumulated in lysosomes leading to a transient blockage of the autophagic flux. Since CXCR4 turnover is mainly regulated by autophagy, we tested the effect of SiO2-NPs on chemotactic migration of hMSCs along the SDF1α/CXCR4 axis that plays a pivotal role in directing MSC homing to sites of injury. Our results showed that SiO2-NP internalization augmented CXCR4 surface levels. We demonstrated that SiO2-NP-dependent CXCR4 increase was transient, and it reversed at the same time as lysosomal compartment normalization. Furthermore, the autophagy inhibitor Bafilomycin-A1 reproduced CXCR4 overexpression in control hMSCs confirming the direct effect of the autophagic degradation blockage on CXCR4 expression. Chemotaxis assays showed that SiO2-NPs increased hMSC migration toward SDF1α. In contrast, migration improvement was not observed in TNFα/TNFR axis, due to the proteasome-dependent TNFR regulation. Overall, our findings demonstrated that SiO2-NP internalization increases the chemotactic behaviour of hMSCs acting on the SDF1α/CXCR4 axis, unmasking a high potential to improve hMSC migration to sites of injury and therapeutic efficacy upon cell injection in vivo.
Collapse
Affiliation(s)
- Emanuela Vitale
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.V.); (D.R.); (S.P.); (M.L.I.); (C.G.)
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.V.); (D.R.); (S.P.); (M.L.I.); (C.G.)
| | - Sadia Perveen
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.V.); (D.R.); (S.P.); (M.L.I.); (C.G.)
| | - Ivana Miletto
- Department of Science and Technological Innovation, University of Eastern Piedmont, 15121 Alessandria, Italy;
| | - Marco Lo Iacono
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.V.); (D.R.); (S.P.); (M.L.I.); (C.G.)
| | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.V.); (D.R.); (S.P.); (M.L.I.); (C.G.)
- Correspondence:
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.V.); (D.R.); (S.P.); (M.L.I.); (C.G.)
| |
Collapse
|
31
|
Pelliccia F, Pasceri V, Zimarino M, De Luca G, De Caterina R, Mehran R, Dangas G. Endothelial progenitor cells in coronary atherosclerosis and percutaneous coronary intervention: A systematic review and meta-analysis. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2022; 42:94-99. [DOI: 10.1016/j.carrev.2022.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/19/2022]
|
32
|
Yan J, Li X, Long W, Yuan T, Xian S. Association Between Obesity and Lower Short- and Long-Term Mortality in Coronary Care Unit Patients: A Cohort Study of the MIMIC-III Database. Front Endocrinol (Lausanne) 2022; 13:855650. [PMID: 35444615 PMCID: PMC9013888 DOI: 10.3389/fendo.2022.855650] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Obesity has long been considered an independent risk factor for cardiovascular diseases (CVD), even in the COVID-19 pandemic. However, recent studies have found that a certain degree of obesity may be beneficial for patients who have already suffered from CVD, which is called the "obesity paradox". Our objective was to investigate whether the obesity paradox existed in coronary care unit (CCU) patients and the relationship between body mass index (BMI) and short- and long-term mortality. METHODS We performed a cohort analysis of 3,502 adult CCU patients from the Medical Information Mart for Intensive Care III (MIMIC-III) database. The patients were divided into four groups according to the WHO BMI categories. Both multivariable logistic regression and Cox regression were used to reveal the relation between BMI and mortality. Subgroup analyses were performed based on Simplified Acute Physiology Score (SAPS) and age. RESULTS After adjusting for confounders, obese patients had 33% and 30% lower mortality risk at 30-day and 1-year (OR 0.67, 95% CI 0.51 to 0.89; HR 0.70, 95% CI 0.59 to 0.83; respectively) compared with normal-weight patients, while the underweight group were opposite, with 141% and 81% higher in short- and long-term (OR 2.41, 95% CI 1.37 to 4.12; HR 1.81, 95% CI 1.34 to 2.46; respectively). Overweight patients did not have a significant survival advantage at 30-day (OR 0.91, 95% CI 0.70 to 1.17), but did have a 22% lower mortality risk at 1-year (HR 0.78; 95% CI 0.67 to 0.91). The results were consistent after being stratified by SAPS and age. CONCLUSION Our study supports that obesity improved survival at both 30-day and 1-year after CCU admission, and the obesity paradox existed in CCU patients.
Collapse
Affiliation(s)
- Junlue Yan
- The First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyuan Li
- Department of Community Health, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Wenjie Long
- Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianhui Yuan
- Geriatrics Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Shaoxiang Xian, ; Tianhui Yuan,
| | - Shaoxiang Xian
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Cardiovascular Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Shaoxiang Xian, ; Tianhui Yuan,
| |
Collapse
|
33
|
Sen A, Vincent V, Thakkar H, Abraham R, Ramakrishnan L. Beneficial Role of Vitamin D on Endothelial Progenitor Cells (EPCs) in Cardiovascular Diseases. J Lipid Atheroscler 2022; 11:229-249. [PMID: 36212746 PMCID: PMC9515729 DOI: 10.12997/jla.2022.11.3.229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/23/2022] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the world. Endothelial progenitor cells (EPCs) are currently being explored in the context of CVD risk. EPCs are bone marrow derived progenitor cells involved in postnatal endothelial repair and neovascularization. A large body of evidence from clinical, animal, and in vitro studies have shown that EPC numbers in circulation and their functionality reflect endogenous vascular regenerative capacity. Traditionally vitamin D is known to be beneficial for bone health and calcium metabolism and in the last two decades, its role in influencing CVD and cancer risk has generated significant interest. Observational studies have shown that low vitamin D levels are associated with an adverse cardiovascular risk profile. Still, Mendelian randomization studies and randomized control trials (RCTs) have not shown significant effects of vitamin D on cardiovascular events. The criticism regarding the RCTs on vitamin D and CVD is that they were not designed to investigate cardiovascular outcomes in vitamin D-deficient individuals. Overall, the association between vitamin D and CVD remains inconclusive. Recent clinical and experimental studies have demonstrated the beneficial role of vitamin D in increasing the circulatory level of EPC as well as their functionality. In this review we present evidence supporting the beneficial role of vitamin D in CVD through its modulation of EPC homeostasis.
Collapse
Affiliation(s)
- Atanu Sen
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Vinnyfred Vincent
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Himani Thakkar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ransi Abraham
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Lakshmy Ramakrishnan
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
34
|
Hess DA, Verma S, Bhatt D, Bakbak E, Terenzi DC, Puar P, Cosentino F. Vascular repair and regeneration in cardiometabolic diseases. Eur Heart J 2021; 43:450-459. [PMID: 34849704 DOI: 10.1093/eurheartj/ehab758] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/27/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
Chronic cardiometabolic assaults during type 2 diabetes (T2D) and obesity induce a progenitor cell imbalance in the circulation characterized by overproduction and release of pro-inflammatory monocytes and granulocytes from the bone marrow alongside aberrant differentiation and mobilization of pro-vascular progenitor cells that generate downstream progeny for the coordination of blood vessel repair. This imbalance can be detected in the peripheral blood of individuals with established T2D and severe obesity using multiparametric flow cytometry analyses to discern pro-inflammatory vs. pro-angiogenic progenitor cell subsets identified by high aldehyde dehydrogenase activity, a conserved progenitor cell protective function, combined with lineage-restricted cell surface marker analyses. Recent evidence suggests that progenitor cell imbalance can be reversed by treatment with pharmacological agents or surgical interventions that reduce hyperglycaemia or excess adiposity. In this state-of-the-art review, we present current strategies to assess the progression of pro-vascular regenerative cell depletion in peripheral blood samples of individuals with T2D and obesity and we summarize novel clinical data that intervention using sodium-glucose co-transporter 2 inhibition or gastric bypass surgery can efficiently restore cell-mediated vascular repair mechanisms associated with profound cardiovascular benefits in recent outcome trials. Collectively, this thesis generates a compelling argument for early intervention using current pharmacological agents to prevent or restore imbalanced circulating progenitor content and maintain vascular regenerative cell trafficking to sites of ischaemic damage. This conceptual advancement may lead to the design of novel therapeutic approaches to prevent or reverse the devastating cardiovascular comorbidities currently associated with T2D and obesity.
Collapse
Affiliation(s)
- David A Hess
- Department of Pharmacology and Toxicology, University of Toronto, 27 King's College Circle, Toronto, ON M5S 3J3, Canada.,Division of Vascular Surgery, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada.,Molecular Medicine Research Laboratories, Krembil Centre for Stem Cells Biology, Robarts Research Institute, University of Western Ontario, 1151 Richmond Street North, London, ON N6H 0E8, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street North, London, ON N6H 0E8, Canada
| | - Subodh Verma
- Department of Pharmacology and Toxicology, University of Toronto, 27 King's College Circle, Toronto, ON M5S 3J3, Canada.,Division of Cardiovascular Surgery, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada.,Institute of Medical Sciences, University of Toronto, 27 King's College Circle, Toronto, ON M5S 3J3, Canada.,Department of Surgery, University of Toronto, 27 King's College Circle, Toronto, ON M5S 3J3, Canada
| | - Deepak Bhatt
- Department of Cardiovascular Medicine, Harvard Medical School, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Ehab Bakbak
- Department of Pharmacology and Toxicology, University of Toronto, 27 King's College Circle, Toronto, ON M5S 3J3, Canada.,Division of Cardiovascular Surgery, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Daniella C Terenzi
- Division of Cardiovascular Surgery, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada.,Institute of Medical Sciences, University of Toronto, 27 King's College Circle, Toronto, ON M5S 3J3, Canada
| | - Pankaj Puar
- Division of Cardiovascular Surgery, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm SE171 77, Sweden
| |
Collapse
|
35
|
Almuwaqqat Z, Kim JH, Garcia M, Ko YA, Moazzami K, Lima B, Sullivan S, Alkhalaf J, Mehta A, Shah AJ, Hussain MS, Pearce BD, Bremner JD, Waller EK, Vaccarino V, Quyyumi AA. Associations Between Inflammation, Cardiovascular Regenerative Capacity, and Cardiovascular Events: A Cohort Study. Arterioscler Thromb Vasc Biol 2021; 41:2814-2822. [PMID: 34551591 PMCID: PMC8675629 DOI: 10.1161/atvbaha.121.316574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022]
Abstract
Objective Circulating progenitor cells possess immune modulatory properties and might mitigate inflammation that is characteristic of patients with coronary artery disease. We hypothesized that patients with fewer circulating progenitor cells (CPCs) will have higher inflammatory markers and worse outcomes. Approach and Results Patients with stable coronary artery disease were enrolled in a prospective study enumerating CPCs as CD (cluster of differentiation)-34-expressing mononuclear cells (CD34+) and inflammation as levels of IL (interleukin)-6 and high-sensitivity CRP (C-reactive protein) levels. Patients were followed for 5 years for the end points of death and myocardial infarction with repeat inflammatory biomarkers measured after a median of 2 years. In the entire cohort of 392 patients, IL-6 and high-sensitivity CRP levels remained unchanged (0.3+/-2.4 pg/mL and 0.1+/-1.0 mg/L; P=0.45) after 2 years. CPC counts (log-transformed) were inversely correlated with the change in IL-6 levels (r, -0.17; P<0.001). Using linear regression, IL-6 and high-sensitivity CRP levels declined by -0.59 (95% CI, -0.90 to -0.20) pg/mL and -0.13 (-0.28 to 0.01) mg/L per 1 log higher CPC counts after adjustment for the demographic and clinical variables, as well as medications. Using Cox models adjusted for these risk factors, a rise in 1 pg/mL of IL-6 was associated with a 11% (95% CI, 9-13) greater risk of death/myocardial infarction. We found that the change in IL6 level partly (by 40%) mediated the higher risk of adverse events among those with low CPC counts. Conclusions Reduced cardiovascular regenerative capacity is independently associated with progressive inflammation in patients with coronary artery disease that in turn is associated with poor outcomes.
Collapse
Affiliation(s)
- Zakaria Almuwaqqat
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Jeong Hwan Kim
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Mariana Garcia
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Kasra Moazzami
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Bruno Lima
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Samaah Sullivan
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Jamil Alkhalaf
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Anurag Mehta
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Amit J. Shah
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
- Atlanta VA Medical Center, Decatur, Georgia
| | - Mohammad S. Hussain
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Brad D. Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - J. Douglas Bremner
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
- Atlanta VA Medical Center, Decatur, Georgia
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Edmund K. Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Viola Vaccarino
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Arshed A. Quyyumi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
36
|
Chambers SE, Pathak V, Pedrini E, Soret L, Gendron N, Guerin CL, Stitt AW, Smadja DM, Medina RJ. Current concepts on endothelial stem cells definition, location, and markers. Stem Cells Transl Med 2021; 10 Suppl 2:S54-S61. [PMID: 34724714 PMCID: PMC8560200 DOI: 10.1002/sctm.21-0022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic vascular disease is a major cause of mortality and morbidity worldwide, and regeneration of blood vessels in perfusion-deficient tissues is a worthwhile therapeutic goal. The idea of delivering endothelial stem/progenitor cells to repair damaged vasculature, reperfuse hypoxic tissue, prevent cell death, and consequently diminish tissue inflammation and fibrosis has a strong scientific basis and clinical value. Various labs have proposed endothelial stem/progenitor cell candidates. This has created confusion, as there are profound differences between these cell definitions based on isolation methodology, characterization, and reparative biology. Here, a stricter definition based on stem cell biology principles is proposed. Although preclinical studies have often been promising, results from clinical trials have been highly contradictory and served to highlight multiple challenges associated with disappointing therapeutic benefit. This article reviews recent accomplishments in the field and discusses current difficulties when developing endothelial stem cell therapies. Emerging evidence that disputes the classic view of the bone marrow as the source for these cells and supports the vascular wall as the niche for these tissue-resident endothelial stem cells is considered. In addition, novel markers to identify endothelial stem cells, including CD157, EPCR, and CD31low VEGFR2low IL33+ Sox9+ , are described.
Collapse
Affiliation(s)
- Sarah E.J. Chambers
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Varun Pathak
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Edoardo Pedrini
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Lou Soret
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Nicolas Gendron
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Coralie L. Guerin
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Cytometry Platform, Institut CurieParisFrance
| | - Alan W. Stitt
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - David M. Smadja
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Reinhold J. Medina
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| |
Collapse
|
37
|
Could a Multi-Marker and Machine Learning Approach Help Stratify Patients with Heart Failure? MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57100996. [PMID: 34684033 PMCID: PMC8538712 DOI: 10.3390/medicina57100996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022]
Abstract
Half of the patients with heart failure (HF) have preserved ejection fraction (HFpEF). To date, there are no specific markers to distinguish this subgroup. The main objective of this work was to stratify HF patients using current biochemical markers coupled with clinical data. The cohort study included HFpEF (n = 24) and heart failure with reduced ejection fraction (HFrEF) (n = 34) patients as usually considered in clinical practice based on cardiac imaging (EF ≥ 50% for HFpEF; EF < 50% for HFrEF). Routine blood tests consisted of measuring biomarkers of renal and heart functions, inflammation, and iron metabolism. A multi-test approach and analysis of peripheral blood samples aimed to establish a computerized Machine Learning strategy to provide a blood signature to distinguish HFpEF and HFrEF. Based on logistic regression, demographic characteristics and clinical biomarkers showed no statistical significance to differentiate the HFpEF and HFrEF patient subgroups. Hence a multivariate factorial discriminant analysis, performed blindly using the data set, allowed us to stratify the two HF groups. Consequently, a Machine Learning (ML) strategy was developed using the same variables in a genetic algorithm approach. ML provided very encouraging explorative results when considering the small size of the samples applied. The accuracy and the sensitivity were high for both validation and test groups (69% and 100%, 64% and 75%, respectively). Sensitivity was 100% for the validation and 75% for the test group, whereas specificity was 44% and 55% for the validation and test groups because of the small number of samples. Lastly, the precision was acceptable, with 58% in the validation and 60% in the test group. Combining biochemical and clinical markers is an excellent entry to develop a computer classification tool to diagnose HFpEF. This translational approach is a springboard for improving new personalized treatment methods and identifying “high-yield” populations for clinical trials.
Collapse
|
38
|
Associations between increased circulating endothelial progenitor cell levels and anxiety/depressive severity, cognitive deficit and function disability among patients with major depressive disorder. Sci Rep 2021; 11:18221. [PMID: 34521977 PMCID: PMC8440504 DOI: 10.1038/s41598-021-97853-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 05/31/2021] [Indexed: 12/11/2022] Open
Abstract
The association of major depressive disorder (MDD) with cardiovascular diseases (CVDs) through endothelial dysfunction is bidirectional. Circulating endothelial progenitor cells (cEPCs), essential for endothelial repair and function, are associated with risks of various CVDs. Here, the relationship of cEPC counts with MDD and the related clinical presentations were investigated in 50 patients with MDD and 46 healthy controls. In patients with MDD, a battery of clinical domains was analysed: depressed mood with Hamilton Depression Rating Scale (HAMD) and Montgomery–Åsberg Depression Rating Scale (MADRS), anxiety with Hamilton Anxiety Rating Scale (HAMA), cognitive dysfunction and deficit with Digit Symbol Substitution Test (DSST) and Perceived Deficits Questionnaire-Depression (PDQ-D), somatic symptoms with Depressive and Somatic Symptom Scale (DSSS), quality of life with 12-Item Short Form Health Survey (SF-12) and functional disability with Sheehan Disability Scale (SDS). Immature and mature cEPC counts were measured through flow cytometry. Increased mature and immature cEPC counts were significantly associated with higher anxiety after controlling the confounding effect of systolic blood pressure, and potentially associated with more severe depressive symptoms, worse cognitive performance and increased cognitive deficit, higher social disability, and worse mental health outcomes. Thus, cEPCs might have pleiotropic effects on MDD-associated symptoms and psychosocial outcomes.
Collapse
|
39
|
Jayaraman S, Gnanasampanthapandian D, Rajasingh J, Palaniyandi K. Stem Cell-Derived Exosomes Potential Therapeutic Roles in Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:723236. [PMID: 34447796 PMCID: PMC8382889 DOI: 10.3389/fcvm.2021.723236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Owing to myocardial abnormalities, cardiac ailments are considered to be the major cause of morbidity and mortality worldwide. According to a recent study, membranous vesicles that are produced naturally, termed as "exosomes", have emerged as the potential candidate in the field of cardiac regenerative medicine. A wide spectrum of stem cells has also been investigated in the treatment of cardiovascular diseases (CVD). Exosomes obtained from the stem cells are found to be cardioprotective and offer great hope in the treatment of CVD. The basic nature of exosomes is to deal with the intracellular delivery of both proteins and nucleic acids. This activity of exosomes helps us to rely on them as the attractive pharmaceutical delivery agents. Most importantly, exosomes derived from microRNAs (miRNAs) hold great promise in assessing the risk of CVD, as they serve as notable biomarkers of the disease. Exosomes are small, less immunogenic, and lack toxicity. These nanovesicles harbor immense potential as a therapeutic entity and would provide fruitful benefits if consequential research were focused on their upbringing and development as a useful diagnostic and therapeutic tool in the field of medicine.
Collapse
Affiliation(s)
- Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Dhanavathy Gnanasampanthapandian
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| | - Johnson Rajasingh
- Department of Bioscience Research & Medicine-Cardiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kanagaraj Palaniyandi
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| |
Collapse
|
40
|
Fadini GP, Vigili de Kreutzenberg S, Avogaro A. Where diabetes care meets cardiovascular research: our cardiovascular perspective at a Centre devoted to diabetes research and care. Eur Heart J 2021; 42:2417-2419. [PMID: 33755089 DOI: 10.1093/eurheartj/ehab177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, Section of Metabolic Disease, University Hospital of Padova, Via Giustiniani 2, Padua 35128, Italy.,Veneto Institute of Molecular Medicine, Laboratory of Experimental Diabetology, Padua 35128, Italy
| | - Saula Vigili de Kreutzenberg
- Department of Medicine, Section of Metabolic Disease, University Hospital of Padova, Via Giustiniani 2, Padua 35128, Italy
| | - Angelo Avogaro
- Department of Medicine, Section of Metabolic Disease, University Hospital of Padova, Via Giustiniani 2, Padua 35128, Italy
| |
Collapse
|
41
|
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
|
42
|
Albiero M, Tedesco S, Amendolagine FI, D'Anna M, Migliozzi L, Zuccolotto G, Rosato A, Cappellari R, Avogaro A, Fadini GP. Inhibition of SGLT2 Rescues Bone Marrow Cell Traffic for Vascular Repair: Role of Glucose Control and Ketogenesis. Diabetes 2021; 70:1767-1779. [PMID: 33903150 DOI: 10.2337/db20-1045] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/21/2021] [Indexed: 11/13/2022]
Abstract
The mechanisms by which sodium-glucose cotransporter 2 inhibitors (SGLT2i) improve cardiovascular outcomes in people with diabetes are incompletely understood. Recent studies show that SGLT2i may increase the levels of circulating cells with vascular regenerative capacity, at least in part by lowering glycemia. In this study, we used mice with streptozotocin-induced diabetes treated with the SGLT2i dapagliflozin at a dose that reduced glucose levels by 20%. Dapagliflozin improved the diabetes-associated defect of hematopoietic stem cell mobilization after stimulation with granulocyte colony-stimulating factor. Dapagliflozin rescued the traffic of bone marrow (BM)-derived cells to injured carotid arteries and improved endothelial healing in diabetic mice. Defective homing of CD49d+ granulocytes was causally linked with impaired endothelial repair and was reversed by dapagliflozin. The effects of dapagliflozin were mimicked by a similar extent of glucose reduction achieved with insulin therapy and by a ketone drink that artificially elevated β-hydroxybutyrate. Inhibition of endothelial repair by resident cells using the CXCR4 antagonist AMD3100 did not abolish the vascular effect of dapagliflozin, indirectly supporting that endothelial healing by dapagliflozin was mediated by recruitment of circulating cells. In summary, we show that dapagliflozin improved the traffic of BM-derived hematopoietic cells to the site of vascular injury, providing a hitherto unappreciated mechanism of vascular protection.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Serena Tedesco
- 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
| | - Ludovica Migliozzi
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gaia Zuccolotto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto-Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto-Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy
| | - Roberta Cappellari
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, 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
|
43
|
Vilahur G, Nguyen PH, Badimon L. Impact of Diabetes Mellitus on the Potential of Autologous Stem Cells and Stem Cell-Derived Microvesicles to Repair the Ischemic Heart. Cardiovasc Drugs Ther 2021; 36:933-949. [PMID: 34251593 DOI: 10.1007/s10557-021-07208-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2021] [Indexed: 10/20/2022]
Abstract
Ischemic heart disease remains the leading cause of morbidity and mortality worldwide. Despite the advances in medical management and catheter-based therapy, mortality remains high, as does the risk of developing heart failure. Regenerative therapies have been widely used as an alternative option to repair the damaged heart mainly because of their paracrine-related beneficial effects. Although cell-based therapy has been demonstrated as feasible and safe, randomized controlled trials and meta-analyses show little consistent benefit from treatments with adult-derived stem cells. Mounting evidence from our group and others supports that cardiovascular risk factors and comorbidities impair stem cell potential thus hampering their autologous use. This review aims to better understand the influence of diabetes on stem cell potential. For this purpose, we will first discuss the most recent advances in the mechanistic understanding of the effects of diabetes on stem cell phenotype, function, and molecular fingerprint to further elaborate on diabetes-induced alterations in stem cell extracellular vesicle profile. Although we acknowledge that multiple sources of stem or progenitor cells are used for regenerative purposes, we will focus on bone marrow hematopoietic stem/progenitor cells, mesenchymal stem cells residing in the bone marrow, and adipose tissue and briefly discuss endothelial colony-forming cells.
Collapse
Affiliation(s)
- Gemma Vilahur
- Cardiovascular-Program ICCC, IR-Hospital Santa Creu I Sant Pau, IIB Sant Pau, C/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain.,Ciber CV - ISCIII, Madrid, Spain
| | - Phuong Hue Nguyen
- Cardiovascular-Program ICCC, IR-Hospital Santa Creu I Sant Pau, IIB Sant Pau, C/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular-Program ICCC, IR-Hospital Santa Creu I Sant Pau, IIB Sant Pau, C/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain. .,Ciber CV - ISCIII, Madrid, Spain. .,Cardiovascular Research Chair UAB, Barcelona, Spain.
| |
Collapse
|
44
|
Mehta A, Meng Q, Li X, Desai SR, D’Souza MS, Ho AH, Islam SJ, Dhindsa DS, Almuwaqqat Z, Nayak A, Alkhoder AA, Hooda A, Varughese A, Ahmad SF, Mokhtari A, Hesaroieh I, Sperling LS, Ko YA, Waller EK, Quyyumi AA. Vascular Regenerative Capacity and the Obesity Paradox in Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2021; 41:2097-2108. [PMID: 33853349 PMCID: PMC8147702 DOI: 10.1161/atvbaha.120.315703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/17/2021] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anurag Mehta
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Qi Meng
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA (Q.M., X.L., Y.-A.K.)
| | - Xiaona Li
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA (Q.M., X.L., Y.-A.K.)
| | - Shivang R. Desai
- Department of Medicine (S.R.D., M.S.D., A.H.H.), Emory University School of Medicine, Atlanta, GA
| | - Melroy S. D’Souza
- Department of Medicine (S.R.D., M.S.D., A.H.H.), Emory University School of Medicine, Atlanta, GA
| | - Annie H. Ho
- Department of Medicine (S.R.D., M.S.D., A.H.H.), Emory University School of Medicine, Atlanta, GA
| | - Shabatun J. Islam
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Devinder S. Dhindsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Zakaria Almuwaqqat
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Aditi Nayak
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Ayman A. Alkhoder
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Ananya Hooda
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Anil Varughese
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Syed F. Ahmad
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Ali Mokhtari
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Iraj Hesaroieh
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Laurence S. Sperling
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| | - Yi-An Ko
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA (Q.M., X.L., Y.-A.K.)
| | - Edmund K. Waller
- Department of Hematology and Oncology, Winship Cancer Institute (E.K.W.), Emory University School of Medicine, Atlanta, GA
| | - Arshed A. Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute (A. Mehta, S.J.I., D.S.D., Z.A., A.N., A.A.A., A.H., A.V., S.F.A., A. Mokhtari, I.H., L.S.S., Y.-A.K., A.A.Q.), Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
45
|
Brain-heart connections in stress and cardiovascular disease: Implications for the cardiac patient. Atherosclerosis 2021; 328:74-82. [PMID: 34102426 PMCID: PMC8254768 DOI: 10.1016/j.atherosclerosis.2021.05.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/05/2021] [Accepted: 05/27/2021] [Indexed: 12/25/2022]
Abstract
The influence of psychological stress on the physiology of the cardiovascular system, and on the etiology and outcomes of cardiovascular disease (CVD) has been the object of intense investigation. As a whole, current knowledge points to a "brain-heart axis" that is especially important in individuals with pre-existing CVD. The use of acute psychological stress provocation in the laboratory has been useful to clarify the effects of psychological stress on cardiovascular physiology, immune function, vascular reactivity, myocardial ischemia, neurobiology and cardiovascular outcomes. An emerging paradigm is that dynamic perturbations of physiological and molecular pathways during stress or negative emotions are important in influencing cardiovascular outcomes, and that some patient subgroups, such as women, patients with an early-onset myocardial infarction, and patients with adverse psychosocial exposures, may be at especially high risk for these effects. This review summarizes recent knowledge on mind-body connections in CVD among cardiac patients and highlights important pathways of risk which could become the object of future intervention efforts. As a whole, this research suggests that an integrated study of mind and body is necessary to fully understand the determinants and consequences of CVD.
Collapse
|
46
|
Kim YS, Cho HH, Cho DI, Jeong HY, Lim SY, Jun JH, Kim MR, Kang BG, Cho M, Kang HJ, Kang WS, Oh GT, Ahn Y. The adipokine Retnla deficiency increases responsiveness to cardiac repair through adiponectin-rich bone marrow cells. Cell Death Dis 2021; 12:307. [PMID: 33753732 PMCID: PMC7985519 DOI: 10.1038/s41419-021-03593-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Resistin-like alpha (Retnla) is a member of the resistin family and known to modulate fibrosis and inflammation. Here, we investigated the role of Retnla in the cardiac injury model. Myocardial infarction (MI) was induced in wild type (WT), Retnla knockout (KO), and Retnla transgenic (TG) mice. Cardiac function was assessed by echocardiography and was significantly preserved in the KO mice, while worsened in the TG group. Angiogenesis was substantially increased in the KO mice, and cardiomyocyte apoptosis was markedly suppressed in the KO mice. By Retnla treatment, the expression of p21 and the ratio of Bax to Bcl2 were increased in cardiomyocytes, while decreased in cardiac fibroblasts. Interestingly, the numbers of cardiac macrophages and unsorted bone marrow cells (UBCs) were higher in the KO mice than in the WT mice. Besides, phosphorylated histone H3(+) cells were more frequent in bone marrow of KO mice. Moreover, adiponectin in UBCs was notably higher in the KO mice compared with WT mice. In an adoptive transfer study, UBCs were isolated from KO mice to transplant to the WT infarcted heart. Cardiac function was better in the KO-UBCs transplanted group in the WT-UBCs transplanted group. Taken together, proliferative and adiponectin-rich bone marrow niche was associated with substantial cardiac recovery by suppression of cardiac apoptosis and proliferation of cardiac fibroblast.
Collapse
Affiliation(s)
- Yong Sook Kim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Biomedical Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyang Hee Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Department of Molecular Medicine, Graduate School, Chonnam National University, Gwangju, Republic of Korea
| | - Dong Im Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Biomedical Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hye-Yun Jeong
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Soo Yeon Lim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Ju Hee Jun
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Mi Ra Kim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Bo Gyeong Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Meeyoung Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hye-Jin Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Wan Seok Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Youngkeun Ahn
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea. .,Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea. .,Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea.
| |
Collapse
|
47
|
Wang M, Li Y, Zhang R, Zhang S, Feng H, Kong Z, Aiziretiaili N, Luo Z, Cai Q, Hong Y, Liu Y. Adiponectin-Transfected Endothelial Progenitor Cells Have Protective Effects After 2-Hour Middle-Cerebral Artery Occlusion in Rats With Type 2 Diabetes Mellitus. Front Neurol 2021; 12:630681. [PMID: 33746885 PMCID: PMC7966523 DOI: 10.3389/fneur.2021.630681] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/28/2021] [Indexed: 02/05/2023] Open
Abstract
Objectives: This present study aimed to examine the effects of adiponectin-transfected endothelial progenitor cells (LV-APN-EPCs) on cerebral ischemia–reperfusion injury in rats with type 2 diabetes mellitus (T2DM) and to explore the underlying mechanisms. Methods: Seventy male Sprague–Dawley rats with T2DM were randomly divided into sham, phosphate-buffered saline (PBS), LV-APN-EPCs, LV-EPCs, and EPCs groups. Transient middle cerebral artery occlusion (MCAO) was induced by the intraluminal suture method. After 1 h of reperfusion, the five interventions were performed by tail-vein injections. The modified neurological severity score (mNSS) was used to assess neurological function before and on days 1, 7, and 14 after MCAO. After 14 days, magnetic resonance imaging scanning, hematoxylin and eosin staining, terminal dUTP nick-end labeling staining, Western blotting analysis, cluster of differentiation (CD) 31 immunofluorescence, and enzyme-linked immunosorbent assay were used to evaluate infarct rate, morphological damage, cell apoptosis, and microvessel density. Results: Compared with PBS, LV-EPCs, and EPCs groups, the LV-APN-EPCs group showed significantly lower mNSS score, lower infarct rate, and less morphological damage (all P < 0.05). In addition, compared with other groups, the LV-APN-EPCs group had significantly increased levels of B cell lymphoma/leukemia-2 (Bcl-2) protein, CD31+ microvessels, endothelial nitric oxide synthase, and vascular endothelial growth factor, and decreased levels of Bcl-2-associated X protein and neuronal apoptosis in the peri-infarct cortex (all P < 0.05). Conclusion: These results suggest that LV-APN-EPCs exert protective effects against cerebral ischemia–reperfusion injury in T2DM rats by increasing angiogenesis.
Collapse
Affiliation(s)
- Meiyao Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Li
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Renwei Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuaimei Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongliang Feng
- Guangdong Mental Health Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhaohong Kong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nadire Aiziretiaili
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhengjin Luo
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qi Cai
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Hong
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yumin Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
48
|
Alexander Y, Osto E, Schmidt-Trucksäss A, Shechter M, Trifunovic D, Duncker DJ, Aboyans V, Bäck M, Badimon L, Cosentino F, De Carlo M, Dorobantu M, Harrison DG, Guzik TJ, Hoefer I, Morris PD, Norata GD, Suades R, Taddei S, Vilahur G, Waltenberger J, Weber C, Wilkinson F, Bochaton-Piallat ML, Evans PC. Endothelial function in cardiovascular medicine: a consensus paper of the European Society of Cardiology Working Groups on Atherosclerosis and Vascular Biology, Aorta and Peripheral Vascular Diseases, Coronary Pathophysiology and Microcirculation, and Thrombosis. Cardiovasc Res 2021; 117:29-42. [PMID: 32282914 PMCID: PMC7797212 DOI: 10.1093/cvr/cvaa085] [Citation(s) in RCA: 180] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/08/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells (ECs) are sentinels of cardiovascular health. Their function is reduced by the presence of cardiovascular risk factors, and is regained once pathological stimuli are removed. In this European Society for Cardiology Position Paper, we describe endothelial dysfunction as a spectrum of phenotypic states and advocate further studies to determine the role of EC subtypes in cardiovascular disease. We conclude that there is no single ideal method for measurement of endothelial function. Techniques to measure coronary epicardial and micro-vascular function are well established but they are invasive, time-consuming, and expensive. Flow-mediated dilatation (FMD) of the brachial arteries provides a non-invasive alternative but is technically challenging and requires extensive training and standardization. We, therefore, propose that a consensus methodology for FMD is universally adopted to minimize technical variation between studies, and that reference FMD values are established for different populations of healthy individuals and patient groups. Newer techniques to measure endothelial function that are relatively easy to perform, such as finger plethysmography and the retinal flicker test, have the potential for increased clinical use provided a consensus is achieved on the measurement protocol used. We recommend further clinical studies to establish reference values for these techniques and to assess their ability to improve cardiovascular risk stratification. We advocate future studies to determine whether integration of endothelial function measurements with patient-specific epigenetic data and other biomarkers can enhance the stratification of patients for differential diagnosis, disease progression, and responses to therapy.
Collapse
Affiliation(s)
- Yvonne Alexander
- Centre for Bioscience, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Elena Osto
- Institute of Clinical Chemistry, University and University Hospital Zurich, University Heart Center, Zurich, Switzerland
- Laboratory of Translational Nutrition Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Arno Schmidt-Trucksäss
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland
| | - Michael Shechter
- Leviev Heart Center, Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Danijela Trifunovic
- Cardiology Department, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Victor Aboyans
- Department of Cardiology, Dupuytren University Hospital, Inserm U-1094, Limoges University, Limoges, France
| | - Magnus Bäck
- Department of Cardiology, Center for Molecular Medicine, Karolinska University Hospital, Solna, Stockholm, Sweden
- INSERM U1116, Université de Lorraine, Centre Hospitalier Régional Universitaire de Nancy, Vandoeuvre les Nancy, France
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Francesco Cosentino
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Marco De Carlo
- Catheterization Laboratory, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Maria Dorobantu
- ‘CarolDavila’ University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Department of Medicine, Jagiellonian University Collegium Medicum, Cracow, Poland
| | - Imo Hoefer
- Laboratory of Clinical Chemistry and Hematology, University Medical Centre Utrecht, The Netherlands
| | - Paul D Morris
- Department of Infection, Immunity and Cardiovascular Disease, Bateson Centre & INSIGNEO Institute, University of Sheffield, Sheffield S10 2RX, UK
- Insigneo Institute for In Silico Medicine, Sheffield, UK
| | - Giuseppe D Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Rosa Suades
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gemma Vilahur
- Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany
- SRH Central Hospital Suhl, Suhl, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillian-Universität (LMU) München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Fiona Wilkinson
- Centre for Bioscience, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | | | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, Bateson Centre & INSIGNEO Institute, University of Sheffield, Sheffield S10 2RX, UK
- Insigneo Institute for In Silico Medicine, Sheffield, UK
| |
Collapse
|
49
|
Crea F. The complex biology of the arterial wall: much more than endothelial dysfunction and atherosclerosis. Eur Heart J 2020; 41:4215-4218. [PMID: 33295616 DOI: 10.1093/eurheartj/ehaa926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
50
|
Crea F. The growing non-pharmacological armamentarium for the treatment of cardiovascular diseases: from drug-coated balloons to drug-eluting stents, extracorporeal membrane oxygenation, and stem cells. Eur Heart J 2020; 41:3593-3597. [PMID: 33216880 DOI: 10.1093/eurheartj/ehaa854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|