1
|
Ibrahim M, Ba-Essa EM, Alvarez JA, Baker J, Bruni V, Cahn A, Ceriello A, Cosentino F, Davies MJ, De Domenico F, Eckel RH, Friedman AN, Goldney J, Hamtzany O, Isaacs S, Karadeniz S, Leslie RD, Lingvay I, McLaughlin S, Mobarak O, Del Prato S, Prattichizzo F, Rizzo M, Rötzer RD, le Roux CW, Schnell O, Seferovic PM, Somers VK, Standl E, Thomas A, Tuccinardi D, Valensi P, Umpierrez GE. Obesity and its management in primary care setting. J Diabetes Complications 2025; 39:109045. [PMID: 40305970 DOI: 10.1016/j.jdiacomp.2025.109045] [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: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Obesity is a worldwide epidemic affecting adults and children, regardless of their socioeconomic status. Significant progress has been made in understanding the genetic causes contributing to obesity, shedding light on a portion of cases worldwide. In young children with severe obesity however, recessive mutations, i.e., leptin or leptin receptor deficiency should be sought. Much more has been learned about the far-reaching impact of obesity on complications, including cardiovascular disease, liver and kidney dysfunction, diabetes, inflammation, hypertension, sleep, cancer, and the eye. Preventive strategies, particularly in children, are crucial for reducing obesity rates and mitigating its long-term complications. While dietary modifications and lifestyle changes remain the cornerstone of obesity prevention or treatment, recent advancements have introduced highly effective pharmacological options complementing weight-reduction surgery. Newer medications, like incretin-based therapies including glucagon-like peptide-1 agonists (GLP-1RA), have demonstrated remarkable efficacy in promoting weight loss, offering new insights into margining obesity-related conditions. Primary care providers, whether treating adults or children, play a pivotal role in preventing obesity, initiating treatment, and making onward referrals to specialists to assist in managing obesity and obesity-related complications.
Collapse
Affiliation(s)
| | | | - Jessica A Alvarez
- Division of Endocrinology, Lipids, and Metabolism, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Vincenzo Bruni
- Bariatric Surgery Unit, Campus Bio-Medico University, Rome, Italy
| | - Avivit Cahn
- The Diabetes Unit, Department of Endocrinology and Metabolism, Hadassah Hebrew University Hospital, Jerusalem, Israel; The faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | - Francesco Cosentino
- Unit of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Melanie J Davies
- Diabetes Research Centre, University of Leicester, Leicester, UK; NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Francesco De Domenico
- Research Unit of Endocrinology and Diabetes, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Robert H Eckel
- University of Colorado Anschutz Medical Campus and University of Colorado Hospital, Aurora, Colorado, USA
| | - Allon N Friedman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Jonathan Goldney
- Diabetes Research Centre, University of Leicester, Leicester, UK; NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Omer Hamtzany
- Division of Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Scott Isaacs
- Division of Endocrinology, Lipids, and Metabolism, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Richard David Leslie
- Blizard Institute, Centre of Immunobiology, Barts and the London School of Medicine, Queen Mary, University of London, London, UK
| | - Ildiko Lingvay
- Department of Internal Medicine/ Endocrinology and Peter O'Donnell Jr School of Public Health, UT Southwestern Medical Center at Dallas, USA
| | - Sue McLaughlin
- Department of Pharmacy and Nutrition Services, Nebraska Medicine, Department of Pediatric Endocrinology, Children's Nebraska, Omaha, NE, USA; Public Health Department, Winnebago Comprehensive Healthcare System, Winnebago, NE, USA
| | - Omar Mobarak
- Alfaisal University College of Medicine, Riyadh, Saudi Arabia
| | - Stefano Del Prato
- University of Pisa and Sant'Anna School of Advanced Studies, Pisa, Italy
| | | | - Manfredi Rizzo
- School of Medicine, Promise Department, University of Palermo, Italy; College of Medicine, Ras Al Khaimah Medical and Health Sciences University, United Arab Emirates
| | | | - Carel W le Roux
- Diabetes complications Research Centre, University College Dublin, Ireland
| | - Oliver Schnell
- Forschergruppe Diabetes eV at the Helmholtz Centre, Munich, Neuherberg, Germany
| | - Petar M Seferovic
- Academician, Serbian Academy of Sciences and Arts, Professor, University of Belgrade Faculty of Medicine and Belgrade University Medical Center, Serbia
| | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Eberhard Standl
- Forschergruppe Diabetes eV at the Helmholtz Centre, Munich, Neuherberg, Germany
| | | | - Dario Tuccinardi
- Research Unit of Endocrinology and Diabetes, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Paul Valensi
- Polyclinique d'Aubervilliers, Aubervilliers and Paris Nord University, Bobigny, France
| | - Guillermo E Umpierrez
- Division of Endocrinology, Lipids, and Metabolism, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Wassaifi S, Kaeffer B, Zarrouk S. Cellular Phenotypic Transformation During Atherosclerosis: The Potential Role of miRNAs as Biomarkers. Int J Mol Sci 2025; 26:2083. [PMID: 40076710 PMCID: PMC11900927 DOI: 10.3390/ijms26052083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 03/14/2025] Open
Abstract
Cellular phenotypic transformation is a key process that occurs during the development and progression of atherosclerosis. Within the arterial wall, endothelial cells, vascular smooth muscle cells, and macrophages undergo phenotypic changes that contribute to the pathogenesis of atherosclerosis. miRNAs have emerged as potential biomarkers for cellular phenotypic changes during atherosclerosis. Monitoring miR-155-5p, miR-210-3p, and miR-126-3p or 5p levels could provide valuable insights into disease progression, risk of complications, and response to therapeutic interventions. Moreover, miR-92a-3p's elevated levels in atherosclerotic plaques present opportunities for predicting disease progression and related complications. Baseline levels of miR-33a/b hold the potential for predicting responses to cholesterol-lowering therapies, such as statins, and the likelihood of dyslipidemia-related complications. Additionally, the assessment of miR-122-5p levels may offer insights into the efficacy of low-density-lipoprotein-lowering therapies. Understanding the specific miRNA-mediated regulatory mechanisms involved in cellular phenotypic transformations can provide valuable insights into the pathogenesis of atherosclerosis and potentially identify novel therapeutic targets.
Collapse
Affiliation(s)
- Souhir Wassaifi
- LR99E10 Human Genetics Laboratory, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia;
| | - Bertrand Kaeffer
- UMR 1280, PhAN, INRAE, Nantes Université, F-44000 Nantes, France;
| | - Sinda Zarrouk
- LR99E10 Human Genetics Laboratory, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia;
- Institut Pasteur Tunis, University of Tunis El Manar, Tunis 1068, Tunisia
| |
Collapse
|
3
|
Schröder S, Epple R, Fischer A, Schettler VJJ. Effective exosomes reduction in hypercholesterinemic patients suffering from cardiovascular diseases by lipoprotein apheresis: Exosomes apheresis. Ther Apher Dial 2024; 28:863-870. [PMID: 38837319 DOI: 10.1111/1744-9987.14172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/21/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024]
Abstract
INTRODUCTION Extracellular vesicles (EVs) have been identified as playing a role in atherosclerosis. METHODS A group of 37 hypercholesterolemic patients with atherosclerotic cardiovascular diseases (ASCVD) and 9 patients requiring hemodialysis (HD) were selected for the study. RESULTS EVs were comparably reduced by various LA methods (Thermo: 87.66% ± 3.64, DALI: 87.96% ± 4.81, H.E.L.P.: 83.38% ± 11.98; represented as SEM). However, LDL-C (66%; 55%; 75%) and Lp(a) (72%; 67%; 79%) were less effectively reduced by DALI. There was no significant difference in the reduction of EVs when comparing different techniques, such as hemoperfusion (DALI; n = 13), a precipitation (H.E.L.P.; n = 5), and a double filtration procedure (Thermofiltration; n = 19). Additionally, no effect of hemodialysis on EVs reduction was found. CONCLUSIONS The study suggests that EVs can be effectively removed by various LA procedures, and this effect appears to be independent of the specific LA procedure used, as compared to hemodialysis.
Collapse
Affiliation(s)
- Sophie Schröder
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany
| | - Robert Epple
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany
| | - Andre Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | | |
Collapse
|
4
|
Wolnik J, Adamska P, Oleksy A, Sanetra AM, Palus-Chramiec K, Lewandowski MH, Dulak J, Biniecka M. A novel 3D cardiac microtissue model for investigation of cardiovascular complications in rheumatoid arthritis. Stem Cell Res Ther 2024; 15:382. [PMID: 39468575 PMCID: PMC11520781 DOI: 10.1186/s13287-024-03956-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disease that affects not only the joints but also has significant cardiovascular (CV) manifestations. The mechanistic interplay between RA and cardiovascular complications is not yet well understood due to the lack of relevant in vitro models. In this study, we established RA cardiac microtisses (cMTs) from iPSC-derived cardiomyocytes (CMs), endothelial cells (ECs) and cardiac fibroblasts (CFs) to investigate whether this fully human 3D multicellular system could serve as a platform to elucidate the connection between RA and CV disorders. METHODS PBMC and FLS from healthy and RA donors were reprogrammed to hiPSCs with Sendai vectors. hiPSCs pluripotency was assessed by IF, FACS, spontaneous embryoid bodies formation and teratoma assay. hiPSCs were differentiated to cardiac derivatives such as CMs, ECs and CFs, followed by cell markers characterizations (IF, FACS, qRT-PCR) and functional assessments. 3D cMTs were generated by aggregation of 70% CMs, 15% ECs and 15% CFs. After 21 days in culture, structural and metabolic properties of 3D cMTs were examined by IF, qRT-PCR and Seahorse bioanalyzer. RESULTS hiPSCs demonstrated typical colony-like morphology, normal karyotype, presence of pluripotency markers, and ability to differentiate into cells originating from all three germ layers. hiPSC-CMs showed spontaneous beating and expression of cardiac markers (cTnT, MYL7, NKX2.5, MYH7). hiPSC-ECs formed sprouting spheres and tubes and expressed CD31 and CD144. hiPSC-CFs presented spindle-shaped morphology and expression of vimentin, collagen 1 and DDR2. Self-aggregation of CMs/ECs/CFs allowed development of contracting 3D cMTs, demonstrating spherical organization of the cells, which partially resembled the cardiac muscle, both in structure and function. IF analysis confirmed the expression of cTnT, CD31, CD144 and DDR2 in generated 3D cMTs. RA cMTs exhibited significantly greater formation of capillary-like structures, mimicking enhanced vascularization-key RA feature-compared to control cMTs. Seahorse examination of cMTs revealed changes in mitochondrial and glycolytic rates in the presence of metabolic substrates and inhibitors. CONCLUSIONS The cMTs model may represent an advanced human stem cell-based platform for modeling CV complications in RA. The highly developed capillary-like structures observed within RA cMTs highlight a critical feature of inflammation-induced CV dysfunction in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jan Wolnik
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Patrycja Adamska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Aleksandra Oleksy
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Anna Magdalena Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Marian Henryk Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Monika Biniecka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
- Silesian Park of Medical Technology, Kardio-Med Silesia, Zabrze, Poland.
| |
Collapse
|
5
|
Pingili A, Vempati R, Vemula M, Lakkimsetti M, Madhavaram H, Nanjundappa A, Gummadi J, Singh S, Desai R, Sunkara P. Prevalence of cardiovascular events among transgender adults with obesity: A population-based analysis. OBESITY PILLARS 2024; 11:100125. [PMID: 39252793 PMCID: PMC11381998 DOI: 10.1016/j.obpill.2024.100125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Although obesity and its impact on cardiovascular (CV) events have been extensively studied in the cisgender population, little is known about its impact on CV events in transgender individuals. Our study aimed to establish the prevalence of obesity and CV events in transgender adults. METHODS We conducted a retrospective cohort comparative study utilizing the U.S. National Inpatient Sample 2020 database. We identified admissions of transgender patients with administrative codes. Later, these patients were divided into obesity and non-obesity cohorts. Multivariable regression analysis was then performed for in-hospital all-cause mortality, acute myocardial infarction, acute ischemic stroke, cardiac arrest, pulmonary embolism and, major adverse cardiovascular and cerebrovascular events (MACCE). RESULTS In 2020, 19,345 transgender patients were admitted; 16,390 (84.7 %) had no obesity, and 2,955 (15.3 %) had obesity. The median age was 31 years in the non-obesity cohort and 37 years in the obesity cohort. Transgender men comprised 54.5 % of the non-obesity cohort and 47.9 % of the obesity cohort. Common baseline conditions in the non-obesity and obesity cohorts, respectively, included hypertension (20.7 % vs. 43.5 %), diabetes (10.2 % vs. 32.5 %), chronic pulmonary disease (18.9 % vs. 27.7 %), and hyperlipidemia (11.5 % vs. 25 %). MACCE was observed in 2.3 % of the non-obesity cohort compared to 5.4 % in the obesity cohort, and cardiac arrest occurred in 0.2 % of the non-obesity cohort versus 1.2 % in the obesity cohort. A statistically significant association was found in MACCE [odds ratio (OR) 2.1, 95 % confidence interval (CI) 1.24-3.55, p = 0.006] and cardiac arrest [OR 3.92, 95 % CI 1.11-12.63, p = 0.022] among transgender patients with obesity. CONCLUSION We observed increased odds of MACCE and cardiac arrest in transgender patients with obesity, possibly due to obesity and CV risk factors like hypertension, diabetes, and hyperlipidemia. Further large-scale comparative studies are needed to better understand obesity's impact on CV outcomes in the transgender population.
Collapse
Affiliation(s)
- Adhvithi Pingili
- Department of Internal Medicine, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Roopeessh Vempati
- Department of Internal Medicine, Trinity Health Oakland Hospital, Pontiac, MI, USA
| | - Madhusha Vemula
- Department of General Medicine, Malla Reddy Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Mohit Lakkimsetti
- Department of General Medicine, Mamata Medical College, Khammam, Telangana, India
| | - Hasmitha Madhavaram
- Department of Internal Medicine, Morristown Medical Centre, Morristown, NJ, USA
| | | | - Jyotsna Gummadi
- Department of Medicine, MedStar Franklin Square Medical Centre, Baltimore, MD, USA
| | - Sandeep Singh
- University Hospitals of North Midlands MHS Trust, Stroke-on-Trent, UK
| | - Rupak Desai
- Independent Researcher, Outcomes Research, Atlanta, GA, USA
| | - Praveena Sunkara
- Department of Internal Medicine, Passion Health Primary Care, Denton, TX, USA
| |
Collapse
|
6
|
Pingili A, Vempati R, Vemula M, Lakkimsetti M, Madhavaram H, Nanjundappa A, Gummadi J, Singh S, Desai R, Sunkara P. Prevalence of cardiovascular events among transgender adults with obesity: A population-based analysis. OBESITY PILLARS 2024; 11:100125. [DOI: 10.1016/j.obpill.2024.100125 | pmid: 39252793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
|
7
|
Lei S, Liu C, Zheng TX, Fu W, Huang MZ. The relationship of redox signaling with the risk for atherosclerosis. Front Pharmacol 2024; 15:1430293. [PMID: 39148537 PMCID: PMC11324460 DOI: 10.3389/fphar.2024.1430293] [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: 05/09/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
Oxidative balance plays a pivotal role in physiological homeostasis, and many diseases, particularly age-related conditions, are closely associated with oxidative imbalance. While the strategic role of oxidative regulation in various diseases is well-established, the specific involvement of oxidative stress in atherosclerosis remains elusive. Atherosclerosis is a chronic inflammatory disorder characterized by plaque formation within the arteries. Alterations in the oxidative status of vascular tissues are linked to the onset, progression, and outcome of atherosclerosis. This review examines the role of redox signaling in atherosclerosis, including its impact on risk factors such as dyslipidemia, hyperglycemia, inflammation, and unhealthy lifestyle, along with dysregulation, vascular homeostasis, immune system interaction, and therapeutic considerations. Understanding redox signal transduction and the regulation of redox signaling will offer valuable insights into the pathogenesis of atherosclerosis and guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sujuan Lei
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chen Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Tian-Xiang Zheng
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Mei-Zhou Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| |
Collapse
|
8
|
Zhang L, Xiang Y, Cao C, Tan J, Li F, Yang X. Ciliary neurotrophic factor promotes the development of homocysteine-induced vascular endothelial injury through inflammation mediated by the JAK2/STAT3 signaling pathway. Exp Cell Res 2024; 440:114103. [PMID: 38848951 DOI: 10.1016/j.yexcr.2024.114103] [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: 04/25/2024] [Revised: 05/22/2024] [Accepted: 05/26/2024] [Indexed: 06/09/2024]
Abstract
Elevated homocysteine (Hcy) levels have been recognized as significant risk factor for cardiovascular and cerebrovascular diseases, closely related to endothelial injury. While expression of Ciliary Neurotrophic Factor (CNTF) significantly increases during Hcy-induced vascular endothelial cell injury, the precise molecular pathways through which CNTF operates remain to be clarified. To induce vascular endothelial cell injury, human umbilical vein endothelial cells (HUVECs) were treated with Hcy. Cell viability and apoptosis in HUVECs were assessed using the CCK-8 assay and flow cytometry. Western blot analysis determined the expression levels of the JAK2-STAT3 pathway, inflammation-related factors (IL-1β, NLRP3, ICAM-1, VCAM-1), and apoptosis-related factors (cleaved Caspase-3 and Bax). Immunofluorescence staining and western blotting were employed to examine CD31 and α-SMA expression. Knockdown of CNTF was achieved using lentiviral interference, and its effects on inflammation and cell injury were evaluated. Chromatin immunoprecipitation (ChIP) and dual luciferase reporter analysis were conducted to investigate the interaction between the MAFK and CNTF promoters. Our results indicated that Hcy induced high expression of CNTF and activated the JAK2-STAT3 signaling pathway, thereby upregulating factors associated with inflammation and cell apoptosis. Inhibiting CNTF alleviated Hcy-induced inflammation and cell injury. MAFK was identified as a transcription factor promoting CNTF transcription, and its overexpression exacerbated inflammation and cell injury in Hcy-treated HUVECs through the CNTF-JAK2-STAT3 axis, which could be reversed by knocking down CNTF. Activation of MAFK leads to CNTF upregulation, which activates the JAK2-STAT3 signaling pathway, regulating inflammation and inducing injury in Hcy-exposed vascular endothelial cells. Targeting CNTF or its upstream regulator MAFK may represent potential therapeutic strategies for mitigating endothelial dysfunction associated with hyperhomocysteinemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Yan Xiang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China; Ouyang Road Community Health Service Center, Hongkou District, Shanghai, China
| | - Chengxiu Cao
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Jiaorong Tan
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Fei Li
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Xin Yang
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China.
| |
Collapse
|
9
|
Gasparotto M, Di Pierro G, Toffoli B, Grillo A, Bressan M, Fiorentin M, Di Luozzo L, Fischetti F, Zen M, Fabris B, Bernardi S, Tomietto P. Preliminary Study on Pulse Wave Changes in Patients with Inflammatory Arthropathies Treated with bDMARDs. J Clin Med 2024; 13:2684. [PMID: 38731213 PMCID: PMC11084438 DOI: 10.3390/jcm13092684] [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: 03/30/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Patients with inflammatory arthropathies exhibit an increased cardiovascular disease (CVD) risk as compared to the general population, which is not fully quantified by the conventional CVD risk scores. Biotechnological disease-modifying drugs (bDMARDs) have proved beneficial to reduce the overall CVD risk in these patients, although CVD remains a major cause of increased mortality. Since it has been shown that pulse wave parameters and in particular carotid-femoral pulse wave velocity (cfPWV) are predictors of CVD risk, the aim of this study was to evaluate their changes in patients with inflammatory arthropathies before and after bDMARD therapy. Methods: Pulse wave parameters were evaluated with applanation tonometry in patients with ankylosing spondylitis (AS), psoriatic arthritis (PsA), and rheumatoid arthritis (RA), before and after two years of bDMARD therapy. Results: At baseline, cfPWV was significantly associated with age (p < 0.001) and, among pulse wave parameters, the subendocardial viability ratio was negatively associated with C-reactive protein (CRP) (p = 0.04) and the HAQ-disability index (p = 0.03). At baseline, PsA patients showed a higher percentage of male subjects, higher CRP, and the highest cfPWV values (p = 0.048). After two years, pulse wave parameters improved in the AS and RA groups, but not in the PsA group. Conclusions: Our data confirm that pulse wave parameters are potentially reversible after bDMARD therapy, as they improved in AS and RA patients. In PsA patients, there were no changes, which may be due to the higher percentage of male subjects and higher baseline cfPWV values.
Collapse
Affiliation(s)
- Michela Gasparotto
- UCO Medicina Clinica, ASUGI, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (M.G.); (A.G.); (F.F.) (B.F.); (P.T.)
- Department of Medicine, Padua University Hospital, University of Padua, 35122 Padova, Italy;
| | - Giuliano Di Pierro
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Barbara Toffoli
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Andrea Grillo
- UCO Medicina Clinica, ASUGI, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (M.G.); (A.G.); (F.F.) (B.F.); (P.T.)
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Marco Bressan
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Marco Fiorentin
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Lorenzo Di Luozzo
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Fabio Fischetti
- UCO Medicina Clinica, ASUGI, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (M.G.); (A.G.); (F.F.) (B.F.); (P.T.)
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Margherita Zen
- Department of Medicine, Padua University Hospital, University of Padua, 35122 Padova, Italy;
| | - Bruno Fabris
- UCO Medicina Clinica, ASUGI, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (M.G.); (A.G.); (F.F.) (B.F.); (P.T.)
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Stella Bernardi
- UCO Medicina Clinica, ASUGI, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (M.G.); (A.G.); (F.F.) (B.F.); (P.T.)
- Department of Medical Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (G.D.P.); (B.T.); (M.B.); (M.F.); (L.D.L.)
| | - Paola Tomietto
- UCO Medicina Clinica, ASUGI, Cattinara Teaching Hospital, Strada di Fiume 447, 34149 Trieste, Italy; (M.G.); (A.G.); (F.F.) (B.F.); (P.T.)
| |
Collapse
|
10
|
Artac I, Karakayali M, Omar T, Ilis D, Arslan A, Hakan Sahin M, Kina S, Karabag Y, Rencuzogullari I. Predictive Value of the Naples Prognostic Score on Long-Term Outcomes in Patients with Peripheral Artery Disease Revascularized via Percutaneous Intervention. Ann Vasc Surg 2024; 102:121-132. [PMID: 38307231 DOI: 10.1016/j.avsg.2023.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/15/2023] [Accepted: 11/01/2023] [Indexed: 02/04/2024]
Abstract
BACKGROUND Lower extremity peripheral artery disease (PAD) is the third most common clinical manifestation of atherosclerosis after coronary artery disease and stroke. Despite successful endovascular treatment (EVT), mortality and morbidity rates still remain higher in patients with PAD. Naples prognostic score (NPS) is a novel scoring system, reflects the patient's nutritional and immunological statuses as well as systemic inflammatory responses. In this study, we aimed to investigate the relationship between NPS and long-term outcomes in patients with PAD. METHODS The population of this retrospective study consisted of 629 PAD patients who underwent EVT at Kafkas University Hospital between 2020 and 2023. For each patient, the NPS was calculated and then patients were divided into 3 groups based on their NPS. The primary end point of the study was the rate of major adverse cardiovascular (MACEs) and limb events (MALEs), that is, all-cause death or development of critical limb ischemia with consequent amputation. RESULTS Of a total of 629 patients, 62 were classified into group 0 (NPS 0), 315 into group 1 (NPS 1 or 2), and 252 into group 2 (NPS 3 or 4). The distribution of patients' baseline characteristics, angiographic features and MACEs and MALEs according to the NPS groups was analyzed. Significant adverse outcomes differences were observed among the 3 groups (P < 0.001). Multivariate logistic regression analysis revealed that age, diabetes mellitus, chronic kidney disease, lowest preprocedure ankle-brachial index, left ventricular ejection fraction and NPS (hazard ratio 1.916, 95% confidence interval [CI] 1.530-2.398, P < 0.001) were independent predictors of MACE whereas diabetes mellitus, presence of previous PAD, hemoglobin level, in-hospital acute thrombotic occlusion and NPS (odds ratio 1.963, 95% CI 1.489-2.588, P < 0.001) were independent predictors of MALE. CONCLUSIONS The inflammatory and nutritional state reflected by NPS levels was strongly associated with all-cause mortality and amputation after EVT in patients with PAD. Furthermore, NPS was found to be an independent predictor of these clinical outcomes.
Collapse
Affiliation(s)
- Inanc Artac
- Department of Cardiology, Kafkas University Faculty of Medicine, Kars, Turkey.
| | - Muammer Karakayali
- Department of Cardiology, Kafkas University Faculty of Medicine, Kars, Turkey
| | - Timor Omar
- Department of Cardiology, Kafkas University Faculty of Medicine, Kars, Turkey
| | - Dogan Ilis
- Department of Cardiology, Kafkas University Faculty of Medicine, Kars, Turkey
| | - Ayca Arslan
- Department of Cardiology, Kafkas University Faculty of Medicine, Kars, Turkey
| | - Mehmet Hakan Sahin
- Department of Cardiology, Kafkas University Faculty of Medicine, Kars, Turkey
| | - Soner Kina
- Department of Anesthesiology and Reanimation, Kafkas University Faculty of Medicine, Kars, Turkey
| | - Yavuz Karabag
- Department of Cardiology, Kafkas University Faculty of Medicine, Kars, Turkey
| | | |
Collapse
|
11
|
Darapaneni H, Lakhanpal S, Chhayani H, Parikh K, Patel M, Gupta V, Anamika F, Munjal R, Jain R. Shedding light on weight loss: A narrative review of medications for treating obesity. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:3-11. [PMID: 37752761 DOI: 10.2478/rjim-2023-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Indexed: 09/28/2023]
Abstract
Obesity and overweight are the major risk factors for numerous chronic diseases, including cardiovascular diseases such as heart disease and stroke, which are the leading causes of death worldwide. The prevalence of obesity has dramatically risen in both developed and developing countries, making it a significant public health concern and a global crisis. Despite lifestyle modifications being the first-line treatment, the high risk of relapse has led to a growing interest in non-invasive pharmacotherapeutic interventions to achieve and maintain weight loss and reverse the growth of the obesity epidemic. Cardiovascular diseases and cancer account for the highest mortality rates among other comorbidities associated with obesity and overweight. Excess and abnormally deposited adipose tissue secretes various inflammatory mediators, leading to cardiovascular diseases and cancers. Weight loss of 5-10% significantly reduces cardiometabolic risk. Medications currently approved in the USA for long-term management of obesity are orlistat, naltrexone, bupropion, phentermine/topiramate, and Glucagon Like Peptide-1 (GLP-1) agonists such as liraglutide and semaglutide. The benefit-to-risk of medications, comorbidities, and individual responses should guide the treatment decisions. The article provides a comprehensive overview and discussion of several weight loss medications used previously and currently, including their efficacy, mechanisms of action, and side effects.
Collapse
Affiliation(s)
| | | | | | - Kinna Parikh
- G.M.E.R.S. Medical College, Gandhinagar, Gujarat, India
| | - Meet Patel
- Tianjin Medical University, Tianjin, China
| | - Vasu Gupta
- Dayanand Medical College and Hospital, Ludhiana, India
| | - Fnu Anamika
- University College of Medical Sciences, New Delhi, India
| | | | - Rohit Jain
- Penn state Milton S. Hershey Medical Center, Hershey, USA
| |
Collapse
|
12
|
Kirwan R, Mazidi M, Butler T, Perez de Heredia F, Lip GYH, Davies IG. The association of appendicular lean mass and grip strength with low-density lipoprotein, very low-density lipoprotein, and high-density lipoprotein particle diameter: a Mendelian randomization study of the UK Biobank cohort. EUROPEAN HEART JOURNAL OPEN 2024; 4:oeae019. [PMID: 38595990 PMCID: PMC11003544 DOI: 10.1093/ehjopen/oeae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 04/11/2024]
Abstract
Aims Reduced muscle mass and reduced strength are frequently associated with both alterations in blood lipids and poorer cardiometabolic outcomes in epidemiological studies; however, a causal association cannot be determined from such observations. Two-sample Mendelian randomization (MR) was applied to assess the association of genetically determined appendicular lean mass (ALM) and handgrip strength (HGS) with serum lipid particle diameter. Methods and results Mendelian randomization was implemented using summary-level data from the largest genome-wide association studies on ALM (n = 450 243), HGS (n = 223 315), and lipoprotein [low-density lipoprotein (LDL), very LDL (VLDL), and high-density lipoprotein (HDL)] particle diameters (n = 115 078). Inverse variance-weighted (IVW) method was used to calculate the causal estimates. Weighted median-based method, MR-Egger, and leave-one-out method were applied as sensitivity analysis. Greater ALM had a statistically significant positive effect on HDL particle diameter (MR-Egger: β = 0.055, SE = 0.031, P = 0.081; IVW: β = 0.068, SE = 0.014, P < 0.001) and a statistically significant negative effect on VLDL particle diameter (MR-Egger: β = -0.114, SE = 0.039, P = 0.003; IVW: β = -0.081, SE = 0.017, P < 0.001). Similarly, greater HGS had a statistically significant positive effect on HDL particle diameter (MR-Egger: β = 0.433, SE = 0.184, P = 0.019; IVW: β = 0.121, SE = 0.052, P = 0.021) and a statistically significant negative effect on VLDL particle diameter (MR-Egger: β = -0.416, SE = 0.163, P = 0.011; IVW: β = -0.122, SE = 0.046, P = 0.009). There was no statistically significant effect of either ALM or HGS on LDL particle diameter. Conclusion There were potentially causal associations between both increasing ALM and HGS and increasing HDL particle size and decreasing VLDL particle size. These causal associations may offer possibilities for interventions aimed at improving cardiovascular disease risk profile.
Collapse
Affiliation(s)
- Richard Kirwan
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool, UK
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Mohsen Mazidi
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, Liverpool Heart and Chest Hospital, Liverpool, UK
- Clinical Trial Service Unit, Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Roosevelt Dr., Doll Bldg, Oxford, OX3 7LF, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Tom Butler
- School of Applied Health and Social Care and Social Work, Faculty of Health, Social Care and Medicine, Edge Hill University, Ormskirk, UK
| | - Fatima Perez de Heredia
- School of Biological and Environmental Sciences, Liverpool John Moores University, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, Liverpool Heart and Chest Hospital, Liverpool, UK
- Danish Center for Clinical Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Ian G Davies
- Research Institute of Sport and Exercise Science, Liverpool John Moores University, Liverpool, UK
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, Liverpool Heart and Chest Hospital, Liverpool, UK
| |
Collapse
|
13
|
Walker LE, Stewart A, Pirmohamed SM, Meschia JF, Kinne FB. Stroke Pharmacogenetics. STROKE GENETICS 2024:423-508. [DOI: 10.1007/978-3-031-41777-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
14
|
Edsfeldt A, Singh P, Matthes F, Tengryd C, Cavalera M, Bengtsson E, Dunér P, Volkov P, Karadimou G, Gisterå A, Orho-Melander M, Nilsson J, Sun J, Gonçalves I. Transforming growth factor-β2 is associated with atherosclerotic plaque stability and lower risk for cardiovascular events. Cardiovasc Res 2023; 119:2061-2073. [PMID: 37200403 PMCID: PMC10478752 DOI: 10.1093/cvr/cvad079] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 01/27/2023] [Accepted: 02/21/2023] [Indexed: 05/20/2023] Open
Abstract
AIMS Transforming growth factor-beta (TGF-β) exists in three isoforms TGF-β1, -β2, and -β3. TGF-β1 has been suggested to be important for maintaining plaque stability, yet the role of TGF-β2 and -β3 in atherosclerosis remains to be investigated.This study explores the association of the three isoforms of TGF-β with plaque stability in the human atherosclerotic disease. METHODS AND RESULTS TGF-β1, -β2, and -β3 proteins were quantified in 223 human carotid plaques by immunoassays. Indications for the endarterectomy were: symptomatic carotid plaque with stenosis >70% or without symptoms and >80% stenosis. Plaque mRNA levels were assessed by RNA sequencing. Plaque components and extracellular matrix were measured histologically and biochemically. Matrix metalloproteinases and monocyte chemoattractant protein-1 (MCP-1) was measured with immunoassays. The effect of TGF-β2 on inflammation and protease activity was investigated in vitro using THP-1 and RAW264.7 macrophages. Patients were followed longitudinally for cardiovascular (CV) events.TGF-β2 was the most abundant isoform and was increased at both protein and mRNA levels in asymptomatic plaques. TGF-β2 was the main determinant separating asymptomatic plaques in an Orthogonal Projections to Latent Structures Discriminant Analysis. TGF-β2 correlated positively to features of plaque stability and inversely to markers of plaque vulnerability. TGF-β2 was the only isoform inversely correlated to the matrix-degrading matrix metalloproteinase-9 and inflammation in the plaque tissue. In vitro, TGF-β2 pre-treatment reduced MCP-1 gene and protein levels as well as matrix metalloproteinase-9 gene levels and activity. Patients with plaques with high TGF-β2 levels had a lower risk to suffer from future CV events. CONCLUSIONS TGF-β2 is the most abundant TGF-β isoform in human plaques and may maintain plaque stability by decreasing inflammation and matrix degradation.
Collapse
Affiliation(s)
- Andreas Edsfeldt
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Pratibha Singh
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Frank Matthes
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | | | - Michele Cavalera
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms—Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Pontus Dunér
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Petr Volkov
- Department of Clinical Sciences, LUDC Bioinformatics Unit, Malmö, Lund University, Lund, Sweden
- Data Science and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Glykeria Karadimou
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Anton Gisterå
- Department of Medicine, Center for Molecular Medicine, Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Nilsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Jiangming Sun
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Isabel Gonçalves
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
15
|
Mureșan AV, Tomac A, Opriș DR, Bandici BC, Coșarcă CM, Covalcic DC, Hălmaciu I, Akácsos-Szász OZ, Rădulescu F, Lázár K, Stoian A, Tilinca MC. Inflammatory Markers Used as Predictors of Subclinical Atherosclerosis in Patients with Diabetic Polyneuropathy. Life (Basel) 2023; 13:1861. [PMID: 37763265 PMCID: PMC10532684 DOI: 10.3390/life13091861] [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: 08/01/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND peripheral arterial disease (PAD) is identified late in diabetic patients because, in the majority of cases, it is associated with diabetic peripheral neuropathy, resulting in little or no symptoms, or symptoms that are completely neglected. METHODS In this study were enrolled all patients over 18 years of age, with diabetes mellitus type II for more than a year with poor glycemic control, diagnosed with diabetic polyneuropathy admitted to the Diabetology Department, Emergency County Hospital of Targu Mures, Romania between January 2020 and March 2023. We divided the patients into two groups, based on the presence or absence of subclinical atherosclerosis in the lower limb, named "SA" and "non-SA". RESULTS Patients in the SA group were older (p = 0.01) and had a higher incidence of IHD (p = 0.03), history of MI (p = 0.02), and diabetic nephropathy (p = 0.01). Moreover, patients with subclinical atherosclerosis had a higher BMI (p < 0.0001) and a longer duration of diabetes (p < 0.0001). Among all patients, the systemic inflammatory markers, MLR (r = 0.331, p < 0.001), NLR (r = 0.517, p < 0.001), PLR (r = 0.296, p < 0.001), SII (r = 0.413, p < 0.001), as well as BMI (r = 0.241, p < 0.001) and HbA1C (r = 0.489, p < 0.001), demonstrated a strong positive correlation with the diabetes duration. The multivariate logistic regression analysis showed that older patients (OR: 2.58, p < 0.001), the male gender (OR: 2.30, p = 0.006), a higher baseline levels of BMI (OR: 7.71, p < 0.001), and the duration of diabetes (OR: 8.65, p < 0.001) are predictors of subclinical atherosclerosis in DN patients. Additionally, the high baseline levels of all systemic inflammatory markers (for all: p < 0.001) and poor diabetes management (OR: 10.4, p < 0.001 for HbA1C; OR: 10.78, p < 0.001 for admission glucose) are independent predictors of SA. CONCLUSIONS the inflammatory markers, NLR, MLR, PLR, and SII, being cheap and easy to collect in routine medical practice from the standard blood tests, could be an important step in predicting vascular outcomes in diabetic patients and the disease's progression, playing a key role in follow-up visits in type-2 diabetic patients and PAD patients.
Collapse
Affiliation(s)
- Adrian Vasile Mureșan
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania; (A.V.M.); (C.M.C.)
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Alexandru Tomac
- Clinic of Plastic Surgery, Emergency Clinical Hospital Saint Spiridon, 700111 Iasi, Romania;
| | - Diana Roxana Opriș
- Emergency Institute for Cardiovascular Diseases and Transplantation (IUBCVT) of Targu Mures, 540136 Targu Mures, Romania
| | - Bogdan Corneliu Bandici
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania; (A.V.M.); (C.M.C.)
| | - Cătălin Mircea Coșarcă
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania; (A.V.M.); (C.M.C.)
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Diana Carina Covalcic
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania; (A.V.M.); (C.M.C.)
| | - Ioana Hălmaciu
- Department of Radiology, Mures County Emergency Hospital, 540136 Targu Mures, Romania;
| | - Orsolya-Zsuzsa Akácsos-Szász
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Flavia Rădulescu
- Clinical Department of Endocrinology, Mures County Emergency Hospital, 540136 Targu Mures, Romania; (F.R.)
- Department of Scientific Research Methodology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Krisztina Lázár
- Clinical Department of Endocrinology, Mures County Emergency Hospital, 540136 Targu Mures, Romania; (F.R.)
| | - Adina Stoian
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Mariana Cornelia Tilinca
- Department of Diabetes, Nutrition and Metabolic Diseases, Mures County Emergency Hospital, 540136 Targu Mures, Romania;
- Department of Internal Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| |
Collapse
|
16
|
Parsons AJ, Stein KR, Atanasoff KE, Ophir SI, Casado JP, Tortorella D. The CD46 ectodomain participates in human cytomegalovirus infection of epithelial cells. J Gen Virol 2023; 104:001892. [PMID: 37668349 PMCID: PMC10484303 DOI: 10.1099/jgv.0.001892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/18/2023] [Indexed: 09/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) primary infections are typically asymptomatic in healthy individuals yet can cause increased morbidity and mortality in organ transplant recipients, AIDS patients, neonates, and the elderly. The successful, widespread dissemination of this virus among the population can be attributed in part to its wide cellular tropism and ability to establish life-long latency. HCMV infection is a multi-step process that requires numerous cellular and viral factors. The viral envelope consists of envelope protein complexes that interact with cellular factors; such interactions dictate virus recognition and attachment to different cell types, followed by fusion either at the cell membrane or within an endocytic vesicle. Several HCMV entry factors, including neuropilin-2 (Nrp2), THBD, CD147, OR14I1, and CD46, are characterized as participating in HCMV pentamer-specific entry of non-fibroblast cells such as epithelial, trophoblast, and endothelial cells, respectively. This study focuses on characterizing the structural elements of CD46 that impact HCMV infection. Infectivity studies of wild-type and CD46 knockout epithelial cells demonstrated that levels of CD46 expressed on the cell surface were directly related to HCMV infectivity. Overexpression of CD46 isomers BC1, BC2, and C2 enhanced infection. Further, CD46 knockout epithelial cells expressing CD46 deletion and chimeric molecules identified that the intact ectodomain was critical for rescue of HCMV infection in CD46 knockout cells. Collectively, these data support a model that the extracellular domain of CD46 participates in HCMV infection due to its surface expression.
Collapse
Affiliation(s)
- Andrea J. Parsons
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kathryn R. Stein
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristina E. Atanasoff
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sabrina I. Ophir
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jailene Paredes Casado
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Domenico Tortorella
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
17
|
Simeunovic A, Brunborg C, Heier M, Seljeflot I, Dahl-Jørgensen K, Margeirsdottir HD. Sustained low-grade inflammation in young participants with childhood onset type 1 diabetes: The Norwegian atherosclerosis and childhood diabetes (ACD) study. Atherosclerosis 2023; 379:117151. [PMID: 37349194 DOI: 10.1016/j.atherosclerosis.2023.05.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND AND AIMS Persons with type 1 diabetes (T1D) have increased mortality from cardiovascular disease. Early inflammation is important in the development of atherosclerosis. We aimed to evaluate the extent of inflammation and difference in mean over a five-year period in young persons with T1D compared to healthy controls. METHODS The Norwegian Atherosclerosis and Childhood Diabetes (ACD) study is a prospective population-based cohort study on atherosclerosis development in childhood-onset T1D compared to healthy controls, with follow-ups every fifth year. The original study cohort consisted of 314 children with T1D on intensive insulin treatment and 120 healthy controls of similar age. Circulating levels of VCAM-1, TNA-α, P-selectin, E-selectin, CRP, IL-6, IL-18, MCP-1, MMP-9 and TIMP-1 were measured by ELISAs at baseline and at the five-year follow-up. RESULTS The group with T1D had mean age 13.7 (SD = 2.8) years, disease duration 5.6 (SD = 3.4) years and HbA1c 68 (SD = 13.1) mmol/mol at baseline. Levels of almost all inflammatory markers were significantly increased in the group with T1D compared to controls, and significant mean-difference between the two groups over the five-year period was observed in four markers: IL-18, P-selectin, E-selectin and TIMP-1. CONCLUSIONS The early low-grade inflammation present in young individuals with T1D five years after diagnosis is sustained at ten-year disease duration, with moderate changes for most markers of inflammation over time. The evolving inflammatory profile indicates an accelerated chain of events in the progression of early atheromatosis in T1D.
Collapse
Affiliation(s)
- Aida Simeunovic
- Department of Paediatric and Adolescent Medicine, Akershus University Hospital, Lørenskog, Norway; Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway.
| | - Cathrine Brunborg
- Centre for Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital, Oslo, Norway
| | - Martin Heier
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| | - Ingebjørg Seljeflot
- University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Center for Clinical Heart Research and Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Knut Dahl-Jørgensen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| | - Hanna Dis Margeirsdottir
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| |
Collapse
|
18
|
Lin W, Huang F, Yuan Y, Li Q, Lin Z, Zhu W, Lin B, Zhu P. Endothelial exosomes work as a functional mediator to activate macrophages. Front Immunol 2023; 14:1169471. [PMID: 37575264 PMCID: PMC10416261 DOI: 10.3389/fimmu.2023.1169471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Intercellular communication is essential for almost all physiological and pathological processes. Endothelial cell (EC)-derived exosomes, working as mediators for intercellular information exchange, are involved in the pathophysiological mechanisms of atherosclerosis. However, the effect of inflamed endothelial exosomes on the function of macrophages (Mϕ) is poorly defined. This study aims to unravel how exosomes derived from tumor necrosis factor-α (TNF-α)-stimulated ECs (exo-T) affect Mϕ in vitro. Methods and results Exosomes derived from untreated ECs (exo) and exo-T were identified by using TEM, NTA, and western blot, and we observed that PKH67-labeled exo/exo-T were taken up by Mϕ. Exposure to exo-T for 24 h not only skewed Mϕ to the M1 subtype and exacerbated lipid deposition, but also promoted Mϕ apoptosis, while it did not significantly affect Mϕ migration, as detected by RT-qPCR, Dil-ox-LDL uptake assay, flow cytometry, wound healing assay, and transwell assay, respectively. In addition, exo/exo-T-related microRNA-Seq revealed 104 significantly differentially expressed microRNAs (DE-miRNAs). The target genes of DE-miRNAs were mainly enriched functionally in metabolic pathways, MAPK signaling pathway, etc., as determined using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. We further demonstrated by immunoblotting that exo-T intervention improves the phosphorylation of MAPK/NF-κB-related proteins. Discussion and conclusion Collectively, this study reveals that inflamed endothelial exosomes (TNF-α-stimulated EC-derived exosomes) work as a functional mediator to affect Mϕ function and may activate Mϕ through MAPK/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Wenwen Lin
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, China
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, China
- Fujian Provincial Center of Geriatrics, Fuzhou, China
| | - Feng Huang
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, China
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, China
- Fujian Provincial Center of Geriatrics, Fuzhou, China
| | - Yin Yuan
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, China
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, China
- Fujian Provincial Center of Geriatrics, Fuzhou, China
| | - Qiaowei Li
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, China
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, China
- Fujian Provincial Center of Geriatrics, Fuzhou, China
| | - Zhong Lin
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, China
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, China
- Fujian Provincial Center of Geriatrics, Fuzhou, China
| | - Wenqing Zhu
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, China
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, China
- Fujian Provincial Center of Geriatrics, Fuzhou, China
| | - Binbin Lin
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, China
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, China
- Fujian Provincial Center of Geriatrics, Fuzhou, China
| | - Pengli Zhu
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Provincial Key Laboratory of Geriatrics, Fuzhou, China
- Fujian Provincial Institute of Clinical Geriatrics, Fuzhou, China
- Fujian Provincial Center of Geriatrics, Fuzhou, China
| |
Collapse
|
19
|
Penn MS, MacRae C, Goldfaden RF, Choksi RR, Smith S, Wrenn D, Saghir MX, Klemes AB. Association of chronic neutrophil activation with risk of mortality. PLoS One 2023; 18:e0288712. [PMID: 37471318 PMCID: PMC10358907 DOI: 10.1371/journal.pone.0288712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/03/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Levels of free myeloperoxidase (MPO), a cardiovascular risk marker, have been reported to decline with standard care. Whether such declines signify decreased risk of mortality remains unknown. DESIGN Cox proportional hazard models were generated using data from a retrospective cohort study of prospectively collected measures. PARTICIPANTS Patients (3,658) who had MPO measurements and LDL-C ≥ 90 mg/dL during 2011-2015 were selected based on a stratified random sampling on MPO risk level. Baseline MPO was either low (<470 pmol/L), moderate (470-539 pmol/L), or high (≥540 pmol/L). MAIN OUTCOMES AND MEASURES First occurrence of MACE (myocardial infarction, stroke, coronary revascularization, or all-cause death). RESULTS Mean age was 66.5 years, and 64.7% were women. During a mean 6.5-year follow-up, crude incidence per 1000 patient years was driven by death. The incidence and all-cause death was highest for patients with high MPO (21.2; 95% CI, 19.0-23.7), then moderate (14.6; 95% CI, 11.5-18.5) and low (2.3; 95% CI, 1.2-4.6) MPO. After adjusting for age, sex, and cardiovascular risk factors, risk of cardiovascular death did not differ significantly between patients with high and low MPO (HR, 1.57; 95% CI, 0.56-4.39), but patients with high MPO had greater risk of non-cardiovascular (HR, 6.15; 95% CI, 2.27-16.64) and all-cause (HR, 3.83; 95% CI, 1.88-7.78) death. During follow-up, a 100 pmol/L decrease in MPO correlated with a 5% reduction in mortality (HR, 0.95; 95% CI, 0.93-0.97) over 5 years. CONCLUSIONS Free circulating MPO is a strong marker of risk of mortality. Monitoring changes in MPO levels over time may provide insight into changes in physiology that mark a patient for increased risk of mortality.
Collapse
Affiliation(s)
- Marc S Penn
- Summa Health Heart and Vascular Institute, Summa Health, Akron, Ohio, United States of America
- Quest Center of Excellence for Cardiometabolic Testing at Cleveland HeartLab, Cleveland, Ohio, United States of America
| | - Calum MacRae
- Department of Medicine, One Brave Idea - American Heart Association, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rebecca F Goldfaden
- East Coast Institute of Research, Jacksonville, Florida, United States of America
| | - Rushab R Choksi
- East Coast Institute of Research, Jacksonville, Florida, United States of America
| | - Steven Smith
- Department of Pharmacotherapy & Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida, United States of America
| | - David Wrenn
- Quest Center of Excellence for Cardiometabolic Testing at Cleveland HeartLab, Cleveland, Ohio, United States of America
| | - Mouris X Saghir
- Quest Center of Excellence for Cardiometabolic Testing at Cleveland HeartLab, Cleveland, Ohio, United States of America
| | | |
Collapse
|
20
|
Sobolevskaya EV, Shumkov OA, Smagin MA, Guskov AE, Malysheva AV, Atuchin VV, Nimaev VV. Markers of Restenosis after Percutaneous Transluminal Balloon Angioplasty in Patients with Critical Limb Ischemia. Int J Mol Sci 2023; 24:ijms24109096. [PMID: 37240440 DOI: 10.3390/ijms24109096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/14/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Among cardiovascular diseases, chronic obliterating lesions of the arteries of lower extremities, which are one of the important problems of modern healthcare, are distinguished. In most cases, the cause of damage to the arteries of lower extremities is atherosclerosis. The most severe form is chronic ischemia, characterized by pain at rest and ischemic ulcers, ultimately increasing the risk of limb loss and cardiovascular mortality. Therefore, patients with critical limb ischemia need limb revascularization. Percutaneous transluminal balloon angioplasty is one of the least invasive and safe approaches, with advantages for patients with comorbidities. However, after this procedure, restenosis is still possible. Early detection of changes in the composition of some molecules as markers of restenosis will help screen patients at the risk of restenosis, as well as find ways to apply efforts for further directions of inhibition of this process. The purpose of this review is to provide the most important and up-to-date information on the mechanisms of restenosis development, as well as possible predictors of their occurrence. The information collected in this publication may be useful in predicting outcomes after surgical treatment and will also find new ways for the target implication to the mechanisms of development of restenosis and atherosclerosis.
Collapse
Affiliation(s)
- Elvira V Sobolevskaya
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Oleg A Shumkov
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Mikhail A Smagin
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Andrey E Guskov
- Laboratory of Scientometrics and Scientific Communications, Russian Research Institute of Economics, Politics and Law in Science and Technology, Moscow 127254, Russia
| | - Alexandra V Malysheva
- Laboratory of Scientometrics and Scientific Communications, Russian Research Institute of Economics, Politics and Law in Science and Technology, Moscow 127254, Russia
| | - Victor V Atuchin
- Laboratory of Optical Materials and Structures, Institute of Semiconductor Physics, SB RAS, Novosibirsk 630090, Russia
- Research and Development Department, Kemerovo State University, Kemerovo 650000, Russia
- Department of Industrial Machinery Design, Novosibirsk State Technical University, Novosibirsk 630073, Russia
- R&D Center "Advanced Electronic Technologies", Tomsk State University, Tomsk 634034, Russia
| | - Vadim V Nimaev
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| |
Collapse
|
21
|
Park JY, Park HM, Kim S, Jeon KB, Lim CM, Hong JT, Yoon DY. Human IL-32θA94V mutant attenuates monocyte-endothelial adhesion by suppressing the expression of ICAM-1 and VCAM-1 via binding to cell surface receptor integrin αVβ3 and αVβ6 in TNF-α-stimulated HUVECs. Front Immunol 2023; 14:1160301. [PMID: 37228610 PMCID: PMC10203490 DOI: 10.3389/fimmu.2023.1160301] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Interleukin-32 (IL-32), first reported in 2005, and its isoforms have been the subject of numerous studies investigating their functions in virus infection, cancer, and inflammation. IL-32θ, one of the IL-32 isoforms, has been shown to modulate cancer development and inflammatory responses. A recent study identified an IL-32θ mutant with a cytosine to thymine replacement at position 281 in breast cancer tissues. It means that alanine was also replaced to valine at position 94 in amino acid sequence (A94V). In this study, we investigated the cell surface receptors of IL-32θA94V and evaluated their effect on human umbilical vein endothelial cells (HUVECs). Recombinant human IL-32θA94V was expressed, isolated, and purified using Ni-NTA and IL-32 mAb (KU32-52)-coupled agarose columns. We observed that IL-32θA94V could bind to the integrins αVβ3 and αVβ6, suggesting that integrins act as cell surface receptors for IL-32θA94V. IL-32θA94V significantly attenuated monocyte-endothelial adhesion by inhibiting the expression of Intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in tumor necrosis factor (TNF)-α-stimulated HUVECs. IL-32θA94V also reduced the TNF-α-induced phosphorylation of protein kinase B (AKT) and c-jun N-terminal kinases (JNK) by inhibiting phosphorylation of focal adhesion kinase (FAK). Additionally, IL-32θA94V regulated the nuclear translocation of nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1), which are involved in ICAM-1 and VCAM-1 expression. Monocyte-endothelial adhesion mediated by ICAM-1 and VCAM-1 is an important early step in atherosclerosis, which is a major cause of cardiovascular disease. Our findings suggest that IL-32θA94V binds to the cell surface receptors, integrins αVβ3 and αVβ6, and attenuates monocyte-endothelial adhesion by suppressing the expression of ICAM-1 and VCAM-1 in TNF-α-stimulated HUVECs. These results demonstrate that IL-32θA94V can act as an anti-inflammatory cytokine in a chronic inflammatory disease such as atherosclerosis.
Collapse
Affiliation(s)
- Jae-Young Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Hyo-Min Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Seonhwa Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Kyeong-Bae Jeon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Chae-Min Lim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Cormican S, Negi N, Naicker SD, Islam MN, Fazekas B, Power R, Griffin TP, Dennedy MC, MacNeill B, Malone AF, Griffin MD. Chronic Kidney Disease Is Characterized by Expansion of a Distinct Proinflammatory Intermediate Monocyte Subtype and by Increased Monocyte Adhesion to Endothelial Cells. J Am Soc Nephrol 2023; 34:793-808. [PMID: 36799882 PMCID: PMC10125648 DOI: 10.1681/asn.0000000000000083] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/13/2022] [Indexed: 01/27/2023] Open
Abstract
SIGNIFICANCE STATEMENT CKD is accompanied by abnormal inflammation, which contributes to progressive loss of functional renal tissue and accelerated cardiovascular disease. Although studies have documented that dysregulation of monocyte maturation and function is associated with CKD and its complications, it is not well characterized. This study reveals that a distinctive human monocyte subtype with high propensity for releasing proinflammatory mediators and activating endothelial cells is increased in adults with CKD compared with adults with high cardiovascular risk and normal kidney function. It also demonstrates that human monocyte adhesion to endothelial layers and responses to specific inflammatory migration signals are enhanced in CKD. These findings offer insights into the mechanisms of CKD-associated intravascular and localized inflammation and may suggest potential targets for therapeutic interventions. BACKGROUND Cardiovascular disease (CVD) in patients with CKD is associated with increased circulating intermediate monocytes (IMs). Dysregulation of monocyte maturation and function is associated with CKD and its complications, but it is incompletely characterized. METHODS To explore monocyte repertoire abnormalities in CKD, we studied properties of monocyte subpopulations, including IM subpopulations distinguished by HLA-DR expression level, in individuals with or without CKD. Using flow cytometry, we profiled monocyte populations in blood samples from adults with CKD, healthy volunteers (HVs), and patient controls (PCs) with high CVD risk. Monocyte subpopulations were also derived from single-cell RNA-sequencing profiles of paired blood and biopsy samples from kidney transplant recipients. We quantified intracellular cytokine production, migration, and endothelial adhesion in ex vivo assays of PBMCs. RESULTS Of four predefined blood monocyte subpopulations, only HLA-DR hi IMs were increased in individuals with CKD compared with HVs and PCs. In HVs and patients with CKD, LPS-stimulated HLA-DR hi IMs isolated from blood produced higher amounts of TNF and IL-1 β than other monocyte populations. Single-cell analysis revealed four monocyte clusters common to blood and kidneys, including an HLA-DR hi IM-like cluster that was enriched in kidneys versus blood. Migration toward CCL5 and CX3CL1 and adhesion to primary endothelial cell layers were increased in monocyte subpopulations in individuals with CKD compared with HVs. Monocyte adhesion to endothelial cells was partly dependent on CX3CR1/CX3CL1 interaction. CONCLUSIONS CKD is associated with an increased number of a distinctive proinflammatory IM subpopulation and abnormalities of monocyte migration and endothelial adhesion. Dysregulated monocyte maturation and function may represent targetable factors contributing to accelerated CVD in CKD.
Collapse
Affiliation(s)
- Sarah Cormican
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- Nephrology Department, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| | - Neema Negi
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Serika D. Naicker
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Md Nahidul Islam
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Barbara Fazekas
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Rachael Power
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Tomás P. Griffin
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - M. Conall Dennedy
- Department of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Briain MacNeill
- Cardiology Department, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| | - Andrew F. Malone
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew D. Griffin
- CÚRAM Centre for Research in Medical Devices, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- Nephrology Department, Galway University Hospitals, Saolta University Health Care Group, Galway, Ireland
| |
Collapse
|
23
|
The Effects of Human Papillomavirus Infection and Vaccination on Cardiovascular Diseases, NHANES 2003-2016. Am J Med 2023; 136:294-301.e2. [PMID: 36252711 DOI: 10.1016/j.amjmed.2022.09.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Human papillomavirus (HPV) infection has been proposed to be an unconventional risk factor for cardiovascular diseases. We investigated the association between HPV infection and cardiovascular diseases among women with or without HPV vaccination. METHODS This cross-sectional study included 9,353 women aged between 20 to 59 years old who were tested for vaginal HPV DNA in the National Health and Nutrition Examination Survey (NHANES) 2003-2016. Cardiovascular diseases were defined as the presence of self-reported coronary heart diseases, heart attacks, angina pectoris, and stroke. The association between HPV and cardiovascular diseases was studied using logistic regression, with adjustment for the potential confounders. RESULTS A total of 40.8% of women were HPV DNA positive; 3.0% had cardiovascular diseases; and 9.0% of women received the HPV vaccine. The presence of vaginal HPV infection was associated with cardiovascular diseases (odd ratio [OR] = 1.66, 95% confidence interval [CI] 1.28-2.16), which remained significant (OR = 1.54, 95% CI 1.15-2.08) after adjustment for sociodemographic characteristics, lifestyle behaviors, medical history, family history of cardiovascular diseases, and antihypertensive drugs. The association was absent among those who were vaccinated against HPV (OR= 0.50, 95% CI 0.07-3.51) but present among those who were not (OR = 1.63, 95% CI 1.18-2.25). CONCLUSIONS There was an association between HPV infection and cardiovascular diseases. This association was not significant among women vaccinated against HPV. The effect of HPV vaccination on cardiovascular diseases requires further investigation.
Collapse
|
24
|
Hypotheses on Atherogenesis Triggering: Does the Infectious Nature of Atherosclerosis Development Have a Substruction? Cells 2023; 12:cells12050707. [PMID: 36899843 PMCID: PMC10001176 DOI: 10.3390/cells12050707] [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: 12/19/2022] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Since the end of the 20th century, it has been clear that atherosclerosis is an inflammatory disease. However, the main triggering mechanism of the inflammatory process in the vascular walls is still unclear. To date, many different hypotheses have been put forward to explain the causes of atherogenesis, and all of them are supported by strong evidence. Among the main causes of atherosclerosis, which underlies these hypotheses, the following can be mentioned: lipoprotein modification, oxidative transformation, shear stress, endothelial dysfunction, free radicals' action, homocysteinemia, diabetes mellitus, and decreased nitric oxide level. One of the latest hypotheses concerns the infectious nature of atherogenesis. The currently available data indicate that pathogen-associated molecular patterns from bacteria or viruses may be an etiological factor in atherosclerosis. This paper is devoted to the analysis of existing hypotheses for atherogenesis triggering, and special attention is paid to the contribution of bacterial and viral infections to the pathogenesis of atherosclerosis and cardiovascular disease.
Collapse
|
25
|
Kuneman JH, van Rosendael SE, van der Bijl P, van Rosendael AR, Kitslaar PH, Reiber JH, Jukema JW, Leon MB, Ajmone Marsan N, Knuuti J, Bax JJ. Pericoronary Adipose Tissue Attenuation in Patients With Acute Coronary Syndrome Versus Stable Coronary Artery Disease. Circ Cardiovasc Imaging 2023; 16:e014672. [PMID: 36802444 PMCID: PMC9946175 DOI: 10.1161/circimaging.122.014672] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Pericoronary adipose tissue (PCAT) attenuation has been associated with coronary inflammation and can be evaluated with coronary computed tomography angiography. The aims of this study were to compare the PCAT attenuation across precursors of culprit and nonculprit lesions of patients with acute coronary syndrome versus stable coronary artery disease (CAD). METHODS In this case-control study, patients with suspected CAD who underwent coronary computed tomography angiography were included. Patients who developed an acute coronary syndrome within 2 years after the coronary computed tomography angiography scan were identified, and patients with stable CAD (defined as any coronary plaque ≥30% luminal diameter stenosis) were 1:2 propensity score matched for age, sex, and cardiac risk factors. The mean PCAT attenuation was analyzed at lesion level and compared between precursors of culprit lesions, nonculprit lesions, and stable coronary plaques. RESULTS In total, 198 patients (age 62±10 years, 65% male) were selected, including 66 patients who developed an acute coronary syndrome and 132 propensity matched patients with stable CAD. Overall, 765 coronary lesions were analyzed (culprit lesion precursors: n=66; nonculprit lesion precursors: n=207; and stable lesions: n=492). Culprit lesion precursors had larger total plaque volume, fibro-fatty plaque volume, and low-attenuation plaque volume compared to nonculprit and stable lesions. The mean PCAT attenuation was significantly higher across culprit lesion precursors compared to nonculprit and stable lesions (-63.8±9.7 Hounsfield units versus -68.8±10.6 Hounsfield units versus -69.6±10.6 Hounsfield units, respectively; P<0.001), whereas the mean PCAT attenuation around nonculprit and stable lesions was not significantly different (P=0.99). CONCLUSIONS The mean PCAT attenuation is significantly increased across culprit lesion precursors in patients with acute coronary syndrome, compared to nonculprit lesions of these patients and to lesions of patients with stable CAD, which may suggest a higher intensity of inflammation. PCAT attenuation on coronary computed tomography angiography may be a novel marker to identify high-risk plaques.
Collapse
Affiliation(s)
- Jurrien H. Kuneman
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.H.K., S.E.v.R., P.v.d.R., A.R.v.R., J.W.J., N.A.M., J.K., J.J.B.)
| | - Sophie E. van Rosendael
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.H.K., S.E.v.R., P.v.d.R., A.R.v.R., J.W.J., N.A.M., J.K., J.J.B.)
| | - Pieter van der Bijl
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.H.K., S.E.v.R., P.v.d.R., A.R.v.R., J.W.J., N.A.M., J.K., J.J.B.)
| | - Alexander R. van Rosendael
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.H.K., S.E.v.R., P.v.d.R., A.R.v.R., J.W.J., N.A.M., J.K., J.J.B.)
| | - Pieter H. Kitslaar
- Division of Image Processing, Department of Radiology, Leiden University Medical Centre, The Netherlands (P.H.K.).,Medis Medical Imaging, Leiden, The Netherlands (P.H.K.)
| | - Johan H.C. Reiber
- Department of Radiology, Leiden University Medical Center, The Netherlands (J.H.C.R.)
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.H.K., S.E.v.R., P.v.d.R., A.R.v.R., J.W.J., N.A.M., J.K., J.J.B.).,Netherlands Heart Institute, Utrecht, The Netherlands (J.W.J.)
| | - Martin B. Leon
- Department of Cardiology, Columbia University Irving Medical Center/New York-Presbyterian Hospital and Cardiovascular Research Foundation, NY (M.B.L.)
| | - Nina Ajmone Marsan
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.H.K., S.E.v.R., P.v.d.R., A.R.v.R., J.W.J., N.A.M., J.K., J.J.B.)
| | - Juhani Knuuti
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.H.K., S.E.v.R., P.v.d.R., A.R.v.R., J.W.J., N.A.M., J.K., J.J.B.).,Turku PET Centre, Turku University Hospital and University of Turku, Finland (J.K.)
| | - Jeroen J. Bax
- Department of Cardiology, Leiden University Medical Center, The Netherlands (J.H.K., S.E.v.R., P.v.d.R., A.R.v.R., J.W.J., N.A.M., J.K., J.J.B.).,Heart Center, Turku University Hospital and University of Turku, Finland (J.J.B.)
| |
Collapse
|
26
|
Association between cardiovascular diseases and periodontal disease: more than what meets the eye. Drug Target Insights 2023; 17:31-38. [PMID: 36761891 PMCID: PMC9906023 DOI: 10.33393/dti.2023.2510] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/10/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) are inflammatory diseases of coronary arteries accompanying atheroma formation that can spawn impairment and, in severe cases, death. CVDs are the leading cause of death in the world. In recent decades, investigators have focused their impact on CVD by periodontal disease (PD). PD is a risk factor that can trigger the formation, maturation, and instability of atheroma in the arteries. Two mechanisms have been proposed to explain this relationship: periodontopathic pathogens explicitly invade the circulation or indirectly increase systemic levels of inflammatory mediators. It has been suggested that improvement in disease state has a positive effect on others. This review summarizes evidence from epidemiological studies as well as researches focusing on potential causation channels to deliver a comprehensive representation of the relationship between PD and CVD.
Collapse
|
27
|
Wu J, Fang C, Wei L, Liu Y, Xu H, Wang X, Yuan L, Wu X, Xu Y, Zhang A. Spotlight on clinical strategies of Chronic Internal Carotid Artery Occlusion: Endovascular interventions and external-intracarotid bypasses compared to conservative treatment. Front Surg 2022; 9:971066. [PMID: 36425889 PMCID: PMC9679017 DOI: 10.3389/fsurg.2022.971066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/18/2022] [Indexed: 10/11/2023] Open
Abstract
Chronic internal carotid artery occlusion (CICAO) has high prevalence and incidence rates, and patients with CICAO can be completely asymptomatic, experience a devastating stroke or die. It is important to note that CICAO causes cerebrovascular accidents. Currently, the external carotid-internal carotid (EC-IC) bypass technique is used to treat CICAO. However, many clinical studies showed that EC-IC bypass was not beneficial for many patients with CICAO. Meanwhile, endovascular intervention treatment options for CICAO are evolving, and an increasing number of patients are undergoing endovascular intervention therapy. Accordingly, a review comparing both techniques is warranted. For this review, we searched PubMed and collected relevant case study reports comparing endovascular interventional therapy and internal and external cervical bypass surgeries to provide strategies for clinical treatment.
Collapse
Affiliation(s)
- Junnan Wu
- Department of Emergency, Dongyang Hospital Affiliated to Wenzhou Medical University, Jinhua, China
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingying Wei
- Department of Emergency, Dongyang Hospital Affiliated to Wenzhou Medical University, Jinhua, China
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Shanghai, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Shanghai, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoya Wu
- Department of Emergency, Dongyang Hospital Affiliated to Wenzhou Medical University, Jinhua, China
| | - Yuanzhi Xu
- Department of Neurosurgery, Huashan Hospital, School of Medicine, Fudan University, Shanghai, China
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Shanghai, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
28
|
Guzmán-Martínez G, Marañón C. Immune mechanisms associated with cardiovascular disease in systemic lupus erythematosus: A path to potential biomarkers. Front Immunol 2022; 13:974826. [PMID: 36420265 PMCID: PMC9677819 DOI: 10.3389/fimmu.2022.974826] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/13/2022] [Indexed: 10/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) patients display an increased risk of cardiovascular disease (CVD). With the improved clinical management of other classical severe manifestation of the disease, CVD is becoming one of the most relevant complications of SLE, and it is an important factor causing morbidity and mortality. Several immune constituents have been shown to be involved in the pathogenesis of atherosclerosis and endothelial damage in SLE patients, including specific circulating cell populations, autoantibodies, and inflammatory mediators. In this review, we summarize the presentation of CVD in SLE and the role of the autoimmune responses present in SLE patients in the induction of atherogenesis, endothelial impairment and cardiac disease. Additionally, we discuss the utility of these immune mediators as early CVD biomarkers and targets for clinical intervention in SLE patients.
Collapse
Affiliation(s)
- Gabriela Guzmán-Martínez
- Atrys Health, Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
- Department of Cardiology, La Paz University Hospital, IdiPaz, Madrid, Spain
| | - Concepción Marañón
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| |
Collapse
|
29
|
Giachi A, Cugno M, Gualtierotti R. Disease-modifying anti-rheumatic drugs improve the cardiovascular profile in patients with rheumatoid arthritis. Front Cardiovasc Med 2022; 9:1012661. [PMID: 36352850 PMCID: PMC9637771 DOI: 10.3389/fcvm.2022.1012661] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/28/2022] [Indexed: 11/28/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease affecting about 0. 5–1% of the adult population and manifesting as persistent synovitis, systemic inflammation and production of autoantibodies. Patients affected by RA not only experience chronic disease progression, but are also burdened by a 1.5-fold increased cardiovascular (CV) risk, which is comparable to the risk experienced by patients with type 2 diabetes mellitus. RA patients also have a higher incidence and prevalence of coronary artery disease (CAD). Although RA patients frequently present traditional CV risk factors such as insulin resistance and active smoking, previous studies have clarified the pivotal role of chronic inflammation–driven by proinflammatory cytokines such as interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-alpha)–in accelerating the process of atherosclerosis and impairing the coagulation system. Over the last years, a number of studies have shown that disease-modifying anti-rheumatic drugs (DMARDs) reducing the inflammatory state in general improve the CV risk, however some drugs may carry some apparent negative effects. Thus, RA is a model of disease in which targeting inflammation may counteract the progression of atherosclerosis and reduce CV risk. Clinical and experimental evidence indicates that the management of RA patients should be tailored based on the positive and negative effects of DMARDs on CV risk together with the individual traditional CV risk profile. The identification of genetic, biochemical and clinical biomarkers, predictive of evolution and response to treatment, will be the next challenge for a precision approach to reduce the burden of the disease.
Collapse
Affiliation(s)
- Andrea Giachi
- UOC Medicina Generale Emostasi e Trombosi, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Cugno
- UOC Medicina Generale Emostasi e Trombosi, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
- *Correspondence: Massimo Cugno
| | - Roberta Gualtierotti
- UOC Medicina Generale Emostasi e Trombosi, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| |
Collapse
|
30
|
Wen Y, Kong Y, Cao G, Xu Y, Zhang C, Zhang J, Xiao P, Wang Y. Di-n-butyl phthalate regulates vascular smooth muscle cells phenotypic switching by MiR-139-5p-MYOCD pathways. Toxicology 2022; 477:153279. [PMID: 35926758 DOI: 10.1016/j.tox.2022.153279] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/20/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022]
Abstract
Di-n-butyl phthalate (DBP) is ubiquitous in environment and has been detected in almost all human bodies. Few data could be found about the effects of DBP on cardiovascular system, though its reproductive toxicities have been studied extensively. This study aimed to explore effects of DBP on phenotypic switching of vascular smooth muscle cells (VSMCs), an essential step during the formation of atherosclerosis (AS). A7r5 cells were employed and exposed to various levels of DBP (10-9, 10-8, 10-7, 10-6, and 10-5 M) or DMSO as control. CCK-8 assay was used to detect the effects of DBP on cell viability. Expressions of mRNA/miRNAs and proteins were measured by qRT-PCR and western blotting, respectively. Bioinformatic analysis and dual-luciferase reporter assay were used to analyze the combination between miR-139-5p and Myocardin (MYOCD). Results revealed that DBP at 10-7 M prompted phenotypic switching from contractile to synthetic of VSMCs by inhibiting contractile VSMCs marker genes via suppressing the expression of MYOCD. Moreover, miR-139c-5p directly targeted MYOCD 3'UTR and modulated MYOCD expression. Besides, DBP inhibited the expression of MYOCD and VSMCs marker genes by upregulating miR-139-5p. Collectively, these data suggested that DBP could promote the phenotypic switching from contractile to synthetic of VSMCs in A7r5 cells through miR-139-5p-MYOCD.
Collapse
Affiliation(s)
- Yun Wen
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Yi Kong
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Guofa Cao
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Yuan Xu
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Chengxiang Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Jingshu Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Pingxi Xiao
- The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yubang Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
31
|
Lim L, Ki YJ, Kim H, Chu B, Choi IY, Choi DH, Song H. Plantamajoside Attenuates Neointima Formation via Upregulation of Tissue Inhibitor of Metalloproteinases in Balloon-Injured Rats. J Med Food 2022; 25:503-512. [PMID: 35483086 DOI: 10.1089/jmf.2021.k.0162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The abnormal change of vascular smooth muscle cell (VSMC) behavior is an important cellular event leading to neointimal hyperplasia in atherosclerosis and restenosis. Plantamajoside (PMS), a phenylethanoid glycoside compound of the Plantago asiatica, has been reported to have anti-inflammatory, antioxidative, and anticancer activities. In this study, the protective effects of PMS against intimal hyperplasia and the mechanisms underlying the regulation of VSMC behavior were investigated. MTT and BrdU assays were performed to evaluate the cytotoxicity and cell proliferative activity of PMS, respectively. Rat aortic VSMC migrations after treatment with the determined concentration of PMS (50 and 150 μM) were evaluated using wound healing and Boyden chamber assays. The inhibitory effects of PMS on intimal hyperplasia were evaluated in balloon-injured (BI) rat carotid artery. PMS suppressed the proliferation in platelet-derived growth factor-BB-induced VSMC, as confirmed from the decrease in cyclin-dependent kinase (CDK)-2, CDK-4, cyclin D1, and proliferating cell nuclear antigen levels. PMS also inhibited VSMC migration, consistent with the downregulated expression and zymolytic activities of matrix metalloproteinase (MMP)2, MMP9, and MMP13. PMS specifically regulated MMP expression through p38 mitogen-activated protein kinase and focal adhesion kinase pathways. Tissue inhibitor of metalloproteinase (TIMP)1 and TIMP2 levels were upregulated via Smad1. TIMPs inhibited the conversion of pro-MMPs to active MMPs. PMS significantly inhibited neointimal formation in BI rat carotid arteries. In conclusion, PMS inhibits VSMC proliferation and migration by upregulating TIMP1 and TIMP2 expression. Therefore, PMS could be a potential therapeutic agent for vascular atherosclerosis and restenosis treatment.
Collapse
Affiliation(s)
- Leejin Lim
- Cancer Mutation Research Center, Chosun University, Gwangju, Korea
| | - Young-Jae Ki
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju, Korea
| | - Hyeonhwa Kim
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju, Korea
| | - Byeongsam Chu
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju, Korea
| | - In Young Choi
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju, Korea
| | - Dong-Hyun Choi
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju, Korea
| | - Heesang Song
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju, Korea.,Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju, Korea
| |
Collapse
|
32
|
Xie Y, Zhang Y, Qin P, Ping Z, Wang C, Peng X, Chen H, Zhao D, Xu S, Wang L, Wei P, Hu F, Lou Y, Hu D. The association between Chinese Visceral Adipose Index and coronary heart disease: A cohort study in China. Nutr Metab Cardiovasc Dis 2022; 32:550-559. [PMID: 34961647 DOI: 10.1016/j.numecd.2021.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/22/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS The purpose of this study is to explore the relationship between Chinese visceral adipose index (CVAI) and the risk of coronary heart disease (CHD) in Chinese through a large cohort study. METHODS AND RESULTS This study included 42,165 adults who were without CHD at baseline and who completed at least one annual follow-up between 2009 and 2016. We used the Cox proportional hazards model to estimate Hazard Ratios (HRs) and 95% Confidence Intervals (CIs) for the association between CVAI and risk of CHD. During the median follow-up of 3.36 years (154,808 person years), 520 participants developed CHD, including 374 males and 146 females. Compared with the first quartile of CVAI, the risk of CHD was significantly increased in the fourth quartile of CVAI in multivariate model (HR [95% CI]: 9.92 [5.45, 18.04], P < 0.001). Sensitivity analysis by excluding incident CHD developed in the first two years of follow-up reinforced our results. Gender stratification analyses showed that the relationship between CVAI and CHD risk was higher in males than that in females. The restricted cubic spline showed a non-linear dose-response relationship between CVAI and CHD risk. In addition, CVAI was associated with CHD risk in the subgroups of participants without T2DM, without hypertension, and without fatty liver. CONCLUSION CVAI was significantly associated with the risk of CHD. Individuals should keep CVAI at normal level to prevent CHD.
Collapse
Affiliation(s)
- Yilin Xie
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China
| | - Yanyan Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China
| | - Pei Qin
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China
| | - Zhao Ping
- Department of Health Management, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping, Beijing 102200, People's Republic of China
| | - Changyi Wang
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Xiaolin Peng
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Hongen Chen
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Dan Zhao
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Shan Xu
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Li Wang
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease, No.7 Huaming Road, Shenzhen, Guangdong 518054, People's Republic of China
| | - Pengfei Wei
- Department of Endocrine, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, People's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China.
| | - Yanmei Lou
- Department of Health Management, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping, Beijing 102200, People's Republic of China.
| | - Dongsheng Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, No.1066 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong 518060, People's Republic of China.
| |
Collapse
|
33
|
Zhang M, Che C, Cheng J, Li P, Yang Y. Ion channels in stem cells and their roles in stem cell biology and vascular diseases. J Mol Cell Cardiol 2022; 166:63-73. [PMID: 35143836 DOI: 10.1016/j.yjmcc.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
Abstract
Stem cell therapy may be a promising option for the treatment of vascular diseases. In recent years, significant progress has been made in stem cell research, especially in the mechanism of stem cell activation, homing and differentiation in vascular repair and reconstruction. Current research on stem cells focuses on protein expression and transcriptional networks. Ion channels are considered to be the basis for the generation of bioelectrical signals, which control the proliferation, differentiation and migration of various cell types. Although heterogeneity of multiple ion channels has been found in different types of stem cells, it is unclear whether the heterogeneous expression of ion channels is related to different cell subpopulations and/or different stages of the cell cycle. There is still a long way to go in clinical treatment by using the regulation of stem cell ion channels. In this review, we reviewed the main ion channels found on stem cells, their expression and function in stem cell proliferation, differentiation and migration, and the research status of stem cells' involvement in vascular diseases.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Chang Che
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| |
Collapse
|
34
|
Could the systemic immune-inflammation index be a predictor to estimate cerebrovascular events in hypertensive patients? Blood Press Monit 2022; 27:33-38. [PMID: 34992205 DOI: 10.1097/mbp.0000000000000560] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVES Hypertension is one of the most important risk factors for cardiovascular and cerebrovascular events. Inflammatory processes occupy an important place in the pathogenesis of hypertension. Many studies have studied inflammatory markers responsible for the onset of hypertension and organ damage. In this study, we investigated whether the systemic immune-inflammation index (SII) (platelet × neutrophil/lymphocyte), - one of the new inflammatory markers - can be used to predict cerebrovascular events in hypertensive patients. METHODS Ambulatory blood pressure monitoring results between January 2019 and June 2020 of approximately 379 patients followed up with hypertension were retrospectively analyzed. These patients were divided into two groups as with or without a previous cerebrovascular event in the analyzed database. In all patients, complete blood count and biochemistry test results just before the cerebrovascular event were found from the database. SII, atherogenic index, neutrophil-lymphocyte ratio were calculated from the complete blood count. Forty-nine patients with stroke (group 1: 12.9%; mean age: 64.3 ± 14.6) and 330 patients without stroke (group 2: 87.1%; mean age: 50.8 ± 14.4). RESULTS Ambulatory blood pressure measurements were lower in group 1. Lipid parameters were also lower in this group. Receiver operating characteristic curve analysis showed that SII had a sensitivity of 85.7% and specificity of 84.8 % for stroke in individuals who participated in the study when the cutoff value of SII was 633.26 × 103 (P = 0.0001) area under curve (95%); 0.898 (0.856-0.941). In multivariate logistic regression analysis, age and SII were significantly associated with a higher risk of stroke. Age, (hazard ratio:1.067; 95% CI, 1.021-1.115), SII (hazard ratio:1.009; 95% CI, 1.000-1.009), respectively. CONCLUSIONS In conclusion, SII is a simple, useful new inflammatory parameter for predicting stroke from hypertension. We found that the high SII levels increase the risk of stroke in hypertensive patients.
Collapse
|
35
|
Pupina N, Avarlaid A, Sadam H, Pihlak A, Jaago M, Tuvikene J, Rähni A, Planken A, Planken M, Kalso E, Tienari PJ, Nieminen JK, Seppänen MRJ, Vaheri A, Lindholm D, Sinisalo J, Pussinen P, Timmusk T, Palm K. Immune response to a conserved enteroviral epitope of the major capsid VP1 protein is associated with lower risk of cardiovascular disease. EBioMedicine 2022; 76:103835. [PMID: 35091341 PMCID: PMC8801986 DOI: 10.1016/j.ebiom.2022.103835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Major cardiac events including myocardial infarction (MI) are associated with viral infections. However, how specific infections contribute to the cardiovascular insults has remained largely unclear. METHODS We employed next generation phage display mimotope-variation analysis (MVA) to explore the link between antibody-based immune response and severe cardiovascular conditions. Here, we used a case-control design, including the first-stage discovery cohort (n = 100), along with cohorts for second-stage discovery (n = 329) and validation (n = 466). FINDINGS We observed strong antibody response to the peptide antigens with Gly-Ile-X-Asp (G-I-X-D) core structure in healthy individuals but not in patients with MI. Analysis of the origin of this epitope linked it with the N-terminus of the VP1 protein of poliovirus 3 (PV3), but also other species of picornaviruses. Consistently, we found low levels of antibody response to the G-I-X-D epitope in individuals with severe cardiac disease complications. INTERPRETATION Our findings imply that antibody response to the G-I-X-D epitope is associated with polio vaccinations and that high antibody levels to this epitope could discriminate healthy individuals from prospective MI patients as a blood-derived biomarker. Together, these findings highlight the importance of epitope-specific antibody response and suggest that protective immunity against the polio- and non-polio enteroviral infections support improved cardiovascular health. FUNDING Estonian Ministry of Education (5.1-4/20/170), Estonian Research Council (PRG573, PRG805), H2020-MSCA-RISE-2016 (EU734791), H2020 PANBioRA (EU760921), European Union through the European Regional Development Fund (Project no. 2014-2020.4.01.15-0012), Helsinki University Hospital grants, Mary and Georg C. Ehrnrooth Foundation, Finnish Eye Foundation, Finska Läkaresällskapet, The Finnish Society of Sciences and Letters, Magnus Ehrnrooth Foundation and Sigrid Jusélius Foundation.
Collapse
Affiliation(s)
| | - Annela Avarlaid
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia
| | - Helle Sadam
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia; Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia
| | - Arno Pihlak
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia
| | - Mariliis Jaago
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia; Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia
| | - Jürgen Tuvikene
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia; Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia; dxlabs LLC, Mäealuse 4, Tallinn 12618, Estonia
| | - Annika Rähni
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia; Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia
| | - Anu Planken
- The North Estonia Medical Center, Tallinn, Estonia
| | | | - Eija Kalso
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital and Department of Pharmacology and SleepWell Research Programme, University of Helsinki, Finland
| | - Pentti J Tienari
- Department of Neurology, Neurocenter, Helsinki University Hospital, and Translational Immunology Research Program, University of Helsinki, Finland
| | - Janne K Nieminen
- Department of Neurology, Neurocenter, Helsinki University Hospital, and Translational Immunology Research Program, University of Helsinki, Finland
| | - Mikko R J Seppänen
- Department of Neurology, Neurocenter, Helsinki University Hospital, and Translational Immunology Research Program, University of Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Medicum, University of Helsinki, Finland
| | - Dan Lindholm
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Finland
| | - Pirkko Pussinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Finland
| | - Tõnis Timmusk
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia; Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia
| | - Kaia Palm
- Protobios LLC, Mäealuse 4, Tallinn 12618, Estonia; Department of Chemistry and Biotechnology, Tallinn University of Technology, Estonia.
| |
Collapse
|
36
|
Snarski P, Sukhanov S, Yoshida T, Higashi Y, Danchuk S, Chandrasekar B, Tian D, Rivera-Lopez V, Delafontaine P. Macrophage-Specific IGF-1 Overexpression Reduces CXCL12 Chemokine Levels and Suppresses Atherosclerotic Burden in Apoe-Deficient Mice. Arterioscler Thromb Vasc Biol 2022; 42:113-126. [PMID: 34852642 PMCID: PMC8792341 DOI: 10.1161/atvbaha.121.316090] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE IGF-1 (insulin-like growth factor 1) exerts pleiotropic effects including promotion of cellular growth, differentiation, survival, and anabolism. We have shown that systemic IGF-1 administration reduced atherosclerosis in Apoe-/- (apolipoprotein E deficient) mice, and this effect was associated with a reduction in lesional macrophages and a decreased number of foam cells in the plaque. Almost all cell types secrete IGF-1, but the effect of macrophage-derived IGF-1 on the pathogenesis of atherosclerosis is poorly understood. We hypothesized that macrophage-derived IGF-1 will reduce atherosclerosis. Approach and Results: We created macrophage-specific IGF-1 overexpressing mice on an Apoe-/- background. Macrophage-specific IGF-1 overexpression reduced plaque macrophages, foam cells, and atherosclerotic burden and promoted features of stable atherosclerotic plaque. Macrophage-specific IGF1 mice had a reduction in monocyte infiltration into plaque, decreased expression of CXCL12 (CXC chemokine ligand 12), and upregulation of ABCA1 (ATP-binding cassette transporter 1), a cholesterol efflux regulator, in atherosclerotic plaque and in peritoneal macrophages. IGF-1 prevented oxidized lipid-induced CXCL12 upregulation and foam cell formation in cultured THP-1 macrophages and increased lipid efflux. We also found an increase in cholesterol efflux in macrophage-specific IGF1-derived peritoneal macrophages. CONCLUSIONS Macrophage IGF-1 overexpression reduced atherosclerotic burden and increased features of plaque stability, likely via a reduction in CXCL12-mediated monocyte recruitment and an increase in ABCA1-dependent macrophage lipid efflux.
Collapse
Affiliation(s)
- Patricia Snarski
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Sergiy Sukhanov
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Tadashi Yoshida
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Yusuke Higashi
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Svitlana Danchuk
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA
| | - Bysani Chandrasekar
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO
| | - Di Tian
- Department of Pathology, Tulane University School of Medicine, New Orleans, LA
| | | | - Patrick Delafontaine
- Section of Cardiology, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA,Department of Physiology, Tulane University School of Medicine, New Orleans, LA,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW The obesity epidemic is on the rise, and while it is well known that obesity is associated with an increase in cardiovascular risk factors such as type 2 diabetes mellitus, hypertension, and obstructive sleep apnea, recent data has highlighted that the degree and type of fat distribution may play a bigger role in the pathogenesis of cardiovascular disease (CVD) than body mass index (BMI) alone. We aim to review updated data on adipose tissue inflammation and distribution and CVD. RECENT FINDINGS We review the pathophysiology of inflammation secondary to adipose tissue, the association of obesity-related adipokines and CVD, and the differences and significance of brown versus white adipose tissue. We delve into the clinical manifestations of obesity-related inflammation in CVD. We discuss the available data on heterogeneity of adipose tissue-related inflammation with a focus on subcutaneous versus visceral adipose tissue, the differential pathophysiology, and clinical CVD manifestations of adipose tissue across sex, race, and ethnicity. Finally, we present the available data on lifestyle modification, medical, and surgical therapeutics on reduction of obesity-related inflammation. Obesity leads to a state of chronic inflammation which significantly increases the risk for CVD. More research is needed to develop non-invasive VAT quantification indices such as risk calculators which include variables such as sex, age, race, ethnicity, and VAT concentration, along with other well-known CVD risk factors in order to comprehensively determine risk of CVD in obese patients. Finally, pre-clinical biomarkers such as pro-inflammatory adipokines should be validated to estimate risk of CVD in obese patients.
Collapse
Affiliation(s)
- Mariam N Rana
- Department of Medicine, University Hospitals, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Ian J Neeland
- Department of Medicine, University Hospitals, 11100 Euclid Ave, Cleveland, OH, 44106, USA.
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Harrington Heart and Vascular Institute, University Hospitals, 11100 Euclid Ave, Cleveland, OH, 44106, USA.
| |
Collapse
|
38
|
Zhu QQ, Li DL, Wang X, Wang YS, Zeng QL, Qiu CY, He YY, Wu ZH, He YJ, Shang T, Zhang HK. The role of RAGE, MAPK and NF-κB pathway in the advanced glycation end-products induced HUVECs dysfunction. VASCULAR INVESTIGATION AND THERAPY 2022. [DOI: 10.4103/2589-9686.360874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
39
|
El Hadri K, Smith R, Duplus E, El Amri C. Inflammation, Oxidative Stress, Senescence in Atherosclerosis: Thioredoxine-1 as an Emerging Therapeutic Target. Int J Mol Sci 2021; 23:ijms23010077. [PMID: 35008500 PMCID: PMC8744732 DOI: 10.3390/ijms23010077] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/19/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular diseases (CVD) worldwide and intimately linked to aging. This pathology is characterized by chronic inflammation, oxidative stress, gradual accumulation of low-density lipoproteins (LDL) particles and fibrous elements in focal areas of large and medium arteries. These fibrofatty lesions in the artery wall become progressively unstable and thrombogenic leading to heart attack, stroke or other severe heart ischemic syndromes. Elevated blood levels of LDL are major triggering events for atherosclerosis. A cascade of molecular and cellular events results in the atherosclerotic plaque formation, evolution, and rupture. Moreover, the senescence of multiple cell types present in the vasculature were reported to contribute to atherosclerotic plaque progression and destabilization. Classical therapeutic interventions consist of lipid-lowering drugs, anti-inflammatory and life style dispositions. Moreover, targeting oxidative stress by developing innovative antioxidant agents or boosting antioxidant systems is also a well-established strategy. Accumulation of senescent cells (SC) is also another important feature of atherosclerosis and was detected in various models. Hence, targeting SCs appears as an emerging therapeutic option, since senolytic agents favorably disturb atherosclerotic plaques. In this review, we propose a survey of the impact of inflammation, oxidative stress, and senescence in atherosclerosis; and the emerging therapeutic options, including thioredoxin-based approaches such as anti-oxidant, anti-inflammatory, and anti-atherogenic strategy with promising potential of senomodulation.
Collapse
|
40
|
Yang X, Sun W, Hou D, Wang T, Li C, Luo Y, Zhang S, Shen L, Liu W, Wu D. The Degree of Plasma Oxidized Low-Density Lipoprotein Level Decrease Is Related to Clinical Outcomes for Patients with Acute Ischemic Stroke. DISEASE MARKERS 2021; 2021:4998823. [PMID: 34950249 PMCID: PMC8692044 DOI: 10.1155/2021/4998823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/05/2021] [Accepted: 11/15/2021] [Indexed: 12/03/2022]
Abstract
OBJECTIVE To investigate the relationship between the decrease of plasma oxidized low-density lipoprotein (oxLDL) levels and clinical outcomes in patients with acute atherosclerosis-related ischemic stroke. METHODS We recruited acute ischemic stroke patients within 3 days of onset consecutively. Plasma oxLDL levels were measured on the second day after admission and before discharge (10-14 days after stroke onset). Initial stroke severity was assessed by the National Institutes of Health Stroke Scale (NIHSS) scores, and infarct volume was measured using diffusion-weighted imaging (DWI) by the ITK-SNAP software. Clinical outcomes were evaluated by DWI volumes in the acute phase, neurological improvement at discharge, and favorable functional prognosis at 90 days. Logistic regression was performed to evaluate the association between oxLDL level decrease and clinical outcomes. RESULTS 207 patients were enrolled in this study. Compared with the mild decrease of the oxLDL level group, patients with a significant decrease of the oxLDL level group were more likely to have a higher ratio of neurological improvement at discharge (55.07% vs. 14.49%, p < 0.01) and favorable functional prognosis at 90 days (91.30% vs. 55.07%, p < 0.01). In multivariable logistic regression, the degree of oxLDL level decrease was related to neurological improvement at discharge and favorable functional prognosis at 90 days (p < 0.01). Patients with significant decrease were more likely to have neurological improvement at discharge (OR = 7.92, 95% CI, 3.14-19.98, and p < 0.01) and favorable functional prognosis at 90 days (OR = 7.46, 95% CI, 2.40-23.23, and p < 0.01) compared to patients with mild decrease of oxLDL level. The DWI volumes in patients with different oxLDL level decrease groups had no statistical difference (p = 0.41), and the Spearman's rho between oxLDL level decrease and DWI infarct volumes was -0.03, but no statistical difference (p = 0.72). CONCLUSIONS The degree of oxLDL level decrease is related to neurological improvement at discharge and favorable functional prognosis at 90 days for patients with acute atherosclerosis-related ischemic stroke, but not with infarct volume in the acute phase.
Collapse
Affiliation(s)
- Xiaoli Yang
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wenbo Sun
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Duanlu Hou
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Tianyao Wang
- Radiology Department, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Chen Li
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yufan Luo
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Shufan Zhang
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Liwei Shen
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wenpeng Liu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Danhong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Kang H, Yu H, Fan J, Cao G. Rotigotine protects against oxidized low-density lipoprotein(ox-LDL)-induced damages in human umbilical vein endothelial cells(HUVECs). Bioengineered 2021; 12:10568-10579. [PMID: 34860135 PMCID: PMC8810014 DOI: 10.1080/21655979.2021.2000224] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Rotigotine is a non-ergoline dopamine agonist that has been licensed for the treatment of Parkinson’s disease. Cardiovascular diseases are the world’s leading cause of death. Ox-LDL- induced endothelial damages are involved in the initiation and progression of cardiovascular diseases. In this study, we assessed the beneficial properties of Rotigotine on ox-LDL-induced insults to HUVECs to highlight its potential use in the treatment of cardiovascular diseases. Our findings show that Rotigotine suppresses the expressions of low-density lipoprotein receptor (LDL-R), proprotein convertase subtilisin/kexin type 9 (PCSK-9), and sterol regulatory element-binding protein (SREBP-2). It also inhibits ox-LDL-induced cholesterol accumulation in endothelial cells (ECs), improves U937 monocytes adhesion, and decreases the representation of NADPH oxidase (NOX-4) and generation of reactive oxygen species (ROS) in endothelial cells (ECs). Furthermore, Rotigotine inhibited the expressions of both vascular cellular adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) in HUVECs and had anti-inflammatory efficacy in ox-LDL-induced cells by inhibiting the expressions of pro-inflammatory cytokines. Notably, Rotigotine inhibits the activation of nuclear factor-kappaB (NF-κB) by preventing nuclear translocation of NF-κB p65 and reducing the luciferase activity of NF-κB reporter. We, therefore, conclude that these effects of Rotigotine on HUVECs suggest that it may play a therapeutic role in cardiovascular diseases.
Collapse
Affiliation(s)
- Hui Kang
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Hui Yu
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jingxiu Fan
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ge Cao
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Russell C, Keshavamurthy S, Saha S. Nutraceuticals in the Management of Cardiovascular Risk Factors: Where is the Evidence? Cardiovasc Hematol Disord Drug Targets 2021; 21:150-161. [PMID: 34852755 DOI: 10.2174/1871529x21666211201104124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/24/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
Cardiovascular disease continues to rise at an alarming rate, and research focuses on possible therapies to reduce the risk and slow down its progression. Several epidemiological studies have indicated that dietary modifications, such as increased consumption of fruits and vegetables play an important role in reducing cardiovascular disease risk factors. Food sources rich in antioxidants, anti-inflammatory, hypolipidemic, and hypoglycemic properties are thought to ameliorate the progression of cardiovascular disease and serve as a potential treatment mode. Many in vivo and in vitro studies using turmeric, cinnamon, mango, blueberries, red wine, chocolate, and extra virgin olive oil have demonstrated significant improvements in cholesterol profiles, toxic reactive oxygen species, inflammation, obesity, and hypertension. In this review, we summarize recent evidence on the cardioprotective effect of different food groups, outline their potential mechanisms involved in slowing down the progression of cardiovascular disease, and highlight the beneficial effects associated with increased consumption.
Collapse
Affiliation(s)
- Cody Russell
- The American University of the Caribbean School of Medicine. 0
| | | | - Sibu Saha
- University of Kentucky College of Medicine, Department of Surgery Professor of Surgery and Bioengineering. 0
| |
Collapse
|
43
|
Miao R, Qi C, Fu Y, Wang Y, Lang Y, Liu W, Zhang Y, Zhang Z, Liu A, Chai H, Zhang Y, Song Y, Lu X. Silencing of circARHGAP12 inhibits the progression of atherosclerosis via miR-630/EZH2/TIMP2 signal axis. J Cell Physiol 2021; 237:1057-1069. [PMID: 34750830 DOI: 10.1002/jcp.30598] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Atherosclerosis (AS) is a common disease that seriously threatens human health. So far, the pathogenesis of AS has not been fully understood. This project investigates the effects of circARHGAP12 on AS and its regulatory mechanism. ApoE-/- knockout mice (ApoE) were adopted and reared with a high-fat diet to construct an AS model. Lentivirus was established to knock down the expression of circARHGAP12 in mice. After 12 weeks, the aorta was removed and the expression of circARHGAP12 was detected. Vascular oil red O staining was used to detect the degree of AS. The expression of inflammatory factors was detected by ELISA. Aortic smooth muscle cells (MASMCs) were cultured to evaluate the effects of circARHGAP12 on the phenotype of MASMCs. RNA pull-down and luciferase assay were used to verify the downstream target genes of circARHGAP12. In addition, the effects of circARHGAP12 on MASMCs proliferation and migration were detected by MTT and transwell assay. Compared with the normal group, the expression of circARHGAP12 in the MASMCs under ox-LDL treatment was elevated, and circARHGAP12 silencing could inhibit AS in vitro and in vivo. The results of the mechanism study showed that circARHGAP12 can directly bind with miR-630. In addition, miR-630 can also target EZH2 to modulate the transcription of TIMP2 and to influence the migration of MASMCs. circARHGAP12 is upregulated in AS. CircARHGAP12 knockdown can inhibit the progression of AS. This study expands on the role of circRNA in AS and provides potential targets for the treatment of AS.
Collapse
Affiliation(s)
- Renying Miao
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chaoran Qi
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yiqun Fu
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanjun Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuchang Lang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wanli Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yifei Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhimin Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ankang Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hao Chai
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yonggan Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Song
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiubo Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
44
|
Benchemoul M, Mateo T, Savery D, Gehin C, Massot B, Ferin G, Vince P, Flesch M. Pulse wave velocity measurement along the ulnar artery in the wrist region using a high frequency ultrasonic array. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:4123-4127. [PMID: 34892134 DOI: 10.1109/embc46164.2021.9629889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A pulse wave velocity (PWV) measurement method performed above a small blood vessel using an ultrasonic probe is studied and reported in this paper. These experimentations are carried out using a high-frequency probe (14-22 MHz), allowing a high level of resolution compatible with the vessel dimensions, combined with an open research ultrasound scanner. High frame-rate (HFR) imaging (10 000 frames per second) is used for a precise PWV estimation. The measurements are performed in-vivo on a healthy volunteer. The probe is placed above the ulnar artery on the wrist in order to make longitudinal scans. In addition to conventional duplex ultrasound evaluation, the measurement of the PWV using this method at this location could strengthen the detection and diagnosis of cardiovascular diseases (CVDs), in particular for arm artery diseases (AADs). Moreover, these experimentations are also carried out within the scope of a demonstration for a potential miniaturized and wearable device (i.e., a probe with fewer elements, typically less than 32, and its associated electronics). The study has shown results coherent with expected PWV and also promising complementary results such as intima-media thickness (IMT) with spatiotemporal resolution on the order of 6.2 μm and 0.1 ms.
Collapse
|
45
|
Giebe S, Hofmann A, Brux M, Lowe F, Breheny D, Morawietz H, Brunssen C. Comparative study of the effects of cigarette smoke versus next generation tobacco and nicotine product extracts on endothelial function. Redox Biol 2021; 47:102150. [PMID: 34601427 PMCID: PMC8531844 DOI: 10.1016/j.redox.2021.102150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 11/29/2022] Open
Abstract
Tobacco smoking and hemodynamic forces are key stimuli for the development of endothelial dysfunction. As an alternative to smoking, next generation tobacco and nicotine products (NGP) are now widely used. However, little is known about their potential pro-inflammatory and atherogenic effects on the endothelium. In this study, we analyzed key parameters of endothelial function after exposure to aqueous smoke extracts (AqE) of a heated tobacco product (HTP), an electronic cigarette (e-cig), a conventional cigarette (3R4F) and pure nicotine. All experiments were performed under atheroprotective high laminar or atherogenic low flow with primary human endothelial cells. Treatment with 3R4F, but not alternative smoking products, reduced endothelial cell viability and wound healing capability via the PI3K/AKT/eNOS(NOS3) pathway. Laminar flow delayed detrimental effects on cell viability by 3R4F treatment. 3R4F stimulation led to activation of NRF2 antioxidant defense system at nicotine concentrations ≥0.56 μg/ml and increased expression of its target genes HMOX1 and NQO1. Treatment with HTP revealed an induction of HMOX1 and NQO1 at dosages with ≥1.68 μg/ml nicotine, whereas e-cig and nicotine exposure had no impact. Analyses of pro-inflammatory genes revealed an increased ICAM1 expression under 3R4F treatment. 3R4F reduced VCAM1 expression in a dose-dependent manner; HTP treatment had similar but milder effects; e-cig and nicotine treatment had no impact. SELE expression was induced by 3R4F under static conditions. High laminar flow prevented this upregulation. Stimulation with laminar flow led to downregulation of CCL2 (MCP-1). From this downregulated level, only 3R4F increased CCL2 expression at higher concentrations. Finally, under static conditions, all components increased adhesion of monocytes to endothelial cells. Interestingly, only stimulation with 3R4F revealed increased monocyte adhesion under atherosclerosis-prone low flow. In conclusion, all product categories activated anti-oxidative or pro-inflammatory patterns. NGP responses were typically lower than in 3R4F exposed cells. Also, 3R4F stimulation led to an impaired endothelial wound healing and induced a pro-inflammatory phenotype compared to NGP treatment.
Collapse
Affiliation(s)
- Sindy Giebe
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Anja Hofmann
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Melanie Brux
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Frazer Lowe
- Group Research & Development, British American Tobacco, Southampton, United Kingdom
| | - Damien Breheny
- Group Research & Development, British American Tobacco, Southampton, United Kingdom
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
46
|
Kang X, Deng Y, Cao Y, Huo Y, Luo J. Zyxin Mediates Vascular Repair via Endothelial Migration Promoted by Forskolin in Mice. Front Physiol 2021; 12:741699. [PMID: 34690814 PMCID: PMC8531502 DOI: 10.3389/fphys.2021.741699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: Endothelial repair upon vascular injury is critical for the protection of vessel integrity and prevention of the development of vascular disorders, but the underlying mechanisms remain poorly understood. In this study, we investigated the role of zyxin and its associated cyclic adenosine monophosphate (cAMP) signaling in the regulation of re-endothelialization after vascular injury. Experimental Approach: In zyxin-/- and wild-type mice, wire injury of the carotid artery was carried out, followed by Evans blue staining, to evaluate the re-endothelialization. Mice with endothelium-specific zyxin knockout were used to further determine its role. An in vitro wound-healing assay was performed in primary human endothelial cells (ECs) expressing zyxin-specific short-hairpin RNAs (shRNAs) or scrambled controls by measuring cell migration and proliferation. The effects of the cAMP signaling agonist forskolin were assessed. Key Results: The re-endothelialization of the injured carotid artery was impaired in zyxin-deficient mice, whereas the rate of cell proliferation was comparable with that in wild-type controls. Furthermore, endothelium-specific deletion of zyxin led to similar phenotypes. Knockdown of zyxin by shRNAs in primary human ECs significantly reduced cell migration in the wound-healing assay. Notably, forskolin enhanced endothelial migration in a dose-dependent manner, and this was dependent on zyxin through its interaction with vasodilator-stimulated phosphoprotein. In addition, forskolin promoted the re-endothelialization of the injured carotid artery, and this was compromised by zyxin deficiency. Conclusion and Implications: This study reveals zyxin as a new player in endothelial repair, which is promoted by forskolin, after vascular injury. Thus, zyxin-mediated signaling might be a potential treatment target for diseases involving vascular injury.
Collapse
Affiliation(s)
- Xuya Kang
- Laboratory of Vascular Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, School of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yanan Deng
- Laboratory of Vascular Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, School of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yang Cao
- Laboratory of Vascular Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, School of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yingqing Huo
- Laboratory of Vascular Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, School of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jincai Luo
- Laboratory of Vascular Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, School of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
47
|
Choi RH, Tatum SM, Symons JD, Summers SA, Holland WL. Ceramides and other sphingolipids as drivers of cardiovascular disease. Nat Rev Cardiol 2021; 18:701-711. [PMID: 33772258 PMCID: PMC8978615 DOI: 10.1038/s41569-021-00536-1] [Citation(s) in RCA: 228] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 02/03/2023]
Abstract
Increases in calorie consumption and sedentary lifestyles are fuelling a global pandemic of cardiometabolic diseases, including coronary artery disease, diabetes mellitus, cardiomyopathy and heart failure. These lifestyle factors, when combined with genetic predispositions, increase the levels of circulating lipids, which can accumulate in non-adipose tissues, including blood vessel walls and the heart. The metabolism of these lipids produces bioactive intermediates that disrupt cellular function and survival. A compelling body of evidence suggests that sphingolipids, such as ceramides, account for much of the tissue damage in these cardiometabolic diseases. In humans, serum ceramide levels are proving to be accurate biomarkers of adverse cardiovascular disease outcomes. In mice and rats, pharmacological inhibition or depletion of enzymes driving de novo ceramide synthesis prevents the development of diabetes, atherosclerosis, hypertension and heart failure. In cultured cells and isolated tissues, ceramides perturb mitochondrial function, block fuel usage, disrupt vasodilatation and promote apoptosis. In this Review, we discuss the body of literature suggesting that ceramides are drivers - and not merely passengers - on the road to cardiovascular disease. Moreover, we explore the feasibility of therapeutic strategies to lower ceramide levels to improve cardiovascular health.
Collapse
Affiliation(s)
- Ran Hee Choi
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
48
|
Physiological significance of pericoronary inflammation in epicardial functional stenosis and global coronary flow reserve. Sci Rep 2021; 11:19026. [PMID: 34561466 PMCID: PMC8463533 DOI: 10.1038/s41598-021-97849-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022] Open
Abstract
Both fractional flow reserve (FFR) and global coronary flow reserve (g-CFR) provide prognostic information in patients with stable coronary artery disease (CAD). Inflammation plays a vital role in impaired endothelial dysfunction and atherosclerotic progression, potentially predicting cardiovascular mortality. This study aimed to evaluate the physiological significance of pericoronary adipose tissue inflammation assessed by CT attenuation (PCATA) in epicardial functional stenosis severity and g-CFR in patients with CAD. A total of 131 CAD patients with a single de novo epicardial coronary stenosis who underwent coronary CT-angiography (CCTA), phase-contrast cine-magnetic resonance imaging (PC-CMR) and FFR measurement were studied. PCATA was assessed using the mean CT attenuation value. G-CFR was obtained by quantifying absolute coronary sinus flow (ml/min/g) by PC-CMR at rest and during maximum hyperemia. Median FFR, g-CFR, and PCATA values were 0.75, 2.59, and − 71.3, respectively. Serum creatinine, NT-proBNP, left ventricular end-diastolic volume, and PCATA were independently associated with g-CFR. PCATA showed a significant incremental predictive efficacy for impaired g-CFR (< 2.0) when added to the clinical risk model. PCATA was significantly associated with g-CFR, independent of FFR. Our results suggest the pathophysiological mechanisms linking perivascular inflammation with g-CFR in CAD patients.
Collapse
|
49
|
Fujiwara Y, Okada S, Uryu K, Maru I, Komohara Y. The extract of Ilex kudingcha inhibits atherosclerosis in apoE-deficient mice by suppressing cholesterol accumulation in macrophages. Biosci Biotechnol Biochem 2021; 85:2177-2184. [PMID: 34369980 DOI: 10.1093/bbb/zbab140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/27/2021] [Indexed: 11/13/2022]
Abstract
It was previously reported that oleanolic acid and ursolic acid, triterpenoid compounds occurring in Ilex kudingcha, ameliorate hyperlipidemia and atherosclerosis in apoE-deficient mice. In the present study, we investigated whether I. kudingcha extract exerts similar inhibitory effects on cholesterol accumulation in human monocyte-derived macrophages (HMDMs) and atherogenesis in apoE-deficient mice. I. kudingcha extract significantly inhibited cholesterol ester (CE) accumulation induced by acetylated LDL (acetyl-LDL) in HMDMs; however, it generated no effect on cell viability in HMDMs. I. kudingcha extract also suppressed CE accumulation in acyl-CoA:cholesterol acyl-transferase (ACAT)-overexpressing Chinese hamster ovary (CHO) cells, thereby indicating that it inhibits ACAT activity. Furthermore, the oral administration of I. kudingcha extract to apoE-deficient mice significantly decreased the levels of serum cholesterol, triglyceride, sLOX-1, as well as the regions of atherosclerotic lesions in the mice. Our study reveals crucial new-found evidence that I. kudingcha extract significantly inhibits ACAT activity and suppresses atherogenesis.
Collapse
Affiliation(s)
- Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
50
|
Pandey M, Cuddihy G, Gordon JA, Cox ME, Wasan KM. Inhibition of Scavenger Receptor Class B Type 1 (SR-B1) Expression and Activity as a Potential Novel Target to Disrupt Cholesterol Availability in Castration-Resistant Prostate Cancer. Pharmaceutics 2021; 13:1509. [PMID: 34575583 PMCID: PMC8467449 DOI: 10.3390/pharmaceutics13091509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
There have been several studies that have linked elevated scavenger receptor class b type 1 (SR-B1) expression and activity to the development and progression of castration-resistant prostate cancer (CRPC). SR-B1 facilitates the influx of cholesterol to the cell from lipoproteins in systemic circulation. This influx of cholesterol may be important for many cellular functions, including the synthesis of androgens. Castration-resistant prostate cancer tumors can synthesize androgens de novo to supplement the loss of exogenous sources often induced by androgen deprivation therapy. Silencing of SR-B1 may impact the ability of prostate cancer cells, particularly those of the castration-resistant state, to maintain the intracellular supply of androgens by removing a supply of cholesterol. SR-B1 expression is elevated in CRPC models and has been linked to poor survival of patients. The overarching belief has been that cholesterol modulation, through either synthesis or uptake inhibition, will impact essential signaling processes, impeding the proliferation of prostate cancer. The reduction in cellular cholesterol availability can impede prostate cancer proliferation through both decreased steroid synthesis and steroid-independent mechanisms, providing a potential therapeutic target for the treatment of prostate cancer. In this article, we discuss and highlight the work on SR-B1 as a potential novel drug target for CRPC management.
Collapse
Affiliation(s)
- Mitali Pandey
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Grace Cuddihy
- College of Pharmacy and Nutrition, University of Saskatchewan, 104 Clinic Place, Saskatoon, SK S7N 2Z4, Canada;
| | - Jacob A. Gordon
- Oncology Bioscience, Oncology R&D, AstraZeneca, Boston, MA 02451, USA;
| | - Michael E. Cox
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Kishor M. Wasan
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| |
Collapse
|