51
|
Echrish J, Pasca MI, Cabrera D, Yang Y, Harper AGS. Developing a Biomimetic 3D Neointimal Layer as a Prothrombotic Substrate for a Humanized In Vitro Model of Atherothrombosis. Biomimetics (Basel) 2024; 9:372. [PMID: 38921252 PMCID: PMC11201422 DOI: 10.3390/biomimetics9060372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Acute cardiovascular events result from clots caused by the rupture and erosion of atherosclerotic plaques. This paper aimed to produce a functional biomimetic hydrogel of the neointimal layer of the atherosclerotic plaque that can support thrombogenesis upon exposure to human blood. A biomimetic hydrogel of the neointima was produced by culturing THP-1-derived foam cells within 3D collagen hydrogels in the presence or absence of atorvastatin. Prothrombin time and platelet aggregation onset were measured after exposure of the neointimal models to platelet-poor plasma and washed platelet suspensions prepared from blood of healthy, medication-free volunteers. Activity of the extrinsic coagulation pathway was measured using the fluorogenic substrate SN-17. Foam cell formation was observed following preincubation of the neointimal biomimetic hydrogels with oxidized LDL, and this was inhibited by pretreatment with atorvastatin. The neointimal biomimetic hydrogel was able to trigger platelet aggregation and blood coagulation upon exposure to human blood products. Atorvastatin pretreatment of the neointimal biomimetic layer significantly reduced its pro-aggregatory and pro-coagulant properties. In the future, this 3D neointimal biomimetic hydrogel can be incorporated as an additional layer within our current thrombus-on-a-chip model to permit the study of atherosclerosis development and the screening of anti-thrombotic drugs as an alternative to current animal models.
Collapse
Affiliation(s)
| | | | - David Cabrera
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK; (D.C.); (Y.Y.)
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK; (D.C.); (Y.Y.)
| | | |
Collapse
|
52
|
Hannan Hazari MA, Laxman Rao K, Tazneem B, Rafeeq S, Fatima SR, Jabeen S, Kavya K. CORRELATION OF COMORBIDITIES AND OUTCOME IN CAD PATIENTS: A NOVEL TANGENTS SCORE STUDY. MILITARY MEDICAL SCIENCE LETTERS 2024. [DOI: 10.31482/mmsl.2024.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
|
53
|
Liu Y, Jiang Z, Yang X, Wang Y, Yang B, Fu Q. Engineering Nanoplatforms for Theranostics of Atherosclerotic Plaques. Adv Healthc Mater 2024; 13:e2303612. [PMID: 38564883 DOI: 10.1002/adhm.202303612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Atherosclerotic plaque formation is considered the primary pathological mechanism underlying atherosclerotic cardiovascular diseases, leading to severe cardiovascular events such as stroke, acute coronary syndromes, and even sudden cardiac death. Early detection and timely intervention of plaques are challenging due to the lack of typical symptoms in the initial stages. Therefore, precise early detection and intervention play a crucial role in risk stratification of atherosclerotic plaques and achieving favorable post-interventional outcomes. The continuously advancing nanoplatforms have demonstrated numerous advantages including high signal-to-noise ratio, enhanced bioavailability, and specific targeting capabilities for imaging agents and therapeutic drugs, enabling effective visualization and management of atherosclerotic plaques. Motivated by these superior properties, various noninvasive imaging modalities for early recognition of plaques in the preliminary stage of atherosclerosis are comprehensively summarized. Additionally, several therapeutic strategies are proposed to enhance the efficacy of treating atherosclerotic plaques. Finally, existing challenges and promising prospects for accelerating clinical translation of nanoplatform-based molecular imaging and therapy for atherosclerotic plaques are discussed. In conclusion, this review provides an insightful perspective on the diagnosis and therapy of atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zeyu Jiang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
54
|
Malý M, Kučerka O, Bechyňská K, Kočí K, Mandys V, Hajšlová J, Kosek V. Plasma lipidome differences in patients with and without significant carotid plaque. Vascul Pharmacol 2024; 155:107377. [PMID: 38705432 DOI: 10.1016/j.vph.2024.107377] [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: 12/10/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Atherosclerosis is a major cause of ischemic stroke, and early detection of advanced atherosclerosis in the carotid artery is important for reducing morbidity and mortality. What is even more important is not only detection of atherosclerosis but early determination whether the patients are at high risk of an event with adverse effects as the size of the plaque does not necessarily reflect its potential to trigger such events. AIM We studied whether plasma lipidomics profile can be used as a diagnostic tool for stratification of stable or unstable plaques without the need of removing the carotid plaque. METHODS This study used liquid chromatography high-resolution tandem mass spectrometry lipidomics to characterize lipid profiles in patients' plasma and found that patients with significant and complicated (vulnerable) atherosclerotic plaque had distinct lipid profiles compared to those with insignificant plaques. RESULTS The lipid classes that were most predictive of vulnerable plaque were lysophosphoethanolamines, fatty acyl esters of hydroxy fatty acids, free fatty acids, plasmalogens, and triacylglycerols. Most of these compounds were found decreased in plasma of patients with unstable plaques which enabled sufficient performance of a statistical model used for patient stratification. CONCLUSIONS Plasma lipidomes measured by liquid chromatography-mass spectrometry show differences in patients with stable and unstable carotid plaques, therefore these compounds could potentially be used as biomarkers for unstable plaque in future clinical diagnosis.
Collapse
Affiliation(s)
- Martin Malý
- Department of Medicine, First Faculty of Medicine, Charles University in Prague and the Military University Hospital, Prague 16902, Czech Republic
| | - Ondřej Kučerka
- Department of Medicine, First Faculty of Medicine, Charles University in Prague and the Military University Hospital, Prague 16902, Czech Republic; Department of Military Internal Medicine and Military Hygiene, Faculty of Military Health Sciences, University of Defence, Hradec Kralove 50002, Czech Republic
| | - Kamila Bechyňská
- University of Chemistry and Technology, Department of Food Chemistry and Analysis, Technická 3, Prague 6 166 28, Czech Republic
| | - Karolína Kočí
- Department of Medicine, First Faculty of Medicine, Charles University in Prague and the Military University Hospital, Prague 16902, Czech Republic
| | - Václav Mandys
- Department of Pathology, Third Faculty of Medicine, Charles University and the University Hospital Kralovske Vinohrady, Prague 100 00, Czech Republic
| | - Jana Hajšlová
- University of Chemistry and Technology, Department of Food Chemistry and Analysis, Technická 3, Prague 6 166 28, Czech Republic
| | - Vít Kosek
- University of Chemistry and Technology, Department of Food Chemistry and Analysis, Technická 3, Prague 6 166 28, Czech Republic.
| |
Collapse
|
55
|
Nenna A, Laudisio A, Taffon C, Fogolari M, Spadaccio C, Ferrisi C, Loreni F, Giacinto O, Mastroianni C, Barbato R, Rose D, Salsano A, Santini F, Angeletti S, Crescenzi A, Antonelli Incalzi R, Chello M, Lusini M. Intestinal Microbiota and Derived Metabolites in Myocardial Fibrosis and Postoperative Atrial Fibrillation. Int J Mol Sci 2024; 25:6037. [PMID: 38892223 PMCID: PMC11173100 DOI: 10.3390/ijms25116037] [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: 05/01/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The high incidence of atrial fibrillation (AFib) following cardiac surgery (postoperative atrial fibrillation, POAF) relies on specific surgical features. However, in the setting of POAF, the role of the microbiome in the modulation of cardiac fibrosis is still not clear. This study aimed to analyze the effect of the microbiome and its main metabolic product (trimethylamine-N-oxide, TMAO) in the fibrosis of myocardial tissue, to investigate its role in POAF. Patients undergoing elective cardiac surgery with cardiopulmonary bypass, central atrio-caval cannulation and no history of AFib, were included. A fragment of the right atrium was analyzed for qualitative and mRNA-quantitative evaluation. A preoperative blood sample was analyzed with enzyme-linked immunosorbent assay (ELISA). A total of 100 patients have been included, with POAF occurring in 38%. Histologically, a higher degree of fibrosis, angiogenesis and inflammation has been observed in POAF. Quantitative evaluation showed increased mRNA expression of collagen-1, collagen-3, fibronectin, and transforming growth factor beta (TGFb) in the POAF group. ELISA analysis showed higher levels of TMAO, lipopolysaccharide and TGFb in POAF, with similar levels of sP-selectin and zonulin. TMAO ≥ 61.8 ng/mL (odds ratio, OR 2.88 [1.35-6.16], p = 0.006), preoperative hemoglobin < 13.1 g/dL (OR 2.37 [1.07-5.24], p = 0.033) and impaired right ventricular function (OR 2.38 [1.17-4.83], p = 0.017) were independent predictors of POAF. Also, TMAO was significantly associated with POAF by means of increased fibrosis. Gut microbiome product TMAO is crucial for myocardial fibrosis, which is a key factor for POAF. Patients in preoperative sinus rhythm who will develop POAF have increased genetic expression of pro-fibrotic genes and enhanced fibrosis in histological staining. Elevated TMAO level (≥61.8 ng/mL) is an independent risk factor for POAF.
Collapse
Affiliation(s)
- Antonio Nenna
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Alice Laudisio
- Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (A.L.); (R.A.I.)
| | - Chiara Taffon
- Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (C.T.); (A.C.)
| | - Marta Fogolari
- Clinical Laboratory, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (M.F.); (S.A.)
| | - Cristiano Spadaccio
- Cardiac Surgery, University of Cincinnati Medical Center, Cincinnati, OH 45219, USA;
- Cardiothoracic Surgery, Lancashire Cardiac Centre, Blackpool Teaching Hospital, Blackpool FY3 8NP, UK;
| | - Chiara Ferrisi
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Francesco Loreni
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Omar Giacinto
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Ciro Mastroianni
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Raffaele Barbato
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - David Rose
- Cardiothoracic Surgery, Lancashire Cardiac Centre, Blackpool Teaching Hospital, Blackpool FY3 8NP, UK;
| | - Antonio Salsano
- Cardiac Surgery, Ospedale Policlinico San Martino, University of Genoa, 16126 Genoa, Italy; (A.S.); (F.S.)
| | - Francesco Santini
- Cardiac Surgery, Ospedale Policlinico San Martino, University of Genoa, 16126 Genoa, Italy; (A.S.); (F.S.)
| | - Silvia Angeletti
- Clinical Laboratory, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (M.F.); (S.A.)
| | - Anna Crescenzi
- Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (C.T.); (A.C.)
| | - Raffaele Antonelli Incalzi
- Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (A.L.); (R.A.I.)
| | - Massimo Chello
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Mario Lusini
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| |
Collapse
|
56
|
Fularski P, Czarnik W, Dąbek B, Lisińska W, Radzioch E, Witkowska A, Młynarska E, Rysz J, Franczyk B. Broader Perspective on Atherosclerosis-Selected Risk Factors, Biomarkers, and Therapeutic Approach. Int J Mol Sci 2024; 25:5212. [PMID: 38791250 PMCID: PMC11121693 DOI: 10.3390/ijms25105212] [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: 03/30/2024] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) stands as the leading cause of mortality worldwide. At its core lies a progressive process of atherosclerosis, influenced by multiple factors. Among them, lifestyle-related factors are highlighted, with inadequate diet being one of the foremost, alongside factors such as cigarette smoking, low physical activity, and sleep deprivation. Another substantial group of risk factors comprises comorbidities. Amongst others, conditions such as hypertension, diabetes mellitus (DM), chronic kidney disease (CKD), or familial hypercholesterolemia (FH) are included here. Extremely significant in the context of halting progression is counteracting the mentioned risk factors, including through treatment of the underlying disease. What is more, in recent years, there has been increasing attention paid to perceiving atherosclerosis as an inflammation-related disease. Consequently, efforts are directed towards exploring new anti-inflammatory medications to limit ASCVD progression. Simultaneously, research is underway to identify biomarkers capable of providing insights into the ongoing process of atherosclerotic plaque formation. The aim of this study is to provide a broader perspective on ASCVD, particularly focusing on its characteristics, traditional and novel treatment methods, and biomarkers that can facilitate its early detection.
Collapse
Affiliation(s)
- Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Witold Czarnik
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Bartłomiej Dąbek
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewa Radzioch
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Alicja Witkowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
57
|
Andercou O, Andrei MC, Gheban D, Marian D, Coman HF, Oprea VA, Mihaileanu FV, Ciocan R, Cucuruz B, Stancu B. Histological Changes in the Popliteal Artery Wall in Patients with Critical Limb Ischemia. Diagnostics (Basel) 2024; 14:989. [PMID: 38786287 PMCID: PMC11119664 DOI: 10.3390/diagnostics14100989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
INTRODUCTION This prospective study aims to illustrate the histopathological arterial changes in the popliteal artery in peripheral arterial disease of the lower limbs. MATERIAL AND METHOD A total of 60 popliteal artery segments taken from patients who had undergone lower limb amputation were examined between April and June 2023. The degree of arterial stenosis, medial calcinosis, and the vasa vasorum changes in the arterial adventitia were quantified. The presence of risk factors for atherosclerosis was also observed. RESULTS Atherosclerotic plaque was found in all of the examined segments. Medial calcinosis was observed in 40 (66.6%) of the arterial segments. A positive association between the degree of arterial stenosis and the vasa vasorum changes in the arterial adventitia was also found (p = 0.025). The level of blood sugar and cholesterol were predictive factors for the severity of atherosclerosis. CONCLUSIONS Atherosclerosis and medial calcinosis are significant in patients who underwent lower limb amputation. Medial calcinosis causes damage to the arterial wall and leads to a reduction in responsiveness to dilator stimuli.
Collapse
Affiliation(s)
- Octavian Andercou
- Department of Surgery, Emergency County Hospital Cluj, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj Napoca, Romania; (F.V.M.); (R.C.); (B.S.)
| | - Maria Cristina Andrei
- Vascular Surgery Department, Satu Mare County Emergency Hospital, 440192 Satu Mare, Romania;
| | - Dan Gheban
- Department of Pathological Anatomy, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj-Napoca, Romania;
| | - Dorin Marian
- Second Surgical Department, Emergency County Hospital Mures, University of Medicine and Pharmacy Emil Palade, 540142 Targu Mures, Romania;
| | - Horațiu F. Coman
- Department of Vascular Surgery, Emergency County Hospital, 400006 Cluj Napoca, Romania;
| | - Valentin Aron Oprea
- Department of Surgery, Emergency Military Hospital Cluj Napoca, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj-Napoca, Romania;
| | - Florin Vasile Mihaileanu
- Department of Surgery, Emergency County Hospital Cluj, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj Napoca, Romania; (F.V.M.); (R.C.); (B.S.)
| | - Razvan Ciocan
- Department of Surgery, Emergency County Hospital Cluj, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj Napoca, Romania; (F.V.M.); (R.C.); (B.S.)
| | - Beatrix Cucuruz
- Department of Vascular Surgery, Martha Maria Hospital Nuremberg, 90491 Nuremberg, Germany;
| | - Bogdan Stancu
- Department of Surgery, Emergency County Hospital Cluj, University of Medicine and Pharmacy Iuliu Hatieganu, 400347 Cluj Napoca, Romania; (F.V.M.); (R.C.); (B.S.)
| |
Collapse
|
58
|
Guillamot M, Subudhi I, Paraskevopoulou V, Prystupa A, Sidhu I, Yeaton A, Laskou M, Hannemann C, Donahoe C, Wiseman D, Aifantis I, Naik S, Weinstock A. Interferon-sensitized hematopoietic progenitors dynamically alter organismal immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590828. [PMID: 38712060 PMCID: PMC11071608 DOI: 10.1101/2024.04.24.590828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Inflammation has enduring impacts on organismal immunity. However, the precise mechanisms by which tissue-restricted inflammation conditions systemic responses are poorly understood. Here, we leveraged a highly compartmentalized model of skin inflammation and identified a surprising type I interferon (IFN)- mediated activation of hematopoietic stem/progenitor cells (HSPCs) that results in profound changes to systemic host responses. Post-inflamed mice were protected from atherosclerosis and had worse outcomes following influenza virus infection. This IFN-mediated HSPC modulation was dependent on IFNAR signaling and could be recapitulated with the administration of recombinant IFNα. Importantly, the transfer of post-inflamed HSPCs was sufficient to transmit the immune suppression phenotype. IFN modulation of HSPCs was rooted both in long-term changes in chromatin accessibility and the emergence of an IFN- responsive functional state from multiple progenitor populations. Collectively, our data reveal the profound and enduring effect of transient inflammation and more specifically type I IFN signaling and set the stage for a more nuanced understanding of HSPC functional modulation by peripheral immune signals.
Collapse
|
59
|
Miceli G, Basso MG, Pintus C, Pennacchio AR, Cocciola E, Cuffaro M, Profita M, Rizzo G, Tuttolomondo A. Molecular Pathways of Vulnerable Carotid Plaques at Risk of Ischemic Stroke: A Narrative Review. Int J Mol Sci 2024; 25:4351. [PMID: 38673936 PMCID: PMC11050267 DOI: 10.3390/ijms25084351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The concept of vulnerable carotid plaques is pivotal in understanding the pathophysiology of ischemic stroke secondary to large-artery atherosclerosis. In macroscopic evaluation, vulnerable plaques are characterized by one or more of the following features: microcalcification; neovascularization; lipid-rich necrotic cores (LRNCs); intraplaque hemorrhage (IPH); thin fibrous caps; plaque surface ulceration; huge dimensions, suggesting stenosis; and plaque rupture. Recognizing these macroscopic characteristics is crucial for estimating the risk of cerebrovascular events, also in the case of non-significant (less than 50%) stenosis. Inflammatory biomarkers, such as cytokines and adhesion molecules, lipid-related markers like oxidized low-density lipoprotein (LDL), and proteolytic enzymes capable of degrading extracellular matrix components are among the key molecules that are scrutinized for their associative roles in plaque instability. Through their quantification and evaluation, these biomarkers reveal intricate molecular cross-talk governing plaque inflammation, rupture potential, and thrombogenicity. The current evidence demonstrates that plaque vulnerability phenotypes are multiple and heterogeneous and are associated with many highly complex molecular pathways that determine the activation of an immune-mediated cascade that culminates in thromboinflammation. This narrative review provides a comprehensive analysis of the current knowledge on molecular biomarkers expressed by symptomatic carotid plaques. It explores the association of these biomarkers with the structural and compositional attributes that characterize vulnerable plaques.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Andrea Roberta Pennacchio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Elena Cocciola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Mariagiovanna Cuffaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Martina Profita
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (G.M.); (M.G.B.); (C.P.); (A.R.P.); (E.C.); (M.C.); (M.P.); (G.R.)
- Internal Medicine and Stroke Care Ward, University Hospital, Policlinico “P. Giaccone”, 90127 Palermo, Italy
| |
Collapse
|
60
|
Hayderi A, Kumawat AK, Shavva VS, Dreifaldt M, Sigvant B, Petri MH, Kragsterman B, Olofsson PS, Sirsjö A, Ljungberg LU. RSAD2 is abundant in atherosclerotic plaques and promotes interferon-induced CXCR3-chemokines in human smooth muscle cells. Sci Rep 2024; 14:8196. [PMID: 38589444 PMCID: PMC11001978 DOI: 10.1038/s41598-024-58592-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 04/01/2024] [Indexed: 04/10/2024] Open
Abstract
In atherosclerotic lesions, monocyte-derived macrophages are major source of interferon gamma (IFN-γ), a pleotropic cytokine known to regulate the expression of numerous genes, including the antiviral gene RSAD2. While RSAD2 was reported to be expressed in endothelial cells of human carotid lesions, its significance for the development of atherosclerosis remains utterly unknown. Here, we harnessed publicly available human carotid atherosclerotic data to explore RSAD2 in lesions and employed siRNA-mediated gene-knockdown to investigate its function in IFN-γ-stimulated human aortic smooth muscle cells (hAoSMCs). Silencing RSAD2 in IFN-γ-stimulated hAoSMCs resulted in reduced expression and secretion of key CXCR3-chemokines, CXCL9, CXCL10, and CXCL11. Conditioned medium from RSAD2-deficient hAoSMCs exhibited diminished monocyte attraction in vitro compared to conditioned medium from control cells. Furthermore, RSAD2 transcript was elevated in carotid lesions where it was expressed by several different cell types, including endothelial cells, macrophages and smooth muscle cells. Interestingly, RSAD2 displayed significant correlations with CXCL10 (r = 0.45, p = 0.010) and CXCL11 (r = 0.53, p = 0.002) in human carotid lesions. Combining our findings, we uncover a novel role for RSAD2 in hAoSMCs, which could potentially contribute to monocyte recruitment in the context of atherosclerosis.
Collapse
Affiliation(s)
- Assim Hayderi
- School of Medical Sciences, Örebro University, Örebro, Sweden.
| | - Ashok K Kumawat
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Vladimir S Shavva
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Center for Bioelectronic Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mats Dreifaldt
- School of Medical Sciences, Örebro University, Örebro, Sweden
- Department of Cardiothoracic Surgery and Vascular Surgery, Örebro University Hospital, Örebro, Sweden
| | - Birgitta Sigvant
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Centre for Clinical Research and Education, Region Värmland, Karlstad, Sweden
| | - Marcelo H Petri
- School of Medical Sciences, Örebro University, Örebro, Sweden
- Department of Cardiothoracic Surgery and Vascular Surgery, Örebro University Hospital, Örebro, Sweden
| | - Björn Kragsterman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Surgery, Västmanlands Hospital Västerås, Västerås, Sweden
| | - Peder S Olofsson
- Laboratory of Immunobiology, Division of Cardiovascular Medicine, Department of Medicine, Center for Bioelectronic Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Allan Sirsjö
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | | |
Collapse
|
61
|
Homma S, Kato K. Validity of Atherosclerotic Calcified Lesions Observed on Low-Dose Computed Tomography and Cardio-Ankle Vascular Index as Surrogate Markers of Atherosclerosis Progression. Angiology 2024; 75:349-358. [PMID: 36787785 DOI: 10.1177/00033197231155963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The significance of atherosclerotic calcified lesions observed on low-dose computed tomography (LDCT) performed during general checkups was investigated. The coronary arteries (CA), ascending aorta and aortic arch (AAAA), descending thoracic aorta (DTA), and abdominal aorta (AA) were examined. Semiquantitative calcified index analysis of the DTA and AA in terms of atherosclerosis risk factors and cardio-ankle vascular index (CAVI) measurements was also performed. We included 1594 participants (mean age: 59.2 years; range: 31-91 years). The prevalence of calcified lesions was 71.0%, 66.6%, 57.2%, and 37.9% in the AA, CA, AAAA, and DTA, respectively. Age-related advances in calcification among participants with no major risk factors, revealed that calcification appeared earliest in the AA, followed by the CA, AAAA, and DTA. Participants with calcified lesions in all arteries had a significantly greater CAVI than those without calcification. The CAVI was negatively correlated with low-density lipoprotein cholesterol levels, particularly in participants without calcified lesions in the DTA. Calcified lesions on LDCT could indicate the end stage of atherosclerotic lesions. The CAVI can be used to assess atherosclerotic changes at all stages of disease progression. A combination of LDCT and CAVI could be used as a routine non-invasive assessment of atherosclerosis.
Collapse
Affiliation(s)
- Satoki Homma
- Health Care Center in Saitama Medical Center of the Japan Community Health Care Organization, Saitama, Japan
- Faculty of Nursing and Medical Care, Keio University & Keio Research Institute at SFC (Shonan Fujisawa Campus), Fujisawa, Japan
| | - Kiyoe Kato
- Center of General Health Check-Up, Saiseikai Central Hospital, Tokyo, Japan
| |
Collapse
|
62
|
Xiao J, Poblete RA, Lerner A, Nguyen PL, Song JW, Sanossian N, Wilcox AG, Song SS, Lyden PD, Saver JL, Wasserman BA, Fan Z. MRI in the Evaluation of Cryptogenic Stroke and Embolic Stroke of Undetermined Source. Radiology 2024; 311:e231934. [PMID: 38652031 PMCID: PMC11070612 DOI: 10.1148/radiol.231934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 04/25/2024]
Abstract
Cryptogenic stroke refers to a stroke of undetermined etiology. It accounts for approximately one-fifth of ischemic strokes and has a higher prevalence in younger patients. Embolic stroke of undetermined source (ESUS) refers to a subgroup of patients with nonlacunar cryptogenic strokes in whom embolism is the suspected stroke mechanism. Under the classifications of cryptogenic stroke or ESUS, there is wide heterogeneity in possible stroke mechanisms. In the absence of a confirmed stroke etiology, there is no established treatment for secondary prevention of stroke in patients experiencing cryptogenic stroke or ESUS, despite several clinical trials, leaving physicians with a clinical dilemma. Both conventional and advanced MRI techniques are available in clinical practice to identify differentiating features and stroke patterns and to determine or infer the underlying etiologic cause, such as atherosclerotic plaques and cardiogenic or paradoxical embolism due to occult pelvic venous thrombi. The aim of this review is to highlight the diagnostic utility of various MRI techniques in patients with cryptogenic stroke or ESUS. Future trends in technological advancement for promoting the adoption of MRI in such a special clinical application are also discussed.
Collapse
Affiliation(s)
- Jiayu Xiao
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Roy A. Poblete
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Alexander Lerner
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Peggy L. Nguyen
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Jae W. Song
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Nerses Sanossian
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Alison G. Wilcox
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Shlee S. Song
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Patrick D. Lyden
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Jeffrey L. Saver
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Bruce A. Wasserman
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| | - Zhaoyang Fan
- From the Departments of Radiology (J.X., A.L., A.G.W., Z.F.),
Neurology (R.A.P., P.L.N., N.S., P.D.L.), Physiology and Neuroscience (P.D.L.),
Biomedical Engineering (Z.F.), and Radiation Oncology (Z.F.), University of
Southern California, 2250 Alcazar St, CSC Room 104, Los Angeles, CA 90033;
Department of Radiology, Hospital of the University of Pennsylvania,
Philadelphia, Pa (J.W.S.); Department of Neurology, Cedars-Sinai Medical Center,
Los Angeles, Calif (S.S.S.); Comprehensive Stroke Center and Department of
Neurology, David Geffen School of Medicine, University of California–Los
Angeles, Los Angeles, Calif (J.L.S.); Department of Diagnostic Radiology and
Nuclear Medicine, University of Maryland–Baltimore, Baltimore, Md
(B.A.W.); and Department of Radiology and Radiological Sciences, Johns Hopkins
University, Baltimore, Md (B.A.W.)
| |
Collapse
|
63
|
Arderiu G, Bejar MT, Civit-Urgell A, Peña E, Badimon L. Crosstalk of human coronary perivascular adipose-derived stem cells with vascular cells: role of tissue factor. Basic Res Cardiol 2024; 119:291-307. [PMID: 38430261 DOI: 10.1007/s00395-024-01037-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
The coronary perivascular adipose tissue (cPVAT) has been associated to the burden of cardiovascular risk factors and to the underlying vessel atherosclerotic plaque severity. Although the "outside to inside" hypothesis of PVAT-derived-adipokine regulation of vessel function is currently accepted, whether the resident mesenchymal stem cells (ASCs) in PVAT have a regulatory role on the underlying vascular arterial smooth muscle cells (VSMCs) is not known. Here, we investigated the interactions between resident PVAT-ASCs and VSMCs. ASCs were obtained from PVAT overlying the left anterior descending (LAD) coronary artery of hearts removed at heart transplant operations. PVAT was obtained both from patients with non-ischemic and ischemic heart disease as the cause of heart transplant. ASCs were isolated from PVAT, phenotypically characterized by flow cytometry, functionally tested for proliferation, and differentiation. Crosstalk between ASCs and VSMCs was investigated by co-culture studies. ASCs were detected in the adventitia of the LAD-PVAT showing differentiation capacity and angiogenic potential. ASCs obtained from PVAT of non-ischemic and ischemic hearts showed different tissue factor (TF) expression levels, different VSMCs recruitment capacity through the axis ERK1/2-ETS1 signaling and different angiogenic potential. Induced upregulation of TF in ASCs isolated from ischemic PVAT rescued their angiogenic capacity in subcutaneously implanted plugs in mice, whereas silencing TF in ASCs decreased the proangiogenic capacity of non-ischemic ASCs. The results indicate for the first time a novel mechanism of regulation of VSMCs by PVAT-ASCs in angiogenesis, mediated by TF expression in ASCs. Regulation of TF in ASCs may become a therapeutic intervention to increase cardiac protection.
Collapse
Affiliation(s)
- Gemma Arderiu
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain.
- Ciber CV, Instituto Carlos III, Madrid, Spain.
| | - Maria Teresa Bejar
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Anna Civit-Urgell
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Barcelona, Spain
| | - Esther Peña
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Ciber CV, Instituto Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Ciber CV, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
64
|
Wu X, Zhang H. Omics Approaches Unveiling the Biology of Human Atherosclerotic Plaques. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:482-498. [PMID: 38280419 PMCID: PMC10988765 DOI: 10.1016/j.ajpath.2023.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall, characterized by the buildup of plaques with the accumulation and transformation of lipids, immune cells, vascular smooth muscle cells, and necrotic cell debris. Plaques with collagen-poor thin fibrous caps infiltrated by macrophages and lymphocytes are considered unstable because they are at the greatest risk of rupture and clinical events. However, the current histologic definition of plaque types may not fully capture the complex molecular nature of atherosclerotic plaque biology and the underlying mechanisms contributing to plaque progression, rupture, and erosion. The advances in omics technologies have changed the understanding of atherosclerosis plaque biology, offering new possibilities to improve risk prediction and discover novel therapeutic targets. Genomic studies have shed light on the genetic predisposition to atherosclerosis, and integrative genomic analyses expedite the translation of genomic discoveries. Transcriptomic, proteomic, metabolomic, and lipidomic studies have refined the understanding of the molecular signature of atherosclerotic plaques, aiding in data-driven hypothesis generation for mechanistic studies and offering new prospects for biomarker discovery. Furthermore, advancements in single-cell technologies and emerging spatial analysis techniques have unveiled the heterogeneity and plasticity of plaque cells. This review discusses key omics-based discoveries that have advanced the understanding of human atherosclerotic plaque biology, focusing on insights derived from omics profiling of human atherosclerotic vascular specimens.
Collapse
Affiliation(s)
- Xun Wu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
65
|
Zhang L, Wu X, Hong L. Endothelial Reprogramming in Atherosclerosis. Bioengineering (Basel) 2024; 11:325. [PMID: 38671747 PMCID: PMC11048243 DOI: 10.3390/bioengineering11040325] [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: 03/01/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Atherosclerosis (AS) is a severe vascular disease that results in millions of cases of mortality each year. The development of atherosclerosis is associated with vascular structural lesions, characterized by the accumulation of immune cells, mesenchymal cells, lipids, and an extracellular matrix at the intimal resulting in the formation of an atheromatous plaque. AS involves complex interactions among various cell types, including macrophages, endothelial cells (ECs), and smooth muscle cells (SMCs). Endothelial dysfunction plays an essential role in the initiation and progression of AS. Endothelial dysfunction can encompass a constellation of various non-adaptive dynamic alterations of biology and function, termed "endothelial reprogramming". This phenomenon involves transitioning from a quiescent, anti-inflammatory state to a pro-inflammatory and proatherogenic state and alterations in endothelial cell identity, such as endothelial to mesenchymal transition (EndMT) and endothelial-to-immune cell-like transition (EndIT). Targeting these processes to restore endothelial balance and prevent cell identity shifts, alongside modulating epigenetic factors, can attenuate atherosclerosis progression. In the present review, we discuss the role of endothelial cells in AS and summarize studies in endothelial reprogramming associated with the pathogenesis of AS.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Xin Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
66
|
Irqsusi M, Dong LA, Rodepeter FR, Ramzan R, Talipov I, Ghazy T, Günther M, Vogt S, Rastan AJ. The Role of Matrix Metalloproteinases in Thoracic Aortic Disease: Are They Indicators for the Pathogenesis of Dissections? Biomedicines 2024; 12:619. [PMID: 38540232 PMCID: PMC10967891 DOI: 10.3390/biomedicines12030619] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 04/16/2025] Open
Abstract
The pathogenesis of aortic aneurysm and dissection continues to be under discussion. Extracellular matrix (ECM) remodeling processes in the aortic wall are hypothesized to be involved in the development of the disorders. Therefore, in a histological study, we investigated the expression of metalloproteases 1 and 9 (MMP1 and MMP9) and their inhibitors (TIMP 1 and TIMP 2) in cardiac surgery patients. In parallel, we studied the aortic roots by echocardiography. Clinical reports of 111 patients (30 women and 81 men) who suffered from aortic aneurysms and aortic dissection were evaluated and studied by transesophageal echocardiography. Seven patients who had coronary heart disease served as "healthy controls". All patients underwent the necessary surgical procedure according to the diagnosed aortic disease in the period from 2007 to 2015. A tissue sample of the aortic biopsies was collected from each patient during surgery. Immunohistochemical staining was performed for MMP1 and MMP9 and TIMP1 and TIMP2 as well. Vascularization was monitored by a CD 31 antibody. In direct comparison, the expressions are not homogeneous. We found the smallest changes in the intima area at all. TIMP 1 and TIMP 2 distribution increases from the lumen of the vessel outward in the wall layers of the aorta. In the case of arteriosclerotic changes, intima had a capillarization, but not in the media. An opposite pattern was found in the dissected aortas. There are differences in the vascularization between the aneurysm and dissection and the different layers, respectively. A different remodeling process of the ECM in comparison to the vascular layers must be hypothesized. Reading the patterns of staining and with regard to the known inhibitory effect of MMP9 on ECM remodeling, but especially TIMP 2 on neoangiogenesis, disturbed nutrition, and dysfunctional vasa vasorum remodeling must be assumed as causes of dissection.
Collapse
|
67
|
Lai Z, Wang C, Liu X, Sun H, Guo Z, Shao J, Li K, Chen J, Wang J, Lei X, Shu K, Feng Y, Kong D, Sun W, Liu B. Characterization of the proteome of stable and unstable carotid atherosclerotic plaques using data-independent acquisition mass spectrometry. J Transl Med 2024; 22:247. [PMID: 38454421 PMCID: PMC10921703 DOI: 10.1186/s12967-023-04723-1] [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: 08/01/2023] [Accepted: 11/13/2023] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Currently, noninvasive imaging techniques and circulating biomarkers are still insufficient to accurately assess carotid plaque stability, and an in-depth understanding of the molecular mechanisms that contribute to plaque instability is still lacking. METHODS We established a clinical study cohort containing 182 patients with carotid artery stenosis. After screening, 39 stable and 49 unstable plaques were included in the discovery group, and quantitative proteomics analysis based on data independent acquisition was performed for these plaque samples. Additionally, 35 plaques were included in the validation group to validate the proteomics results by immunohistochemistry analysis. RESULTS A total of 397 differentially expressed proteins were identified in stable and unstable plaques. These proteins are primarily involved in ferroptosis and lipid metabolism-related functions and pathways. Plaque validation results showed that ferroptosis- and lipid metabolism-related proteins had different expression trends in stable plaques versus unstable fibrous cap regions and lipid core regions. Ferroptosis- and lipid metabolism-related mechanisms in plaque stability were discussed. CONCLUSIONS Our results may provide a valuable strategy for revealing the mechanisms affecting plaque stability and will facilitate the discovery of specific biomarkers to broaden the therapeutic scope.
Collapse
Affiliation(s)
- Zhichao Lai
- Department of Vascular Surgery, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Peking Union Medical College, Shuaifuyuan 1St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Chaonan Wang
- Department of Vascular Surgery, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Peking Union Medical College, Shuaifuyuan 1St, Dongcheng District, Beijing, 100730, People's Republic of China
- Department of Hemangiomas & Vascular Malformations, Plastic Surgery Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| | - Xiaoyan Liu
- Proteomics Research Center, Core Facility of Instruments, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Dongdansantiao 9St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Haidan Sun
- Proteomics Research Center, Core Facility of Instruments, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Dongdansantiao 9St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Zhengguang Guo
- Proteomics Research Center, Core Facility of Instruments, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Dongdansantiao 9St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Jiang Shao
- Department of Vascular Surgery, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Peking Union Medical College, Shuaifuyuan 1St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Kang Li
- Department of Vascular Surgery, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Peking Union Medical College, Shuaifuyuan 1St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Junye Chen
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Jiaxian Wang
- Eight-Year Program of Clinical Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Xiangling Lei
- Eight-Year Program of Clinical Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Keqiang Shu
- Department of Vascular Surgery, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Peking Union Medical College, Shuaifuyuan 1St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Yuyao Feng
- Department of Vascular Surgery, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Peking Union Medical College, Shuaifuyuan 1St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Deqiang Kong
- Department of Vascular Surgery, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Peking Union Medical College, Shuaifuyuan 1St, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Wei Sun
- Proteomics Research Center, Core Facility of Instruments, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Dongdansantiao 9St, Dongcheng District, Beijing, 100730, People's Republic of China.
| | - Bao Liu
- Department of Vascular Surgery, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Peking Union Medical College, Shuaifuyuan 1St, Dongcheng District, Beijing, 100730, People's Republic of China.
| |
Collapse
|
68
|
Henriques J, Amaro AM, Piedade AP. Biomimicking Atherosclerotic Vessels: A Relevant and (Yet) Sub-Explored Topic. Biomimetics (Basel) 2024; 9:135. [PMID: 38534820 DOI: 10.3390/biomimetics9030135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Atherosclerosis represents the etiologic source of several cardiovascular events, including myocardial infarction, cerebrovascular accidents, and peripheral artery disease, which remain the leading cause of mortality in the world. Numerous strategies are being delineated to revert the non-optimal projections of the World Health Organization, by both designing new diagnostic and therapeutic approaches or improving the interventional procedures performed by physicians. Deeply understanding the pathological process of atherosclerosis is, therefore, mandatory to accomplish improved results in these trials. Due to their availability, reproducibility, low expensiveness, and rapid production, biomimicking physical models are preferred over animal experimentation because they can overcome some limitations, mainly related to replicability and ethical issues. Their capability to represent any atherosclerotic stage and/or plaque type makes them valuable tools to investigate hemodynamical, pharmacodynamical, and biomechanical behaviors, as well as to optimize imaging systems and, thus, obtain meaningful prospects to improve the efficacy and effectiveness of treatment on a patient-specific basis. However, the broadness of possible applications in which these biomodels can be used is associated with a wide range of tissue-mimicking materials that are selected depending on the final purpose of the model and, consequently, prioritizing some materials' properties over others. This review aims to summarize the progress in fabricating biomimicking atherosclerotic models, mainly focusing on using materials according to the intended application.
Collapse
Affiliation(s)
- Joana Henriques
- University of Coimbra, CEMMPRE, ARISE, Department of Mechanical Engineering, 3030-788 Coimbra, Portugal
| | - Ana M Amaro
- University of Coimbra, CEMMPRE, ARISE, Department of Mechanical Engineering, 3030-788 Coimbra, Portugal
| | - Ana P Piedade
- University of Coimbra, CEMMPRE, ARISE, Department of Mechanical Engineering, 3030-788 Coimbra, Portugal
| |
Collapse
|
69
|
Koppara T, Dregely I, Nekolla SG, Nährig J, Langwieser N, Bradaric C, Ganter C, Laugwitz KL, Schwaiger M, Ibrahim T. Simultaneous 18-FDG PET and MR imaging in lower extremity arterial disease. Front Cardiovasc Med 2024; 11:1352696. [PMID: 38404725 PMCID: PMC10884315 DOI: 10.3389/fcvm.2024.1352696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
Background Simultaneous positron emission tomography (PET) and magnetic resonance imaging (MRI) is a novel hybrid imaging method integrating the advances of morphological tissue characterization of MRI with the pathophysiological insights of PET applications. Aim This study evaluated the use of simultaneous 18-FDG PET/MR imaging for characterizing atherosclerotic lesions in lower extremity arterial disease (LEAD). Methods Eight patients with symptomatic stenoses of the superficial femoral artery (SFA) under simultaneous acquisition of 18-FDG PET and contrast-enhanced MRI using an integrated whole-body PET/MRI scanner. Invasive plaque characterization of the SFA was performed by intravascular imaging using optical coherence tomography. Histological analysis of plaque specimens was performed after directional atherectomy. Results MRI showed contrast enhancement at the site of arterial stenosis, as assessed on T2-w and T1-w images, compared to a control area of the contralateral SFA (0.38 ± 0.15 cm vs. 0.23 ± 0.11 cm; 1.77 ± 0.19 vs. 1.57 ± 0.15; p-value <0.05). On PET imaging, uptake of 18F-FDG (target-to-background ratio TBR > 1) at the level of symptomatic stenosis was observed in all but one patient. Contrast medium-induced MR signal enhancement was detected in all plaques, whereas FDG uptake in PET imaging was increased in lesions with active fibroatheroma and reduced in fibrocalcified lesions. Conclusion In this multimodal imaging study, we report the feasibility and challenges of simultaneous PET/MR imaging of LEAD, which might offer new perspectives for risk estimation.
Collapse
Affiliation(s)
- Tobias Koppara
- Department of Internal Medicine I, Cardiology and Angiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research)—Partner Site Munich Heart Alliance, Munich, Germany
| | - Isabel Dregely
- Department of Nuclear Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Stephan G. Nekolla
- DZHK (German Center for Cardiovascular Research)—Partner Site Munich Heart Alliance, Munich, Germany
- Department of Nuclear Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Jörg Nährig
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nicolas Langwieser
- Department of Internal Medicine I, Cardiology and Angiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christian Bradaric
- Department of Internal Medicine I, Cardiology and Angiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Carl Ganter
- Institute of Radiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Karl-Ludwig Laugwitz
- Department of Internal Medicine I, Cardiology and Angiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research)—Partner Site Munich Heart Alliance, Munich, Germany
| | - Markus Schwaiger
- DZHK (German Center for Cardiovascular Research)—Partner Site Munich Heart Alliance, Munich, Germany
- Department of Nuclear Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Tareq Ibrahim
- Department of Internal Medicine I, Cardiology and Angiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
70
|
Los J, Mensink FB, Mohammadnia N, Opstal TSJ, Damman P, Volleberg RHJA, Peeters DAM, van Royen N, Garcia-Garcia HM, Cornel JH, El Messaoudi S, van Geuns RJM. Invasive coronary imaging of inflammation to further characterize high-risk lesions: what options do we have? Front Cardiovasc Med 2024; 11:1352025. [PMID: 38370159 PMCID: PMC10871865 DOI: 10.3389/fcvm.2024.1352025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 02/20/2024] Open
Abstract
Coronary atherosclerosis remains a leading cause of morbidity and mortality worldwide. The underlying pathophysiology includes a complex interplay of endothelial dysfunction, lipid accumulation and inflammatory pathways. Multiple structural and inflammatory features of the atherosclerotic lesions have become targets to identify high-risk lesions. Various intracoronary imaging devices have been developed to assess the morphological, biocompositional and molecular profile of the intracoronary atheromata. These techniques guide interventional and therapeutical management and allow the identification and stratification of atherosclerotic lesions. We sought to provide an overview of the inflammatory pathobiology of atherosclerosis, distinct high-risk plaque features and the ability to visualize this process with contemporary intracoronary imaging techniques.
Collapse
Affiliation(s)
- Jonathan Los
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frans B. Mensink
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Tjerk S. J. Opstal
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, Netherlands
| | - Peter Damman
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Denise A. M. Peeters
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Niels van Royen
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Jan H. Cornel
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, Netherlands
- Dutch Network for Cardiovascular Research (WCN), Utrecht, Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | |
Collapse
|
71
|
Al-Shaibi K, Bharadwaj A, Mathur A, Jaikishen A, Riley R. Management of Calcified Coronary Lesions. US CARDIOLOGY REVIEW 2024; 18:e01. [PMID: 39494408 PMCID: PMC11526476 DOI: 10.15420/usc.2022.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 08/15/2023] [Indexed: 11/05/2024] Open
Abstract
With an aging population, coronary calcification is increasingly encountered in modern day interventional practice. Unfortunately, it is associated with lower procedural success and higher rates of periprocedural complications, such as failure to deliver stents, perforations, dissections, and other major adverse cardiac events. Furthermore, suboptimal stent deployment in the setting of severe calcification is associated with both short-and long-term major adverse cardiac events, including stent thrombosis, MI, in-stent restenosis, and target lesion revascularization. A variety of treatment options for these lesions exist, including specialized balloons, atherectomy, and intravascular lithotripsy. While there is currently no universally accepted algorithm for choosing between these treatment strategies, several different algorithms exist, and the optimization of these treatment regimens will continue to evolve in the coming years. This review aims to provide insights on the different therapeutic modalities and an understanding of the current body of evidence.
Collapse
Affiliation(s)
- Khaled Al-Shaibi
- Cardiac Center, King Fahd Armed Forces HospitalJeddah, Saudi Arabia
| | - Aditya Bharadwaj
- Division of Cardiology, Loma Linda University Medical CenterLoma Linda, CA
| | - Atul Mathur
- Division of Cardiology, Fortis Escorts Heart InstituteNew Delhi, India
| | - Ashish Jaikishen
- Division of Cardiology, Fortis Escorts Heart InstituteNew Delhi, India
| | - Robert Riley
- Cardiology Division, Overlake Medical Center and ClinicsBellevue, WA
| |
Collapse
|
72
|
Kotsovilis S, Salagianni M, Varela A, Davos CH, Galani IE, Andreakos E. Comprehensive Analysis of 1-Year-Old Female Apolipoprotein E-Deficient Mice Reveals Advanced Atherosclerosis with Vulnerable Plaque Characteristics. Int J Mol Sci 2024; 25:1355. [PMID: 38279355 PMCID: PMC10816800 DOI: 10.3390/ijms25021355] [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: 12/29/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
Apolipoprotein E-knockout (Apoe-/-) mice constitute the most widely employed animal model of atherosclerosis. Deletion of Apoe induces profound hypercholesterolemia and promotes the development of atherosclerosis. However, despite its widespread use, the Apoe-/- mouse model remains incompletely characterized, especially at late time points and advanced disease stages. Thus, it is unclear how late atherosclerotic plaques compare to earlier ones in terms of lipid deposition, calcification, macrophage accumulation, smooth muscle cell presence, or plaque necrosis. Additionally, it is unknown how cardiac function and hemodynamic parameters are affected at late disease stages. Here, we used a comprehensive analysis based on histology, fluorescence microscopy, and Doppler ultrasonography to show that in normal chow diet-fed Apoe-/- mice, atherosclerotic lesions at the level of the aortic valve evolve from a more cellular macrophage-rich phenotype at 26 weeks to an acellular, lipid-rich, and more necrotic phenotype at 52 weeks of age, also marked by enhanced lipid deposition and calcification. Coronary artery atherosclerotic lesions are sparse at 26 weeks but ubiquitous and extensive at 52 weeks; yet, left ventricular function was not significantly affected. These findings demonstrate that atherosclerosis in Apoe-/- mice is a highly dynamic process, with atherosclerotic plaques evolving over time. At late disease stages, histopathological characteristics of increased plaque vulnerability predominate in combination with frequent and extensive coronary artery lesions, which nevertheless may not necessarily result in impaired cardiac function.
Collapse
Affiliation(s)
- Sotirios Kotsovilis
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (S.K.); (M.S.); (I.E.G.)
| | - Maria Salagianni
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (S.K.); (M.S.); (I.E.G.)
| | - Aimilia Varela
- Cardiovascular Research Laboratory, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (A.V.); (C.H.D.)
| | - Constantinos H. Davos
- Cardiovascular Research Laboratory, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (A.V.); (C.H.D.)
| | - Ioanna E. Galani
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (S.K.); (M.S.); (I.E.G.)
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation, Academy of Athens, GR 11527 Athens, Greece; (S.K.); (M.S.); (I.E.G.)
| |
Collapse
|
73
|
Li S, He RC, Wu SG, Song Y, Zhang KL, Tang ML, Bei YR, Zhang T, Lu JB, Ma X, Jiang M, Qin LJ, Xu Y, Dong XH, Wu J, Dai X, Hu YW. LncRNA PSMB8-AS1 Instigates Vascular Inflammation to Aggravate Atherosclerosis. Circ Res 2024; 134:60-80. [PMID: 38084631 DOI: 10.1161/circresaha.122.322360] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 11/20/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Increasing evidence suggests that long noncoding RNAs play significant roles in vascular biology and disease development. One such long noncoding RNA, PSMB8-AS1, has been implicated in the development of tumors. Nevertheless, the precise role of PSMB8-AS1 in cardiovascular diseases, particularly atherosclerosis, has not been thoroughly elucidated. Thus, the primary aim of this investigation is to assess the influence of PSMB8-AS1 on vascular inflammation and the initiation of atherosclerosis. METHODS We generated PSMB8-AS1 knockin and Apoe (Apolipoprotein E) knockout mice (Apoe-/-PSMB8-AS1KI) and global Apoe and proteasome subunit-β type-9 (Psmb9) double knockout mice (Apoe-/-Psmb9-/-). To explore the roles of PSMB8-AS1 and Psmb9 in atherosclerosis, we fed the mice with a Western diet for 12 weeks. RESULTS Long noncoding RNA PSMB8-AS1 is significantly elevated in human atherosclerotic plaques. Strikingly, Apoe-/-PSMB8-AS1KI mice exhibited increased atherosclerosis development, plaque vulnerability, and vascular inflammation compared with Apoe-/- mice. Moreover, the levels of VCAM1 (vascular adhesion molecule 1) and ICAM1 (intracellular adhesion molecule 1) were significantly upregulated in atherosclerotic lesions and serum of Apoe-/-PSMB8-AS1KI mice. Consistently, in vitro gain- and loss-of-function studies demonstrated that PSMB8-AS1 induced monocyte/macrophage adhesion to endothelial cells and increased VCAM1 and ICAM1 levels in a PSMB9-dependent manner. Mechanistic studies revealed that PSMB8-AS1 induced PSMB9 transcription by recruiting the transcription factor NONO (non-POU domain-containing octamer-binding protein) and binding to the PSMB9 promoter. PSMB9 (proteasome subunit-β type-9) elevated VCAM1 and ICAM1 expression via the upregulation of ZEB1 (zinc finger E-box-binding homeobox 1). Psmb9 deficiency decreased atherosclerotic lesion size, plaque vulnerability, and vascular inflammation in Apoe-/- mice in vivo. Importantly, endothelial overexpression of PSMB8-AS1-increased atherosclerosis and vascular inflammation were attenuated by Psmb9 knockout. CONCLUSIONS PSMB8-AS1 promotes vascular inflammation and atherosclerosis via the NONO/PSMB9/ZEB1 axis. Our findings support the development of new long noncoding RNA-based strategies to counteract atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Shu Li
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Run-Chao He
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Shao-Guo Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangdong, China (S.-G.W.)
| | - Yu Song
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Ke-Lan Zhang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Mao-Lin Tang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Yan-Rou Bei
- Laboratory Medicine Center (Y.-R.B.), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Zhang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Jin-Bo Lu
- Department of Peripheral Vascular Surgery, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen (J.-B.L.)
| | - Xin Ma
- Department of Anesthesiology (X.M.), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Jiang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Liang-Jun Qin
- Department of Pathology, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (L.J.Q.)
| | - Yudan Xu
- Laboratory Medicine Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China (Y.X.)
| | - Xian-Hui Dong
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Jia Wu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Xiaoyan Dai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangdong, China (X.D.)
- Clinical Research Institute, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, China (X.D.)
| | - Yan-Wei Hu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
- Department of Laboratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (Y.-W.H.)
| |
Collapse
|
74
|
Liu JW, Zhang ZH, Lv XS, Xu MY, Ni B, He B, Wang F, Chen J, Zhang JB, Ye ZD, Liu P, Wen JY. Identification of key pyroptosis-related genes and microimmune environment among peripheral arterial beds in atherosclerotic arteries. Sci Rep 2024; 14:233. [PMID: 38167983 PMCID: PMC10761966 DOI: 10.1038/s41598-023-50689-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized with innate and adaptive immunity but also involves pyroptosis. Few studies have explored the role of pyroptosis in advanced atherosclerotic plaques from different vascular beds. Here we try to identify the different underlying function of pyroptosis in the progression of atherosclerosis between carotid arteries and femoral. arteries. We extracted gene expression levels from 55 advanced carotid or femoral atherosclerotic plaques. The pyroptosis score of each sample was calculated by single-sample-gene-set enrichment analysis (ssGSEA). We then divided the samples into two clusters: high pyroptosis scores cluster (PyroptosisScoreH cluster) and low pyroptosis scores cluster (PyroptosisScoreL cluster), and assessed functional enrichment and immune cell infiltration in the two clusters. Key pyroptosis related genes were identified by the intersection between results of Cytoscape and LASSO (Least Absolute Shrinkage and Selection Operator) regression analysis. Finally, all key pyroptosis related genes were validated in vitro. We found all but one of the 29 carotid plaque samples belonged to the PyroptosisScoreH cluster and the majority (19 out of 26) of femoral plaques were part of the PyroptosisScoreL cluster. Atheromatous plaque samples in the PyroptosisScoreL cluster had higher proportions of gamma delta T cells, M2 macrophages, myeloid dendritic cells (DCs), and cytotoxic lymphocytes (CTLs), but lower proportions of endothelial cells (ECs). Immune full-activation pathways (e.g., NOD-like receptor signaling pathway and NF-kappa B signaling pathway) were highly enriched in the PyroptosisScoreH cluster. The key pyroptosis related genes GSDMD, CASP1, NLRC4, AIM2, and IL18 were upregulated in advanced carotid atherosclerotic plaques. We concluded that compared to advanced femoral atheromatous plaques, advanced carotid atheromatous plaques were of higher grade of pyroptosis. GSDMD, CASP1, NLRC4, AIM2, and IL18 were the key pyroptosis related genes, which might provide a new sight in the prevention of fatal strokes in advanced carotid atherosclerosis.
Collapse
Affiliation(s)
- Jing-Wen Liu
- Peking University China-Japan Friendship School of Clinical Medicine, NO. 2 Yinghua Eastern Road, Beijing, China
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
| | - Zhao-Hua Zhang
- Peking University China-Japan Friendship School of Clinical Medicine, NO. 2 Yinghua Eastern Road, Beijing, China
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
| | - Xiao-Shuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
- Graduate School of Peking, Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Ming-Yuan Xu
- Peking University China-Japan Friendship School of Clinical Medicine, NO. 2 Yinghua Eastern Road, Beijing, China
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
| | - Bin Ni
- Peking University China-Japan Friendship School of Clinical Medicine, NO. 2 Yinghua Eastern Road, Beijing, China
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
| | - Bin He
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
| | - Feng Wang
- Graduate School of Peking, Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Jie Chen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
| | - Jian-Bin Zhang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
| | - Zhi-Dong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China
| | - Peng Liu
- Peking University China-Japan Friendship School of Clinical Medicine, NO. 2 Yinghua Eastern Road, Beijing, China.
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China.
| | - Jian-Yan Wen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, NO. 2 Yinghua Eastern Road, Beijing, 10029, China.
| |
Collapse
|
75
|
Iannuzzi J, Conte M. Peripheral Arterial Disease. GERIATRIC MEDICINE 2024:429-450. [DOI: 10.1007/978-3-030-74720-6_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
76
|
Chen J, Wang S, Wang K, Abiri P, Huang Z, Yin J, Jabalera AM, Arianpour B, Roustaei M, Zhu E, Zhao P, Cavallero S, Duarte‐Vogel S, Stark E, Luo Y, Benharash P, Tai Y, Cui Q, Hsiai TK. Machine learning-directed electrical impedance tomography to predict metabolically vulnerable plaques. Bioeng Transl Med 2024; 9:e10616. [PMID: 38193119 PMCID: PMC10771559 DOI: 10.1002/btm2.10616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/05/2023] [Accepted: 10/15/2023] [Indexed: 01/10/2024] Open
Abstract
The characterization of atherosclerotic plaques to predict their vulnerability to rupture remains a diagnostic challenge. Despite existing imaging modalities, none have proven their abilities to identify metabolically active oxidized low-density lipoprotein (oxLDL), a marker of plaque vulnerability. To this end, we developed a machine learning-directed electrochemical impedance spectroscopy (EIS) platform to analyze oxLDL-rich plaques, with immunohistology serving as the ground truth. We fabricated the EIS sensor by affixing a six-point microelectrode configuration onto a silicone balloon catheter and electroplating the surface with platinum black (PtB) to improve the charge transfer efficiency at the electrochemical interface. To demonstrate clinical translation, we deployed the EIS sensor to the coronary arteries of an explanted human heart from a patient undergoing heart transplant and interrogated the atherosclerotic lesions to reconstruct the 3D EIS profiles of oxLDL-rich atherosclerotic plaques in both right coronary and left descending coronary arteries. To establish effective generalization of our methods, we repeated the reconstruction and training process on the common carotid arteries of an unembalmed human cadaver specimen. Our findings indicated that our DenseNet model achieves the most reliable predictions for metabolically vulnerable plaque, yielding an accuracy of 92.59% after 100 epochs of training.
Collapse
Affiliation(s)
- Justin Chen
- Department of Bioengineering, Henry Samueli School of EngineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Shaolei Wang
- Department of Bioengineering, Henry Samueli School of EngineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Kaidong Wang
- Division of Cardiology, Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Parinaz Abiri
- Department of Bioengineering, Henry Samueli School of EngineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Division of Cardiology, Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Zi‐Yu Huang
- Department of Medical EngineeringCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| | - Junyi Yin
- Department of Bioengineering, Henry Samueli School of EngineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Alejandro M. Jabalera
- Department of Bioengineering, Henry Samueli School of EngineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Brian Arianpour
- Department of Bioengineering, Henry Samueli School of EngineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Mehrdad Roustaei
- Department of Bioengineering, Henry Samueli School of EngineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Enbo Zhu
- Division of Cardiology, Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Peng Zhao
- Division of Cardiology, Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Susana Cavallero
- Division of Cardiology, Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Division of Cardiology, Department of MedicineGreater Los Angeles VA Healthcare SystemLos AngelesCaliforniaUSA
| | - Sandra Duarte‐Vogel
- Division of Laboratory Animal Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Elena Stark
- Division of Anatomy, Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Yuan Luo
- Department of Medical EngineeringCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| | - Peyman Benharash
- Division of Cardiothoracic Surgery, Department of Surgery, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Yu‐Chong Tai
- Department of Medical EngineeringCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| | - Qingyu Cui
- Division of Cardiology, Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Tzung K. Hsiai
- Department of Bioengineering, Henry Samueli School of EngineeringUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Division of Cardiology, Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Department of Medical EngineeringCalifornia Institute of TechnologyPasadenaCaliforniaUSA
- Division of Cardiology, Department of MedicineGreater Los Angeles VA Healthcare SystemLos AngelesCaliforniaUSA
| |
Collapse
|
77
|
Baaten CCFMJ, Nagy M, Bergmeier W, Spronk HMH, van der Meijden PEJ. Platelet biology and function: plaque erosion vs. rupture. Eur Heart J 2024; 45:18-31. [PMID: 37940193 PMCID: PMC10757869 DOI: 10.1093/eurheartj/ehad720] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/20/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
The leading cause of heart disease in developed countries is coronary atherosclerosis, which is not simply a result of ageing but a chronic inflammatory process that can lead to acute clinical events upon atherosclerotic plaque rupture or erosion and arterial thrombus formation. The composition and location of atherosclerotic plaques determine the phenotype of the lesion and whether it is more likely to rupture or to erode. Although plaque rupture and erosion both initiate platelet activation on the exposed vascular surface, the contribution of platelets to thrombus formation differs between the two phenotypes. In this review, plaque phenotype is discussed in relation to thrombus composition, and an overview of important mediators (haemodynamics, matrix components, and soluble factors) in plaque-induced platelet activation is given. As thrombus formation on disrupted plaques does not necessarily result in complete vessel occlusion, plaque healing can occur. Therefore, the latest findings on plaque healing and the potential role of platelets in this process are summarized. Finally, the clinical need for more effective antithrombotic agents is highlighted.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, School of Medicine, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
- Blood Research Center, School of Medicine, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
| | - Henri M H Spronk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
- Thrombosis Expertise Center, Heart+ Vascular Center, Maastricht University Medical Center+, P. Debeyelaan 25, Maastricht, the Netherlands
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- Thrombosis Expertise Center, Heart+ Vascular Center, Maastricht University Medical Center+, P. Debeyelaan 25, Maastricht, the Netherlands
| |
Collapse
|
78
|
Juźwik-Kopacz E, Kozak A, Rzepecka-Woźniak E, Woźniak K, Moskała A. Postmortem diagnosis of arterial atherosclerosis in autopsy, postmortem computed tomography and histopathological examinations in medico-legal aspect. ARCHIVES OF FORENSIC MEDICINE AND CRIMINOLOGY 2024; 74:50-63. [PMID: 39450595 DOI: 10.4467/16891716amsik.24.004.19649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 10/26/2024] Open
Abstract
Aims The aim of this study is to provide a retrospective assessment of the possibility of determining the severity of atherosclerosis based on postmortem computed tomography (PMCT), autopsy protocols and histopathological examination results. In the first stage of the study, 200 cases were evaluated of persons over 40 years of age in whom postmortem computed tomography and autopsy were performed. In the second stage, the cases were divided into a study group (cardiovascular deaths) and a control group, as well as divided by age and, in addition to autopsy protocols and PMCT results, histopathological findings were evaluated. Results The results of stage I demonstrated that the best detection of atherosclerosis was in the advanced stage with a predominance of detection in PMCT. Atherosclerosis detection in autopsy was highest in the coronary arteries, aorta and cerebral arteries; while in PMCT it remained equal in all evaluated locations. Autopsy showed higher detection of advanced atherosclerotic lesions in the coronary arteries and aorta compared to PMCT. The results of stage II of the study revealed that attaching the results of the general histopathological examination to the retrospective evaluation does not provide an opportunity to increase the accuracy of the evaluation of atherosclerotic lesions. The results obtained indicate the need for prospective studies. Conclusions Autopsy allows macroscopic evaluation of a very broad spectrum of atherosclerotic lesions, but often without precise determination of their nature, and with limited localization; PMCT allows accurate and reproducible evaluation of calcified atherosclerotic lesions in large and medium-sized vessels, but is unsuitable for the evaluation of non-calcified lesions and small vessels; targeted histopathological examinations allow very accurate, but local assessment of atherosclerotic lesions.
Collapse
Affiliation(s)
- Ewa Juźwik-Kopacz
- Department of Forensic Medicine, Jagiellonian University Medical College, Kraków, Poland
| | | | - Ewa Rzepecka-Woźniak
- Department of Forensic Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Krzysztof Woźniak
- Department of Forensic Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Artur Moskała
- Department of Forensic Medicine, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
79
|
Saba L, Cau R, Murgia A, Nicolaides AN, Wintermark M, Castillo M, Staub D, Kakkos SK, Yang Q, Paraskevas KI, Yuan C, Edjlali M, Sanfilippo R, Hendrikse J, Johansson E, Mossa-Basha M, Balu N, Dichgans M, Saloner D, Bos D, Jager HR, Naylor R, Faa G, Suri JS, Costello J, Auer DP, Mcnally JS, Bonati LH, Nardi V, van der Lugt A, Griffin M, Wasserman BA, Kooi ME, Gillard J, Lanzino G, Mikhailidis DP, Mandell DM, Benson JC, van Dam-Nolen DHK, Kopczak A, Song JW, Gupta A, DeMarco JK, Chaturvedi S, Virmani R, Hatsukami TS, Brown M, Moody AR, Libby P, Schindler A, Saam T. Carotid Plaque-RADS: A Novel Stroke Risk Classification System. JACC Cardiovasc Imaging 2024; 17:62-75. [PMID: 37823860 DOI: 10.1016/j.jcmg.2023.09.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Carotid artery atherosclerosis is highly prevalent in the general population and is a well-established risk factor for acute ischemic stroke. Although the morphological characteristics of vulnerable plaques are well recognized, there is a lack of consensus in reporting and interpreting carotid plaque features. OBJECTIVES The aim of this paper is to establish a consistent and comprehensive approach for imaging and reporting carotid plaque by introducing the Plaque-RADS (Reporting and Data System) score. METHODS A panel of experts recognized the necessity to develop a classification system for carotid plaque and its defining characteristics. Using a multimodality analysis approach, the Plaque-RADS categories were established through consensus, drawing on existing published reports. RESULTS The authors present a universal classification that is applicable to both researchers and clinicians. The Plaque-RADS score offers a morphological assessment in addition to the prevailing quantitative parameter of "stenosis." The Plaque-RADS score spans from grade 1 (indicating complete absence of plaque) to grade 4 (representing complicated plaque). Accompanying visual examples are included to facilitate a clear understanding of the Plaque-RADS categories. CONCLUSIONS Plaque-RADS is a standardized and reliable system of reporting carotid plaque composition and morphology via different imaging modalities, such as ultrasound, computed tomography, and magnetic resonance imaging. This scoring system has the potential to help in the precise identification of patients who may benefit from exclusive medical intervention and those who require alternative treatments, thereby enhancing patient care. A standardized lexicon and structured reporting promise to enhance communication between radiologists, referring clinicians, and scientists.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, University of Cagliari, Cagliari, Italy.
| | - Riccardo Cau
- Department of Radiology, University of Cagliari, Cagliari, Italy
| | | | - Andrew N Nicolaides
- Vascular Screening and Diagnostic Centre, Nicosia, Cyprus; University of Nicosia Medical School, Nicosia, Cyprus; Department of Vascular Surgery, Imperial College, London, United Kingdom
| | - Max Wintermark
- Department of Neuroradiology, The University of Texas MD Anderson Center, Houston, Texas, USA
| | - Mauricio Castillo
- Department of Radiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Daniel Staub
- Vascular Medicine/Angiology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Stavros K Kakkos
- Department of Vascular Surgery, University of Patras Medical School, Patras, Greece
| | - Qi Yang
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | | | - Chun Yuan
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Myriam Edjlali
- Multimodal Biomedical Imaging Laboratory (BioMaps), Paris-Saclay University, CEA, CNRS, Inserm, Frédéric Joliot Hospital Department, Orsay, France; Department of Radiology, APHP, Paris, France
| | | | | | - Elias Johansson
- Clinical Science, Umeå University, Neurosciences, Umeå, Sweden
| | - Mahmud Mossa-Basha
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Niranjan Balu
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - David Saloner
- Department of Radiology and Biomedical Imaging, University of California-San Francisco, San Francisco, California, USA
| | - Daniel Bos
- Department of Radiology and Nuclear Medicine, Erasmus MC Rotterdam, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands; Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Clinical Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
| | - H Rolf Jager
- Lysholm Department of Neuroradiology and the Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, United Kingdom
| | - Ross Naylor
- The Leicester Vascular Institute, Glenfield Hospital, Leicester, United Kingdom
| | - Gavino Faa
- Department of Pathology, University of Cagliari, Cagliari, Italy
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoin, Roseville, California, USA
| | - Justin Costello
- Department of Neuroradiology, Walter Reed National Military Medical Center and Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Dorothee P Auer
- Radiological Sciences, Division of Clinical Neuroscience, and NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
| | - J Scott Mcnally
- Department of Radiology, University of Utah, Salt Lake City, Utah, USA
| | - Leo H Bonati
- Department of Neurology and Stroke Center, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Valentina Nardi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus MC Rotterdam, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maura Griffin
- Vascular Screening and Diagnostic Centre, Nicosia, Cyprus
| | - Bruce A Wasserman
- Department of Radiology, University of Maryland School of Medicine and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - M Eline Kooi
- Department of Radiology and Nuclear Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | | | - Giuseppe Lanzino
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London School, University College London, London, United Kingdom
| | - Daniel M Mandell
- Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - John C Benson
- Department of Radiology Mayo Clinic, Rochester, Minnesota, USA
| | - Dianne H K van Dam-Nolen
- Department of Radiology and Nuclear Medicine, Erasmus MC Rotterdam, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Anna Kopczak
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Jae W Song
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ajay Gupta
- Department of Radiology Weill Cornell Medical College, New York, New York, USA
| | - J Kevin DeMarco
- Walter Reed National Military Medical Center and Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Seemant Chaturvedi
- Department of Neurology, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, Maryland, USA
| | | | - Martin Brown
- Department of Neurology and Neurosurgery, University College London Hospitals, London, United Kingdom
| | - Alan R Moody
- Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andreas Schindler
- Institute of Neuroradiology, University Hospital, LMU Munich, Munich, Germany
| | - Tobias Saam
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany; Die Radiologie, Rosenheim, Germany
| |
Collapse
|
80
|
Saba L, Scicolone R, Johansson E, Nardi V, Lanzino G, Kakkos SK, Pontone G, Annoni AD, Paraskevas KI, Fox AJ. Quantifying Carotid Stenosis: History, Current Applications, Limitations, and Potential: How Imaging Is Changing the Scenario. Life (Basel) 2024; 14:73. [PMID: 38255688 PMCID: PMC10821425 DOI: 10.3390/life14010073] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
Carotid artery stenosis is a major cause of morbidity and mortality. The journey to understanding carotid disease has developed over time and radiology has a pivotal role in diagnosis, risk stratification and therapeutic management. This paper reviews the history of diagnostic imaging in carotid disease, its evolution towards its current applications in the clinical and research fields, and the potential of new technologies to aid clinicians in identifying the disease and tailoring medical and surgical treatment.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, University of Cagliari, 09042 Cagliari, Italy;
| | - Roberta Scicolone
- Department of Radiology, University of Cagliari, 09042 Cagliari, Italy;
| | - Elias Johansson
- Neuroscience and Physiology, Sahlgrenska Academy, 41390 Gothenburg, Sweden;
| | - Valentina Nardi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Giuseppe Lanzino
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA;
| | - Stavros K. Kakkos
- Department of Vascular Surgery, University of Patras, 26504 Patras, Greece;
| | - Gianluca Pontone
- Centro Cardiologico Monzino IRCCS, Via C. Parea 4, 20138 Milan, Italy; (G.P.); (A.D.A.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
| | - Andrea D. Annoni
- Centro Cardiologico Monzino IRCCS, Via C. Parea 4, 20138 Milan, Italy; (G.P.); (A.D.A.)
| | | | - Allan J. Fox
- Department of Medical Imaging, Neuroradiology Section, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada;
| |
Collapse
|
81
|
Al-Sharydah AM, AlZahrani KS, Alghanimi IA, AlAnazi MM, AlHarbi RE. Anatomical Distribution Patterns of Peripheral Arterial Disease in the Upper Extremities According to Patient Characteristics: A Retrospective Cohort Study. Vasc Health Risk Manag 2023; 19:871-883. [PMID: 38173811 PMCID: PMC10762427 DOI: 10.2147/vhrm.s440408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
Purpose Peripheral arterial disease (PAD) greatly affects the patients' quality of life. We aimed to investigate the affected anatomical sites and distribution patterns in upper extremity PAD using computed tomography angiography (CTA). Furthermore, we sought to identify the correlations between patient characteristics and the identified patterns. Patients and Methods This was a retrospective chart review of upper limb CTA findings from patients with symptomatic PAD aged >18 years. Significant variables from univariate logistic regression analysis were further tested using multivariate logistic regression analysis. Statistical significance was set at p < 0.05, with confidence intervals of 95%. Results The mean age of the 102 included patients with upper extremity PAD was 55.45 years. Laterality analysis revealed that the upper left limb segments were more affected than the upper right limb segments (42 vs 63; left-to-right ratio, 3:2). The forearm was the segment most affected by stenotic PAD (62 segments, 3.37%). The arm was the segment most affected by occlusive PAD (14 segments, 0.76%). Diabetes mellitus (DM) and hypertension (HTN) were significant predictors of PAD (p = 0.046). In patients with DM, the occlusive form of PAD was dominant in the arm (18.18%); however, the stenotic form prevailed in the forearm (72.72%). In patients with HTN, the occlusive form of PAD was predominant in the arm (45.45%); however, the stenotic form tended to occur in the arm and forearm (90.90%). Conclusion The distribution patterns of upper extremity PAD are linked to its underlying pathophysiology. HTN and DM are the most frequent comorbidities in patients with upper extremity PAD. Angiographically, PAD in these patients is likely to present as stenosis rather than as occlusion. This is vital for interventionists who deviate from radial arterial access in patients with PAD. Therefore, targeted screening standards are required, and further studies on PAD are warranted.
Collapse
Affiliation(s)
- Abdulaziz Mohammad Al-Sharydah
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al Khobar City, Eastern Province, Saudi Arabia
| | - Khaled Saud AlZahrani
- Radiology Department, King Fahad General Hospital-Jeddah, Ministry of Health, Al Andalus, Jeddah, 23325, Saudi Arabia
| | - Ibrahim Abobaker Alghanimi
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al Khobar City, Eastern Province, Saudi Arabia
| | - Maha Mukhlef AlAnazi
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al Khobar City, Eastern Province, Saudi Arabia
| | - Razan Essam AlHarbi
- Diagnostic and Interventional Radiology Department, King Fahd Hospital of the University, Imam Abdulrahman Bin Faisal University, Al Khobar City, Eastern Province, Saudi Arabia
| |
Collapse
|
82
|
Cui HK, Tang CJ, Gao Y, Li ZA, Zhang J, Li YD. An integrative analysis of single-cell and bulk transcriptome and bidirectional mendelian randomization analysis identified C1Q as a novel stimulated risk gene for Atherosclerosis. Front Immunol 2023; 14:1289223. [PMID: 38179058 PMCID: PMC10764496 DOI: 10.3389/fimmu.2023.1289223] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/05/2023] [Indexed: 01/06/2024] Open
Abstract
Background The role of complement component 1q (C1Q) related genes on human atherosclerotic plaques (HAP) is less known. Our aim is to establish C1Q associated hub genes using single-cell RNA sequencing (scRNA-seq) and bulk RNA analysis to diagnose and predict HAP patients more effectively and investigate the association between C1Q and HAP (ischemic stroke) using bidirectional Mendelian randomization (MR) analysis. Methods HAP scRNA-seq and bulk-RNA data were download from the Gene Expression Omnibus (GEO) database. The C1Q-related hub genes was screened using the GBM, LASSO and XGBoost algorithms. We built machine learning models to diagnose and distinguish between types of atherosclerosis using generalized linear models and receiver operating characteristics (ROC) analyses. Further, we scored the HALLMARK_COMPLEMENT signaling pathway using ssGSEA and confirmed hub gene expression through qRT-PCR in RAW264.7 macrophages and apoE-/- mice. Furthermore, the risk association between C1Q and HAP was assessed through bidirectional MR analysis, with C1Q as exposure and ischemic stroke (IS, large artery atherosclerosis) as outcomes. Inverse variance weighting (IVW) was used as the main method. Results We utilized scRNA-seq dataset (GSE159677) to identify 24 cell clusters and 12 cell types, and revealed seven C1Q associated DEGs in both the scRNA-seq and GEO datasets. We then used GBM, LASSO and XGBoost to select C1QA and C1QC from the seven DEGs. Our findings indicated that both training and validation cohorts had satisfactory diagnostic accuracy for identifying patients with HPAs. Additionally, we confirmed SPI1 as a potential TF responsible for regulating the two hub genes in HAP. Our analysis further revealed that the HALLMARK_COMPLEMENT signaling pathway was correlated and activated with C1QA and C1QC. We confirmed high expression levels of C1QA, C1QC and SPI1 in ox-LDL-treated RAW264.7 macrophages and apoE-/- mice using qPCR. The results of MR indicated that there was a positive association between the genetic risk of C1Q and IS, as evidenced by an odds ratio (OR) of 1.118 (95%CI: 1.013-1.234, P = 0.027). Conclusion The authors have effectively developed and validated a novel diagnostic signature comprising two genes for HAP, while MR analysis has provided evidence supporting a favorable association of C1Q on IS.
Collapse
Affiliation(s)
- Hong-Kai Cui
- Department of Neurological Intervention, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chao-Jie Tang
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Gao
- Department of Neurological Intervention, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zi-Ang Li
- Department of Neurological Intervention, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jian Zhang
- Department of Neurological Intervention, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yong-Dong Li
- Department of Neurological Intervention, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
83
|
Jiang M, Song Y, Ren MX, He RC, Dong XH, Li XH, Lu ZF, Li S, Wu J, Bei YR, Liu F, Long Y, Wu SG, Liu XH, Wu LM, Yang HL, McVey DG, Dai XY, Ye S, Hu YW. LncRNA NIPA1-SO confers atherosclerotic protection by suppressing the transmembrane protein NIPA1. J Adv Res 2023; 54:29-42. [PMID: 36736696 DOI: 10.1016/j.jare.2023.01.017] [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: 04/06/2022] [Revised: 10/10/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are emerging as important players in gene regulation and cardiovascular diseases. However, the roles of lncRNAs in atherosclerosis are poorly understood. In the present study, we found that the levels of NIPA1-SO were decreased while those of NIPA1 were increased in human atherosclerotic plaques. Furthermore, NIPA1-SO negatively regulated NIPA1 expression in human umbilical vein endothelial cells (HUVECs). Mechanistically, NIPA1-SO interacted with the transcription factor FUBP1 and the NIPA1 gene. The effect of NIPA1-SO on NIPA1 protein levels was reversed by the knockdown of FUBP1. NIPA1-SO overexpression increased, whilst NIPA1-SO knockdown decreased BMPR2 levels; these effects were enhanced by the knockdown of NIPA1. The overexpression of NIPA1-SO reduced while NIPA1-SO knockdown increased monocyte adhesion to HUVECs; these effects were diminished by the knockdown of BMPR2. The lentivirus-mediated-overexpression of NIPA1-SO or gene-targeted knockout of NIPA1 in low-density lipoprotein receptor-deficient mice reduced monocyte-endothelium adhesion and atherosclerotic lesion formation. Collectively, these findings revealed a novel anti-atherosclerotic role for the lncRNA NIPA1-SO and highlighted its inhibitory effects on vascular inflammation and intracellular cholesterol accumulation by binding to FUBP1 and consequently repressing NIPA1 expression.
Collapse
Affiliation(s)
- Min Jiang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - Yu Song
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - Mei-Xia Ren
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350001, China; Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian Key Laboratory of Geriatrics, Fujian Provincial Center for Geriatrics, Fuzhou 350013, China
| | - Run-Chao He
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - Xian-Hui Dong
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - Xue-Heng Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhi-Feng Lu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shu Li
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - Jia Wu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - Yan-Rou Bei
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fei Liu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - Yan Long
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - Shao-Guo Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China
| | - Xue-Hui Liu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China
| | - Li-Mei Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China
| | - Hong-Ling Yang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China
| | - David G McVey
- Department of Cardiovascular Sciences & NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester LE3 9QP, UK
| | - Xiao-Yan Dai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| | - Shu Ye
- Cardiovascular Translational Research Programme, National University of Singapore, Singapore; Shantou University Medical College, Shantou, China.
| | - Yan-Wei Hu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou 510620, China; Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
84
|
Tscheuschner L, Tzafriri AR. Cardiovascular Tissue Engineering Models for Atherosclerosis Treatment Development. Bioengineering (Basel) 2023; 10:1373. [PMID: 38135964 PMCID: PMC10740643 DOI: 10.3390/bioengineering10121373] [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: 11/09/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
In the early years of tissue engineering, scientists focused on the generation of healthy-like tissues and organs to replace diseased tissue areas with the aim of filling the gap between organ demands and actual organ donations. Over time, the realization has set in that there is an additional large unmet need for suitable disease models to study their progression and to test and refine different treatment approaches. Increasingly, researchers have turned to tissue engineering to address this need for controllable translational disease models. We review existing and potential uses of tissue-engineered disease models in cardiovascular research and suggest guidelines for generating adequate disease models, aimed both at studying disease progression mechanisms and supporting the development of dedicated drug-delivery therapies. This involves the discussion of different requirements for disease models to test drugs, nanoparticles, and drug-eluting devices. In addition to realistic cellular composition, the different mechanical and structural properties that are needed to simulate pathological reality are addressed.
Collapse
Affiliation(s)
- Linnea Tscheuschner
- Department of Vascular Surgery, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Abraham R. Tzafriri
- Department of Research and Innovation, CBSET Inc., Lexington, MA 02421, USA;
| |
Collapse
|
85
|
Mosquera JV, Auguste G, Wong D, Turner AW, Hodonsky CJ, Alvarez-Yela AC, Song Y, Cheng Q, Lino Cardenas CL, Theofilatos K, Bos M, Kavousi M, Peyser PA, Mayr M, Kovacic JC, Björkegren JLM, Malhotra R, Stukenberg PT, Finn AV, van der Laan SW, Zang C, Sheffield NC, Miller CL. Integrative single-cell meta-analysis reveals disease-relevant vascular cell states and markers in human atherosclerosis. Cell Rep 2023; 42:113380. [PMID: 37950869 DOI: 10.1016/j.celrep.2023.113380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/12/2023] [Accepted: 10/20/2023] [Indexed: 11/13/2023] Open
Abstract
Coronary artery disease (CAD) is characterized by atherosclerotic plaque formation in the arterial wall. CAD progression involves complex interactions and phenotypic plasticity among vascular and immune cell lineages. Single-cell RNA-seq (scRNA-seq) studies have highlighted lineage-specific transcriptomic signatures, but human cell phenotypes remain controversial. Here, we perform an integrated meta-analysis of 22 scRNA-seq libraries to generate a comprehensive map of human atherosclerosis with 118,578 cells. Besides characterizing granular cell-type diversity and communication, we leverage this atlas to provide insights into smooth muscle cell (SMC) modulation. We integrate genome-wide association study data and uncover a critical role for modulated SMC phenotypes in CAD, myocardial infarction, and coronary calcification. Finally, we identify fibromyocyte/fibrochondrogenic SMC markers (LTBP1 and CRTAC1) as proxies of atherosclerosis progression and validate these through omics and spatial imaging analyses. Altogether, we create a unified atlas of human atherosclerosis informing cell state-specific mechanistic and translational studies of cardiovascular diseases.
Collapse
Affiliation(s)
- Jose Verdezoto Mosquera
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA; Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - Gaëlle Auguste
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - Doris Wong
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA; Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - Adam W Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - Chani J Hodonsky
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Yipei Song
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA; Department of Computer Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Qi Cheng
- CVPath Institute, Gaithersburg, MD 20878, USA
| | - Christian L Lino Cardenas
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | | - Maxime Bos
- Department of Epidemiology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Patricia A Peyser
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48019, USA
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London WC2R 2LS, UK; National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Rajeev Malhotra
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - P Todd Stukenberg
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | | | - Sander W van der Laan
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, the Netherlands
| | - Chongzhi Zang
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA; Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Public Health Sciences, University of Virginia, Charlottesville, VA 22908, USA
| | - Nathan C Sheffield
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA; Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Public Health Sciences, University of Virginia, Charlottesville, VA 22908, USA
| | - Clint L Miller
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA; Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Public Health Sciences, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
86
|
Curcio A, Panarello A, Spaccarotella C, Indolfi C. Cardiovascular Prognosis in Patients with Peripheral Artery Disease and Approach to Therapy. Biomedicines 2023; 11:3131. [PMID: 38137352 PMCID: PMC10740501 DOI: 10.3390/biomedicines11123131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Peripheral artery disease (PAD), the pathophysiologic narrowing of the arterial blood vessels of the lower limbs due to atherosclerosis, is estimated to affect more than 200 million people worldwide and its prevalence ranges from 0.9 to 31.3% in people aged ≥50 years. It is an established marker of systemic obstructive atherosclerosis, which depicts patients at higher risk of myocardial infarction and stroke, due to the involvement of coronary and cerebral arteries in the atherosclerotic process. Therefore, identifying PAD, particularly in patients with coronary artery disease, is important to assess the cardiovascular risk score and implement specific therapies and prevention strategies. Since PAD emerged as an important clinical cardiovascular predictor, even more than other typical cardiovascular risk factors, an aggressive strategy to identify and treat PAD patients should be pursued by general practitioners, cardiologists, and vascular surgeons; similarly, preventive strategies should be implemented to improve prognosis and outcomes, particularly in patients suffering from both coronary artery disease and PAD. In this review, we describe the pathophysiology, including limb vasoconstriction after coronary angioplasty, the diagnosis of PAD, prognosis according to cardiovascular events, coronary artery disease, and heart failure. Furthermore, a large section of this review is on management, which spans from risk factors' modification to antithrombotic therapy, and revascularization is provided. Finally, considerations about newer therapeutic options for the "desert foot" are discussed, including gene therapy.
Collapse
Affiliation(s)
- Antonio Curcio
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (A.P.); (C.I.)
| | - Alessandra Panarello
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (A.P.); (C.I.)
| | - Carmen Spaccarotella
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy;
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (A.P.); (C.I.)
| |
Collapse
|
87
|
Patrakka O, Tuomisto S, Pienimäki J, Ollikainen J, Oksala N, Lampinen V, Ojanen MJT, Huhtala H, Hytönen VP, Lehtimäki T, Martiskainen M, Karhunen PJ. Thrombus Aspirates From Patients With Acute Ischemic Stroke Are Infiltrated by Viridans Streptococci. J Am Heart Assoc 2023; 12:e030639. [PMID: 37982253 PMCID: PMC10727284 DOI: 10.1161/jaha.123.030639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/27/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Acute ischemic stroke may be due to embolism from ruptured atherosclerotic carotid arteries. DNA of oral bacteria, mainly the viridans streptococci group, has been detected in thrombus aspirates of patients with ischemic stroke as well as in carotid endarterectomy samples. Because viridans streptococci are known to possess thrombogenic properties, we studied whether their presence in thrombus aspirates and in carotid artery specimens can be confirmed using bacterial immunohistochemistry. METHODS AND RESULTS Thrombus aspirates from 61 patients with ischemic stroke (70.5% men; mean age, 66.8 years) treated with mechanical thrombectomy, as well as carotid endarterectomy samples from 20 symptomatic patients (65.0% men; mean age, 66.2 years) and 48 carotid artery samples from nonstroke autopsy cases (62.5% men; mean age, 66.4 years), were immunostained with an antibody cocktail against 3 species (Streptococcus sanguinis, Streptococcus mitis, and Streptococcus gordonii) of viridans streptococci. Of the thrombus aspirates, 84.8% were immunopositive for viridans streptococci group bacteria, as were 80.0% of the carotid endarterectomy samples, whereas immunopositivity was observed in 31.3% of the carotid artery samples from nonstroke autopsies. Most streptococci were detected inside neutrophil granulocytes, but there were also remnants of bacterial biofilm as well as free bacterial infiltrates in some samples. CONCLUSIONS Oral streptococci were found in aspirated thrombi of patients with acute ischemic stroke as well as in carotid artery samples. Our results suggest that viridans streptococci group bacteria may play a role in the pathophysiology of ischemic stroke.
Collapse
Affiliation(s)
- Olli Patrakka
- Department of Forensic Medicine, Faculty of Medicine and Health TechnologyTampere University and Fimlab LaboratoriesTampereFinland
| | - Sari Tuomisto
- Department of Forensic Medicine, Faculty of Medicine and Health TechnologyTampere University and Fimlab LaboratoriesTampereFinland
| | | | - Jyrki Ollikainen
- Department of NeurologyTampere University HospitalTampereFinland
| | - Niku Oksala
- Vascular CentreTampere University HospitalTampereFinland
- Surgery, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Vili Lampinen
- Department of Forensic Medicine, Faculty of Medicine and Health TechnologyTampere University and Fimlab LaboratoriesTampereFinland
| | - Markus J. T. Ojanen
- Department of Forensic Medicine, Faculty of Medicine and Health TechnologyTampere University and Fimlab LaboratoriesTampereFinland
- Laboratory of Protein Dynamics, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Heini Huhtala
- Faculty of Social SciencesTampere UniversityTampereFinland
| | - Vesa P. Hytönen
- Department of Forensic Medicine, Faculty of Medicine and Health TechnologyTampere University and Fimlab LaboratoriesTampereFinland
- Laboratory of Protein Dynamics, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health TechnologyTampere University, Fimlab Laboratories and Finnish Cardiovascular Research Center TampereTampereFinland
| | - Mika Martiskainen
- Department of Forensic Medicine, Faculty of Medicine and Health TechnologyTampere University and Fimlab LaboratoriesTampereFinland
- National Institute for Health and WelfareHelsinkiFinland
| | - Pekka J. Karhunen
- Department of Forensic Medicine, Faculty of Medicine and Health TechnologyTampere University and Fimlab LaboratoriesTampereFinland
| |
Collapse
|
88
|
Trumble BC, Negrey J, Koebele SV, Thompson RC, Samuel Wann L, Allam AH, Beheim B, Linda Sutherland M, Sutherland JD, Eid Rodriguez D, Michalik DE, Rowan CJ, Lombardi GP, Garcia AR, Cummings DK, Seabright E, Alami S, Kraft TS, Hooper P, Buetow K, Irimia A, Gatz M, Stieglitz J, Gurven MD, Kaplan H, Thomas GS. Testosterone is positively associated with coronary artery calcium in a low cardiovascular disease risk population. Evol Med Public Health 2023; 11:472-484. [PMID: 38145005 PMCID: PMC10746324 DOI: 10.1093/emph/eoad039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 10/11/2023] [Indexed: 12/26/2023] Open
Abstract
Background In industrialized populations, low male testosterone is associated with higher rates of cardiovascular mortality. However, coronary risk factors like obesity impact both testosterone and cardiovascular outcomes. Here, we assess the role of endogenous testosterone on coronary artery calcium in an active subsistence population with relatively low testosterone levels, low cardiovascular risk and low coronary artery calcium scores. Methodology In this cross-sectional community-based study, 719 Tsimane forager-horticulturalists in the Bolivian Amazon aged 40+ years underwent computed tomography (49.8% male, mean age 57.6 years). Results Coronary artery calcium levels were low; 84.5% had no coronary artery calcium. Zero-inflated negative binomial models found testosterone was positively associated with coronary artery calcium for the full sample (Incidence Rate Ratio [IRR] = 1.477, 95% Confidence Interval [CI] 1.001-2.170, P = 0.031), and in a male-only subset (IRR = 1.532, 95% CI 0.993-2.360, P = 0.053). Testosterone was also positively associated with clinically relevant coronary atherosclerosis (calcium >100 Agatston units) in the full sample (Odds Ratio [OR] = 1.984, 95% CI 1.202-3.275, P = 0.007) and when limited to male-only sample (OR = 2.032, 95% CI 1.118-4.816, P = 0.024). Individuals with coronary artery calcium >100 had 20% higher levels of testosterone than those with calcium <100 (t = -3.201, P = 0.007). Conclusions and Implications Among Tsimane, testosterone is positively associated with coronary artery calcium despite generally low normal testosterone levels, minimal atherosclerosis and rare cardiovascular disease (CVD) events. Associations between low testosterone and CVD events in industrialized populations are likely confounded by obesity and other lifestyle factors.
Collapse
Affiliation(s)
- Benjamin C Trumble
- Arizona State University, School of Human Evolution and Social Change, Center for Evolution and Medicine, Institute of Human Origins, Tempe, AZ, USA
| | - Jacob Negrey
- Arizona State University, School of Human Evolution and Social Change, Center for Evolution and Medicine, Institute of Human Origins, Tempe, AZ, USA
| | - Stephanie V Koebele
- Arizona State University, School of Human Evolution and Social Change, Center for Evolution and Medicine, Institute of Human Origins, Tempe, AZ, USA
| | - Randall C Thompson
- Saint Luke’s Mid America Heart Institute, Department of Cardiology, Kansas City, MO, USA
| | - L Samuel Wann
- University of New Mexico, School of Medicine, Albuquerque, NM, USA
| | - Adel H Allam
- Al Azhar University, School of Medicine, Cairo, Egypt
| | - Bret Beheim
- Max Planck Institute for Evolutionary Anthropology, Department of Human Behavior, Ecology and Culture, Leipzig, Germany
| | | | | | | | - David E Michalik
- University of California Irvine, School of Medicine, Irvine, CA, USA
- Miller Women’s and Children’s Hospital Long Beach, CA, USA
| | | | - Guido P Lombardi
- Universidad Peruana Cayetano Heredia, Laboratorio de Paleopatología, Lima, Peru
| | - Angela R Garcia
- Arizona State University, School of Human Evolution and Social Change, Center for Evolution and Medicine, Institute of Human Origins, Tempe, AZ, USA
| | | | - Edmond Seabright
- Mohammed VI Polytechnic University, School of Collective Intelligence, Ben Guerir, Morocco
| | - Sarah Alami
- Mohammed VI Polytechnic University, School of Collective Intelligence, Ben Guerir, Morocco
| | - Thomas S Kraft
- University of Utah, Anthropology Department, Salt Lake City, UT, USA
| | - Paul Hooper
- Chapman University, Economic Science Institute, Orange, CA, USA
| | - Kenneth Buetow
- Arizona State University, School of Human Evolution and Social Change, Center for Evolution and Medicine, Institute of Human Origins, Tempe, AZ, USA
| | - Andrei Irimia
- University of Southern California, Psychology Department, Los Angeles, CA, USA
| | - Margaret Gatz
- University of Southern California, Psychology Department, Los Angeles, CA, USA
| | - Jonathan Stieglitz
- Toulouse Scool of Economics, Institute for Advanced Study Toulouse, Toulouse, France
| | - Michael D Gurven
- University of California Santa Barbara, Department of Anthropology, Santa Barbara, CA, USA
| | - Hillard Kaplan
- Chapman University, Economic Science Institute, Orange, CA, USA
| | - Gregory S Thomas
- MemorialCare Health System, Fountain Valley, CA, USA
- University of California Irvine, Division of Cardiology, Orange, CA, USA
| | | |
Collapse
|
89
|
Yan A, Gotlieb AI. The microenvironment of the atheroma expresses phenotypes of plaque instability. Cardiovasc Pathol 2023; 67:107572. [PMID: 37595697 DOI: 10.1016/j.carpath.2023.107572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Data from histopathology studies of human atherosclerotic tissue specimens and from vascular imaging studies support the concept that the local arterial microenvironment of a stable atheroma promotes destabilizing conditions that result in the transition to an unstable atheroma. Destabilization is characterized by several different plaque phenotypes that cause major clinical events such as acute coronary syndrome and cerebrovascular strokes. There are several rupture-associated phenotypes causing thrombotic vascular occlusion including simple fibrous cap rupture of an atheroma, fibrous cap rupture at site of previous rupture-and-repair of an atheroma, and nodular calcification with rupture. Endothelial erosion without rupture has more recently been shown to be a common phenotype to promote thrombosis as well. Microenvironment features that are linked to these phenotypes of plaque instability are neovascularization arising from the vasa vasorum network leading to necrotic core expansion, intraplaque hemorrhage, and cap rupture; activation of adventitial and perivascular adipose tissue cells leading to secretion of cytokines, growth factors, adipokines in the outer artery wall that destabilize plaque structure; and vascular smooth muscle cell phenotypic switching through transdifferentiation and stem/progenitor cell activation resulting in the promotion of inflammation, calcification, and secretion of extracellular matrix, altering fibrous cap structure, and necrotic core growth. As the technology evolves, studies using noninvasive vascular imaging will be able to investigate the transition of stable to unstable atheromas in real time. A limitation in the field, however, is that reliable and predictable experimental models of spontaneous plaque rupture and/or erosion are not currently available to study the cell and molecular mechanisms that regulate the conversion of the stable atheroma to an unstable plaque.
Collapse
Affiliation(s)
- Angela Yan
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
90
|
Ellenbogen Y, Hendriks EJ, Karadimas S, O'Reilly S, Itsekzon Hayosh Z, Alshahrani R, Agid R, Schaafsma J, Krings T, Nicholson P. Use of the neuroform atlas for stenting of intracranial atherosclerotic disease: Clinical and angiographic outcomes. Interv Neuroradiol 2023:15910199231195134. [PMID: 37817560 DOI: 10.1177/15910199231195134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Intracranial atherosclerotic disease (ICAD) is a potential cause of ischemic stroke. Treatment of ICAD can include intracranial stenting. There are specifically designed stents for this use-case; however, less is known about the off-label use of the Neuroform Atlas stent. In this study, we describe the outcomes of the Neuroform Atlas stent for treatment of ICAD. METHODS Adult patients with symptomatic ICAD failing best medical treatment undergoing elective intracranial stenting using the Neuroform Atlas stent between November 2018 and March 2021 were included. Patient demographics, procedure-related details and clinical and imaging outcomes were analyzed. RESULTS Eighteen patients met the inclusion criteria, with a mean follow-up duration of 9.6 ± 6.8 (standard deviation) months. There were two procedure-related mortalities (one massive intracranial hemorrhage and one groin site complication with sepsis). Fifteen patients were alive at the 6-month follow-up, all with satisfactory stent patency on follow-up imaging without any new ischemic events. Modified Rankin Scale at latest follow-up was 1.9 (interquartile range 5). CONCLUSION In this single-center consecutive series, intracranial stenting with the Neuroform Atlas stent was a safe and effective treatment for symptomatic ICAD patients failing best medical management.
Collapse
Affiliation(s)
- Yosef Ellenbogen
- Division of Neurosurgery, Department of Surgery, University Health Network, Toronto, Ontario, Canada
| | - Eef J Hendriks
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Spyros Karadimas
- Division of Neurosurgery, Department of Surgery, University Health Network, Toronto, Ontario, Canada
| | - Sean O'Reilly
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Zeev Itsekzon Hayosh
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Rabab Alshahrani
- Division of Neurosurgery, Department of Surgery, University Health Network, Toronto, Ontario, Canada
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Ronit Agid
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Joanna Schaafsma
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Timo Krings
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Patrick Nicholson
- Division of Neuroradiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
91
|
Li W, Lin A, Hutton M, Dhaliwal H, Nadel J, Rodor J, Tumanov S, Örd T, Hadden M, Mokry M, Mol BM, Pasterkamp G, Padula MP, Geczy CL, Ramaswamy Y, Sluimer JC, Kaikkonen MU, Stocker R, Baker AH, Fisher EA, Patel S, Misra A. Colchicine promotes atherosclerotic plaque stability independently of inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560632. [PMID: 37873248 PMCID: PMC10592948 DOI: 10.1101/2023.10.03.560632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease which is driven in part by the aberrant trans -differentiation of vascular smooth muscle cells (SMCs). No therapeutic drug has been shown to reverse detrimental SMC-derived cell phenotypes into protective phenotypes, a hypothesized enabler of plaque regression and improved patient outcome. Herein, we describe a novel function of colchicine in the beneficial modulation of SMC-derived cell phenotype, independent of its conventional anti-inflammatory effects. Using SMC fate mapping in an advanced atherosclerotic lesion model, colchicine induced plaque regression by converting pathogenic SMC-derived macrophage-like and osteoblast-like cells into protective myofibroblast-like cells which thickened, and thereby stabilized, the fibrous cap. This was dependent on Notch3 signaling in SMC-derived plaque cells. These findings may help explain the success of colchicine in clinical trials relative to other anti-inflammatory drugs. Thus, we demonstrate the potential of regulating SMC phenotype in advanced plaque regression through Notch3 signaling, in addition to the canonical anti-inflammatory actions of drugs to treat atherosclerosis.
Collapse
|
92
|
Chen D, Zhao Z, Liu P, Liu X, Wang X, Ren Q, Chang B. Adventitial Vasa Vasorum Neovascularization in Femoral Artery of Type 2 Diabetic Patients with Macroangiopathy Is Associated with Macrophages and Lymphocytes as well as the Occurrence of Cardiovascular Events. Thromb Haemost 2023; 123:989-998. [PMID: 37037199 DOI: 10.1055/s-0043-1768162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
OBJECTIVES This study was conducted to assess the relationship between adventitial vasa vasorum neovascularization (VVn) in femoral artery of type 2 diabetic patients with macroangiopathy and the recruitment of macrophages and lymphocytes, and to relate the density of VVn to the occurrence of cardiovascular events. MATERIALS Femoral artery samples were obtained from amputation cases. A total of 55 type 2 diabetic patients with macroangiopathy, 15 autopsy cases with type 2 diabetes without atherosclerosis. METHODS Hematoxylin and eosin (H&E) staining to observe the histopathological features; Victoria blue staining to analyze the histological features; immunohistochemistry (CD34, CD68, CD20, and CD3) to determine the VVn density and the expression of macrophages, B lymphocytes, and T lymphocytes. RESULTS Type 2 diabetic patients with macroangiopathy showed a higher mean adventitial VVn density in femoral artery (48.40 ± 9.39 no./mm2) than patients with type 2 diabetes without atherosclerosis (19.75 ± 6.28 no./mm2) (p < 0.01). In addition, the VVn density was positively associated with the expression of CD68 macrophages (r = 0.62, p < 0.01) and CD20 B lymphocytes (r = 0.59, p < 0.01). Type 2 diabetic patients with high VVn density showed more adverse cardiovascular events (27/35 vs. 8/20 events, p = 0.006). In multivariable analysis adjusted for main risk factors for cardiovascular disease, VVn was still independently associated with adverse cardiovascular events (p = 0.01). CONCLUSION VVn density in type 2 diabetic patients with macroangiopathy is positively correlated with the adventitial immune-inflammatory cell numbers and the development of atherosclerotic lesions. Furthermore, VVn density is associated with adverse cardiovascular events.
Collapse
Affiliation(s)
- Dong Chen
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Zixi Zhao
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Peng Liu
- Department of Surgery, Binhai New Area Hospital of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Xinbang Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Xin Wang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Qiuyue Ren
- Department of Endocrinology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People's Republic of China
| | - Bai Chang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
93
|
Tornifoglio B, Johnston RD, Stone AJ, Kerskens C, Lally C. Microstructural and mechanical insight into atherosclerotic plaques: an ex vivo DTI study to better assess plaque vulnerability. Biomech Model Mechanobiol 2023; 22:1515-1530. [PMID: 36652053 PMCID: PMC10511397 DOI: 10.1007/s10237-022-01671-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/08/2022] [Indexed: 01/19/2023]
Abstract
Non-invasive microstructural characterisation has the potential to determine the stability, or lack thereof, of atherosclerotic plaques and ultimately aid in better assessing plaques' risk to rupture. If linked with mechanical characterisation using a clinically relevant imaging technique, mechanically sensitive rupture risk indicators could be possible. This study aims to provide this link-between a clinically relevant imaging technique and mechanical characterisation within human atherosclerotic plaques. Ex vivo diffusion tensor imaging, mechanical testing, and histological analysis were carried out on human carotid atherosclerotic plaques. DTI-derived tractography was found to yield significant mechanical insight into the mechanical properties of more stable and more vulnerable microstructures. Coupled with insights from digital image correlation and histology, specific failure characteristics of different microstructural arrangements furthered this finding. More circumferentially uniform microstructures failed at higher stresses and strains when compared to samples which had multiple microstructures, like those seen in a plaque cap. The novel findings in this study motivate diagnostic measures which use non-invasive characterisation of the underlying microstructure of plaques to determine their vulnerability to rupture.
Collapse
Affiliation(s)
- B Tornifoglio
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - R D Johnston
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - A J Stone
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Medical Physics and Clinical Engineering, St. Vincent's University Hospital, Dublin, Ireland
| | - C Kerskens
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - C Lally
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland.
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
94
|
Sharifi MA, Wierer M, Dang TA, Milic J, Moggio A, Sachs N, von Scheidt M, Hinterdobler J, Müller P, Werner J, Stiller B, Aherrahrou Z, Erdmann J, Zaliani A, Graettinger M, Reinshagen J, Gul S, Gribbon P, Maegdefessel L, Bernhagen J, Sager HB, Mann M, Schunkert H, Kessler T. ADAMTS-7 Modulates Atherosclerotic Plaque Formation by Degradation of TIMP-1. Circ Res 2023; 133:674-686. [PMID: 37675562 PMCID: PMC7615141 DOI: 10.1161/circresaha.123.322737] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The ADAMTS7 locus was genome-wide significantly associated with coronary artery disease. Lack of the ECM (extracellular matrix) protease ADAMTS-7 (A disintegrin and metalloproteinase-7) was shown to reduce atherosclerotic plaque formation. Here, we sought to identify molecular mechanisms and downstream targets of ADAMTS-7 mediating the risk of atherosclerosis. METHODS Targets of ADAMTS-7 were identified by high-resolution mass spectrometry of atherosclerotic plaques from Apoe-/- and Apoe-/-Adamts7-/- mice. ECM proteins were identified using solubility profiling. Putative targets were validated using immunofluorescence, in vitro degradation assays, coimmunoprecipitation, and Förster resonance energy transfer-based protein-protein interaction assays. ADAMTS7 expression was measured in fibrous caps of human carotid artery plaques. RESULTS In humans, ADAMTS7 expression was higher in caps of unstable as compared to stable carotid plaques. Compared to Apoe-/- mice, atherosclerotic aortas of Apoe-/- mice lacking Adamts-7 (Apoe-/-Adamts7-/-) contained higher protein levels of Timp-1 (tissue inhibitor of metalloprotease-1). In coimmunoprecipitation experiments, the catalytic domain of ADAMTS-7 bound to TIMP-1, which was degraded in the presence of ADAMTS-7 in vitro. ADAMTS-7 reduced the inhibitory capacity of TIMP-1 at its canonical target MMP-9 (matrix metalloprotease-9). As a downstream mechanism, we investigated collagen content in plaques of Apoe-/- and Apoe-/-Adamts7-/- mice after a Western diet. Picrosirius red staining of the aortic root revealed less collagen as a readout of higher MMP-9 activity in Apoe-/- as compared to Apoe-/- Adamts7-/- mice. To facilitate high-throughput screening for ADAMTS-7 inhibitors with the aim of decreasing TIMP-1 degradation, we designed a Förster resonance energy transfer-based assay targeting the ADAMTS-7 catalytic site. CONCLUSIONS ADAMTS-7, which is induced in unstable atherosclerotic plaques, decreases TIMP-1 stability reducing its inhibitory effect on MMP-9, which is known to promote collagen degradation and is likewise associated with coronary artery disease. Disrupting the interaction of ADAMTS-7 and TIMP-1 might be a strategy to increase collagen content and plaque stability for the reduction of atherosclerosis-related events.
Collapse
Affiliation(s)
- M. Amin Sharifi
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Wierer
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Tan An Dang
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Jelena Milic
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Aldo Moggio
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Nadja Sachs
- Vascular Biology and Experimental Vascular Medicine Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Moritz von Scheidt
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Philipp Müller
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Julia Werner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Barbara Stiller
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics and University Heart Centre Lübeck, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics and University Heart Centre Lübeck, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Mira Graettinger
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Jeanette Reinshagen
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Lars Maegdefessel
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
- Vascular Biology and Experimental Vascular Medicine Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Jürgen Bernhagen
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
95
|
Sato M, Neufeld EB, Playford MP, Lei Y, Sorokin AV, Aponte AM, Freeman LA, Gordon SM, Dey AK, Jeiran K, Hamasaki M, Sampson ML, Shamburek RD, Tang J, Chen MY, Kotani K, Anderson JL, Dullaart RP, Mehta NN, Tietge UJ, Remaley AT. Cell-free, high-density lipoprotein-specific phospholipid efflux assay predicts incident cardiovascular disease. J Clin Invest 2023; 133:e165370. [PMID: 37471145 PMCID: PMC10503808 DOI: 10.1172/jci165370] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 07/18/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUNDCellular cholesterol efflux capacity (CEC) is a better predictor of cardiovascular disease (CVD) events than HDL-cholesterol (HDL-C) but is not suitable as a routine clinical assay.METHODSWe developed an HDL-specific phospholipid efflux (HDL-SPE) assay to assess HDL functionality based on whole plasma HDL apolipoprotein-mediated solubilization of fluorescent phosphatidylethanolamine from artificial lipid donor particles. We first assessed the association of HDL-SPE with prevalent coronary artery disease (CAD): study I included NIH severe-CAD (n = 50) and non-CAD (n = 50) participants, who were frequency matched for sex, BMI, type 2 diabetes mellitus, and smoking; study II included Japanese CAD (n = 70) and non-CAD (n = 154) participants. We also examined the association of HDL-SPE with incident CVD events in the Prevention of Renal and Vascular End-stage Disease (PREVEND) study comparing 340 patients with 340 controls individually matched for age, sex, smoking, and HDL-C levels.RESULTSReceiver operating characteristic curves revealed stronger associations of HDL-SPE with prevalent CAD. The AUCs in study I were as follows: HDL-SPE, 0.68; apolipoprotein A-I (apoA-I), 0.62; HDL-C, 0.63; and CEC, 0.52. The AUCs in study II were as follows: HDL-SPE, 0.83; apoA-I, 0.64; and HDL-C, 0.53. Also longitudinally, HDL-SPE was significantly associated with incident CVD events independent of traditional risk factors with ORs below 0.2 per SD increment in the PREVEND study (P < 0.001).CONCLUSIONHDL-SPE could serve as a routine clinical assay for improving CVD risk assessment and drug discovery.TRIAL REGISTRATIONClinicalTrials.gov NCT01621594.FUNDINGNHLBI Intramural Research Program, NIH (HL006095-06).
Collapse
Affiliation(s)
- Masaki Sato
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
- Division of Community and Family Medicine and Department of Clinical Laboratory Medicine, Jichi Medical University, Shimotsuke-City, Tochigi, Japan
- Biochemical Research Laboratory II, Eiken Chemical Co., Ltd., Shimotsuga-gun, Tochigi, Japan
| | - Edward B. Neufeld
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, NHLBI, NIH, Bethesda, Maryland, USA
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Alexander V. Sorokin
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
- Section of Inflammation and Cardiometabolic Diseases, NHLBI, NIH, Bethesda, Maryland, USA
| | - Angel M. Aponte
- Proteomics Core Facility, NHLBI, NIH, Bethesda, Maryland, USA
| | - Lita A. Freeman
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Scott M. Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Amit K. Dey
- Section of Inflammation and Cardiometabolic Diseases, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kianoush Jeiran
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Masato Hamasaki
- Division of Community and Family Medicine and Department of Clinical Laboratory Medicine, Jichi Medical University, Shimotsuke-City, Tochigi, Japan
- Biochemical Research Laboratory II, Eiken Chemical Co., Ltd., Shimotsuga-gun, Tochigi, Japan
| | | | - Robert D. Shamburek
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Jingrong Tang
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Marcus Y. Chen
- Laboratory of Cardiovascular CT, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kazuhiko Kotani
- Division of Community and Family Medicine and Department of Clinical Laboratory Medicine, Jichi Medical University, Shimotsuke-City, Tochigi, Japan
| | - Josephine L.C. Anderson
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Robin P.F. Dullaart
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, NHLBI, NIH, Bethesda, Maryland, USA
| | - Uwe J.F. Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Alan T. Remaley
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
- The NIH Clinical Center and
| |
Collapse
|
96
|
Theofilatos K, Stojkovic S, Hasman M, van der Laan SW, Baig F, Barallobre-Barreiro J, Schmidt LE, Yin S, Yin X, Burnap S, Singh B, Popham J, Harkot O, Kampf S, Nackenhorst MC, Strassl A, Loewe C, Demyanets S, Neumayer C, Bilban M, Hengstenberg C, Huber K, Pasterkamp G, Wojta J, Mayr M. Proteomic Atlas of Atherosclerosis: The Contribution of Proteoglycans to Sex Differences, Plaque Phenotypes, and Outcomes. Circ Res 2023; 133:542-558. [PMID: 37646165 PMCID: PMC10498884 DOI: 10.1161/circresaha.123.322590] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Using proteomics, we aimed to reveal molecular types of human atherosclerotic lesions and study their associations with histology, imaging, and cardiovascular outcomes. METHODS Two hundred nineteen carotid endarterectomy samples were procured from 120 patients. A sequential protein extraction protocol was employed in conjunction with multiplexed, discovery proteomics. To focus on extracellular proteins, parallel reaction monitoring was employed for targeted proteomics. Proteomic signatures were integrated with bulk, single-cell, and spatial RNA-sequencing data, and validated in 200 patients from the Athero-Express Biobank study. RESULTS This extensive proteomics analysis identified plaque inflammation and calcification signatures, which were inversely correlated and validated using targeted proteomics. The inflammation signature was characterized by the presence of neutrophil-derived proteins, such as S100A8/9 (calprotectin) and myeloperoxidase, whereas the calcification signature included fetuin-A, osteopontin, and gamma-carboxylated proteins. The proteomics data also revealed sex differences in atherosclerosis, with large-aggregating proteoglycans versican and aggrecan being more abundant in females and exhibiting an inverse correlation with estradiol levels. The integration of RNA-sequencing data attributed the inflammation signature predominantly to neutrophils and macrophages, and the calcification and sex signatures to smooth muscle cells, except for certain plasma proteins that were not expressed but retained in plaques, such as fetuin-A. Dimensionality reduction and machine learning techniques were applied to identify 4 distinct plaque phenotypes based on proteomics data. A protein signature of 4 key proteins (calponin, protein C, serpin H1, and versican) predicted future cardiovascular mortality with an area under the curve of 75% and 67.5% in the discovery and validation cohort, respectively, surpassing the prognostic performance of imaging and histology. CONCLUSIONS Plaque proteomics redefined clinically relevant patient groups with distinct outcomes, identifying subgroups of male and female patients with elevated risk of future cardiovascular events.
Collapse
Affiliation(s)
- Konstantinos Theofilatos
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Stefan Stojkovic
- Division of Cardiology, Department of Internal Medicine II (S.S., O.H., C.H., J.W., M.M.), Medical University of Vienna, Austria
| | - Maria Hasman
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, the Netherlands (S.W.v.d.L., G.P.)
| | - Ferheen Baig
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Javier Barallobre-Barreiro
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Lukas Emanuel Schmidt
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Siqi Yin
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Xiaoke Yin
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Sean Burnap
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Bhawana Singh
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Jude Popham
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
| | - Olesya Harkot
- Division of Cardiology, Department of Internal Medicine II (S.S., O.H., C.H., J.W., M.M.), Medical University of Vienna, Austria
| | - Stephanie Kampf
- Division of Vascular Surgery, Department of Surgery (S.K., C.N.), Medical University of Vienna, Austria
| | | | - Andreas Strassl
- Division of Cardiovascular and Interventional Radiology, Department of Biomedical Imaging and Image-Guided Therapy (A.S., C.L.), Medical University of Vienna, Austria
| | - Christian Loewe
- Division of Cardiovascular and Interventional Radiology, Department of Biomedical Imaging and Image-Guided Therapy (A.S., C.L.), Medical University of Vienna, Austria
| | - Svitlana Demyanets
- Department of Laboratory Medicine (S.D.), Medical University of Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of Surgery (S.K., C.N.), Medical University of Vienna, Austria
| | - Martin Bilban
- Core Facilities (M.B.), Medical University of Vienna, Austria
| | - Christian Hengstenberg
- Division of Cardiology, Department of Internal Medicine II (S.S., O.H., C.H., J.W., M.M.), Medical University of Vienna, Austria
| | - Kurt Huber
- Third Medical Department, Wilhelminenspital, and Sigmund Freud University, Medical Faculty, Vienna, Austria (K.H.)
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, the Netherlands (S.W.v.d.L., G.P.)
| | - Johann Wojta
- Division of Cardiology, Department of Internal Medicine II (S.S., O.H., C.H., J.W., M.M.), Medical University of Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria (J.W.)
| | - Manuel Mayr
- King’s British Heart Foundation Centre, Kings College London, United Kingdom (K.T., M.H., F.B., J.B.B., L.E.S., S.Y., X.Y., S.B., B.S., J.P., M.M.)
- Division of Cardiology, Department of Internal Medicine II (S.S., O.H., C.H., J.W., M.M.), Medical University of Vienna, Austria
| |
Collapse
|
97
|
Slysz J, Sinha A, DeBerge M, Singh S, Avgousti H, Lee I, Glinton K, Nagasaka R, Dalal P, Alexandria S, Wai CM, Tellez R, Vescovo M, Sunderraj A, Wang X, Schipma M, Sisk R, Gulati R, Vallejo J, Saigusa R, Lloyd-Jones DM, Lomasney J, Weinberg S, Ho K, Ley K, Giannarelli C, Thorp EB, Feinstein MJ. Single-cell profiling reveals inflammatory polarization of human carotid versus femoral plaque leukocytes. JCI Insight 2023; 8:e171359. [PMID: 37471165 PMCID: PMC10544225 DOI: 10.1172/jci.insight.171359] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Femoral atherosclerotic plaques are less inflammatory than carotid plaques histologically, but limited cell-level data exist regarding comparative immune landscapes and polarization at these sites. We investigated intraplaque leukocyte phenotypes and transcriptional polarization in 49 patients undergoing femoral (n = 23) or carotid (n = 26) endarterectomy using single-cell RNA-Seq (scRNA-Seq; n = 13), flow cytometry (n = 24), and IHC (n = 12). Comparative scRNA-Seq of CD45+-selected leukocytes from femoral (n = 9; 35,265 cells) and carotid (n = 4; 30,655 cells) plaque revealed distinct transcriptional profiles. Inflammatory foam cell-like macrophages and monocytes comprised higher proportions of myeloid cells in carotid plaques, whereas noninflammatory foam cell-like macrophages and LYVE1-overexpressing macrophages comprised higher proportions of myeloid cells in femoral plaque (P < 0.001 for all). A significant comparative excess of CCR2+ macrophages in carotid versus plaque was observed by flow cytometry in a separate validation cohort. B cells were more prevalent and exhibited a comparatively antiinflammatory profile in femoral plaque, whereas cytotoxic CD8+ T cells were more prevalent in carotid plaque. In conclusion, human femoral plaques exhibit distinct macrophage phenotypic and transcriptional profiles as well as diminished CD8+ T cell populations compared with human carotid plaques.
Collapse
Affiliation(s)
| | - Arjun Sinha
- Division of Cardiology, Department of Medicine
| | | | | | | | - Inhyeok Lee
- Division of Cardiology, Department of Medicine
| | - Kristofor Glinton
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
| | | | | | - Shaina Alexandria
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| | - Ching Man Wai
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Ricardo Tellez
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
| | | | | | - Xinkun Wang
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Matthew Schipma
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Ryan Sisk
- Division of Cardiology, Department of Medicine
| | - Rishab Gulati
- La Jolla Institute of Immunology, La Jolla, California, USA
| | | | | | - Donald M. Lloyd-Jones
- Division of Cardiology, Department of Medicine
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| | | | | | - Karen Ho
- Division of Vascular Surgery, NUFSM, Chicago, Illinois, USA
| | - Klaus Ley
- Immunology Center of Georgia, Augusta, Georgia, USA
| | - Chiara Giannarelli
- Department of Medicine and
- Department of Pathology, New York University, New York, New York, USA
| | | | - Matthew J. Feinstein
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| |
Collapse
|
98
|
Ciccone MM, Lepera ME, Guaricci AI, Forleo C, Cafiero C, Colella M, Palmirotta R, Santacroce L. Might Gut Microbiota Be a Target for a Personalized Therapeutic Approach in Patients Affected by Atherosclerosis Disease? J Pers Med 2023; 13:1360. [PMID: 37763128 PMCID: PMC10532785 DOI: 10.3390/jpm13091360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
In recent years, the increasing number of studies on the relationship between the gut microbiota and atherosclerosis have led to significant interest in this subject. The gut microbiota, its metabolites (metabolome), such as TMAO, and gut dysbiosis play an important role in the development of atherosclerosis. Furthermore, inflammation, originating from the intestinal tract, adds yet another mechanism by which the human ecosystem is disrupted, resulting in the manifestation of metabolic diseases and, by extension, cardiovascular diseases. The scientific community must understand and elucidate these mechanisms in depth, to gain a better understanding of the relationship between atherosclerosis and the gut microbiome and to promote the development of new therapeutic targets in the coming years. This review aims to present the knowledge acquired so far, to trigger others to further investigate this intriguing topic.
Collapse
Affiliation(s)
- Marco Matteo Ciccone
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Mario Erminio Lepera
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Andrea Igoren Guaricci
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Cinzia Forleo
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Concetta Cafiero
- Area of Molecular Pathology, Anatomic Pathology Unit, Fabrizio Spaziani Hospital, 03100 Frosinone, Italy;
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| | - Raffele Palmirotta
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| |
Collapse
|
99
|
Leković A, Nikolić S. Atheroprotective effect of myocardial bridge sustains in aging: Autopsy study on subjects with dual left anterior descending coronary artery type 3. Cardiovasc Pathol 2023; 66:107553. [PMID: 37321465 DOI: 10.1016/j.carpath.2023.107553] [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: 03/16/2023] [Revised: 05/18/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND The atheroprotective role of the myocardial bridge (MB) on a tunneled segment is already demonstrated in subjects with dual left anterior descending coronary artery (dual LAD) type 3 anomaly, but the dynamics of changes and whether this protective effect sustains during aging is unknown. METHODS The retrospective autopsy study included cases of dual LAD type 3 anomaly identified over 18 years. The severity grade of atherosclerosis in branches of dual LAD was estimated by microscopy. The Spearman's correlation test and Receiver operator characteristics (ROC) curve analyses were performed to determine the relation of subjects' age with a degree of the protective role of the myocardial bridge. RESULTS A total of 32 dual LAD type 3 cases were identified. The systematic heart examination revealed an anomaly prevalence of 2.1%. The age significantly positively correlated with the severity of atherosclerosis in the subepicardial dual LAD branch but not with the severity of atherosclerosis in the intramyocardial dual LAD branch. Subjects aged ≥38 years were likely to have a more severe degree of atherosclerosis in subepicardial than in intramyocardial LAD arteries (AUC 0.81 95% CI 0.59-1; sensitivity 100%, specificity 66.7%). In subjects aged ≥58 years, this difference was likely to be more pronounced (≥2 degree difference; AUC 0.75 95% CI 0.58-0.93; sensitivity 92.9%, specificity 66.7%). CONCLUSION The atheroprotective effect of the myocardial bridge on tunneled segments usually becomes evident throughout the second half of the fourth decade of life and is most pronounced after about 60 years and ceases only in some.
Collapse
Affiliation(s)
- Aleksa Leković
- Institute of Forensic Medicine, University of Belgrade Faculty of Medicine, Belgrade, Serbia.
| | - Slobodan Nikolić
- Institute of Forensic Medicine, University of Belgrade Faculty of Medicine, Belgrade, Serbia.
| |
Collapse
|
100
|
Alonso-Herranz L, Albarrán-Juárez J, Bentzon JF. Mechanisms of fibrous cap formation in atherosclerosis. Front Cardiovasc Med 2023; 10:1254114. [PMID: 37671141 PMCID: PMC10475556 DOI: 10.3389/fcvm.2023.1254114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
The fibrous cap is formed by smooth muscle cells that accumulate beneath the plaque endothelium. Cap rupture is the main cause of coronary thrombosis, leading to infarction and sudden cardiac death. Therefore, the qualities of the cap are primary determinants of the clinical outcome of coronary and carotid atherosclerosis. In this mini-review, we discuss current knowledge about the formation of the fibrous cap, including cell recruitment, clonal expansion, and central molecular signaling pathways. We also examine the differences between mouse and human fibrous caps and explore the impact of anti-atherosclerotic therapies on the state of the fibrous cap. We propose that the cap should be understood as a neo-media to substitute for the original media that becomes separated from the surface endothelium during atherogenesis and that embryonic pathways involved in the development of the arteria media contribute to cap formation.
Collapse
Affiliation(s)
- Laura Alonso-Herranz
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Julián Albarrán-Juárez
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Fog Bentzon
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus and Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| |
Collapse
|