1
|
Buja LM, McDonald MM, Zhao B, Narula N, Narula J, Barth RF. Insights from autopsy-initiated pathological studies of the pathogenesis and clinical manifestations of atherosclerosis and ischemic heart disease: Part II. Ischemic heart disease. Cardiovasc Pathol 2025; 76:107727. [PMID: 39956412 DOI: 10.1016/j.carpath.2025.107727] [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: 12/15/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025] Open
Abstract
CONTEXT Ischemic heart disease (IHD) due to coronary atherosclerosis constitutes the leading cause of morbidity and mortality worldwide. This review was undertaken to retrospectively analyze the lines of research that generated the evidence for our contemporary understanding of atherosclerosis-based coronary artery disease and to provide a rationale for continued support for autopsy-based research in order to make further progress in reduction of the morbidity and mortaility from IHD. OBJECTIVES To analyze the contributions of the autopsy to complement and validate other lines of investigation in determining the complex interactions between coronary artery alterations linked to the major manifestations of coronary atherosclerosis, namely, coronary thrombosis, acute myocardial infarction, and sudden cardiac death. DATA SOURCES Systematic search on PubMed to gather relevant studies concerning autopsy studies and reviews of the pathology and pathogenesis of atherosclerosis, ischemic heart disease, coronary atherosclerosis, coronary thrombosis, myocardial infarction and sudden cardiac death. CONCLUSIONS An extensive search of the published literature has confirmed the continuing importance of the autopsy as a powerful tool to understand the pathogenesis, clinical features, and therapeutic options for the treatment of atherosclerosis and its major manifestation, ischemic heart disease. This has been described in the Part I companion of the present review. Autopsy-initiated studies have documented the prevalence and clinicopathological significance of atherosclerosis in different human populations and its relationship to risk factors. It has been shown that the clinically silent phase of ischemic heart disease (IHD) begins in the first decades of life. Pathological studies have clarified the complex relationship between coronary atherosclerosis, coronary thrombosis, and myocardial ischemic events. These studies also have elucidated the pathological basis of sudden cardiac death. Insights from these studies also have been important in developing and evaluating strategies for continued progress in reducing the morbidity and mortality attributed to atherosclerosis and IHD.
Collapse
Affiliation(s)
- L Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, TX, USA.
| | | | - Bihong Zhao
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, TX, USA
| | - Navneet Narula
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, TX, USA
| | - Jagat Narula
- Division of Cardiology, Department of Internal Medicine, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, TX, USA
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
2
|
O'Donoghue L, Crognale D, Delahunt E, Smolenski A. Effects of exercise on cAMP-mediated platelet inhibition in young women: a pilot study. Eur J Appl Physiol 2025; 125:1283-1296. [PMID: 39636435 DOI: 10.1007/s00421-024-05673-2] [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: 02/14/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE Exercise has been shown to reduce platelet reactivity and increase platelet sensitivity to prostacyclin, an endothelium-derived inhibitor of platelet activation, in middle-aged men and women. It is currently unknown if these beneficial effects can also be observed in young women and the intracellular mechanisms involved have not been identified. In this study, the feasibility of detecting changes in platelet reactivity, prostacyclin sensitivity and cAMP signalling were tested. METHODS 10 well-trained and 10 sedentary but healthy young women participated in this study. Responses of washed platelets to thrombin receptor activating peptide 6, the thromboxane A2 receptor agonist U46619, and prostaglandin E1 were measured by light transmission aggregometry. Expression levels of proteins in the cAMP pathway including phosphorylation of the vasodilator-stimulated phosphoprotein were analysed by western blotting. RESULTS There was no evidence of reduced basal reactivity in platelets from the well-trained group (V ˙ O 2 max = 51.1 ± 3.6 ml/kg/min) compared to the untrained group (V ˙ O 2 max = 31.1 ± 4.7 ml/kg/min). Platelets from the trained group showed evidence of greater sensitivity to the anti-aggregatory effects of prostaglandin E1. The slope of the aggregation curves indicated an overall faster rate of aggregation in the untrained group. Mean phosphorylation levels of vasodilator-stimulating phosphoprotein were consistently higher in the trained group, indicative of increased protein kinase A activity. CONCLUSION Platelets from young women may exhibit an exercise-induced increase in sensitivity to prostacyclin leading to stimulation of the cAMP pathway. A larger study is warranted to explore this vasoprotective effect further.
Collapse
Affiliation(s)
- Lorna O'Donoghue
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin, D02 YN77, Ireland
| | - Domenico Crognale
- Institute for Sport and Health, University College Dublin, Newstead Building C Belfield, Dublin 4, Ireland
| | - Eamonn Delahunt
- School of Public Health, Physiotherapy and Sports Science, UCD, Health Science Centre, Belfield, Dublin 4, Ireland
| | - Albert Smolenski
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin, D02 YN77, Ireland.
| |
Collapse
|
3
|
Buja LM, McDonald MM, Zhao B, Narula N, Narula J, Barth RF. Insights from autopsy-initiated pathological studies of the pathogenesis and clinical manifestations of atherosclerosis and ischemic heart disease: Part I. Atherosclerosis. Cardiovasc Pathol 2025; 76:107726. [PMID: 39971218 DOI: 10.1016/j.carpath.2025.107726] [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: 09/04/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025] Open
Abstract
CONTEXT Ischemic heart disease (IHD) due to coronary atherosclerosis constitutes the leading cause of morbidity and mortality worldwide. This review was undertaken to document the historical basis for our contemporary understanding of atherosclerosis-based disease and to provide a rationale for continued support for autopsy-based research to make further progress in reducing the morbidity and mortality from atherosclerosis-related disease. OBJECTIVES To analyze the contributions of the autopsy-initiated pathological studies to complement and validate other lines of investigation in determining the pathology and pathogenesis of the leading worldwide cause of morbidity and mortality, namely, atherosclerosis and its major complications of coronary atherosclerosis, ischemic heart disease, coronary thrombosis, acute myocardial infarction, and sudden cardiac death. DATA SOURCES Systematic search on PubMed to gather relevant studies concerning autopsy studies and reviews of the pathology and pathogenesis of atherosclerosis, ischemic heart disease, coronary atherosclerosis, coronary thrombosis, myocardial infarction, and sudden cardiac death CONCLUSIONS: Extensive published reports have confirmed the continuing importance of the autopsy as a powerful tool to understand the pathogenesis, clinical features, and therapeutic options for major diseases. This specifically has been shown by the analysis of atherosclerosis and its major manifestation of ischemic heart disease, as presented in this (Part I) and its companion (Part II) review. Autopsy-initiated pathological studies have documented the prevalence and natural history of atherosclerosis in different human populations in relationship to the prevalence of risk factors and established that the clinically silent phase of the disease begins in the first decades of life. Insights from these studies have been essential in developing and evaluating strategies for continued progress in preventing and controlling the disability and death associated with atherosclerotic heart disease.
Collapse
Affiliation(s)
- L Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA.
| | | | - Bihong Zhao
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Navneet Narula
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Jagat Narula
- Division of Cardiology, Department of Internal Medicine, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
4
|
Juricic S, Klac J, Stojkovic S, Tesic M, Jovanovic I, Aleksandric S, Dobric M, Zivkovic S, Maricic B, Simeunovic D, Lasica R, Dikic M, Banovic M, Beleslin B. Molecular and Pathophysiological Mechanisms Leading to Ischemic Heart Disease in Patients with Diabetes Mellitus. Int J Mol Sci 2025; 26:3924. [PMID: 40362167 PMCID: PMC12071796 DOI: 10.3390/ijms26093924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Coronary atherosclerosis in patients with diabetes mellitus is the most significant pathophysiological mechanism responsible for ischemic heart disease. Atherosclerosis in diabetes is premature, more diffuse, and more progressive, and it affects more coronary blood vessels compared to non-diabetics. Atherosclerosis begins with endothelial dysfunction, continues with the formation of fatty streaks in the intima of coronary arteries, and ends with the appearance of an atherosclerotic plaque that expands centrifugally and remodels the coronary artery. If the atherosclerotic plaque is injured, a thrombus forms at the site of the damage, which can lead to vessel occlusion and potentially fatal consequences. Diabetes mellitus and atherosclerosis are connected through several pathological pathways. Among the most significant factors that lead to atherosclerosis in diabetics are hyperglycemia, insulin resistance, oxidative stress, dyslipidemia, and chronic inflammation. Chronic inflammation is currently considered one of the most important factors in the development of atherosclerosis. However, to date, no adequate anti-inflammatory therapeutic measures have been found to prevent the progression of the atherosclerotic process, and they remain a subject of ongoing research. In this review, we summarize the most significant pathophysiological mechanisms that link atherosclerosis and diabetes mellitus.
Collapse
Affiliation(s)
- Stefan Juricic
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
| | - Jovana Klac
- Department of Cardiology, Emergency Center, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (J.K.); (R.L.)
| | - Sinisa Stojkovic
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Milorad Tesic
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ivana Jovanovic
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
| | - Srdjan Aleksandric
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Milan Dobric
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Dedinje Cardiovascular Institute, 11000 Belgrade, Serbia;
| | | | - Bojan Maricic
- Clinic of Cardiology, University Clinical Center Nis, 18000 Nis, Serbia;
| | - Dejan Simeunovic
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ratko Lasica
- Department of Cardiology, Emergency Center, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (J.K.); (R.L.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Miodrag Dikic
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
| | - Marko Banovic
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Branko Beleslin
- Clinic for Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.S.); (M.T.); (I.J.); (S.A.); (D.S.); (M.D.); (M.B.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
5
|
Wu Z, Liu R, Chen J, Cai X, Yi J, Wang J, Wang D, Hu M. An enzymatic cleavage-triggered minimally invasive nanosensor for urine-based detection of early atherosclerosis. SCIENCE ADVANCES 2025; 11:eadu7614. [PMID: 40085714 PMCID: PMC11908493 DOI: 10.1126/sciadv.adu7614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Timely detection of early atherosclerosis (AS) is crucial for improving cardiovascular outcomes, creating a growing demand for diagnostic tools that are simple, sensitive, and cost-effective. Here, we introduce a synthetic nanosensor for early AS detection that leverages the fluorescence and renal clearance properties of carbon quantum dots (CQDs). This nanosensor, designed to respond to the proteolytic activity of AS-associated dysregulated enzymes, entails CQDs as signal reporters to convert AS-associated proteolytic activity to fluorometric readings enabling a sensitive and cost-effective urine-based assay for early AS detection. Our findings demonstrated that the nanosensor provided distinct signals in atherosclerotic versus healthy mice at early AS stages, indicating its diagnostic potential. Moreover, toxicity tests showed no notable adverse effects, supporting its safety for diagnostic applications. This minimally invasive diagnostic approach could facilitate personalized therapy design and continuous efficacy assessment. It is expected that such a modular nanosensor platform can be integrated with simple urine tests to offer cost-effective detection of various diseases.
Collapse
Affiliation(s)
- Zhina Wu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| | - Rui Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Jianai Chen
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xueying Cai
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | | | - Jiasi Wang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China
| |
Collapse
|
6
|
Lim KT, Choe JW, Hwang SS. Association Between Geriatric Oral Health Assessment Index and Cardiovascular Disease in Korean Older Adults. J Prev Med Public Health 2025; 58:103-112. [PMID: 39810541 PMCID: PMC11824635 DOI: 10.3961/jpmph.24.569] [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: 09/30/2024] [Revised: 12/29/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVES This study examined the association between oral health-related quality of life (OHRQoL), as assessed by the Geriatric Oral Health Assessment Index (GOHAI), and cardiovascular disease (CVD) outcomes among Korean older adults. METHODS Data from 5413 participants in the Korean Longitudinal Study of Aging were analyzed. GOHAI scores were categorized as either "poor" (<40) or "not poor" (≥40). Generalized estimating equation models were used to assess the relationship between GOHAI scores and CVD prevalence, with analyses stratified by sex. RESULTS Poor GOHAI score was significantly associated with elevated odds of CVD (odds ratio [OR], 1.13; 95% confidence interval [CI], 1.07 to 1.19; p<0.001). This association was stronger in female (OR, 1.36) compared to male (OR, 1.12). Poor oral health is indicative of systemic inflammation and age-related vulnerabilities, underscoring the utility of the GOHAI as an instrument for early identification of CVD risk. CONCLUSIONS Poor oral health, as measured by the GOHAI, is associated with an increased risk of CVD among older adults, especially female. These findings support the use of the GOHAI as a cost-effective screening tool for the early assessment of CVD risk. Further research is warranted to explore inflammatory biomarkers and sex-specific mechanisms that could inform targeted interventions.
Collapse
Affiliation(s)
- Kyu-Taek Lim
- Department of Preventive Medicine, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Ji-won Choe
- Incorporated Association FREEMED, Seoul, Korea
| | - Seung-sik Hwang
- Department of Preventive Medicine, Graduate School of Public Health, Seoul National University, Seoul, Korea
| |
Collapse
|
7
|
Wang M, Zhou S, Hu Y, Tong W, Zhou H, Ma M, Cai X, Zhang Z, Zhang L, Chen Y. Macrophages overexpressing interleukin-10 target and prevent atherosclerosis: Regression of plaque formation and reduction in necrotic core. Bioeng Transl Med 2025; 10:e10717. [PMID: 39801756 PMCID: PMC11711221 DOI: 10.1002/btm2.10717] [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: 02/01/2024] [Revised: 07/27/2024] [Accepted: 08/08/2024] [Indexed: 01/16/2025] Open
Abstract
Atherosclerosis, a slowly progressing inflammatory disease, is characterized by the presence of monocyte-derived macrophages. Interventions targeting the inflammatory characteristics of atherosclerosis hold promising potential. Although interleukin (IL)-10 is widely acknowledged for its anti-inflammatory effects, systemic administration of IL-10 has limitations due to its short half-life and significant systemic side effects. In this study, we aimed to investigate the effectiveness of an approach designed to overexpress IL-10 in macrophages and subsequently introduce these genetically modified cells into ApoE-/- mice to promote atherosclerosis regression. We engineered RAW264.7 cells to overexpress IL-10 (referred to as IL-10M) using lentivirus vectors. The IL-10M exhibited robust IL-10 secretion, maintained phagocytic function, improved mitochondrial membrane potentials, reduced superoxide production and showed a tendency toward the M2 phenotype when exposed to inflammatory stimuli. IL-10M can selectively target plaques in ApoE-/- mice and has the potential to reduce plaque area and necrotic core at both early and late stages of plaque progression. Moreover, there was a significant reduction in MMP9, a biomarker associated with plaque rupture, in IL-10M-treated plaques from both the early and late intervention groups. Additionally, the administration of IL-10M showed no obvious side effects. This study serves as proof that cell therapy based on anti-inflammatory macrophages might be a promising strategy for the intervention of atherosclerosis.
Collapse
Affiliation(s)
- Mingyi Wang
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Shanshan Zhou
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| | - Yingyun Hu
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- The Medical School of Nankai UniversityTianjinChina
| | - Wei Tong
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| | - Hao Zhou
- Department of CardiologyNo. 966 Hospital of Joint Logisties ForceDandongChina
| | - Mingrui Ma
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Xingxuan Cai
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- The Second Medical School of Southern Medical UniversityGuangzhouChina
| | - Zhengbin Zhang
- Medical School of Chinese PLABeijingChina
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
| | - Luo Zhang
- Medical School of Chinese PLABeijingChina
- Research Center of BioengineeringThe Medical Innovation Research Division of PLA General HospitalBeijingChina
| | - Yundai Chen
- Senior Department of CardiologyThe Sixth Medical Center of PLA General HospitalBeijingChina
- Department of CardiologyThe First Medical Center of PLA General HospitalBeijingChina
| |
Collapse
|
8
|
Kumar S, Sharma V, Yadav S. TLR4 Targeting: A Promising Therapeutic Approach Across Multiple Human Diseases. Curr Protein Pept Sci 2025; 26:241-258. [PMID: 39722483 DOI: 10.2174/0113892037324425241018061548] [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: 05/22/2024] [Revised: 08/31/2024] [Accepted: 09/23/2024] [Indexed: 12/28/2024]
Abstract
TLR4 stands at the forefront of innate immune responses, recognizing various pathogen- associated molecular patterns and endogenous ligands, thus serving as a pivotal mediator in the immune system's defense against infections and tissue damage. Beyond its canonical role in infection, emerging evidence highlights TLR4's involvement in numerous non-infectious human diseases, ranging from metabolic disorders to neurodegenerative conditions and cancer. Targeting TLR4 signaling pathways presents a promising therapeutic approach with broad applicability across these diverse pathological states. In metabolic disorders such as obesity and diabetes, dysregulated TLR4 activation contributes to chronic low-grade inflammation and insulin resistance, driving disease progression. In cardiovascular diseases, TLR4 signaling promotes vascular inflammation and atherogenesis, implicating its potential as a therapeutic target to mitigate cardiovascular risk. Neurodegenerative disorders, including Alzheimer's and Parkinson's diseases, exhibit aberrant TLR4 activation linked to neuroinflammation and neuronal damage, suggesting TLR4 modulation as a strategy to attenuate neurodegeneration. Additionally, in cancer, TLR4 signaling within the tumor microenvironment promotes tumor progression, metastasis, and immune evasion, underscoring its relevance as a target for anticancer therapy. Advances in understanding TLR4 signaling cascades and their contributions to disease pathogenesis have spurred the development of various pharmacological agents targeting TLR4. These agents range from small molecule inhibitors to monoclonal antibodies, with some undergoing preclinical and clinical evaluations. Furthermore, strategies involving TLR4 modulation through dietary interventions and microbiota manipulation offer additional avenues for therapeutic exploration. Hence, targeting TLR4 holds significant promise as a therapeutic strategy across a spectrum of human diseases, offering the potential to modulate inflammation, restore immune homeostasis, and impede disease progression.
Collapse
Affiliation(s)
- Sakshi Kumar
- Department of Pharmacy, Galgotias College, Greater Noida, Uttar Pradesh, 201310, India
| | - Vikram Sharma
- Department of Pharmacy, Galgotias College, Greater Noida, Uttar Pradesh, 201310, India
| | - Shikha Yadav
- School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, 201310, India
| |
Collapse
|
9
|
Li J, Li Y, Lin X, Lv C, Zhang X, Chen J. Evaluation of Aortic Hemodynamics Using Four-Dimensional Flow of Magnetic Resonance Imaging in Rabbits with Liver Fibrosis. J Magn Reson Imaging 2024; 60:2604-2612. [PMID: 38520716 DOI: 10.1002/jmri.29363] [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: 12/21/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Liver fibrosis (LF) precipitates systemic hemodynamic alterations, however, its impact on the aorta remaining undefined. PURPOSE To assess aorta hemodynamics changes during LF development in a rabbit model. STUDY TYPE Prospective, experimental. ANIMAL MODEL Thirty 7-month-old male rabbits underwent bile duct ligation (BDL) to induce LF. FIELD STRENGTH/SEQUENCE Biweekly four-dimensional (4D) flow imaging incorporating a 3D gradient-echo at 3.0 T scanner for 14 weeks post-BDL. ASSESSMENT Histopathological exams for 2-5 rabbits were performed at each time point, following each MRI scan. LF was graded using the Metavir scale by a pathologist. 4D flow was analyzed by two radiologists using dedicated postprocessing software. They recorded 4D flow parameters at four aorta sections (aortic sinus, before and after bifurcation of aortic arch, and descending aorta). STATISTICAL TESTS The linear mixed model; Bonferroni correction; Pearson correlation coefficient (r); receiver operating characteristic (ROC) curve; Delong test. The level of significance was set at P < 0.05. RESULTS Following BDL, the wall shear stress (WSS) (0.23-0.32 Pa), energy loss (EL) (0.27-1.55 mW) of aorta significantly increased at the second week for each plane, peaking at the sixth week (WSS: 0.35-0.49 Pa, EL: 0.57-2.0 mW). So did the relative pressure difference (RPD) (second week: 1.67 ± 1.63 mmHg, sixth week: 2.43 ± 0.63 mmHg) in plane 2. Notably, the RPD in plane 2 at the second week displayed the highest area under ROC curve of 0.998 (specificity: 1, sensitivity: 0.967). LF were found at the second, fourth, and sixth week after BDL, with grade F2, F3, and F4, respectively. The RPD in plane 2 was most strongly correlated with the severity of LF (r = 0.86). DATA CONCLUSIONS The occurrence of LF could increase WSS, EL, and RPD of aorta as early as the second week following BDL. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jiali Li
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuansheng Li
- School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Lin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Cheng Lv
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoyong Zhang
- Clinical Science, Philips Healthcare, Chengdu, Sichuan, China
| | - Jing Chen
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
10
|
Yu M, Wu H, Hu H, Cheng Y, Qin Y, Yang K, Hu C, Guo W, Kong Y, Zhao W, Cheng X, Jiang H, Wang S. Emerging near-infrared targeting diagnostic and therapeutic strategies for ischemic cardiovascular and cerebrovascular diseases. Acta Biomater 2024:S1742-7061(24)00682-2. [PMID: 39577483 DOI: 10.1016/j.actbio.2024.11.027] [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: 07/28/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Ischemic cardiovascular and cerebrovascular diseases (ICCDs), including thrombosis, ischemic stroke and atherosclerosis, represent a significant threat to human health, and there is an urgent requirement for the implementation of emerging diagnostic and therapeutic approaches to improve symptoms and prognosis. As a promising noninvasive modality offering high spatial and temporal resolution with favorable biocompatible properties, near-infrared (NIR) light has demonstrated a vast and profound potential in the biomedical field in recent years. Meanwhile, nanomedicine carriers are undergoing rapid development due to their high specific surface area, elevated drug loading capacity, and unique physicochemical properties. The combination of NIR light with targeted nanoprobes modified with different functional components not only maintains the high penetration depth of NIR irradiation in biological tissues but also significantly enhances the targeting specificity at the lesion site. This strategy allows for the realization of on-demand drug release and photothermal effects, thus inspiring promising avenues for the diagnosis and treatment of ICCDs. However, the clinical translation of NIR imaging and therapy is still hindered by significant obstacles. The existing literature has provided a comprehensive overview of the advancements in NIR-based nanomedicine research. However, there is a notable absence of reviews that summarize the NIR-mediated targeting strategies against ICCDs in imaging and therapy. Therefore, this review concludes the application of the emerging targeting probes combined with NIR radiation for ICCDs classified by molecular targets, analyzes the current challenges, and provides improvement strategies and prospects for further clinical translation. STATEMENT OF SIGNIFICANCE: Ischemic cardiovascular and cerebrovascular diseases (ICCDs) represent a significant threat to human health. Recently, near-infrared (NIR) light combined with targeting probes have been employed for the diagnosis and treatment of ICCDs, offering exceptional advantages including rapid feedback, high penetration depth, on-demand drug release, and favorable biocompatibility. However, there is a notable absence of reviews that summarize the NIR light-mediated targeting strategies for the imaging and therapy of ICCDs. Therefore, this review summarizes the emerging targeting probes combined with NIR light classified by molecular targets, and the proposes potential improvement strategies for clinical translation. This review elucidates the potential and current status of NIR-based techniques in ICCDs, while also serving as a reference point for additional targeted therapeutic strategies for ICCDs.
Collapse
Affiliation(s)
- Mengran Yu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Huijun Wu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Haoyuan Hu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Ye Cheng
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Youran Qin
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Kaiqing Yang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - ChangHao Hu
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Wei Guo
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Yuxuan Kong
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Weiwen Zhao
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Xueqin Cheng
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China
| | - Hong Jiang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China.
| | - Songyun Wang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430061, China.
| |
Collapse
|
11
|
McGonagle D, Giryes S. An immunology model for accelerated coronary atherosclerosis and unexplained sudden death in the COVID-19 era. Autoimmun Rev 2024; 23:103642. [PMID: 39313122 DOI: 10.1016/j.autrev.2024.103642] [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: 07/28/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
The immunological basis for cardiac deaths remote from potential triggering viral infection, including SARS-CoV-2 infection, remains enigmatic. Cardiac surface inflammation, including the pericardium, epicardium and superficial myocardium with associated coronary artery vasculitis in infant Kawasaki Disease (KD) and multisystem inflammatory syndrome in children (MIS-C) is well recognised. In this perspective, we review the evidence pointing towards prominent post-viral infection related epicardial inflammation in older subjects, resulting in atherosclerotic plaque destabilisation with seemingly unrelated myocardial infarction that may be temporally distant from the actual infectious triggers. Cardiac surface inflammation in the relatively immune cell rich tissues in the territory though where the coronary arteries traverse is common in the adult post-COVD pneumonic phase and is also well described after vaccination including pre-COVID era vaccinations. Immunologically, the pericardium/epicardium tissue was known to be critical for coronary artery territory atherosclerotic disease prior to the COVID-19 era and may be linked to the involvement of the coronary artery vasa vasorum that physiologically oxygenates the coronary artery walls. We highlight how viral infection or vaccination-associated diffuse epicardial tissue inflammation adjacent to the coronary artery vasa vasorum territory represents a critical unifying concept for seemingly unrelated fatal coronary artery atherosclerotic disease, that could occur soon after or remote from infection or vaccination in adults. Mechanistically, such epicardial inflammation impacting coronary artery vasa vasorum immunity acts as gateways towards the slow destabilisation of pre-existing atherosclerotic plaques, with resultant myocardial infarction and other cardiac pathology. This model offers immunologists and academic cardiologists an immunopathological roadmap between innocuous viral infections or vaccinations and seemingly temporally remote "unrelated" atherosclerotic disease with excess cardiac deaths.
Collapse
Affiliation(s)
- Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom; National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, United Kingdom.
| | - Sami Giryes
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom; B. Shine Rheumatology Institute, Rambam Healthcare Campus, Haifa, Israel
| |
Collapse
|
12
|
Ren L, Pushpakumar S, Almarshood H, Das SK, Sen U. Epigenetic DNA Methylation and Protein Homocysteinylation: Key Players in Hypertensive Renovascular Damage. Int J Mol Sci 2024; 25:11599. [PMID: 39519150 PMCID: PMC11546175 DOI: 10.3390/ijms252111599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Hypertension has been a threat to the health of people, the mechanism of which, however, remains poorly understood. It is clinically related to loss of nephron function, glomerular sclerosis, or necrosis, resulting in renal functional declines. The mechanisms underlying hypertension's development and progression to organ damage, including hypertensive renal damage, remain to be fully elucidated. As a developing approach, epigenetics has been postulated to elucidate the phenomena that otherwise cannot be explained by genetic studies. The main epigenetic hallmarks, such as DNA methylation, histone acetylation, deacetylation, noncoding RNAs, and protein N-homocysteinylation have been linked with hypertension. In addition to contributing to endothelial dysfunction and oxidative stress, biologically active gases, including NO, CO, and H2S, are crucial regulators contributing to vascular remodeling since their complex interplay conducts homeostatic functions in the renovascular system. Importantly, epigenetic modifications also directly contribute to the pathogenesis of kidney damage via protein N-homocysteinylation. Hence, epigenetic modulation to intervene in renovascular damage is a potential therapeutic approach to treat renal disease and dysfunction. This review illustrates some of the epigenetic hallmarks and their mediators, which have the ability to diminish the injury triggered by hypertension and renal disease. In the end, we provide potential therapeutic possibilities to treat renovascular diseases in hypertension.
Collapse
Affiliation(s)
- Lu Ren
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Hebah Almarshood
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| | - Swapan K. Das
- Department of Internal Medicine, Section on Endocrinology and Metabolism, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA; (L.R.)
| |
Collapse
|
13
|
Saleh C, Budincevic H. Carotid intima-media thickness and autism spectrum disorder. INTERNATIONAL JOURNAL OF DEVELOPMENTAL DISABILITIES 2024; 70:1123-1125. [PMID: 39801530 PMCID: PMC11721853 DOI: 10.1080/20473869.2024.2416321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/18/2024] [Indexed: 01/16/2025]
Affiliation(s)
| | - Hrvoje Budincevic
- Stroke and Intensive Care Unit, Department of Neurology, Sveti Duh University Hospital, Zagreb, Croatia
- Faculty of Medicine, Department of Neurology and Neurosurgery, J.J. Strossmayer University of Osijek, Osijek, Croatia
| |
Collapse
|
14
|
Greco F, Bertagna G, Quercioli L, Pucci A, Rocchiccioli S, Ferrari M, Recchia FA, McDonnell LA. Lipids associated with atherosclerotic plaque instability revealed by mass spectrometry imaging of human carotid arteries. Atherosclerosis 2024; 397:118555. [PMID: 39159550 DOI: 10.1016/j.atherosclerosis.2024.118555] [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: 11/23/2023] [Revised: 06/22/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND AND AIMS Lipids constitute one of the main components of atherosclerosis lesions and are the mediators of many mechanisms involved in plaque progression and stability. Here we tested the hypothesis that lipids known to be involved in plaque development exhibited associations with plaque vulnerability. We used spatial lipidomics to overcome plaque heterogeneity and to compare lipids from specific regions of symptomatic and asymptomatic human carotid atherosclerotic plaques. METHODS Carotid atherosclerotic plaques were collected from symptomatic and asymptomatic patients. Plaque lipids were analyzed with the spatial lipidomics technique matrix-assisted laser desorption/ionization mass spectrometry imaging, and histology and immunofluorescence were used to segment the plaques into histomolecularly distinct regions. RESULTS Macrophage-rich regions from symptomatic lesions were found to be enriched in phosphatidylcholines (synthesized to counteract excess free cholesterol), while the same region from asymptomatic plaques were enriched in polyunsaturated cholesteryl esters and triglycerides, characteristic of functional lipid droplets. Vascular smooth muscle cells (VSMCs) of the fibrous cap of asymptomatic plaques were enriched in lysophosphatidylcholines and cholesteryl esters, know to promote VSMC proliferation and migration, crucial for the buildup of the fibrous cap stabilizing the plaque. CONCLUSIONS The investigation of the region-specific lipid composition of symptomatic and asymptomatic human atherosclerotic plaques revealed specific lipid markers of plaque outcome, which could be linked to known biological characteristics of stable plaques.
Collapse
Affiliation(s)
- Francesco Greco
- Centro Health and BioMedLab, Scuola Superiore Sant'Anna, Pisa, Italy; Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme (PI), Italy; Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giulia Bertagna
- Azienda Ospedaliero Universitaria Pisana, Department of Vascular Surgery, Pisa, Italy
| | - Laura Quercioli
- Azienda Ospedaliero Universitaria Pisana, Department of Vascular Surgery, Pisa, Italy
| | - Angela Pucci
- Department of Histopathology, University Hospital, Pisa, Italy
| | | | - Mauro Ferrari
- Azienda Ospedaliero Universitaria Pisana, Department of Vascular Surgery, Pisa, Italy
| | - Fabio A Recchia
- Institute of Clinical Physiology, National Research Council, Pisa, Italy; Aging & Cardiovascular Discovery Center, Lewis Katz School of Medicine, Philadelphia, USA; Scuola Superiore Sant'Anna, Pisa, Italy
| | - Liam A McDonnell
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme (PI), Italy.
| |
Collapse
|
15
|
Senderovic A, Galijasevic S. The Role of Inducible Nitric Oxide Synthase in Assessing the Functional Level of Coronary Artery Lesions in Chronic Coronary Syndrome. Cardiol Res 2024; 15:330-339. [PMID: 39420980 PMCID: PMC11483113 DOI: 10.14740/cr1700] [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: 07/10/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Chronic coronary syndrome (CCS) is a long-term manifestation of coronary artery disease, marked by stable but recurring chest pain and myocardial ischemia due to the gradual buildup of atherosclerotic plaques in the coronary arteries. It is a metabolic disorder of coronary arteries characterized by oxidative stress, endothelial dysfunction, inflammation, and hyperlipidemia. The imbalance in oxidative-antioxidative status contributes to stable ischemic heart disease. Oxidative stress involves reactive oxygen and nitrogen species, leading to low-density lipoprotein (LDL) oxidation. Endothelial dysfunction, marked by reduced nitric oxide (NO) bioavailability, is an early onset of CCS, affecting vasodilation, cell proliferation, and inflammatory responses. Enzyme myeloperoxidase (MPO), traditionally considered protective, plays a dual role in initiating and progressing inflammatory diseases. MPO interacts with NO, modulating its catalytic activity. Elevated NO levels inhibit MPO through a reversible complex formation, preventing NO-induced inhibition by inducible nitric oxide synthase (iNOS). MPO also inactivates endothelial nitric oxide synthase (eNOS) and reacts with L-arginine, hindering NO synthesis. The interplay between MPO and NO significantly influences inflammation sites, impacting peroxidation rates and oxidation reactions. Peroxynitrite, a reactive species, contributes to nitration of tyrosine residues and lipid peroxidation. Mechanistic pathways suggest MPO enhances iNOS catalytic activity, influencing CCS development. iNOS, implicated in inflammation and atherosclerosis, is connected to NO regulation. This review analyzes the complex interplay of MPO, iNOS, and NO that affects plaque morphology, oxidative stress, and inflammation, contributing to atherosclerosis progression. Therefore, it is possible that the phenotypes of atherosclerotic plaques, focal and diffuse coronary artery disease, could be defined by the relationship between MPO and iNOS.
Collapse
Affiliation(s)
- Admina Senderovic
- Public Institution of Health Centers of the Canton of Sarajevo, Laboratory Diagnostics Service of the Ilidza, Health Center, Ilidza, Bosnia and Herzegovina
- Sarajevo Medical School, University Sarajevo School of Science and Technology, Sarajevo, Bosnia and Hercegovina
| | - Semira Galijasevic
- Sarajevo Medical School, University Sarajevo School of Science and Technology, Sarajevo, Bosnia and Hercegovina
| |
Collapse
|
16
|
Gomez M, Montalvo S, Sanchez A, Conde D, Ibarra-Mejia G, Peñailillo LE, Gurovich AN. Effects of Different Eccentric Cycling Intensities on Brachial Artery Endothelial Shear Stress and Blood Flow Patterns. RESEARCH QUARTERLY FOR EXERCISE AND SPORT 2024:1-11. [PMID: 39302246 DOI: 10.1080/02701367.2024.2404139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 08/31/2024] [Indexed: 09/22/2024]
Abstract
Eccentric exercise has gained attention as a novel exercise modality that increases muscle performance at a lower metabolic demand. However, vascular responses to eccentric cycling (ECC) are unknown, thus gaining knowledge regarding endothelial shear stress (ESS) during ECC may be crucial for its application in patients. The purpose of this study was to explore ECC-induced blood flow patterns and ESS across three different intensities in ECC. Eighteen young, apparently healthy subjects were recruited for two laboratory visits. Maximum oxygen consumption, power output, and blood lactate (BLa) threshold were measured to determine workload intensities. Blood flow patterns in the brachial artery were measured via ultrasound imaging and Doppler on an eccentric ergometer during a 5 min workload steady exercise test at low (BLa of 0-2 mmol/L), moderate (BLa 2-4 mmol/L), and high intensity (BLa levels > 4 mmol/L). There was a significant increase in the antegrade ESS in an intensity-dependent manner (baseline: 44.2 ± 8.97; low: 55.6 ± 15.2; moderate: 56.0 ± 10.5; high: 70.7 ± 14.9, all dynes/cm2, all p values < 0.0002) with the exception between low and moderate and Re (AU) showed turbulent flow at all intensities. Regarding retrograde flow, ESS also increased in an intensity-dependent manner (baseline 9.72 ± 4.38; low: 12.5 ± 3.93; moderate: 15.8 ± 5.45; high: 15.7 ± 6.55, all dynes/cm2, all p values < 0.015) with the exception between high and moderate and Re (AU) showed laminar flow in all intensities. ECC produced exercise-induced blood flow patterns that are intensity-dependent. This suggests that ECC could be beneficial as a modulator of endothelial homeostasis.
Collapse
|
17
|
Saleh C. Letter to the Editor regarding, Wang Y, Zhang G, Wu J, Wang A, Zhang X, Zhang J, Wu S, Zhao X, Zhang Q. The impact of cumulative exposure to diverse lipid profiles on carotid intima-media thickness among an asymptomatic population. J Stroke Cerebrovasc Dis. 2023 Dec;32(12):107443. J Stroke Cerebrovasc Dis 2024; 33:107853. [PMID: 39181685 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 07/08/2024] [Indexed: 08/27/2024] Open
|
18
|
Vail KJ, Bourland JD, Dugan GO, Chen BJ, Clarkson TB, Cline JM, Meléndez GC. Exogenous Growth Hormone Exacerbates Post-Irradiation Atherosclerosis in Susceptible Epicardial Coronary Arteries. Toxicol Pathol 2024; 52:308-318. [PMID: 39254136 PMCID: PMC11521112 DOI: 10.1177/01926233241277454] [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] [Indexed: 09/11/2024]
Abstract
Cardiac exposure to ionizing radiation can damage both the microvasculature and coronary arteries, as well as increase the long-term risk of heart disease, myocardial fibrosis, and conduction abnormalities. Therapeutic agents capable of promoting recovery from radiation injury to the heart are limited. Growth hormone is linked to improved cardiac function following injury. Here, we leveraged a cynomolgus macaque model to determine the long-term outcomes of recombinant human growth hormone (rhGH) therapy on the heart following low-dose ionizing radiation. Macaques were exposed to 2 Gy radiation, treated with rhGH for one month, and assessed after 2 years. Overall, plasma lipid profile, cardiac function, and coronary artery disease were similar between rhGH and placebo treated animals. However, a subgroup of rhGH-treated animals exhibited more extensive atherosclerotic plaques in the coronary arteries. Together, these findings indicate that transient human growth hormone therapy subsequent to a single low dose of ionizing radiation involving the heart does not result in long-term changes to plasma cholesterol but may promote exacerbated coronary artery disease in a subset of individuals.
Collapse
Affiliation(s)
- Krystal J. Vail
- Tulane National Primate Research Center, Covington, Louisiana, USA
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - J. Daniel Bourland
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Gregory O. Dugan
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Benny J. Chen
- Duke University Medical Center, Durham, North Carolina, USA
| | - Thomas B. Clarkson
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - J. Mark Cline
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Giselle C. Meléndez
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
19
|
Barkas F, Sener YZ, Golforoush PA, Kheirkhah A, Rodriguez-Sanchez E, Novak J, Apellaniz-Ruiz M, Akyea RK, Bianconi V, Ceasovschih A, Chee YJ, Cherska M, Chora JR, D'Oria M, Demikhova N, Kocyigit Burunkaya D, Rimbert A, Macchi C, Rathod K, Roth L, Sukhorukov V, Stoica S, Scicali R, Storozhenko T, Uzokov J, Lupo MG, van der Vorst EPC, Porsch F. Advancements in risk stratification and management strategies in primary cardiovascular prevention. Atherosclerosis 2024; 395:117579. [PMID: 38824844 DOI: 10.1016/j.atherosclerosis.2024.117579] [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: 03/20/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 06/04/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of morbidity and mortality worldwide, highlighting the urgent need for advancements in risk assessment and management strategies. Although significant progress has been made recently, identifying and managing apparently healthy individuals at a higher risk of developing atherosclerosis and those with subclinical atherosclerosis still poses significant challenges. Traditional risk assessment tools have limitations in accurately predicting future events and fail to encompass the complexity of the atherosclerosis trajectory. In this review, we describe novel approaches in biomarkers, genetics, advanced imaging techniques, and artificial intelligence that have emerged to address this gap. Moreover, polygenic risk scores and imaging modalities such as coronary artery calcium scoring, and coronary computed tomography angiography offer promising avenues for enhancing primary cardiovascular risk stratification and personalised intervention strategies. On the other hand, interventions aiming against atherosclerosis development or promoting plaque regression have gained attention in primary ASCVD prevention. Therefore, the potential role of drugs like statins, ezetimibe, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, omega-3 fatty acids, antihypertensive agents, as well as glucose-lowering and anti-inflammatory drugs are also discussed. Since findings regarding the efficacy of these interventions vary, further research is still required to elucidate their mechanisms of action, optimize treatment regimens, and determine their long-term effects on ASCVD outcomes. In conclusion, advancements in strategies addressing atherosclerosis prevention and plaque regression present promising avenues for enhancing primary ASCVD prevention through personalised approaches tailored to individual risk profiles. Nevertheless, ongoing research efforts are imperative to refine these strategies further and maximise their effectiveness in safeguarding cardiovascular health.
Collapse
Affiliation(s)
- Fotios Barkas
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.
| | - Yusuf Ziya Sener
- Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | - Azin Kheirkhah
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elena Rodriguez-Sanchez
- Division of Cardiology, Department of Medicine, Department of Physiology, and Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Jan Novak
- 2(nd) Department of Internal Medicine, St. Anne's University Hospital in Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Maria Apellaniz-Ruiz
- Genomics Medicine Unit, Navarra Institute for Health Research - IdiSNA, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ralph Kwame Akyea
- Centre for Academic Primary Care, School of Medicine, University of Nottingham, United Kingdom
| | - Vanessa Bianconi
- Department of Medicine and Surgery, University of Perugia, Italy
| | - Alexandr Ceasovschih
- Internal Medicine Department, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore
| | - Mariia Cherska
- Cardiology Department, Institute of Endocrinology and Metabolism, Kyiv, Ukraine
| | - Joana Rita Chora
- Unidade I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; Universidade de Lisboa, Faculdade de Ciências, BioISI - Biosystems & Integrative Sciences Institute, Lisboa, Portugal
| | - Mario D'Oria
- Division of Vascular and Endovascular Surgery, Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Nadiia Demikhova
- Sumy State University, Sumy, Ukraine; Tallinn University of Technology, Tallinn, Estonia
| | | | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l'institut du Thorax, Nantes, France
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| | - Krishnaraj Rathod
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; Barts Interventional Group, Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Vasily Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - Svetlana Stoica
- "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania; Institute of Cardiovascular Diseases Timisoara, Timisoara, Romania
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Tatyana Storozhenko
- Cardiovascular Center Aalst, OLV Clinic, Aalst, Belgium; Department of Prevention and Treatment of Emergency Conditions, L.T. Malaya Therapy National Institute NAMSU, Kharkiv, Ukraine
| | - Jamol Uzokov
- Republican Specialized Scientific Practical Medical Center of Therapy and Medical Rehabilitation, Tashkent, Uzbekistan
| | | | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336, Munich, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074, Aachen, Germany
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Godbole S, Solomon JL, Johnson M, Srivastava A, Carsons SE, Belilos E, De Leon J, Reiss AB. Treating Cardiovascular Disease in the Inflammatory Setting of Rheumatoid Arthritis: An Ongoing Challenge. Biomedicines 2024; 12:1608. [PMID: 39062180 PMCID: PMC11275112 DOI: 10.3390/biomedicines12071608] [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: 04/29/2024] [Revised: 06/30/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Despite progress in treating rheumatoid arthritis, this autoimmune disorder confers an increased risk of developing cardiovascular disease (CVD). Widely used screening protocols and current clinical guidelines are inadequate for the early detection of CVD in persons with rheumatoid arthritis. Traditional CVD risk factors alone cannot be applied because they underestimate CVD risk in rheumatoid arthritis, missing the window of opportunity for prompt intervention to decrease morbidity and mortality. The lipid profile is insufficient to assess CVD risk. This review delves into the connection between systemic inflammation in rheumatoid arthritis and the premature onset of CVD. The shared inflammatory and immunologic pathways between the two diseases that result in subclinical atherosclerosis and disrupted cholesterol homeostasis are examined. The treatment armamentarium for rheumatoid arthritis is summarized, with a particular focus on each medication's cardiovascular effect, as well as the mechanism of action, risk-benefit profile, safety, and cost. A clinical approach to CVD screening and treatment for rheumatoid arthritis patients is proposed based on the available evidence. The mortality gap between rheumatoid arthritis and non-rheumatoid arthritis populations due to premature CVD represents an urgent research need in the fields of cardiology and rheumatology. Future research areas, including risk assessment tools and novel immunotherapeutic targets, are highlighted.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (J.L.S.); (M.J.); (A.S.); (S.E.C.); (E.B.); (J.D.L.)
| |
Collapse
|
21
|
Muchira JM, Mogos MF, Park C, Logan J, Piano MR. A Mother-Child Dyadic Approach to Evaluating Subclinical Cardiovascular Disease in Young Children: A Feasibility Study. J Cardiovasc Nurs 2024; 39:307-317. [PMID: 36066587 DOI: 10.1097/jcn.0000000000000944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Cardiovascular (CV) risk factors can be transmitted from mothers to their children. However, it is challenging to measure and identify subclinical CV risk in young children using traditional CV risk methods and metrics. OBJECTIVE The purpose of this study was to determine the feasibility of recruiting mother-child dyads and measuring arterial stiffness (pulse wave velocity, augmentation index/pressure), blood pressure (BP), BP circadian pattern, specifically nocturnal BP dipping, and CV health metrics in mothers and in children aged 1 to 5 years. METHODS All BP and arterial stiffness measures were obtained using the noninvasive automated oscillometric Mobil-O-Graph device. Also measured were blood cholesterol level; glucose level; body mass index (BMI); and smoking, diet, and physical activity history. Descriptive statistics were used for assessing recruitment feasibility and Pearson correlations for mother-child associations. RESULTS Thirty-five mother-child dyads completed the protocol. Recruitment reach was 89% and retention rate was 80%. Mothers were 34.3 ± 5.4 years old with a mean systolic BP (SBP) of 114.6 ± 9.5 mm Hg and BMI of 26.0 ± 6.5. Children were 3 ± 1.4 years old with a mean SBP of 103.3 ± 9.4 mm Hg and BMI z -scores of -0.3 ± 1.5. Arterial stiffness parameters were within normal ranges for mothers and children. Twenty-three percent of mothers did not exhibit nocturnal dipping (<10% decrease between day and nighttime SBP). Maternal SBP was positively correlated with child BMI z -scores ( r = 0.42, P = .022) as well as mother-child augmentation pressure ( r = 0.51, P = .010). CONCLUSIONS Our findings support using a mother-child approach and novel noninvasive approaches to assess and target CV risk in mothers and their young children.
Collapse
|
22
|
Islam KN, Nguyen ID, Islam R, Pirzadah H, Malik H. Roles of Hydrogen Sulfide (H2S) as a Potential Therapeutic Agent in Cardiovascular Diseases: A Narrative Review. Cureus 2024; 16:e64913. [PMID: 39156383 PMCID: PMC11330631 DOI: 10.7759/cureus.64913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/20/2024] Open
Abstract
Cardiovascular disease (CVD) stands as one of the leading causes of morbidity and mortality worldwide, and the continued search for novel therapeutics is vital for addressing this global health challenge. Over the past decade, hydrogen sulfide (H₂S) has garnered significant attention in the field of medical research, as it has been proven to be a cardioprotective gaseous signaling molecule. It joins nitric oxide and carbon monoxide as endogenously produced gasotransmitters. As for its mechanism, H₂S functions through the posttranslational addition of a sulfur group to cysteine residues on target proteins in a process called sulfhydration. As a result, the observed physiological effects of H₂S can include vasodilation, anti-apoptosis, anti-inflammation, antioxidant effects, and regulation of ion channels. Various studies have observed the cardioprotective benefits of H₂S in diseases such as myocardial infarction, ischemia-reperfusion injury, cardiac remodeling, heart failure, arrhythmia, and atherosclerosis. In this review, we discuss the mechanisms and therapeutic potential of H₂S in various CVDs.
Collapse
Affiliation(s)
- Kazi N Islam
- Department of Agricultural Research Development Program, Central State University, Wilberforce, USA
| | - Ivan D Nguyen
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Rahib Islam
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Humza Pirzadah
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Hassan Malik
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| |
Collapse
|
23
|
Saleh C. Comment on Bažadona et al. The Interconnection between Carotid Intima-Media Thickness and Obesity: Anthropometric, Clinical and Biochemical Correlations. Medicina 2023, 59, 1512. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:981. [PMID: 38929598 PMCID: PMC11205296 DOI: 10.3390/medicina60060981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
In a recently published paper in Medicina, Bažadona et al [...].
Collapse
|
24
|
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
|
25
|
Ding D, Zhao Y, Jia Y, Niu M, Li X, Zheng X, Chen H. Identification of novel genes associated with atherosclerosis in Bama miniature pig. Animal Model Exp Med 2024; 7:377-387. [PMID: 38720469 PMCID: PMC11228093 DOI: 10.1002/ame2.12412] [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: 12/28/2023] [Accepted: 03/20/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Atherosclerosis is a chronic cardiovascular disease of great concern. However, it is difficult to establish a direct connection between conventional small animal models and clinical practice. The pig's genome, physiology, and anatomy reflect human biology better than other laboratory animals, which is crucial for studying the pathogenesis of atherosclerosis. METHODS We used whole-genome sequencing data from nine Bama minipigs to perform a genome-wide linkage analysis, and further used bioinformatic tools to filter and identify underlying candidate genes. Candidate gene function prediction was performed using the online prediction tool STRING 12.0. Immunohistochemistry and immunofluorescence were used to detect the expression of proteins encoded by candidate genes. RESULTS We mapped differential single nucleotide polymorphisms (SNPs) to genes and obtained a total of 102 differential genes, then we used GO and KEGG pathway enrichment analysis to identify four candidate genes, including SLA-1, SLA-2, SLA-3, and TAP2. nsSNPs cause changes in the primary and tertiary structures of SLA-I and TAP2 proteins, the primary structures of these two proteins have undergone amino acid changes, and the tertiary structures also show slight changes. In addition, immunohistochemistry and immunofluorescence results showed that the expression changes of TAP2 protein in coronary arteries showed a trend of increasing from the middle layer to the inner layer. CONCLUSIONS We have identified SLA-I and TAP2 as potential susceptibility genes of atherosclerosis, highlighting the importance of antigen processing and immune response in atherogenesis.
Collapse
Affiliation(s)
- Dengfeng Ding
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Yuqiong Zhao
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Yunxiao Jia
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Miaomiao Niu
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Xuezhuang Li
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Xinou Zheng
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| | - Hua Chen
- Laboratory Animal CenterChinese PLA General HospitalBeijingChina
| |
Collapse
|
26
|
Rammos A, Bechlioulis A, Kekiopoulou A, Kekiopoulos P, Katsouras CS, Sioka C. Myocardial Perfusion Imaging and C-Reactive Protein in Myocardial Ischemia: A Retrospective Single-Center Study. Life (Basel) 2024; 14:261. [PMID: 38398769 PMCID: PMC10890337 DOI: 10.3390/life14020261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Inflammation is an important mechanism in atherosclerosis and plaque formation. C-reactive protein (CRP) is a common inflammatory biomarker associated with the risk of coronary heart disease. We investigated the relationship of CRP with findings from myocardial perfusion imaging (MPI). METHODS In this retrospective study, 102 consecutive patients (mean age 71 years, 68% males) who underwent MPI (for diagnostic reasons or quantification of myocardial ischemia) and CRP determination (upper limit: 6 mg/L) within 1 month from MPI were included. The patients had no infection or recent acute coronary syndrome. RESULTS The median CRP level was 4 mg/L (2, 10) among the study population. Patients with raised CRP had higher summed stress score (SSS) (p = 0.006) and summed rest score (SRS) (p = 0.001) and higher risk for SSS > 3 (OR 9.25, 95% CI 2.03-42.13, p = 0.001) compared to those with low CRP. The association of SSS and SRS with CRP levels was more evident in patients over 70 years (p = 0.027 and p = 0.005, respectively). No significant difference in summed difference score was shown. The two groups had no difference in other risk factors (p > 0.05 for all comparisons). CONCLUSION a high level of CRP was associated with the presence and extent of stress-induced myocardial ischemia in MPI.
Collapse
Affiliation(s)
- Aidonis Rammos
- 2nd Department of Cardiology, University Hospital of Ioannina, 45110 Ioannina, Greece
| | - Aris Bechlioulis
- 2nd Department of Cardiology, University Hospital of Ioannina, 45110 Ioannina, Greece
| | - Areti Kekiopoulou
- Department of Nuclear Medicine, University Hospital of Ioannina, 45110 Ioannina, Greece
| | - Pavlos Kekiopoulos
- Department of Nuclear Medicine, University Hospital of Ioannina, 45110 Ioannina, Greece
| | - Christos S. Katsouras
- 2nd Department of Cardiology, University Hospital of Ioannina, 45110 Ioannina, Greece
| | - Chrissa Sioka
- Department of Nuclear Medicine, University Hospital of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
27
|
Khatiwada N, Hong Z. Potential Benefits and Risks Associated with the Use of Statins. Pharmaceutics 2024; 16:214. [PMID: 38399268 PMCID: PMC10892755 DOI: 10.3390/pharmaceutics16020214] [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: 10/30/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
HMG-CoA reductase inhibitors, commonly known as statins, are the primary treatment choice for cardiovascular diseases, which stand as the leading global cause of mortality. Statins also offer various pleiotropic effects, including improved endothelial function, anti-inflammatory properties, reduced oxidative stress, anti-thrombotic effects, and the stabilization of atherosclerotic plaques. However, the usage of statins can be accompanied by a range of adverse effects, such as the development of type 2 diabetes mellitus, muscular symptoms, liver toxicity, kidney diseases, cataracts, hemorrhagic strokes, and psychiatric complications. These issues are referred to as statin-associated symptoms (SAS) and are relatively infrequent in clinical trials, making it challenging to attribute them to statin use definitively. Therefore, these symptoms can lead to significant problems, necessitating dose adjustments or discontinuation of statin therapy. This review aims to provide a comprehensive overview of the mechanism of action, potential advantages, and associated risks of statin utilization in clinical settings.
Collapse
Affiliation(s)
| | - Zhongkui Hong
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX 79409, USA;
| |
Collapse
|
28
|
Xing L, Kong F, Wang C, Li L, Peng S, Wang D, Li C. The amelioration of a purified Pleurotus abieticola polysaccharide on atherosclerosis in ApoE -/- mice. Food Funct 2024; 15:79-95. [PMID: 38031758 DOI: 10.1039/d3fo02740f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
In this study, a polysaccharide known as PAPS2 was eluted from Pleurotus abieticola fruiting bodies using 0.1 M NaCl solutions. PAPS2 has a Mw of 19.64 kDa and its backbone is mainly composed of →6)-α-D-Galp-(1→, →6)-β-D-Glcp-(1→ and →2,6)-α-D-Galp-(1→ residues, and its branches mainly end with β-D-Manp-(1→, which is attached at C2 of →2,6)-α-D-Galp-(1→. PAPS2 elicited several effects in high-fat diet (HFD)-fed ApoE-/- mice. It significantly reduced the body weight, liver index, and serum levels of total cholesterol (TC) and triglycerides (TGs), and it alleviated lipid accumulation in the aorta. Intestinal microflora analysis showed that PAPS2 suppressed the abundances of Adlercreutzia, Turicibacter, and Helicobacter and enriched that of Roseburia. It also influenced lipid metabolism, suggesting that it reduced the levels of TGs, lysophosphatidylcholine (LPC), phosphatidylcholine (PC), and ceramide (Cer). Moreover, it suppressed oxidative response by increasing nuclear factor erythroid 2 (Nrf2)-related factor expression and activating the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) to reduce the level of reactive oxygen species (ROS). Meanwhile, it showed anti-inflammatory effects partially related to the inhibition of toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) signaling induced by lipopolysaccharide (LPS) in RAW 264.7 cells, as well as in the aorta of HFD-fed ApoE-/- mice. This study provides experimental evidence of the auxiliary applicability of PAPS2 in atherosclerosis treatment.
Collapse
Affiliation(s)
- Lei Xing
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
- College of Plant Protection, Jilin Agricultural University, Changchun, 130118, China
| | - Fange Kong
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
- College of Plant Protection, Jilin Agricultural University, Changchun, 130118, China
| | - Chunxia Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
- College of Plant Protection, Jilin Agricultural University, Changchun, 130118, China
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
- College of Plant Protection, Jilin Agricultural University, Changchun, 130118, China
| | - Shichao Peng
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
- College of Plant Protection, Jilin Agricultural University, Changchun, 130118, China
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
| | - Changtian Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
29
|
Grant JK, Orringer CE. Coronary and Extra-coronary Subclinical Atherosclerosis to Guide Lipid-Lowering Therapy. Curr Atheroscler Rep 2023; 25:911-920. [PMID: 37971683 DOI: 10.1007/s11883-023-01161-8] [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] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW To discuss and review the technical considerations, fundamentals, and guideline-based indications for coronary artery calcium scoring, and the use of other non-invasive imaging modalities, such as extra-coronary calcification in cardiovascular risk prediction. RECENT FINDINGS The most robust evidence for the use of CAC scoring is in select individuals, 40-75 years of age, at borderline to intermediate 10-year ASCVD risk. Recent US recommendations support the use of CAC scoring in varying clinical scenarios. First, in adults with very high CAC scores (CAC ≥ 1000), the use of high-intensity statin therapy and, if necessary, guideline-based add-on LDL-C lowering therapies (ezetimibe, PCSK9-inhibitors) to achieve a ≥ 50% reduction in LDL-C and optimally an LDL-C < 70 mg/dL is recommended. In patients with a CAC score ≥ 100 at low risk of bleeding, the benefits of aspirin use may outweigh the risk of bleeding. Other applications of CAC scoring include risk estimation on non-contrast CT scans of the chest, risk prediction in younger patients (< 40 years of age), its value as a gatekeeper for the decision to perform nuclear stress testing, and to aid in risk stratification in patients presenting with low-risk chest pain. There is a correlation between extra-coronary calcification (e.g., breast arterial calcification, aortic calcification, and aortic valve calcification) and incident ASCVD events. However, its role in informing lipid management remains unclear. Identification of coronary calcium in selected patients is the single best non-invasive imaging modality to identify future ASCVD risk and inform lipid-lowering therapy decision-making.
Collapse
Affiliation(s)
- Jelani K Grant
- Johns Hopkins Ciccarone Center for the Prevention of Heart Disease, Baltimore, MD, USA
| | - Carl E Orringer
- NCH Rooney Heart Institute, 399 9th Street North, Suite 300, Naples, FL, 34102, USA.
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Francis GA. The Greatly Under-Represented Role of Smooth Muscle Cells in Atherosclerosis. Curr Atheroscler Rep 2023; 25:741-749. [PMID: 37665492 PMCID: PMC10564813 DOI: 10.1007/s11883-023-01145-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
PURPOSE OF REVIEW This article summarizes previous and recent research on the fundamental role of arterial smooth muscle cells (SMCs) as drivers of initial and, along with macrophages, later stages of human atherosclerosis. RECENT FINDINGS Studies using human tissues and SMC lineage-tracing mice have reinforced earlier observations that SMCs drive initial atherogenesis in humans and contribute a multitude of phenotypes including foam cell formation hitherto attributed primarily to macrophages in atherosclerosis. Arterial smooth muscle cells (SMCs) are the primary cell type in human pre-atherosclerotic intima and are responsible for the retention of lipoproteins that drive the development of atherosclerosis. Despite this, images of atherogenesis still depict the process as initially devoid of SMCs, primarily macrophage driven, and indicate only relatively minor roles such as fibrous cap formation to intimal SMCs. This review summarizes historical and recent observations regarding the importance of SMCs in the formation of a pre-atherosclerotic intima, initial and later foam cell formation, and the phenotypic changes that give rise to multiple different roles for SMCs in human and mouse lesions. Potential SMC-specific therapies in atherosclerosis are presented.
Collapse
Affiliation(s)
- Gordon A Francis
- Centre for Heart Lung Innovation, Providence Research, St. Paul's Hospital, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
33
|
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
|
34
|
Wei J, Zhang Y, Li H, Wang F, Yao S. Toll-like receptor 4: A potential therapeutic target for multiple human diseases. Biomed Pharmacother 2023; 166:115338. [PMID: 37595428 DOI: 10.1016/j.biopha.2023.115338] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
The immune response plays a pivotal role in the pathogenesis of diseases. Toll-like receptor 4 (TLR4), as an intrinsic immune receptor, exhibits widespread in vivo expression and its dysregulation significantly contributes to the onset of various diseases, encompassing cardiovascular disorders, neoplastic conditions, and inflammatory ailments. This comprehensive review centers on elucidating the architectural and distributive characteristics of TLR4, its conventional signaling pathways, and its mode of action in diverse disease contexts. Ultimately, this review aims to propose novel avenues and therapeutic targets for clinical intervention.
Collapse
Affiliation(s)
- Jinrui Wei
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Yan Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Haopeng Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Fuquan Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, China.
| |
Collapse
|
35
|
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
|
36
|
Koh JH, Lee BW, Kim WU. Changes in the cholesterol profile of patients with rheumatoid arthritis treated with biologics or Janus kinase inhibitors. JOURNAL OF RHEUMATIC DISEASES 2023; 30:234-242. [PMID: 37736594 PMCID: PMC10509638 DOI: 10.4078/jrd.2023.0030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 09/23/2023]
Abstract
Objective To assess the effects of biological and targeted synthetic disease-modifying antirheumatic drugs (DMARDs) on lipid profiles in patients with moderate-to-severe rheumatoid arthritis (RA). Methods This retrospective single-center observational study included patients with RA taking a tumor necrosis factor-α inhibitor (TNFi), abatacept, tocilizumab, or a Janus kinase inhibitor (JAKi) for at least 6 months. Changes in lipid profile were assessed at 6 months after the start of treatment, and associations between changes in lipid profiles and clinical efficacy, concomitant medications, and comorbidities were evaluated. Results This study included 114 patients treated with TNFi, 81 with abatacept, 103 with tocilizumab, and 89 with JAKi. The mean percentage change (from baseline to 6 months) in total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and non-HDL-C levels was higher in those taking tocilizumab and JAKi than in those taking TNFi and abatacept. A significant change in non-HDL-C was associated with JAKi (versus TNFi odds ratio [OR], 3.228; 95% confidence interval [CI], 1.536~6.785), tocilizumab (versus TNFi OR, 2.203; 95% CI, 1.035~4.689), and statins (OR, 0.487; 95% CI, 0.231~1.024). However, changes in disease activity in 28 joints were not associated with a significant change in non-HDL-C. Conclusion Tocilizumab- and JAKi-associated increases in serum non-HDL-C levels were observed regardless of changes in disease activity. Statins are recommended for RA patients showing a significant increase in cholesterol levels after initiating biological and targeted synthetic DMARDs.
Collapse
Affiliation(s)
- Jung Hee Koh
- Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bong-Woo Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
González‐López P, Álvarez‐Villarreal M, Ruiz‐Simón R, López‐Pastor AR, de Ceniga MV, Esparza L, Martín‐Ventura JL, Escribano Ó, Gómez‐Hernández A. Role of miR-15a-5p and miR-199a-3p in the inflammatory pathway regulated by NF-κB in experimental and human atherosclerosis. Clin Transl Med 2023; 13:e1363. [PMID: 37605307 PMCID: PMC10442475 DOI: 10.1002/ctm2.1363] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/26/2023] [Accepted: 08/05/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) prevalence has significantly increased in the last decade and atherosclerosis development is the main trigger. MicroRNAs (miRNAs) are non-coding RNAs that negatively regulate gene expression of their target and their levels are frequently altered in CVDs. METHODS By RT-qPCR, we analysed miR-9-5p, miR-15a-5p, miR-16-5p and miR-199a-3p levels in aorta from apolipoprotein knockout (ApoE-/- ) mice, an experimental model of hyperlipidemia-induced atherosclerosis, and in human aortic and carotid atherosclerotic samples. By in silico studies, Western blot analysis and immunofluorescence studies, we detected the targets of the altered miRNAs. RESULTS Our results show that miR-15a-5p and miR-199a-3p are significantly decreased in carotid and aortic samples from patients and mice with atherosclerosis. In addition, we found an increased expression in targets of both miRNAs that participate in the inflammatory pathway of nuclear factor kappa B (NF-κB), such as IKKα, IKKβ and p65. In human vein endothelial cells (HUVECs) and vascular smooth muscle cells (VSMCs), the overexpression of miR-15a-5p or miR-199a-3p decreased IKKα, IKKβ and p65 protein levels as well as NF-κB activation. On the other hand, miR-15a-5p and miR-199a-3p overexpression reduced ox-LDL uptake and the inflammation regulated by NF-κB in VSMCs. Moreover, although miR-15a-5p and miR-199a-3p were significantly increased in exosomes from patients with advanced carotid atherosclerosis, only in the ROC analyses for miR-15a-5p, the area under the curve was 0.8951 with a p value of .0028. CONCLUSIONS Our results suggest that the decrease of miR-199a-3p and miR-15a-5p in vascular samples from human and experimental atherosclerosis could be involved in the NF-κB activation pathway, as well as in ox-LDL uptake by VSMCs, contributing to inflammation and progression atherosclerosis. Finally, miR-15a-5p could be used as a novel diagnostic biomarker for advanced atherosclerosis.
Collapse
Affiliation(s)
- Paula González‐López
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Marta Álvarez‐Villarreal
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Rubén Ruiz‐Simón
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Andrea R. López‐Pastor
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Melina Vega de Ceniga
- Department of Angiology and Vascular SurgeryHospital of Galdakao‐UsansoloGaldakaoBizkaiaSpain
- Biocruces Bizkaia Health Research InstituteBarakaldoBizkaiaSpain
| | - Leticia Esparza
- Department of Angiology and Vascular SurgeryHospital of Galdakao‐UsansoloGaldakaoBizkaiaSpain
- Biocruces Bizkaia Health Research InstituteBarakaldoBizkaiaSpain
| | | | - Óscar Escribano
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Almudena Gómez‐Hernández
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| |
Collapse
|
40
|
Ho TWW, Henry A, Lee WL. LDL Transcytosis by the Arterial Endothelium-Atherosclerosis by a Thousand Cuts? Curr Atheroscler Rep 2023; 25:457-465. [PMID: 37358804 DOI: 10.1007/s11883-023-01118-x] [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] [Accepted: 06/01/2023] [Indexed: 06/27/2023]
Abstract
PURPOSE OF REVIEW The accumulation of LDL in the arterial intima is an initiating event in atherosclerosis. After decades of controversy, it is now clear that transcytosis of LDL across an intact endothelial monolayer contributes to its intimal deposition. We review recent observations in this field and address the question of whether LDL transcytosis can be manipulated therapeutically. RECENT FINDINGS The development of a live-cell imaging method for studying transcytosis using total internal reflection fluorescence (TIRF) microscopy has catalyzed recent discoveries. LDL transcytosis is mediated by SR-BI and ALK1. Estrogen down-regulates SR-BI and inhibits LDL transcytosis, while the nuclear structural protein HMGB1 promotes LDL transcytosis. LDL transcytosis by ALK1 is independent of the receptor's kinase activity and is antagonized by BMP9, ALK1's canonical ligand. Inflammation stimulates LDL transcytosis. Identifying the function and mechanisms of LDL transcytosis may ultimately permit its therapeutic manipulation.
Collapse
Affiliation(s)
- Tse Wing Winnie Ho
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Canada
| | - Andria Henry
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Warren L Lee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Canada.
- Department of Biochemistry, University of Toronto, Toronto, Canada.
- Department of Medicine and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada.
- St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON, M5B 1W8, Canada.
| |
Collapse
|
41
|
van Dijk RA, Kleemann R, Schaapherder AF, van den Bogaerdt A, Hedin U, Matic L, Lindeman JH. Validating human and mouse tissues commonly used in atherosclerosis research with coronary and aortic reference tissue: similarities but profound differences in disease initiation and plaque stability. JVS Vasc Sci 2023; 4:100118. [PMID: 37810738 PMCID: PMC10551657 DOI: 10.1016/j.jvssci.2023.100118] [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: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 10/10/2023] Open
Abstract
Objective Characterization of the atherosclerotic process fully relies on histological evaluation and staging through a consensus grading system. So far, a head-to-head comparison of atherosclerotic process in experimental models and tissue resources commonly applied in atherosclerosis research with the actual human atherosclerotic process is missing. Material and Methods Aspects of the atherosclerotic process present in established murine atherosclerosis models and human carotid endarterectomy specimen were systematically graded using the modified American Heart Association histological classification (Virmani classification). Aspects were aligned with the atherosclerotic process observed in human coronary artery and aortic atherosclerosis reference tissues that were available through biobanks based on human tissue/organ donor material. Results Apart from absent intraplaque hemorrhages in aortic lesions, the histological characteristics of the different stages of human coronary and aortic atherosclerosis are similar. Carotid endarterectomy samples all represent end-stage "fibrous calcified plaque" lesions, although secondary, progressive, and vulnerable lesions with gross morphologies similar to coronary/aortic lesions occasionally present along the primary lesions. For the murine lesions, clear histological parallels were observed for the intermediate lesion types ("pathological intimal thickening," and "early fibroatheroma"). However, none of the murine lesions studied progressed to an equivalent of late fibroatheroma or beyond. Notable contrasts were observed for disease initiation: whereas disease initiation in humans is characterized by a mesenchymal cell influx in the intima, the earliest murine lesions are exclusively intimal, with subendothelial accumulation foam cells. A mesenchymal (and medial) response are absent. In fact, it is concluded that the stage of "adaptive intimal thickening" is absent in all mouse models included in this study. Conclusions The Virmani classification for coronary atherosclerosis can be applied for systematically grading experimental and clinical atherosclerosis. Application of this histological grading tool shows clear parallels for intermediate human and murine atherosclerotic lesions. However, clear contrasts are observed for disease initiation, and late stage atherosclerotic lesions. Carotid endarterectomy all represent end-stage fibrous calcified plaque lesions, although secondary earlier lesions may present in a subset of samples.
Collapse
Affiliation(s)
- Rogier A. van Dijk
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Kleemann
- The Netherlands Organization for Applied Scientific Research (TNO), Department of Metabolic Health Research, TNO Metabolic Health Research, Leiden, The Netherlands
| | | | | | - Ulf Hedin
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
| | - Ljubica Matic
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
| | - Jan H.N. Lindeman
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
42
|
Wang B, Liang B, Huang Y, Li Z, Zhang B, Du J, Ye R, Xian H, Deng Y, Xiu J, Yang X, Ichihara S, Ichihara G, Zhong Y, Huang Z. Long-Chain Acyl Carnitines Aggravate Polystyrene Nanoplastics-Induced Atherosclerosis by Upregulating MARCO. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205876. [PMID: 37144527 PMCID: PMC10323628 DOI: 10.1002/advs.202205876] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 04/10/2023] [Indexed: 05/06/2023]
Abstract
Exposure to micro- and nanoplastics (MNPs) is common because of their omnipresence in environment. Recent studies have revealed that MNPs may cause atherosclerosis, but the underlying mechanism remains unclear. To address this bottleneck, ApoE-/- mice are exposed to 2.5-250 mg kg-1 polystyrene nanoplastics (PS-NPs, 50 nm) by oral gavage with a high-fat diet for 19 weeks. It is found that PS-NPs in blood and aorta of mouse exacerbate the artery stiffness and promote atherosclerotic plaque formation. PS-NPs activate phagocytosis of M1-macrophage in the aorta, manifesting as upregulation of macrophage receptor with collagenous structure (MARCO). Moreover, PS-NPs disrupt lipid metabolism and increase long-chain acyl carnitines (LCACs). LCAC accumulation is attributed to the PS-NP-inhibited hepatic carnitine palmitoyltransferase 2. PS-NPs, as well as LCACs alone, aggravate lipid accumulation via upregulating MARCO in the oxidized low-density lipoprotein-activated foam cells. Finally, synergistic effects of PS-NPs and LCACs on increasing total cholesterol in foam cells are found. Overall, this study indicates that LCACs aggravate PS-NP-induced atherosclerosis by upregulating MARCO. This study offers new insight into the mechanisms underlying MNP-induced cardiovascular toxicity, and highlights the combined effects of MNPs with endogenous metabolites on the cardiovascular system, which warrant further study.
Collapse
Affiliation(s)
- Bo Wang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Boxuan Liang
- Affiliated Dongguan People's HospitalSouthern Medical UniversityDongguan523059China
| | - Yuji Huang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Zhiming Li
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Bingli Zhang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Jiaxin Du
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Rongyi Ye
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Hongyi Xian
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Yanhong Deng
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Jiancheng Xiu
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Xingfen Yang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Sahoko Ichihara
- Department of Environmental and Preventive MedicineSchool of MedicineJichi Medical UniversityTochigi329‐0498Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental HealthFaculty of Pharmaceutical SciencesTokyo University of ScienceNoda278‐8510Japan
| | - Yizhou Zhong
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| | - Zhenlie Huang
- NMPA Key Laboratory for Safety Evaluation of CosmeticsGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
43
|
Driban M, Kedia N, Arora S, Chhablani J. Novel pharmaceuticals for the management of retinal vein occlusion and linked disorders. Expert Rev Clin Pharmacol 2023; 16:1125-1139. [PMID: 37933706 DOI: 10.1080/17512433.2023.2277882] [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: 08/09/2023] [Accepted: 10/27/2023] [Indexed: 11/08/2023]
Abstract
INTRODUCTION Retinal vein occlusion (RVO) is the second leading cause of blindness from retinal vascular disease behind diabetic retinopathy. Anti-vascular endothelial growth factor (VEGF) and glucocorticoid therapy are the cornerstones of pharmaceutical treatment for RVO. There is considerable interest in developing new pharmaceuticals in and out of these two classes to reduce costs, lower injection burden, and treat the occlusion itself, rather than the complications. AREAS COVERED In this review, we discuss novel pharmaceuticals for the treatment of RVO outside of current standard of care. We performed a comprehensive literature search encompassing pharmaceuticals that have recently been approved or have shown promising results in early clinical trials or animal models. EXPERT OPINION Anti-VEGF therapy remains the most efficacious treatment for RVO with a very favorable side effect profile. New biosimilars reduce costs while maintaining efficacy. Novel glucocorticoids may be a useful therapy in patients for whom anti-VEGF therapy has failed, or as an adjunct. Pharmaceuticals in other drug classes, particularly those with neuroprotective or regenerative properties, as well as those geared toward treating the occlusion itself, represent exciting options for early RVO therapy, but are likely years away from clinical relevance.
Collapse
Affiliation(s)
- Matthew Driban
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nikita Kedia
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Supriya Arora
- Bahamas Vision Center and Princess Margaret Hospital, Nassau, New Providence, Bahamas
| | - Jay Chhablani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
44
|
Allalou A, Peng J, Robinson GA, Marruganti C, D’Aiuto F, Butler G, Jury EC, Ciurtin C. Impact of puberty, sex determinants and chronic inflammation on cardiovascular risk in young people. Front Cardiovasc Med 2023; 10:1191119. [PMID: 37441710 PMCID: PMC10333528 DOI: 10.3389/fcvm.2023.1191119] [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: 03/21/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
Worrying trends of increased cardiovascular disease (CVD) risk in children, adolescents and young people in the Modern Era have channelled research and public health strategies to tackle this growing epidemic. However, there are still controversies related to the dynamic of the impact of sex, age and puberty on this risk and on cardiovascular health outcomes later in life. In this comprehensive review of current literature, we examine the relationship between puberty, sex determinants and various traditional CVD-risk factors, as well as subclinical atherosclerosis in young people in general population. In addition, we evaluate the role of chronic inflammation, sex hormone therapy and health-risk behaviours on augmenting traditional CVD-risk factors and health outcomes, ultimately aiming to determine whether tailored management strategies for this age group are justified.
Collapse
Affiliation(s)
- Amal Allalou
- University College London Medical School, University College London, London, United Kingdom
| | - Junjie Peng
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London, United Kingdom
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - George A. Robinson
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London, United Kingdom
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Crystal Marruganti
- Eastman Dental Hospital, University College London Hospital, London, United Kingdom
| | - Francesco D’Aiuto
- Eastman Dental Hospital, University College London Hospital, London, United Kingdom
| | - Gary Butler
- Department of Paediatric Endocrinology, University College London Hospital, London, United Kingdom
- Institute of Child Health, University College London, London, United Kingdom
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London, United Kingdom
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
45
|
German CA, Liao JK. Understanding the molecular mechanisms of statin pleiotropic effects. Arch Toxicol 2023; 97:1529-1545. [PMID: 37084080 PMCID: PMC10119541 DOI: 10.1007/s00204-023-03492-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/30/2023] [Indexed: 04/22/2023]
Abstract
Statins represent the cornerstone of pharmacotherapy for the prevention of atherosclerotic cardiovascular disease. These medications not only reduce low-density lipoprotein cholesterol (LDL-C) via inhibition of 3-hydroxy-3-methylglutarate attached to CoA reductase, the key rate-limiting step in the cholesterol biosynthetic pathway, but also upregulate expression of the low-density lipoprotein receptor, improving serum clearance. Given LDL-C is a causal risk factor for the development of atherosclerosis, these complementary mechanisms largely explain why statin therapy leads to reductions in major adverse cardiovascular events. However, decades of basic and clinical research have suggested that statins may exert other effects independent of LDL-C lowering, termed pleiotropic effects, which have become a topic of debate among the scientific community. While some literature suggests statins may improve plaque stability, reduce inflammation and thrombosis, decrease oxidative stress, and improve endothelial function and vascular tone, other studies have suggested potential harmful pleiotropic effects related to increased risk of muscle-related side effects, diabetes, hemorrhagic stroke, and cognitive decline. Furthermore, the introduction of newer, non-statin LDL-C lowering therapies, including ezetimibe, proprotein convertase subtilisin/Kexin Type 9, and bempedoic acid, have challenged the statin pleiotropy theory. This review aims to provide a historical background on the development of statins, explore the mechanistic underpinnings of statin pleiotropy, review the available literature, and provide up to date examples that suggest statins may exert effects outside of LDL-C lowering and the cardiovascular system.
Collapse
Affiliation(s)
- Charles A German
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA.
| | - James K Liao
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
46
|
Torun SG, Munoz PDM, Crielaard H, Verhagen HJM, Kremers GJ, van der Steen AFW, Akyildiz AC. Local Characterization of Collagen Architecture and Mechanical Failure Properties of Fibrous Plaque Tissue of Atherosclerotic Human Carotid Arteries. Acta Biomater 2023; 164:293-302. [PMID: 37086826 DOI: 10.1016/j.actbio.2023.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/24/2023]
Abstract
Atherosclerotic plaque rupture in carotid arteries is a major cause of cerebrovascular events. Plaque rupture is the mechanical failure of the heterogeneous fibrous plaque tissue. Local characterization of the tissue's failure properties and the collagen architecture are of great importance to have insights in plaque rupture for clinical event prevention. Previous studies were limited to average rupture properties and global structural characterization, and did not provide the necessary local information. In this study, we assessed the local collagen architecture and failure properties of fibrous plaque tissue, by analyzing 30 tissue strips from 18 carotid plaques. Our study framework entailed second harmonic generation imaging for local collagen orientation and dispersion, and uniaxial tensile testing and digital image correlation for local tissue mechanics. The results showed that 87% of the imaged locations had collagen orientation close to the circumferential direction (0°) of the artery, and substantial dispersion locally. All regions combined, median [Q1:Q3] of the predominant angle measurements was -2° [-16°:16°]. The stretch ratio measurements clearly demonstrated a nonuniform stretch ratio distribution in the tissue under uniaxial loading. The rupture initiation regions had significantly higher stretch ratios (1.26 [1.15-1.40]) than the tissue average stretch ratio (1.11 [1.10-1.16]). No significant difference in collagen direction and dispersion was identified between the rupture regions and the rest of the tissue. The presented study forms an initial step towards gaining better insights into the characterization of local structural and mechanical fingerprints of fibrous plaque tissue in order to aid improved assessment of plaque rupture risk. STATEMENT OF SIGNIFICANCE: Plaque rupture risk assessment, critical to prevent cardiovascular events, requires knowledge on local failure properties and structure of collagenous plaque tissue. Our current knowledge is unfortunately limited to tissue's overall ultimate failure properties with scarce information on collagen architecture. In this study, local failure properties and collagen architecture of fibrous plaque tissue were obtained. We found predominant circumferential alignment of collagen fibers with substantial local dispersion. The tissue showed nonuniform stretch distribution under uniaxial tensile loading, with high stretches at rupture spots. This study highlights the significance of local mechanical and structural assessment for better insights into plaque rupture and the potential use of local stretches as risk marker for plaque rupture for patient-specific clinical applications.
Collapse
Affiliation(s)
- Su Guvenir Torun
- Department of Biomedical Engineering, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Pablo de Miguel Munoz
- Department of Biomedical Engineering, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| | - Hanneke Crielaard
- Department of Biomedical Engineering, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hence J M Verhagen
- Department of Vascular Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gert-Jan Kremers
- Erasmus Optical Imaging Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Antonius F W van der Steen
- Department of Biomedical Engineering, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| | - Ali C Akyildiz
- Department of Biomedical Engineering, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands.
| |
Collapse
|
47
|
Liu Y. Mathematical modeling and simulation of atherosclerotic formation and progress at arterial bifurcations. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2022.104266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
48
|
Li W, Luo J, Peng F, Liu R, Bai X, Wang T, Zhang X, Zhu J, Li XY, Wang Z, Liu W, Wang J, Zhang L, Chen X, Xue T, Ding C, Wang C, Jiao L. Spatial metabolomics identifies lipid profiles of human carotid atherosclerosis. Atherosclerosis 2023; 364:20-28. [PMID: 36459728 DOI: 10.1016/j.atherosclerosis.2022.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/31/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS Carotid atherosclerosis is an important cause of ischemic stroke. Lipids play a key role in the progression of atherosclerosis. To date, the spatial lipid profile of carotid atherosclerotic plaques related to histology has not been systematically investigated. METHODS Carotid atherosclerosis samples from 12 patients were obtained and classified into four classical pathological stages (preatheroma, atheroma, fibroatheroma and complicated lesion) by histological staining. Desorption electrospray ionization-mass spectrometry imaging (DESI-MSI) was used to investigate the lipid profile of carotid atherosclerosis, and correlated it with histological information. Bioinformatics technology was used to process MSI data among different pathological stages of atherosclerosis lesions. RESULTS A total of 55 lipids (26 throughout cross-section regions [TCSRs], 13 in lipid-rich regions [LRRs], and 16 in collagen-rich regions [CRRs]) were initially identified in carotid plaque from one patient. Subsequently, 32 of 55 lipids (12 in TCSRs, eight in LRRs, and 12 in CRRs) were further screened in 11 patients. Pathway enrichment analysis showed that multiple metabolic pathways, such as fat digestion and absorption, cholesterol metabolism, lipid and atherosclerosis, were enriched in TCSRs; sphingolipid signaling pathway, necroptosis pathway were enriched in LRRs; and glycerophospholipid metabolism, ether lipid metabolism pathway were mainly enriched in CRRs. CONCLUSIONS This study comprehensively showed the spatial lipid metabolism footprint in human carotid atherosclerotic plaques. The lipid profiles and related metabolism pathways in three regions of plaque with disease progression were different markedly, suggesting that the different metabolic mechanisms in these regions of carotid plaque may be critical in atherosclerosis progression.
Collapse
Affiliation(s)
- Wei Li
- Department of Stroke Center, Central Hospital Affiliated to Shandong First Medical University, China; Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Fangda Peng
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China
| | - Ruiting Liu
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Xuesong Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Junge Zhu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xu-Ying Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zhanjun Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wubin Liu
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Xianyang Chen
- Zhongguancun Biological and Medical Big Data Center, Beijing, China; BaoFeng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Teng Xue
- BaoFeng Key Laboratory of Genetics and Metabolism, Beijing, China; Zhongyuanborui Key Laborotory of Genetics and Metabolism, Guangdong-Macao In-depth Cooperation Zone in Hengqin, China
| | - Chunguang Ding
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China.
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; National Clinical Research Center for Geriatric Diseases, Beijing, China.
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Beijing, 100053, China; Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
49
|
Jiménez-Castilla L, Opazo-Ríos L, Marin-Royo G, Orejudo M, Rodrigues-Diez R, Ballesteros-Martínez C, Soto-Catalán M, Caro-Ordieres T, Artaiz I, Suarez-Cortés T, Zazpe A, Hernández G, Cortés M, Tuñón J, Briones AM, Egido J, Gómez-Guerrero C. The Synthetic Flavonoid Hidrosmin Improves Endothelial Dysfunction and Atherosclerotic Lesions in Diabetic Mice. Antioxidants (Basel) 2022; 11:antiox11122499. [PMID: 36552707 PMCID: PMC9774734 DOI: 10.3390/antiox11122499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
In diabetes, chronic hyperglycemia, dyslipidemia, inflammation and oxidative stress contribute to the progression of macro/microvascular complications. Recently, benefits of the use of flavonoids in these conditions have been established. This study investigates, in two different mouse models of diabetes, the vasculoprotective effects of the synthetic flavonoid hidrosmin on endothelial dysfunction and atherogenesis. In a type 2 diabetes model of leptin-receptor-deficient (db/db) mice, orally administered hidrosmin (600 mg/kg/day) for 16 weeks markedly improved vascular function in aorta and mesenteric arteries without affecting vascular structural properties, as assessed by wire and pressure myography. In streptozotocin-induced type 1 diabetic apolipoprotein E-deficient mice, hidrosmin treatment for 7 weeks reduced atherosclerotic plaque size and lipid content; increased markers of plaque stability; and decreased markers of inflammation, senescence and oxidative stress in aorta. Hidrosmin showed cardiovascular safety, as neither functional nor structural abnormalities were noted in diabetic hearts. Ex vivo, hidrosmin induced vascular relaxation that was blocked by nitric oxide synthase (NOS) inhibition. In vitro, hidrosmin stimulated endothelial NOS activity and NO production and downregulated hyperglycemia-induced inflammatory and oxidant genes in vascular smooth muscle cells. Our results highlight hidrosmin as a potential add-on therapy in the treatment of macrovascular complications of diabetes.
Collapse
Affiliation(s)
- Luna Jiménez-Castilla
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Lucas Opazo-Ríos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad de Las Américas, Concepción-Talcahuano 4301099, Chile
- Correspondence: (L.O.-R.); (R.R.-D.); (C.G.-G.); Tel.: +56-920463280 (L.O.-R.); +34-686826139 (R.R.-D.); +34-915504800 (ext. 3126) (C.G.-G.)
| | - Gema Marin-Royo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
| | - Macarena Orejudo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
| | - Raquel Rodrigues-Diez
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Department of Physiology, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Correspondence: (L.O.-R.); (R.R.-D.); (C.G.-G.); Tel.: +56-920463280 (L.O.-R.); +34-686826139 (R.R.-D.); +34-915504800 (ext. 3126) (C.G.-G.)
| | - Constanza Ballesteros-Martínez
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, 28029 Madrid, Spain
| | - Manuel Soto-Catalán
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
| | - Teresa Caro-Ordieres
- Department of Research, Development, and Innovation, FAES Farma, 48940 Bilbao, Spain
| | - Inés Artaiz
- Department of Research, Development, and Innovation, FAES Farma, 48940 Bilbao, Spain
| | - Tatiana Suarez-Cortés
- Department of Research, Development, and Innovation, FAES Farma, 48940 Bilbao, Spain
| | - Arturo Zazpe
- Department of Research, Development, and Innovation, FAES Farma, 48940 Bilbao, Spain
| | - Gonzalo Hernández
- Department of Research, Development, and Innovation, FAES Farma, 48940 Bilbao, Spain
| | - Marcelino Cortés
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - José Tuñón
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Ana M. Briones
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Carmen Gómez-Guerrero
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
- Correspondence: (L.O.-R.); (R.R.-D.); (C.G.-G.); Tel.: +56-920463280 (L.O.-R.); +34-686826139 (R.R.-D.); +34-915504800 (ext. 3126) (C.G.-G.)
| |
Collapse
|
50
|
Balraj J, Murugesan T, Dhanapal AR, Kalieswaran V, Jairaman K, Archunan G, Jayaraman A. Bioconversion of lovastatin to simvastatin by Streptomyces carpaticus toward the inhibition of HMG-CoA activity. Biotechnol Appl Biochem 2022. [PMID: 36524308 DOI: 10.1002/bab.2429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
The aim of this study was the modification of lovastatin by microbes to improve its potential. Actinobacteria exhibit staggering diversity in terms of their biosynthetic capability for specialized metabolites which has been traced back to the presence of specialized gene clusters. The objective of the study is to exploit the potential of Actinobacteria strain(s), which can biotransform lovastatin to simvastatin, which might be a more potent therapeutic agent than lovastatin. We have screened 40 Actinobacteria strains and assessed their biotransformation potential primarily through thin layer chromatography (TLC) analysis, followed by high performance thin layer chromatography and high performance liquid chromatography analysis. One strain C7 (CTL S12) has been identified as a potential Actinobacteria that favored the simvastatin biotransformation. The morphological and biochemical analysis together with 16S rRNA sequencing coupled with phylogenetic analysis confirmed the ideal strain (C7) as Streptomyces carpaticus. Successively, the purified simvastatin from S. carpaticus was characterized by liquid chromatography-mass spectrometry (LC-MS), infrared spectrometry, nuclear magnetic resonance, and HMG-CoA assay. In the LC-MS analysis, a peak at 419.24 m/z confirmed the elemental composition of simvastatin (C25 H39 O5 ). In HMG-CoA assay, the IC50 of simvastatin was 50 μg/ml, and the inhibitory potential was 1.36 times higher compared to that of lovastatin. Thus, the biotransformation of simvastatin from lovastatin by S. carpaticus is reported for the first time.
Collapse
Affiliation(s)
- Janani Balraj
- Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Thandeeswaran Murugesan
- Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India.,Bharathiar Cancer Theranostics Research Centre, RUSA2.0, 641046, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Anand Raj Dhanapal
- Centre for Plant Tissue Culture and Central Instrumentation Laboratory, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India.,Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Vidhya Kalieswaran
- Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Karunyadevi Jairaman
- Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Angayarkanni Jayaraman
- Department of Microbial Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|