1
|
Todorova VK, Azhar G, Stone A, Malapati SJ, Che Y, Zhang W, Makhoul I, Wei JY. Neutrophil Biomarkers Can Predict Cardiotoxicity of Anthracyclines in Breast Cancer. Int J Mol Sci 2024; 25:9735. [PMID: 39273682 PMCID: PMC11395913 DOI: 10.3390/ijms25179735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Doxorubicin (DOX), a commonly used anticancer agent, causes cardiotoxicity that begins with the first dose and may progress to heart failure years after treatment. An inflammatory response associated with neutrophil recruitment has been recognized as a mechanism of DOX-induced cardiotoxicity. This study aimed to validate mRNA expression of the previously identified biomarkers of DOX-induced cardiotoxicity, PGLYRP1, CAMP, MMP9, and CEACAM8, and to assay their protein expression in the peripheral blood of breast cancer patients. Blood samples from 40 breast cancer patients treated with DOX-based chemotherapy were collected before and after the first chemotherapy cycle and > 2 years after treatment. The protein and gene expression of PGLYRP1/Tag7, CAMP/LL37, MMP9/gelatinase B, and CEACAM8/CD66b were determined using ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of each candidate biomarker. Patients with cardiotoxicity (n = 20) had significantly elevated levels of PGLYRP1, CAMP, MMP9, and CEACAM8 at baseline, after the first dose of DOX-based chemotherapy, and at > 2 years after treatment relative to patients without cardiotoxicity (n = 20). The first dose of DOX induced significantly higher levels of all examined biomarkers in both groups of patients. At > 2 years post treatment, the levels of all but MMP9 dropped below the baseline. There was a good correlation between the expression of mRNA and the target proteins. We demonstrate that circulating levels of PGLYRP1, CAMP, MMP9, and CEACAM8 can predict the cardiotoxicity of DOX. This novel finding may be of value in the early identification of patients at risk for cardiotoxicity.
Collapse
Affiliation(s)
- Valentina K Todorova
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Gohar Azhar
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Annjanette Stone
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Sindhu J Malapati
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yingni Che
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wei Zhang
- Department of Mathematics and Statistics, University of Arkansas at Little Rock, Little Rock, AR 72205, USA
| | - Issam Makhoul
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
2
|
Sorysz D, Dziewierz A, Gawlik K, Opalińska M, Sowa Staszczak A, Grochowska A, Malinowski KP, Maruszak N, Bagieński M, Dudek D. Temporal changes in biomarker levels and their association with the early degeneration stage of transcatheter aortic valves in 18F-fluorodeoxyglucose and 18F-sodium fluoride positron emission tomography studies. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2024; 20:329-337. [PMID: 39464587 PMCID: PMC11506396 DOI: 10.5114/aic.2024.142403] [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: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction As transcatheter aortic valve implantation (TAVI) indications expand, understanding the valve degeneration process and potential influencing biomarkers becomes increasingly important. Aim To investigate temporal changes in biomarker levels and their potential association with 18F-fluorodeoxyglucose (18F-FDG) and 18F-sodium fluoride (18F-NaF) uptake, assessed using positron emission tomography/computed tomography (PET/CT) studies as markers for native aortic annulus calcifications and early-stage TAVI valve degeneration. Material and methods A total of 71 TAVI patients underwent blood sampling and transthoracic echocardiography at baseline (pre-TAVI) and 6, 12, 18, and 24 months after the procedure. PET/CT using 18F-NaF and 18F-FDG was performed at 6 and 24 months. Serum levels of matrix metalloproteinase-3 (MMP-3), matrix metalloproteinase-9 (MMP-9), and osteopontin (OPN) were measured. In addition, plasma levels of osteoprotegerin (OPG), lipoprotein a (Lp(a)), and oxidized LDL (ox-LDL) were assessed. Results Finally, 31 patients (median age: 84.0 years) completed the study. Valve function improved after TAVI and remained stable during follow-up. Over 24 months, OPN levels decreased (p = 0.010), while MMP-3 and MMP-9 levels increased (p = 0.046 and p = 0.041). MMP-3 and MMP-9 showed multiple positive correlations across time points. OPN, ox-LDL, and OPG demonstrated significant negative correlations with follow-up effective orifice area index and effective orifice area (EOA). No significant correlations were found between biomarkers and PET/CT uptake. Conclusions Significant biomarker changes over 24 months and negative correlations with EOA suggest potential roles in aortic valve function. However, no correlations between biomarkers and PET/CT results were observed.
Collapse
Affiliation(s)
- Danuta Sorysz
- 2 Department of Cardiology, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- Clinical Department of Cardiology and Cardiovascular Interventions, University Hospital, Krakow, Poland
| | - Artur Dziewierz
- 2 Department of Cardiology, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- Clinical Department of Cardiology and Cardiovascular Interventions, University Hospital, Krakow, Poland
| | - Katarzyna Gawlik
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Marta Opalińska
- Clinical Department of Endocrinology, Oncological Endocrinology and Nuclear Medicine, University Hospital, Krakow, Poland
- Department of Endocrinology, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Sowa Staszczak
- Clinical Department of Endocrinology, Oncological Endocrinology and Nuclear Medicine, University Hospital, Krakow, Poland
- Department of Endocrinology, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Grochowska
- Department of Radiology, University Hospital, Krakow, Poland
| | - Krzysztof Piotr Malinowski
- Department of Bioinformatics and Telemedicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Digital Medicine and Robotics, Jagiellonian University Medical College, Krakow, Poland
| | - Natalia Maruszak
- 2 Department of Cardiology, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Bagieński
- Intensive Cardiac Care Unit, University Hospital, Krakow, Poland
| | - Dariusz Dudek
- Center for Digital Medicine and Robotics, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
3
|
Qiu Y, Ouyang Z, Zhong J, Jin L, Qin Y, Zeng Y. Syndecan-1 as a predictor of vulnerable atherosclerotic plaques. Front Cell Dev Biol 2024; 12:1415788. [PMID: 39175877 PMCID: PMC11338802 DOI: 10.3389/fcell.2024.1415788] [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: 04/26/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Aims Cardiovascular disease remains a major global health concern, with atherosclerosis (AS) being a significant contributor. Vulnerable plaques play a critical role in acute cardiovascular events. Syndecan-1 (SDC-1), a vital membrane proteoglycan in the vascular endothelial glycocalyx, is believed to be associated with plaque progression. However, its precise relationship with severity and vulnerability of atherosclerotic plaque remains unclear. This study aimed to investigate SDC-1 expression and its potential correlation with plaque vulnerability in ApoE-/- atherosclerosis mouse model. Methods and results Eight-week-old mice were induced into the AS model using a high-fat diet (HFD) and/or partial ligation of the left common carotid artery (PLCA), with a chow diet (CD) control group. After 16 weeks, plaques in the aortic root showed the following order: HFD + PLCA group > HFD group > CD + PLCA group > CD group. Immunohistochemistry revealed heightened accumulation of lipid/foam cells and CD68-labeled macrophages in the plaques, elevated vascular endothelial growth factor (VEGF), and matrix Metalloproteinase-9 (MMP-9) in the HFD + PLCA group's plaques, along with reduced collagen and α-SMA-labeled smooth muscle cells, resulting in the highest vulnerability index value. Immunohistofluorescence analysis of frozen plaque sections showed significantly higher SDC-1 expression in the AS mice group compared to the CD group, both positively correlated with plaque vulnerability. Serum analysis demonstrated elevated levels of SDC1, sphingosine 1-phosphate (S1P), and VEGF-A in the AS mice, all positively correlated with plaque vulnerability. Multivariate analysis identified SDC1 as an independent predictor of plaque vulnerability. Conclusion This study enhances our understanding of plaque vulnerability mechanisms and presents SDC1 as a potential biomarker for atherosclerosis. These findings underscore the importance of addressing modifiable risk factors, such as diet and hemodynamics and suggest the utility of serum SDC1 as a valuable clinical marker. Ultimately, these insights may lead to more effective strategies in combating cardiovascular diseases and improving patient outcomes.
Collapse
Affiliation(s)
- Yan Qiu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China
| | - Zhi Ouyang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jian Zhong
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Singh R, Tasnim S, Chandra S, Pp R, Choudhary A, Dawar R, Goyal P, Meena MK, Bhattacharjee J, Tyagi S. Risk stratification analysis of recurrent myocardial infarction in Indian population using inflammatory, lipid, thrombotic and extracellular matrix remodeling markers. Glob Cardiol Sci Pract 2024; 2024:e202425. [PMID: 39351476 PMCID: PMC11439428 DOI: 10.21542/gcsp.2024.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/22/2024] [Indexed: 10/04/2024] Open
Abstract
OBJECTIVE Atherosclerosis is a chronic condition characterized by impaired lipid homeostasis and chronic inflammatory pathology in large and mid-sized arteries. Myocardial infarction is caused by coronary artery thrombosis in a ruptured or unstable atherosclerotic plaque. Despite the emphasis on known triggering factors, such as hypertension and dyslipidemia, adverse events following MI, such as recurrence and mortality, are still high. Therefore, it is imperative to assess potential determinants of plaque instability. We evaluated markers of inflammation, extracellular matrix (ECM) remodeling, thrombosis, and lipids in first-time and recurrent MI (RMI). METHODS Two hundred patients diagnosed with MI within the first 24 h of the event were included in the study and categorized as first-time or recurrent MI. Serum levels of NF-κB, hs-CRP, TNF-α, IFN γ, IL-6, VCAM-1,MMP-9, stromelysin, TIMP-1, MCP-1, PAPP-A, vWF, D-dimer, PLA2, PON-1, Apo-B, Apo-A1, ox-LDL, and anti-oxidized LDL antibodies were analyzed by ELISA. We performed a multivariate logistic regression analysis for risk stratification. RESULTS The mean age of first-time MI patients was 52.4 ± 25 years and that of recurrent MI patients was 55.9 ± 24.6 years. RMI patients showed significant (p¡0.05) upregulation of markers of inflammation (TNF-α), endothelial adhesion (VCAM-1), ECM remodeling (MMP-9, PAPP-A), and antioxidant PON-1 enzyme. First-time MI patients had significantly higher serum IL-6 and D-dimer levels than RMI patients. Risk categorization for RMI was determined at 0.5 cut-off utilizing proteomic indicators at 95% confidence interval. CONCLUSION Non-lipid factors provide substantial insights into plaque instability. Multiple markers of inflammation, thrombosis, extracellular matrix remodeling, and paroxonase-1 are reliable indicators of recurrent myocardial infarction.
Collapse
Affiliation(s)
- Ritu Singh
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Sana Tasnim
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Sudhir Chandra
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Roshnara Pp
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Ankita Choudhary
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Rajni Dawar
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Parul Goyal
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Mukesh Kumar Meena
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Jayashree Bhattacharjee
- Department of Biochemistry, Lady Hardinge Medical College, Connaught Place, New Delhi, India
| | - Sanjay Tyagi
- Department of Cardiology, GB Pant hospital, Raj Ghat, New Delhi, India
| |
Collapse
|
5
|
Lv F, Fang H, Huang L, Wang Q, Cao S, Zhao W, Zhou Z, Zhou W, Wang X. Curcumin Equipped Nanozyme-Like Metal-Organic Framework Platform for the Targeted Atherosclerosis Treatment with Lipid Regulation and Enhanced Magnetic Resonance Imaging Capability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309062. [PMID: 38696653 PMCID: PMC11234396 DOI: 10.1002/advs.202309062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/21/2024] [Indexed: 05/04/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) has become the leading cause of death worldwide, and early diagnosis and treatment of atherosclerosis (AS) are crucial for reducing the occurrence of acute cardiovascular events. However, early diagnosis of AS is challenging, and oral anti-AS drugs suffer from limitations like imprecise targeting and low bioavailability. To overcome the aforementioned shortcomings, Cur/MOF@DS is developed, a nanoplatform integrating diagnosis and treatment by loading curcumin (Cur) into metal-organic frameworks with nanozymes and magnetic resonance imaging (MRI) properties. In addition, the surface-modification of dextran sulfate (DS) enables PCN-222(Mn) effectively target scavenger receptor class A in macrophages or foam cells within the plaque region. This nanoplatform employs mechanisms that effectively scavenge excessive reactive oxygen species in the plaque microenvironment, promote macrophage autophagy and regulate macrophage polarization to realize lipid regulation. In vivo and in vitro experiments confirm that this nanoplatform has outstanding MRI performance and anti-AS effects, which may provide a new option for early diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Fanzhen Lv
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Huaqiang Fang
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Li Huang
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Qingqing Wang
- School of PharmacyNanchang UniversityNanchangJiangxi330006China
| | - Shuangyuan Cao
- The National Engineering Research Center for Bioengineering Drugs and the TechnologiesInstitute of Translational MedicineNanchang UniversityNanchangJiangxi330006China
| | - Wenpeng Zhao
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Zhibin Zhou
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Weimin Zhou
- Department of Vascular Surgerythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangJiangxi330006China
| | - Xiaolei Wang
- School of PharmacyNanchang UniversityNanchangJiangxi330006China
- The National Engineering Research Center for Bioengineering Drugs and the TechnologiesInstitute of Translational MedicineNanchang UniversityNanchangJiangxi330006China
| |
Collapse
|
6
|
Deng X, Wang J, Yu S, Tan S, Yu T, Xu Q, Chen N, Zhang S, Zhang M, Hu K, Xiao Z. Advances in the treatment of atherosclerosis with ligand-modified nanocarriers. EXPLORATION (BEIJING, CHINA) 2024; 4:20230090. [PMID: 38939861 PMCID: PMC11189587 DOI: 10.1002/exp.20230090] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/08/2023] [Indexed: 06/29/2024]
Abstract
Atherosclerosis, a chronic disease associated with metabolism, poses a significant risk to human well-being. Currently, existing treatments for atherosclerosis lack sufficient efficiency, while the utilization of surface-modified nanoparticles holds the potential to deliver highly effective therapeutic outcomes. These nanoparticles can target and bind to specific receptors that are abnormally over-expressed in atherosclerotic conditions. This paper reviews recent research (2018-present) advances in various ligand-modified nanoparticle systems targeting atherosclerosis by specifically targeting signature molecules in the hope of precise treatment at the molecular level and concludes with a discussion of the challenges and prospects in this field. The intention of this review is to inspire novel concepts for the design and advancement of targeted nanomedicines tailored specifically for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiujiao Deng
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
- Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouChina
| | - Jinghao Wang
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
| | - Shanshan Yu
- Department of PharmacyZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Suiyi Tan
- Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouChina
| | - Tingting Yu
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
| | - Qiaxin Xu
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
| | - Nenghua Chen
- Department of PharmacyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ming‐Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical, ScienceNational Institutes for Quantum Science and TechnologyChibaJapan
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia MedicaChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical, ScienceNational Institutes for Quantum Science and TechnologyChibaJapan
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic DiseasesJinan UniversityGuangzhouChina
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical TranslationJinan UniversityGuangzhouChina
| |
Collapse
|
7
|
Hachuła M, Kosowski M, Basiak M, Okopień B. Influence of Dulaglutide on Serum Biomarkers of Atherosclerotic Plaque Instability: An Interventional Analysis of Cytokine Profiles in Diabetic Subjects-A Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:908. [PMID: 38929525 PMCID: PMC11205508 DOI: 10.3390/medicina60060908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: The rise in global diabetes cases, reaching a staggering 529 million in 2021 from 108 million in 1980, underscores the urgency of addressing its complications, notably macrovascular ones like coronary artery, cerebrovascular, and peripheral artery diseases, which contribute to over 50% of diabetes mortality. Atherosclerosis, linked to hyperglycemia-induced endothelial dysfunction, is pivotal in cardiovascular disease development. Cytokines, including pentraxin 3 (PTX3), copeptin, lipoprotein(a) [Lp(a)], and matrix metalloproteinase-9 (MMP-9), influence atherosclerosis progression and plaque vulnerability. Inhibiting atherosclerosis progression is crucial, especially in diabetic individuals. Glucagon-like peptide 1 receptor agonists (GLP-1 RAs), increasingly used for type 2 diabetes, show promise in reducing the cardiovascular risk, sparking interest in their effects on atherogenesis. This study sought to examine the effects of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) on biomarkers that indicate the instability of atherosclerotic plaques. These biomarkers include pentraxin 3 (PTX3), copeptin (CPC), matrix metalloproteinase-9 (MMP-9), and lipoprotein(a) [Lp(a)]. Materials and Methods: A total of 34 participants, ranging in age from 41 to 81 years (with an average age of 61), who had been diagnosed with type 2 diabetes mellitus (with a median HbA1c level of 8.8%), dyslipidemia, and verified atherosclerosis using B-mode ultrasonography, were included in the study. All subjects were eligible to initiate treatment with a GLP-1 RA-dulaglutide. Results: Significant reductions in anthropometric parameters, blood pressure, fasting glucose levels, and HbA1c levels were observed posttreatment. Moreover, a notable decrease in biochemical markers associated with atherosclerotic plaque instability, particularly PTX3 and MMP-9 (p < 0.001), as well as Lp(a) (p < 0.05), was evident following the GLP-1 RA intervention. Conclusions: These findings underscore the potential of GLP-1 RAs in mitigating atherosclerosis progression and plaque vulnerability, thus enhancing cardiovascular outcomes in individuals with type 2 diabetes mellitus.
Collapse
Affiliation(s)
| | | | - Marcin Basiak
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland; (M.H.); (M.K.); (B.O.)
| | | |
Collapse
|
8
|
Zivkovic M, Stankovic A, Koncar I, Kolakovic A, Boskovic M, Djuric T. The MMP-9 promoter genetic variant rs3918242, mRNA and protein expression in advanced carotid plaque tissue. Mol Biol Rep 2024; 51:540. [PMID: 38642151 DOI: 10.1007/s11033-024-09458-w] [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: 02/21/2024] [Accepted: 03/18/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND The MMP-9 is a known player in atherosclerosis, yet associations of the MMP-9 -1562 C/T variant (rs3918242) with various atherosclerotic phenotypes and tissue mRNA expression are still contradictory. This study aimed to investigate the MMP-9 -1562 C/T variant, its mRNA and protein expression in carotid plaque (CP) tissue, as a risk factor for CP presence and as a marker of different plaque phenotypes (hyperechoic and hypoechoic) in patients undergoing carotid endarterectomy. The MnSOD as an MMP-9 negative regulator was also studied in relation to CP phenotypes. METHODS AND RESULTS Genotyping of 770 participants (285 controls/485 patients) was done by tetra-primer ARMS PCR. The MMP-9 mRNA expression in 88 human CP tissues was detected by TaqMan® technology. The protein levels of MMP-9 and MnSOD were assessed by Western blot analysis. The MMP-9 -1562 C/T variant was not recognized as a risk factor for plaque presence or in predisposing MMP-9 mRNA and protein levels in plaque tissue. Patients with hypoechoic plaques had significantly lower MMP-9 mRNA and protein levels than those with hyperechoic plaque (p = 0.008, p = 0.003, respectively). MnSOD protein level was significantly higher in hypoechoic plaque compared to hyperechoic (p = 0.039). MMP-9 protein expression in CP tissue was significantly affected by sex and plaque type interaction (p = 0.009). CONCLUSIONS Considering the differences of MMP-9 mRNA and protein expression in CP tissue regarding different plaque phenotypes and the observed sex-specific effect, the role of MMP-9 in human atherosclerotic plaques should be further elucidated.
Collapse
Grants
- 451-03-66/2024-03/200017 The Ministry of Science, Technological Development and Innovation, Republic of Serbia
- 451-03-66/2024-03/200017 The Ministry of Science, Technological Development and Innovation, Republic of Serbia
- 451-03-66/2024-03/200017 The Ministry of Science, Technological Development and Innovation, Republic of Serbia
- 451-03-66/2024-03/200017 The Ministry of Science, Technological Development and Innovation, Republic of Serbia
- 451-03-66/2024-03/200017 The Ministry of Science, Technological Development and Innovation, Republic of Serbia
- 451-03-66/2024-03/200017 The Ministry of Science, Technological Development and Innovation, Republic of Serbia
Collapse
Affiliation(s)
- Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, VINCA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, P.O. Box 522, Vinca, Belgrade, 11351, Serbia.
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, VINCA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, P.O. Box 522, Vinca, Belgrade, 11351, Serbia
| | - Igor Koncar
- Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Dr Koste Todorovica 8, Belgrade, 11000, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, 11000, Serbia
| | - Ana Kolakovic
- Laboratory for Radiobiology and Molecular Genetics, VINCA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, P.O. Box 522, Vinca, Belgrade, 11351, Serbia
| | - Maja Boskovic
- Laboratory for Radiobiology and Molecular Genetics, VINCA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, P.O. Box 522, Vinca, Belgrade, 11351, Serbia
| | - Tamara Djuric
- Laboratory for Radiobiology and Molecular Genetics, VINCA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, P.O. Box 522, Vinca, Belgrade, 11351, Serbia
| |
Collapse
|
9
|
Ramírez-Carracedo R, Hernández I, Moreno-Gómez-Toledano R, Díez-Mata J, Tesoro L, González-Cucharero C, Jiménez-Guirado B, Alcharani N, Botana L, Saura M, Zamorano JL, Zaragoza C. NOS3 prevents MMP-9, and MMP-13 induced extracellular matrix proteolytic degradation through specific microRNA-targeted expression of extracellular matrix metalloproteinase inducer in hypertension-related atherosclerosis. J Hypertens 2024; 42:685-693. [PMID: 38406874 PMCID: PMC10906209 DOI: 10.1097/hjh.0000000000003679] [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: 07/31/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Endothelial nitric oxide synthase (NOS3) elicits atheroprotection by preventing extracellular matrix (ECM) proteolytic degradation through inhibition of extracellular matrix metalloproteinase inducer (EMMPRIN) and collagenase MMP-13 by still unknown mechanisms. METHODS C57BL/6 mice lacking ApoE , NOS3, and/or MMP13 were fed with a high-fat diet for 6 weeks. Entire aortas were extracted and frozen to analyze protein and nucleic acid expression. Atherosclerotic plaques were detected by ultrasound imaging, Oil Red O (ORO) staining, and Western Blot. RNA-seq and RT-qPCR were performed to evaluate EMMPRIN, MMP-9, and EMMPRIN-targeting miRNAs. Mouse aortic endothelial cells (MAEC) were incubated to assess the role of active MMP-13 over MMP-9. One-way ANOVA or Kruskal-Wallis tests were performed to determine statistical differences. RESULTS Lack of NOS3 in ApoE null mice fed with a high-fat diet increased severe plaque accumulation, vessel wall widening, and high mortality, along with EMMPRIN-induced expression by upregulation of miRNAs 46a-5p and 486-5p. However, knocking out MMP-13 in ApoE/NOS3 -deficient mice was sufficient to prevent mortality (66.6 vs. 26.6%), plaque progression (23.1 vs. 8.8%), and MMP-9 expression, as confirmed in murine aortic endothelial cell (MAEC) cultures, in which MMP-9 was upregulated by incubation with active recombinant MMP-13, suggesting MMP-9 as a new target of MMP-13 in atherosclerosis. CONCLUSION We describe a novel mechanism by which the absence of NOS3 may worsen atherosclerosis through EMMPRIN-induced ECM proteolytic degradation by targeting the expression of miRNAs 146a-5p and 485-5p. Focusing on NOS3 regulation of ECM degradation could be a promising approach in the management of atherosclerosis.
Collapse
Affiliation(s)
- Rafael Ramírez-Carracedo
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Ignacio Hernández
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos
| | - Rafael Moreno-Gómez-Toledano
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
- Universidad de Alcalá, Unidad de Fisiología, Departamento de Biología de Sistemas, Alcalá de Henares
| | - Javier Díez-Mata
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Laura Tesoro
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Claudia González-Cucharero
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Beatriz Jiménez-Guirado
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Nunzio Alcharani
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Laura Botana
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
| | - Marta Saura
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos
- Universidad de Alcalá, Unidad de Fisiología, Departamento de Biología de Sistemas, Alcalá de Henares
| | - Jose L. Zamorano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos
- Departamento de Cardiología, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Carlos Zaragoza
- Unidad Mixta de Investigación Cardiovascular, Departamento de Cardiología, Universidad Francisco de Vitoria, Hospital Ramón y Cajal (IRYCIS)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos
| |
Collapse
|
10
|
Ji T, Yan D, Huang Y, Luo M, Zhang Y, Xu T, Gao S, Zhang L, Ruan L, Zhang C. Fibulin 1, targeted by microRNA-24-3p, promotes cell proliferation and migration in vascular smooth muscle cells, contributing to the development of atherosclerosis in APOE -/- mice. Gene 2024; 898:148129. [PMID: 38184021 DOI: 10.1016/j.gene.2024.148129] [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: 09/27/2023] [Revised: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Extracellular matrix (ECM) and vascular smooth muscle cells (VSMCs) are the main components of atherosclerosis (AS) plaque. VSMCs participate in plaque formation through phenotypic transformation. The complex interplay between ECM and VSMCs plays vital roles in the progression of AS throughout the disease. An in-depth investigation into the functions of ECM-related molecules in VSMC development might contribute to deciphering the complexity of AS pathogenesis. In this study, the roles and molecular mechanisms of the ECM-related molecule Fibulin-1 (FBLN1) in the development of AS and VSMCs were explored using RNA sequencing, bioinformatics analysis, and cell experiments. Furthermore, the expression of FBLN1, as determined by western blot analysis, immunohistochemistry, and real-time quantitative PCR, was significantly increased in AS vascular samples compared to normal vascular samples. Silencing the FBLN1 through AAV viral injection in mice revealed an improvement in AS. Functional analyses revealed that FBLN1 promoted VSMC proliferation, migration, and wound healing. Combined with RNA sequencing and TargetScan7.2 prediction data, 22 microRNAs (miRNAs) were found to have the potential for direct interaction with the FBLN1 3'UTR in VSMCs. Among these 22 miRNAs, it was demonstrated that microRNA-24-3p (miR-24-3p) could negatively regulate FBLN1 expression by directly binding to the FBLN1 3'UTR. Moreover, miR-24-3p inhibited cell proliferation, migration, and wound healing, and suppressed the expression of Ki67, matrix metalloproteinase-2 and -9 (MMP2/9) by targeting FBLN1 in VSMCs. Meanwhile, inhibition of FBLN1 expression could restrain VSMC phenotypic transformation. In conclusion, miR-24-3p inhibited VSMC proliferation and migration by targeting FBLN1. Additionally, multiple miRNAs with the potential to interact with the FBLN1 3'UTR were identified. These findings might deepen our understanding of ECM gene regulatory networks and the complex etiology of AS.
Collapse
Affiliation(s)
- Tianyi Ji
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Dan Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yi Huang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Mandi Luo
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yucong Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ting Xu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shangbang Gao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Sciences and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Le Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lei Ruan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
11
|
Del Moral LE, Lerma C, González-Pacheco H, Chávez-Lázaro AC, Massó F, Rodriguez E. Correlation of Plasmatic Amyloid Beta Peptides (Aβ-40, Aβ-42) with Myocardial Injury and Inflammatory Biomarkers in Acute Coronary Syndrome. J Clin Med 2024; 13:1117. [PMID: 38398429 PMCID: PMC10889335 DOI: 10.3390/jcm13041117] [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: 01/15/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Background/Objective: Amyloid beta (β) -40 levels increase with age and inflammation states and appear to be associated with clinical manifestations of acute coronary syndrome (ACS). We investigated the correlation of Aβ peptides with myocardial injury and inflammation biomarkers in patients with or without ST elevation myocardial infarction (STEMI, NSTEMI). Methods: This singe-center, cross-sectional, observational, and correlation study included 65 patients with ACS (n = 34 STEMI, 29 males, age = 58 ± 12 years; n = 31 NSTEMI, 22 males, age = 60 ± 12 years) who were enrolled in the coronary care unit within 12 h after symptom onset from February 2022 to May 2023. Aβ peptide levels and biochemical parameters were assessed. Results: NSTEMI patients had a higher prevalence of hypertension (p = 0.039), diabetes (p = 0.043), smoking (p = 0.003), and prior myocardial infarction (p = 0.010) compared to STEMI patients. We observed a higher level of Aβ-42 in NSTEMI (p = 0.001) but no difference in Aβ-40 levels. We also found a correlation between age and NT-proBNP with both Aβ peptides (Aβ-40, Aβ-42) (p = 0.001, p = 0.002 respectively). Conclusions: Our results show that patients with NSTEMI had a higher prevalence of cardiovascular risk factors (hypertension, diabetes, smoking, and prior myocardial infarction). Considering these results, we propose that Aβ-42 can add value to risk stratification in NSTEMI patients.
Collapse
Affiliation(s)
- Luis Eduardo Del Moral
- Translacional Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (L.E.D.M.); (A.C.C.-L.); (F.M.)
| | - Claudia Lerma
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Héctor González-Pacheco
- Coronary Care Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Alan Cristhian Chávez-Lázaro
- Translacional Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (L.E.D.M.); (A.C.C.-L.); (F.M.)
| | - Felipe Massó
- Translacional Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (L.E.D.M.); (A.C.C.-L.); (F.M.)
| | - Emma Rodriguez
- Translacional Research Unit, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (L.E.D.M.); (A.C.C.-L.); (F.M.)
| |
Collapse
|
12
|
Liuizė (Abramavičiūtė) A, Mongirdienė A. TGF-β Isoforms and GDF-15 in the Development and Progression of Atherosclerosis. Int J Mol Sci 2024; 25:2104. [PMID: 38396781 PMCID: PMC10889676 DOI: 10.3390/ijms25042104] [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: 01/10/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The effect of oxidised lipoproteins on the endothelium, monocytes, platelets, and macrophages is a key factor in the initiation and development of atherosclerosis. Antioxidant action, lipoprotein metabolism, and chronic inflammation are the fields of research interest for better understanding the development of the disease. All the fields are related to inflammation and hence to the secretion of cytokines, which are being investigated as potential diagnostic markers for the onset of atherosclerosis. Pathways of vascular damage are crucial for the development of new laboratory readouts. The very early detection of endothelial cell damage associated with the onset of atherosclerosis, allowing the initiation of therapy, remains a major research goal. This article summarises the latest results on the relationship of tumour growth factor beta (TGF-β) isoforms and growth differentiation factor 15 (GDF-15) to the pathogenesis of atherosclerosis: which cells involved in atherosclerosis produce them, which effectors stimulate their synthesis and secretion, how they influence atherosclerosis development, and the relationship between the levels of TGF-β and GDF-15 in the blood and the development and extent of atherosclerosis.
Collapse
Affiliation(s)
| | - Aušra Mongirdienė
- Department of Biochemistry, Medical Academy, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
| |
Collapse
|
13
|
Mosleh MM, Sohn MJ, Kim HS. Endothelial marker profiles in cerebral radiation-induced vasculopathy: A comparative immunohistochemical analysis. Medicine (Baltimore) 2024; 103:e37130. [PMID: 38306519 PMCID: PMC10843420 DOI: 10.1097/md.0000000000037130] [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: 11/22/2023] [Accepted: 01/10/2024] [Indexed: 02/04/2024] Open
Abstract
Radiation therapy results in radiation-induced vasculopathy, characterized by alterations in the vascular architecture stemming from radiation exposure. The exact molecular pathways and associated pathologies of this condition have yet to be comprehensively understood. This study aimed to identify specific markers' roles in cerebral vascular endothelial injury pathogenesis after radiosurgery and explore their unique expression patterns in diverse pathologies post-stereotactic radiosurgery. A retrospective cohort study was conducted to assess the expression profiles of endothelial markers via immunohistochemical analysis in 25 adult patients (13 males and 12 females) who had undergone neurosurgical resection for various central nervous system pathologies following stereotactic radiosurgery or radiotherapy from 2001 to 2015. Our findings revealed strong immunohistochemical expression of ICAM-1 and E-selectin across various disease states, while MMP-9, PAI-1, and eNOS exhibited moderate expression levels. In contrast, VCAM-1 and P-Selectin had the weakest expression across all groups. Notably, while individual markers showed significant variations in expression levels when comparing different diseases (P < .001), no substantial differences were found in the overall immunohistochemical expression patterns across the 5 distinct pathologies studied (P = .407, via 2-way ANOVA). Despite the varied long-term effects of radiotherapy on the vascular endothelium, a common thread of inflammation runs through the pathology of these conditions. The distinct patterns of marker expression identified in our study suggest that different markers play unique roles in the development of radiation-induced vasculopathy. These findings offer insights that could lead to the development of novel preventive strategies and treatments.
Collapse
Affiliation(s)
- Mohammad Mohsen Mosleh
- Department of Biomedical Science, Graduate School of Medicine, Inje University, Busanjin-gu, Busan, Korea
| | - Moon-Jun Sohn
- Department of Biomedical Science, Graduate School of Medicine, Inje University, Busanjin-gu, Busan, Korea
- Department of Neurosurgery and Neuroscience & Radiosurgery Hybrid Research Center, Inje University Ilsan Paik Hospital, College of Medicine, Ilsanseo-gu, Goyang City, Gyeonggi-do, Korea
| | - Han Seong Kim
- Department of Pathology, Inje University Ilsan Paik Hospital, Inje University Ilsan Paik Hospital, College of Medicine, Ilsanseo-gu, Goyang City, Gyeonggi-do, Korea
| |
Collapse
|
14
|
Woon CK, Omar E, Siew SF, Nawawi HM, Kasim NAM, Chainchel Singh MK. The effect of post-mortem computed tomography angiography (PMCTA) on biomarkers of coronary artery disease. J Forensic Leg Med 2024; 102:102654. [PMID: 38310784 DOI: 10.1016/j.jflm.2024.102654] [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: 10/03/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
Coronary atherosclerosis is due to build-up of plaque within the coronary arteries. Post-mortem computed tomography (PMCT) allows non or minimally invasive visualization of abnormalities prior to an autopsy, however PMCT-angiography (PMCTA) greatly enhances relevant findings, especially in viewing the cardiovascular system which is important in the diagnosis of coronary atherosclerosis. Contrast media used in PMCTA however has been reported to cause distortion of tissue which may interfere with post-mortem investigation outcomes. A cross sectional study to investigate the effect of PMCTA on tissue biomarkers in coronary arteries was performed involving cases brought in dead to the Institute and Accident and Emergency Unit. Sixty-three autopsy cases were included in this study, whereby 18 cases underwent PMCT while 45 cases underwent PMCTA. The subjects subsequently had a conventional autopsy where coronary artery sections were collected for standard histological examination and immunohistochemistry examination for endothelial inflammatory (CD36), prothrombogenic (TPA) and plaque stability (MMP-9) markers. The subjects consisted of 55 males and 8 females with a mean age ±SD of 49 ± 18.11 years. There were no significant differences in the coronary artery endothelial expression of CD36, MMP-9 and TPA between PMCT and PMCTA subjects. PMCTA does not alter CD36, TPA and MMP-9 markers supporting the safe use of PMCTA in post-mortem examinations.
Collapse
Affiliation(s)
- Choy Ker Woon
- Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Jalan Hospital 47000, Sungai Buloh, Selangor, Malaysia; Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia
| | - Effat Omar
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia; Department of Pathology, Faculty of Medicine, Universiti Teknologi MARA, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia
| | - Sheue Feng Siew
- National Institute of Forensic Medicine (IPFN), Hospital Kuala Lumpur, Ministry of Health Malaysia, Jalan Pahang, 50586, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Hapizah M Nawawi
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia; Department of Pathology, Faculty of Medicine, Universiti Teknologi MARA, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia
| | - Noor Alicezah M Kasim
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia; Department of Pathology, Faculty of Medicine, Universiti Teknologi MARA, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia
| | - Mansharan Kaur Chainchel Singh
- Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia; National Institute of Forensic Medicine (IPFN), Hospital Kuala Lumpur, Ministry of Health Malaysia, Jalan Pahang, 50586, Wilayah Persekutuan Kuala Lumpur, Malaysia; Department of Radiology, Hospital Al-Sultan Abdullah, Universiti Teknologi MARA, 42300, Bandar Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
15
|
Elieh-Ali-Komi D, Bot I, Rodríguez-González M, Maurer M. Cellular and Molecular Mechanisms of Mast Cells in Atherosclerotic Plaque Progression and Destabilization. Clin Rev Allergy Immunol 2024; 66:30-49. [PMID: 38289515 DOI: 10.1007/s12016-024-08981-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 03/28/2024]
Abstract
Mast cells (MCs) are commonly recognized for their crucial involvement in the pathogenesis of allergic diseases, but over time, it has come to light that they also play a role in the pathophysiology of non-allergic disorders including atherosclerosis. The involvement of MCs in the pathology of atherosclerosis is supported by their accumulation in atherosclerotic plaques upon their progression and the association of intraplaque MC numbers with acute cardiovascular events. MCs that accumulate within the atherosclerotic plaque release a cocktail of mediators through which they contribute to neovascularization, plaque progression, instability, erosion, rupture, and thrombosis. At a molecular level, MC-released proteases, especially cathepsin G, degrade low-density lipoproteins (LDL) and mediate LDL fusion and binding of LDL to proteoglycans (PGs). Through a complicated network of chemokines including CXCL1, MCs promote the recruitment of among others CXCR2+ neutrophils, therefore, aggravating the inflammation of the plaque environment. Additionally, MCs produce extracellular traps which worsen inflammation and contribute to atherothrombosis. Altogether, evidence suggests that MCs actively, via several underlying mechanisms, contribute to atherosclerotic plaque destabilization and acute cardiovascular syndromes, thus, making the study of interventions to modulate MC activation an interesting target for cardiovascular medicine.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Marcus Maurer
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany.
| |
Collapse
|
16
|
He J, Gao Y, Yang C, Guo Y, Liu L, Lu S, He H. Navigating the landscape: Prospects and hurdles in targeting vascular smooth muscle cells for atherosclerosis diagnosis and therapy. J Control Release 2024; 366:261-281. [PMID: 38161032 DOI: 10.1016/j.jconrel.2023.12.047] [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: 09/21/2023] [Revised: 12/02/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Vascular smooth muscle cells (VSMCs) have emerged as pivotal contributors throughout all phases of atherosclerotic plaque development, effectively dispelling prior underestimations of their prevalence and significance. Recent lineage tracing studies have unveiled the clonal nature and remarkable adaptability inherent to VSMCs, thereby illuminating their intricate and multifaceted roles in the context of atherosclerosis. This comprehensive review provides an in-depth exploration of the intricate mechanisms and distinctive characteristics that define VSMCs across various physiological processes, firmly underscoring their paramount importance in shaping the course of atherosclerosis. Furthermore, this review offers a thorough examination of the significant strides made over the past two decades in advancing imaging techniques and therapeutic strategies with a precise focus on targeting VSMCs within atherosclerotic plaques, notably spotlighting meticulously engineered nanoparticles as a promising avenue. We envision the potential of VSMC-targeted nanoparticles, thoughtfully loaded with medications or combination therapies, to effectively mitigate pro-atherogenic VSMC processes. These advancements are poised to contribute significantly to the pivotal objective of modulating VSMC phenotypes and enhancing plaque stability. Moreover, our paper also delves into recent breakthroughs in VSMC-targeted imaging technologies, showcasing their remarkable precision in locating microcalcifications, dynamically monitoring plaque fibrous cap integrity, and assessing the therapeutic efficacy of medical interventions. Lastly, we conscientiously explore the opportunities and challenges inherent in this innovative approach, providing a holistic perspective on the potential of VSMC-targeted strategies in the evolving landscape of atherosclerosis research and treatment.
Collapse
Affiliation(s)
- Jianhua He
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Yu Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Can Yang
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yujie Guo
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lisha Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Shan Lu
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
17
|
Huang J, Chen C, Liang H, Dong W, Li L, Ma H. Development of a fluorescence immunochromatography method for quantitative measurement of matrix metalloproteinase-9. Pract Lab Med 2024; 38:e00356. [PMID: 38292924 PMCID: PMC10825512 DOI: 10.1016/j.plabm.2024.e00356] [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: 10/15/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024] Open
Abstract
Objective Abnormal serum matrix metalloproteinase-9 (MMP-9) levels are closely related to the occurrence and development of many diseases. This study aimed to establish a fluorescence immunochromatography (FIC) method using the lanthanide fluorescent element europium(III) (Eu3+) for the quantitative measurement of MMP-9 in serum. Design & Methods The FIC method for quantifying MMP-9 was optimized and established, and the FIC test strips (FICTS) were assembled and subsequently evaluated for sensitivity, specificity and precision. Furthermore, the reference interval and clinical sensitivity/specificity were estimated using clinical healthy/positive serum samples, and a commercial ELISA was used for comparison. Results We successfully established an FIC method and prepared FICTS. The analytical sensitivity of the FICTS was 0.92 ng/mL, with a linearity range of 0-1000 ng/mL. The cross-reactivity of the 7 common serum interferents was less than 1.56%. All recoveries of the intra-array and inter-array samples ranged from 102.50% to 110.99%, and all CVs were less than 5%. The reference interval of the FICTS was >161.15 ng/mL. The clinical sensitivity was 96.00%, and the specificity was 97.5%. The results of 270 clinical serum samples were highly coincident with the clinical diagnostic results. Pearson correlation analysis and Bland‒Altman plots indicated that the FICTS and commercial ELISA results were consistent with the quantitative MMP-9 concentration. Conclusions The designed FIC method and test strips may be suitable for point-of-care quantitative measurement of MMP-9, which provides a new method for screening for atherosclerosis, xerophthalmia, etc.
Collapse
Affiliation(s)
- Jingyan Huang
- Department of Ophthalmology and Optometry, Affiliated Hospital of Beihua University, Jilin, 132011, China
| | - Cuicui Chen
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
- Jinan Laide Bio-technology Co., Ltd., Jinan, 271100, China
| | - Huankun Liang
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| | - Wenqi Dong
- Guangzhou Zhenda Biopharmaceutical Technology Co., Ltd., Guangzhou, 510663, China
| | - Laiqing Li
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
- Jinan Laide Bio-technology Co., Ltd., Jinan, 271100, China
| | - Hongyan Ma
- Department of Cardiology, Affiliated Hospital of Beihua University, Jilin, 132011, China
| |
Collapse
|
18
|
Smit V, de Mol J, Schaftenaar FH, Depuydt MAC, Postel RJ, Smeets D, Verheijen FWM, Bogers L, van Duijn J, Verwilligen RAF, Grievink HW, Bernabé Kleijn MNA, van Ingen E, de Jong MJM, Goncalves L, Peeters JAHM, Smeets HJ, Wezel A, Polansky JK, de Winther MPJ, Binder CJ, Tsiantoulas D, Bot I, Kuiper J, Foks AC. Single-cell profiling reveals age-associated immunity in atherosclerosis. Cardiovasc Res 2023; 119:2508-2521. [PMID: 37390467 PMCID: PMC10676459 DOI: 10.1093/cvr/cvad099] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/07/2023] [Accepted: 05/12/2023] [Indexed: 07/02/2023] Open
Abstract
AIMS Aging is a dominant driver of atherosclerosis and induces a series of immunological alterations, called immunosenescence. Given the demographic shift towards elderly, elucidating the unknown impact of aging on the immunological landscape in atherosclerosis is highly relevant. While the young Western diet-fed Ldlr-deficient (Ldlr-/-) mouse is a widely used model to study atherosclerosis, it does not reflect the gradual plaque progression in the context of an aging immune system as occurs in humans. METHODS AND RESULTS Here, we show that aging promotes advanced atherosclerosis in chow diet-fed Ldlr-/- mice, with increased incidence of calcification and cholesterol crystals. We observed systemic immunosenescence, including myeloid skewing and T-cells with more extreme effector phenotypes. Using a combination of single-cell RNA-sequencing and flow cytometry on aortic leucocytes of young vs. aged Ldlr-/- mice, we show age-related shifts in expression of genes involved in atherogenic processes, such as cellular activation and cytokine production. We identified age-associated cells with pro-inflammatory features, including GzmK+CD8+ T-cells and previously in atherosclerosis undefined CD11b+CD11c+T-bet+ age-associated B-cells (ABCs). ABCs of Ldlr-/- mice showed high expression of genes involved in plasma cell differentiation, co-stimulation, and antigen presentation. In vitro studies supported that ABCs are highly potent antigen-presenting cells. In cardiovascular disease patients, we confirmed the presence of these age-associated T- and B-cells in atherosclerotic plaques and blood. CONCLUSIONS Collectively, we are the first to provide comprehensive profiling of aged immunity in atherosclerotic mice and reveal the emergence of age-associated T- and B-cells in the atherosclerotic aorta. Further research into age-associated immunity may contribute to novel diagnostic and therapeutic tools to combat cardiovascular disease.
Collapse
Affiliation(s)
- Virginia Smit
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Jill de Mol
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Frank H Schaftenaar
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Marie A C Depuydt
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Rimke J Postel
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Diede Smeets
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Fenne W M Verheijen
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Laurens Bogers
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Janine van Duijn
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Robin A F Verwilligen
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Hendrika W Grievink
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL Leiden, The Netherlands
| | - Mireia N A Bernabé Kleijn
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Eva van Ingen
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Maaike J M de Jong
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Lauren Goncalves
- Department of Surgery, Haaglanden Medical Center—location Westeinde, Lijnbaan 32, 2515 VA The Hague, The Netherlands
| | - Judith A H M Peeters
- Department of Surgery, Haaglanden Medical Center—location Westeinde, Lijnbaan 32, 2515 VA The Hague, The Netherlands
| | - Harm J Smeets
- Department of Surgery, Haaglanden Medical Center—location Westeinde, Lijnbaan 32, 2515 VA The Hague, The Netherlands
| | - Anouk Wezel
- Department of Surgery, Haaglanden Medical Center—location Westeinde, Lijnbaan 32, 2515 VA The Hague, The Netherlands
| | - Julia K Polansky
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Menno P J de Winther
- Amsterdam University Medical Centers—location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Lazarettgasse 14, AKH BT25.2, 1090 Vienna, Austria
| | - Dimitrios Tsiantoulas
- Department of Laboratory Medicine, Medical University of Vienna, Lazarettgasse 14, AKH BT25.2, 1090 Vienna, Austria
| | - Ilze Bot
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Amanda C Foks
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
19
|
Hou Q, Jiang J, Na K, Zhang X, Liu D, Jing Q, Yan C, Han Y. Bioinformatics analyses of potentially common pathogenic networks for primary Sjögren's syndrome complicated with acute myocardial infarction. Sci Rep 2023; 13:19276. [PMID: 37935719 PMCID: PMC10630427 DOI: 10.1038/s41598-023-45896-5] [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: 07/09/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023] Open
Abstract
Both primary Sjögren's syndrome (pSS) and acute myocardial infarction (AMI) are intricately linked. However, their common mechanism is not fully understood. Herein, we examined the underlying network of molecular action associated with developing this complication. Datasets were downloaded from the GEO database. We performed enrichment and protein-protein interaction analyses and screened key genes. We used external datasets to confirm the diagnostic performance for these hub genes. Transcription factor and microRNA regulatory networks were constructed for the validated hub genes. Finally, drug prediction and molecular docking validation were performed. We identified 62 common DEGs, many of which were enriched regarding inflammation and immune response. 5 DEGs were found as key hub genes (IGSF6, MMP9, S100A8, MNDA, and NCF2). They had high diagnostic performance in external datasets. Functional enrichment of these five hub genes showed that they were associated with the adaptive immune response. The Type 1T helper cell showed the most association among all cell types related to AMI and pSS. We identified 36 common TFs and 49 identical TF-miRNAs. The drugs, including Benzo, dexamethasone, and NADP, were predicted as potential therapeutic agents. Herein, we revealed common networks involving pSS and AMI etiologies. Knowledge of these networks and hub genes can enhance research into their associated mechanism and the development of future robust therapy.
Collapse
Affiliation(s)
- Qingbin Hou
- Department of Internal Medicine (Cardiovascular), the Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jinping Jiang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
- Department of Cardiology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Kun Na
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaolin Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Quanmin Jing
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Yaling Han
- Department of Internal Medicine (Cardiovascular), the Second Clinical Medical College, Shanxi Medical University, Taiyuan, China.
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
20
|
Luca AC, David SG, David AG, Țarcă V, Pădureț IA, Mîndru DE, Roșu ST, Roșu EV, Adumitrăchioaiei H, Bernic J, Cojocaru E, Țarcă E. Atherosclerosis from Newborn to Adult-Epidemiology, Pathological Aspects, and Risk Factors. Life (Basel) 2023; 13:2056. [PMID: 37895437 PMCID: PMC10608492 DOI: 10.3390/life13102056] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality and morbidity throughout the world, accounting for 16.7 million deaths each year. The underlying pathological process for the majority of cardiovascular diseases is atherosclerosis, a slowly progressing, multifocal, chronic, immune-inflammatory disease that involves the intima of large and medium-sized arteries. The process of atherosclerosis begins in childhood as fatty streaks-an accumulation of lipids, inflammatory cells, and smooth muscle cells in the arterial wall. Over time, a more complex lesion develops into an atheroma and characteristic fibrous plaques. Atherosclerosis alone is rarely fatal; it is the further changes that render fibrous plaques vulnerable to rupture; plaque rupture represents the most common cause of coronary thrombosis. The prevalence of atherosclerosis is increasing worldwide and more than 50% of people with circulatory disease die of it, mostly in modern societies. Epidemiological studies have revealed several environmental and genetic risk factors that are associated with the early formation of a pathogenic foundation for atherosclerosis, such as dyslipidemia, hypertension, diabetes mellitus, obesity, and smoking. The purpose of this review is to bring together the current information concerning the origin and progression of atherosclerosis in childhood as well as the identification of known risk factors for atherosclerotic cardiovascular disease in children.
Collapse
Affiliation(s)
- Alina Costina Luca
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Simona Georgiana David
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Alexandru Gabriel David
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Viorel Țarcă
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ioana-Alexandra Pădureț
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Dana Elena Mîndru
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Solange Tamara Roșu
- Nursing Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Eduard Vasile Roșu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Heidrun Adumitrăchioaiei
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Jana Bernic
- Discipline of Pediatric Surgery, “Nicolae Testemițanu” State University of Medicine and Pharmacy, 2025 Chisinau, Moldova;
| | - Elena Cojocaru
- Department of Morphofunctional Sciences I—Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Elena Țarcă
- Surgery II Department—Pediatric Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| |
Collapse
|
21
|
Meteva D, Vinci R, Seppelt C, Abdelwahed YS, Pedicino D, Nelles G, Skurk C, Haghikia A, Rauch-Kröhnert U, Gerhardt T, Straessler E, Zhao Y, Golla F, Joner M, Rai H, Kratzer A, Arnal HG, Liuzzo G, Klotsche J, Crea F, Landmesser U, Leistner DM, Kränkel N. Toll-like receptor 2, hyaluronan, and neutrophils play a key role in plaque erosion: the OPTICO-ACS study. Eur Heart J 2023; 44:3892-3907. [PMID: 37381760 DOI: 10.1093/eurheartj/ehad379] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND AND AIMS In one-third of patients with acute coronary syndrome (ACS), thrombosis occurs despite an intact fibrous cap (IFC) (IFC-ACS, 'plaque erosion'). Recent studies emphasize neutrophils as the immediate inflammatory response in this pathology, but their exact molecular activation patterns are still poorly understood and may represent future therapeutic targets. METHODS AND RESULTS Thirty-two patients with IFC-ACS and matched patients with ACS with ruptured fibrous cap (RFC) (RFC-ACS) from the OPTICO-ACS study were included, and blood samples were collected from the local site of the culprit lesion and the systemic circulation. Neutrophil surface marker expression was quantified by flow cytometry. Neutrophil cytotoxicity towards endothelial cells was examined in an ex vivo co-culture assay. Secretion of active matrix metalloproteinase 9 (MMP9) by neutrophils was evaluated using zymography in supernatants and in plasma samples. Optical coherence tomography (OCT)-embedded thrombi were used for immunofluorescence analysis. Toll-like receptor 2 (TLR2) expression was higher on neutrophils from IFC-ACS than RFC-ACS patients. TLR2 stimulation increased the release of active MMP9 from local IFC-ACS-derived neutrophils, which also aggravated endothelial cell death independently of TLR2. Thrombi of IFC-ACS patients exhibited more hyaluronidase 2 with concomitant increase in local plasma levels of the TLR2 ligand: hyaluronic acid. CONCLUSION The current study provides first in-human evidence for distinct TLR2-mediated neutrophil activation in IFC-ACS, presumably triggered by elevated soluble hyaluronic acid. Together with disturbed flow conditions, neutrophil-released MMP9 might be promoting endothelial cell loss-triggered thrombosis and therefore providing a potential future target for a phenotype-specific secondary therapeutic approach in IFC-ACS.
Collapse
Affiliation(s)
- Denitsa Meteva
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Ramona Vinci
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Department of Cardiovascular Sciences, IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo Francesco Vito 1, Rome 00168, Italy
| | - Claudio Seppelt
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Department of Cardiology and Angiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60598, Germany
| | - Youssef S Abdelwahed
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Daniela Pedicino
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Department of Cardiovascular Sciences, IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo Francesco Vito 1, Rome 00168, Italy
| | - Gregor Nelles
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Carsten Skurk
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Arash Haghikia
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
| | - Ursula Rauch-Kröhnert
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Teresa Gerhardt
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
| | - Elisabeth Straessler
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Yingjie Zhao
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Felix Golla
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Michael Joner
- Department of Cardiology and ISAR Research Centre, German Heart Centre Munich, Lazarettstrasse 36, Munich 80636, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Munich, Munich 80636, Germany
| | - Himanshu Rai
- Cardiovascular Research Institute Dublin, Mater Private Network, 73 Eccles Street, Dublin D07 YH66, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, 123 St. Stephan's Green, Dublin D02 YN77, Ireland
| | - Adelheid Kratzer
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Hector Giral Arnal
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| | - Giovanna Liuzzo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Department of Cardiovascular Sciences, IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo Francesco Vito 1, Rome 00168, Italy
| | - Jens Klotsche
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- German Rheumatism Research Centre (DRFZ) and Institute for Social Medicine, Epidemiology and Health Economy, Charitė University Medicine Berlin, Campus Charite Mitte, Charitėplatz 1, Berlin 10117, Germany
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Largo Francesco Vito 1, Rome 00168, Italy
- Department of Cardiovascular Sciences, IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo Francesco Vito 1, Rome 00168, Italy
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
| | - David M Leistner
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, Berlin 10178, Germany
- Department of Cardiology and Angiology, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main 60598, Germany
| | - Nicolle Kränkel
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Hindenburgdamm 30, Berlin 12203, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, Berlin 10117, Germany
- DZHK (German Centre for Cardiovascular Research) partner Site Berlin, Berlin 12203, Germany
| |
Collapse
|
22
|
Shi SY, Jia F, Wang MF, Zhou YF, Li JJ. Impacts of Non-alcoholic Fatty Liver Disease on Acute Coronary Syndrome: Evidence and Controversies. Curr Atheroscler Rep 2023; 25:751-768. [PMID: 37768409 PMCID: PMC10564833 DOI: 10.1007/s11883-023-01146-7] [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] [Accepted: 08/23/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW Acute coronary syndrome (ACS) and non-alcoholic fatty liver disease (NAFLD) are two clinically common disease entities that share numerous risk factors. This review aimed to discuss the impacts of NAFLD on ACS. RECENT FINDINGS In an era of improved control of traditional risk factors, the substantial burden of cardiometabolic abnormalities has caused widespread concern. NAFLD is considered the hepatic component of metabolic syndrome, which can exert an impact on human health beyond the liver. Accumulating studies have demonstrated that NAFLD is closely related to cardiovascular disease, especially coronary artery disease. Interestingly, although recent data have suggested an association between NAFLD and the incidence and outcomes of ACS, the results are not consistent. In this review, we comprehensively summarized evidence and controversies regarding whether NAFLD is a contributor to either the development of ACS or worse outcomes in patients with ACS. The potential pathophysiological and molecular mechanisms involved in the impacts of NAFLD on ACS were also elucidated.
Collapse
Affiliation(s)
- Shun-Yi Shi
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Fang Jia
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Meng-Fei Wang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ya-Feng Zhou
- Department of Cardiology, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, China
| | - Jian-Jun Li
- Cardio-Metabolism Center, Fu Wai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 10037, China.
| |
Collapse
|
23
|
O'Hagan R, Hsu LY, Li H, Hong CG, Parel PM, Berg AR, Manyak GA, Bui V, Patel NH, Florida EM, Teague HL, Playford MP, Zhou W, Dey D, Chen MY, Mehta NN, Sorokin AV. Longitudinal association of epicardial and thoracic adipose tissues with coronary and cardiac characteristics in psoriasis. Heliyon 2023; 9:e20732. [PMID: 37867905 PMCID: PMC10585224 DOI: 10.1016/j.heliyon.2023.e20732] [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: 07/19/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Background s: Psoriasis is a disease of systemic inflammation associated with increased cardiometabolic risk. Epicardial adipose tissue (EAT) and thoracic adipose tissue (TAT) are contributing factors for atherosclerosis and cardiac dysfunction. We strove to assess the longitudinal impact of the EAT and TAT on coronary and cardiac characteristics in psoriasis. Methods The study consisted of 301 patients with baseline coronary computed tomography angiography (CTA), of which 139 had four-year follow up scans. EAT and TAT volumes from non-contrast computed tomography scans were quantified by an automated segmentation framework. Coronary plaque characteristics and left ventricular (LV) mass were quantified by CTA. Results When stratified by baseline EAT and TAT volume quartiles, a stepwise significant increase in cardiometabolic parameters was observed. EAT and TAT volumes associated with fibro-fatty burden (FFB) (TAT: ρ = 0.394, P < 0.001; EAT: ρ = 0.459, P < 0.001) in adjusted models. Only EAT had a significant four-year time-dependent association with FFB in fully adjusted models (β = 0.307 P = 0.003), whereas only TAT volume associated with myocardial injury in fully adjusted models (TAT: OR = 1.57 95 % CI = (1.00-2.60); EAT: OR = 1.46 95 % CI = (0.91-2.45). Higher quartiles of EAT and TAT had increased LV mass and developed strong correlation (TAT: ρ = 0.370, P < 0.001; EAT: ρ = 0.512, P < 0.001). Conclusions Our study is the first to explore how both EAT and TAT volumes associate with increased cardiometabolic risk profile in an inflamed psoriasis cohorts and highlight the need for further studies on its use as a potential prognostic tool for high-risk coronary plaques and cardiac dysfunction.
Collapse
Affiliation(s)
- Ross O'Hagan
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Li-Yueh Hsu
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Haiou Li
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christin G. Hong
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Philip M. Parel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander R. Berg
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grigory A. Manyak
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vy Bui
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nidhi H. Patel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth M. Florida
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heather L. Teague
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wunan Zhou
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marcus Y. Chen
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander V. Sorokin
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
24
|
Sharifi MA, Wierer M, Dang TA, Milic J, Moggio A, Sachs N, von Scheidt M, Hinterdobler J, Müller P, Werner J, Stiller B, Aherrahrou Z, Erdmann J, Zaliani A, Graettinger M, Reinshagen J, Gul S, Gribbon P, Maegdefessel L, Bernhagen J, Sager HB, Mann M, Schunkert H, Kessler T. ADAMTS-7 Modulates Atherosclerotic Plaque Formation by Degradation of TIMP-1. Circ Res 2023; 133:674-686. [PMID: 37675562 PMCID: PMC7615141 DOI: 10.1161/circresaha.123.322737] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The ADAMTS7 locus was genome-wide significantly associated with coronary artery disease. Lack of the ECM (extracellular matrix) protease ADAMTS-7 (A disintegrin and metalloproteinase-7) was shown to reduce atherosclerotic plaque formation. Here, we sought to identify molecular mechanisms and downstream targets of ADAMTS-7 mediating the risk of atherosclerosis. METHODS Targets of ADAMTS-7 were identified by high-resolution mass spectrometry of atherosclerotic plaques from Apoe-/- and Apoe-/-Adamts7-/- mice. ECM proteins were identified using solubility profiling. Putative targets were validated using immunofluorescence, in vitro degradation assays, coimmunoprecipitation, and Förster resonance energy transfer-based protein-protein interaction assays. ADAMTS7 expression was measured in fibrous caps of human carotid artery plaques. RESULTS In humans, ADAMTS7 expression was higher in caps of unstable as compared to stable carotid plaques. Compared to Apoe-/- mice, atherosclerotic aortas of Apoe-/- mice lacking Adamts-7 (Apoe-/-Adamts7-/-) contained higher protein levels of Timp-1 (tissue inhibitor of metalloprotease-1). In coimmunoprecipitation experiments, the catalytic domain of ADAMTS-7 bound to TIMP-1, which was degraded in the presence of ADAMTS-7 in vitro. ADAMTS-7 reduced the inhibitory capacity of TIMP-1 at its canonical target MMP-9 (matrix metalloprotease-9). As a downstream mechanism, we investigated collagen content in plaques of Apoe-/- and Apoe-/-Adamts7-/- mice after a Western diet. Picrosirius red staining of the aortic root revealed less collagen as a readout of higher MMP-9 activity in Apoe-/- as compared to Apoe-/- Adamts7-/- mice. To facilitate high-throughput screening for ADAMTS-7 inhibitors with the aim of decreasing TIMP-1 degradation, we designed a Förster resonance energy transfer-based assay targeting the ADAMTS-7 catalytic site. CONCLUSIONS ADAMTS-7, which is induced in unstable atherosclerotic plaques, decreases TIMP-1 stability reducing its inhibitory effect on MMP-9, which is known to promote collagen degradation and is likewise associated with coronary artery disease. Disrupting the interaction of ADAMTS-7 and TIMP-1 might be a strategy to increase collagen content and plaque stability for the reduction of atherosclerosis-related events.
Collapse
Affiliation(s)
- M. Amin Sharifi
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Wierer
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Tan An Dang
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Jelena Milic
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Aldo Moggio
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Nadja Sachs
- Vascular Biology and Experimental Vascular Medicine Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Moritz von Scheidt
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Philipp Müller
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Julia Werner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Barbara Stiller
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics and University Heart Centre Lübeck, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics and University Heart Centre Lübeck, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Mira Graettinger
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Jeanette Reinshagen
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Lars Maegdefessel
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
- Vascular Biology and Experimental Vascular Medicine Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Jürgen Bernhagen
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
25
|
Kadoglou N, Moulakakis KG, Mantas G, Spathis A, Gkougkoudi E, Mylonas SN, Kakisis J, Liapis C. Novel Biomarkers and Imaging Indices for the "Vulnerable Patient" with Carotid Stenosis: A Single-Center Study. Biomolecules 2023; 13:1427. [PMID: 37759829 PMCID: PMC10526466 DOI: 10.3390/biom13091427] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND We investigated the relationship of matrix metalloproteinases (MMPs), cardio-ankle vascular index (CAVI), and Gray-Scale Median (GSM) score with the severity and vulnerability of carotid atherosclerosis and major adverse cardiovascular events (MACE) during follow-up of carotid artery revascularization. METHODS We enrolled 262 patients undergoing carotid revascularization therapy (GRT), 109 asymptomatic patients with low-grade carotid stenosis (40-70%) receiving conservative treatment (GCT), and 92 age- and sex-matched control subjects without carotid atherosclerosis (GCO). All participants underwent carotid ultrasound and we assessed at baseline clinical parameters, metabolic profile, CAVI, GSM, and circulating levels of hsCRP, MMP-3,-7,-9, and TIMP-1. RESULTS Both GRT and GCT presented with elevated CAVI, MMPs, and TIMP-1 levels compared to GCO (p < 0.001). The escalation highly correlated to the presence of symptoms or paralleled the degree of carotid stenosis (p < 0.001). During follow-up (mean duration: 55 months), 51 GRT patients experienced MACE unrelated to the revascularization procedure. Within GRT, diabetes (HR: 2.07; CI: 1.55-2.78, p < 0.001), smoking (HR: 1.67; CI: 1.35-1.95, p < 0.001), high CAVI (HR: 1.22; CI: 1.09-1.43, p = 0.023) and MMP-9 (HR: 1.44; CI: 1.29-2.15, p = 0.005), and low GSM (HR: 1.40; CI: 1.16-2.12, p = 0.002) independently predicted MACE occurrences, despite the optimum medical therapy. CONCLUSIONS Novel imaging and biochemical biomarkers were positively associated with atherosclerosis severity, while CAVI, MMP-9, and low GSM showed a positive, independent relationship with MACE after carotid revascularization, describing "vulnerable patients".
Collapse
Affiliation(s)
| | - Konstantinos G. Moulakakis
- Vascular Surgery Department, Patras University Hospital, University of Patras, Rio, 265 04 Patra, Greece;
| | - George Mantas
- Department of Vascular Surgery, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, 115 27 Athens, Greece; (G.M.); (J.K.); (C.L.)
| | - Aris Spathis
- 2nd Department of Pathology, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | | | - Spyridon N. Mylonas
- Department of Vascular and Endovascular Surgery, University Hospital of Cologne, 50937 Cologne, Germany;
| | - John Kakisis
- Department of Vascular Surgery, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, 115 27 Athens, Greece; (G.M.); (J.K.); (C.L.)
| | - Christos Liapis
- Department of Vascular Surgery, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, 115 27 Athens, Greece; (G.M.); (J.K.); (C.L.)
- Department of Vascular and Endovascular Surgery, Athens Medical Center, 106 73 Athens, Greece
| |
Collapse
|
26
|
Slysz J, Sinha A, DeBerge M, Singh S, Avgousti H, Lee I, Glinton K, Nagasaka R, Dalal P, Alexandria S, Wai CM, Tellez R, Vescovo M, Sunderraj A, Wang X, Schipma M, Sisk R, Gulati R, Vallejo J, Saigusa R, Lloyd-Jones DM, Lomasney J, Weinberg S, Ho K, Ley K, Giannarelli C, Thorp EB, Feinstein MJ. Single-cell profiling reveals inflammatory polarization of human carotid versus femoral plaque leukocytes. JCI Insight 2023; 8:e171359. [PMID: 37471165 PMCID: PMC10544225 DOI: 10.1172/jci.insight.171359] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Femoral atherosclerotic plaques are less inflammatory than carotid plaques histologically, but limited cell-level data exist regarding comparative immune landscapes and polarization at these sites. We investigated intraplaque leukocyte phenotypes and transcriptional polarization in 49 patients undergoing femoral (n = 23) or carotid (n = 26) endarterectomy using single-cell RNA-Seq (scRNA-Seq; n = 13), flow cytometry (n = 24), and IHC (n = 12). Comparative scRNA-Seq of CD45+-selected leukocytes from femoral (n = 9; 35,265 cells) and carotid (n = 4; 30,655 cells) plaque revealed distinct transcriptional profiles. Inflammatory foam cell-like macrophages and monocytes comprised higher proportions of myeloid cells in carotid plaques, whereas noninflammatory foam cell-like macrophages and LYVE1-overexpressing macrophages comprised higher proportions of myeloid cells in femoral plaque (P < 0.001 for all). A significant comparative excess of CCR2+ macrophages in carotid versus plaque was observed by flow cytometry in a separate validation cohort. B cells were more prevalent and exhibited a comparatively antiinflammatory profile in femoral plaque, whereas cytotoxic CD8+ T cells were more prevalent in carotid plaque. In conclusion, human femoral plaques exhibit distinct macrophage phenotypic and transcriptional profiles as well as diminished CD8+ T cell populations compared with human carotid plaques.
Collapse
Affiliation(s)
| | - Arjun Sinha
- Division of Cardiology, Department of Medicine
| | | | | | | | - Inhyeok Lee
- Division of Cardiology, Department of Medicine
| | - Kristofor Glinton
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
| | | | | | - Shaina Alexandria
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| | - Ching Man Wai
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Ricardo Tellez
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
| | | | | | - Xinkun Wang
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Matthew Schipma
- Northwestern University Sequencing Core, Chicago, Illinois, USA
| | - Ryan Sisk
- Division of Cardiology, Department of Medicine
| | - Rishab Gulati
- La Jolla Institute of Immunology, La Jolla, California, USA
| | | | | | - Donald M. Lloyd-Jones
- Division of Cardiology, Department of Medicine
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| | | | | | - Karen Ho
- Division of Vascular Surgery, NUFSM, Chicago, Illinois, USA
| | - Klaus Ley
- Immunology Center of Georgia, Augusta, Georgia, USA
| | - Chiara Giannarelli
- Department of Medicine and
- Department of Pathology, New York University, New York, New York, USA
| | | | - Matthew J. Feinstein
- Division of Cardiology, Department of Medicine
- Department of Pathology, and
- Department of Preventive Medicine at Northwestern University Feinberg School of Medicine (NUFSM), Chicago, Illinois, USA
| |
Collapse
|
27
|
Alonso-Herranz L, Albarrán-Juárez J, Bentzon JF. Mechanisms of fibrous cap formation in atherosclerosis. Front Cardiovasc Med 2023; 10:1254114. [PMID: 37671141 PMCID: PMC10475556 DOI: 10.3389/fcvm.2023.1254114] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023] Open
Abstract
The fibrous cap is formed by smooth muscle cells that accumulate beneath the plaque endothelium. Cap rupture is the main cause of coronary thrombosis, leading to infarction and sudden cardiac death. Therefore, the qualities of the cap are primary determinants of the clinical outcome of coronary and carotid atherosclerosis. In this mini-review, we discuss current knowledge about the formation of the fibrous cap, including cell recruitment, clonal expansion, and central molecular signaling pathways. We also examine the differences between mouse and human fibrous caps and explore the impact of anti-atherosclerotic therapies on the state of the fibrous cap. We propose that the cap should be understood as a neo-media to substitute for the original media that becomes separated from the surface endothelium during atherogenesis and that embryonic pathways involved in the development of the arteria media contribute to cap formation.
Collapse
Affiliation(s)
- Laura Alonso-Herranz
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Julián Albarrán-Juárez
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Fog Bentzon
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus and Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| |
Collapse
|
28
|
Canela VH, Bowen WS, Ferreira RM, Syed F, Lingeman JE, Sabo AR, Barwinska D, Winfree S, Lake BB, Cheng YH, Gaut JP, Ferkowicz M, LaFavers KA, Zhang K, Coe FL, Worcester E, Jain S, Eadon MT, Williams JC, El-Achkar TM. A spatially anchored transcriptomic atlas of the human kidney papilla identifies significant immune injury in patients with stone disease. Nat Commun 2023; 14:4140. [PMID: 37468493 PMCID: PMC10356953 DOI: 10.1038/s41467-023-38975-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 05/24/2023] [Indexed: 07/21/2023] Open
Abstract
Kidney stone disease causes significant morbidity and increases health care utilization. In this work, we decipher the cellular and molecular niche of the human renal papilla in patients with calcium oxalate (CaOx) stone disease and healthy subjects. In addition to identifying cell types important in papillary physiology, we characterize collecting duct cell subtypes and an undifferentiated epithelial cell type that was more prevalent in stone patients. Despite the focal nature of mineral deposition in nephrolithiasis, we uncover a global injury signature characterized by immune activation, oxidative stress and extracellular matrix remodeling. We also identify the association of MMP7 and MMP9 expression with stone disease and mineral deposition, respectively. MMP7 and MMP9 are significantly increased in the urine of patients with CaOx stone disease, and their levels correlate with disease activity. Our results define the spatial molecular landscape and specific pathways contributing to stone-mediated injury in the human papilla and identify associated urinary biomarkers.
Collapse
Affiliation(s)
- Victor Hugo Canela
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William S Bowen
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ricardo Melo Ferreira
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Farooq Syed
- Center for Diabetes and Metabolic Diseases, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James E Lingeman
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Angela R Sabo
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daria Barwinska
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Seth Winfree
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Blue B Lake
- San Diego Institute of Science, Altos Labs, San Diego, CA, USA
| | - Ying-Hua Cheng
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joseph P Gaut
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - Michael Ferkowicz
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kaice A LaFavers
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kun Zhang
- San Diego Institute of Science, Altos Labs, San Diego, CA, USA
| | - Fredric L Coe
- Department of Medicine, Division of Nephrology, University of Chicago, Chicago, IL, USA
| | - Elaine Worcester
- Department of Medicine, Division of Nephrology, University of Chicago, Chicago, IL, USA
| | - Sanjay Jain
- Department of Medicine, Division of Nephrology, Washington University, St. Louis, MO, USA.
| | - Michael T Eadon
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - James C Williams
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Tarek M El-Achkar
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Medicine, Indianapolis VA Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
29
|
Kotlyarov S. The Role of Smoking in the Mechanisms of Development of Chronic Obstructive Pulmonary Disease and Atherosclerosis. Int J Mol Sci 2023; 24:8725. [PMID: 37240069 PMCID: PMC10217854 DOI: 10.3390/ijms24108725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Tobacco smoking is a major cause of chronic obstructive pulmonary disease (COPD) and atherosclerotic cardiovascular disease (ASCVD). These diseases share common pathogenesis and significantly influence each other's clinical presentation and prognosis. There is increasing evidence that the mechanisms underlying the comorbidity of COPD and ASCVD are complex and multifactorial. Smoking-induced systemic inflammation, impaired endothelial function and oxidative stress may contribute to the development and progression of both diseases. The components present in tobacco smoke can have adverse effects on various cellular functions, including macrophages and endothelial cells. Smoking may also affect the innate immune system, impair apoptosis, and promote oxidative stress in the respiratory and vascular systems. The purpose of this review is to discuss the importance of smoking in the mechanisms underlying the comorbid course of COPD and ASCVD.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
30
|
Abstract
Vascular age is determined by functional and structural changes in the arterial wall. When measured by its proxy, pulse wave velocity, it has been shown to predict cardiovascular and total mortality. Disconcordance between chronological and vascular age might represent better or worse vascular health. Cell senescence is caused by oxidative stress and sustained cell replication. Senescent cells acquire senescence-associated secretory phenotype. Oxidative stress, endothelial dysfunction, dysregulation of coagulation and leucocyte infiltration are observed in the aging endothelium. All of these mechanisms lead to increased vascular calcification and stiffness. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can involve the vascular endothelium. It enters cells using angiotensin-converting enzyme 2 (ACE-2) receptors, which are abundant in endothelial cells. The damage this virus does to the endothelium can be direct or indirect. Indirect damage is caused by hyperinflammation. Direct damage results from effects on ACE-2 receptors. The reduction of ACE-2 levels seen during coronavirus disease 2019 (COVID-19) infection might cause vasoconstriction and oxidative stress. COVID-19 and vascular aging share some pathways. Due to the novelty of the virus, there is an urgent need for studies that investigate its long-term effects on vascular health.
Collapse
Affiliation(s)
- Ignas Badaras
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania,Ignas Badaras, Faculty of Medicine, Vilnius
University, M. K. Ciurlionio g. 21/27, LT-03101, Vilnius 01513, Lithuania.
| | - Agnė Laučytė-Cibulskienė
- Department of Nephrology, Skåne University
Hospital, Malmö, Sweden,Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
31
|
Aggarwal A, Jennings CL, Manning E, Cameron SJ. Platelets at the Vessel Wall in Non-Thrombotic Disease. Circ Res 2023; 132:775-790. [PMID: 36927182 PMCID: PMC10027394 DOI: 10.1161/circresaha.122.321566] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/15/2023] [Indexed: 03/18/2023]
Abstract
Platelets are small, anucleate entities that bud from megakaryocytes in the bone marrow. Among circulating cells, platelets are the most abundant cell, traditionally involved in regulating the balance between thrombosis (the terminal event of platelet activation) and hemostasis (a protective response to tissue injury). Although platelets lack the precise cellular control offered by nucleate cells, they are in fact very dynamic cells, enriched in preformed RNA that allows them the capability of de novo protein synthesis which alters the platelet phenotype and responses in physiological and pathological events. Antiplatelet medications have significantly reduced the morbidity and mortality for patients afflicted with thrombotic diseases, including stroke and myocardial infarction. However, it has become apparent in the last few years that platelets play a critical role beyond thrombosis and hemostasis. For example, platelet-derived proteins by constitutive and regulated exocytosis can be found in the plasma and may educate distant tissue including blood vessels. First, platelets are enriched in inflammatory and anti-inflammatory molecules that may regulate vascular remodeling. Second, platelet-derived microparticles released into the circulation can be acquired by vascular endothelial cells through the process of endocytosis. Third, platelets are highly enriched in mitochondria that may contribute to the local reactive oxygen species pool and remodel phospholipids in the plasma membrane of blood vessels. Lastly, platelets are enriched in proteins and phosphoproteins which can be secreted independent of stimulation by surface receptor agonists in conditions of disturbed blood flow. This so-called biomechanical platelet activation occurs in regions of pathologically narrowed (atherosclerotic) or dilated (aneurysmal) vessels. Emerging evidence suggests platelets may regulate the process of angiogenesis and blood flow to tumors as well as education of distant organs for the purposes of allograft health following transplantation. This review will illustrate the potential of platelets to remodel blood vessels in various diseases with a focus on the aforementioned mechanisms.
Collapse
Affiliation(s)
- Anu Aggarwal
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio
| | - Courtney L. Jennings
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio
| | - Emily Manning
- Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Scott J. Cameron
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, Ohio
- Heart Vascular and Thoracic Institute, Department of Cardiovascular Medicine, Section of Vascular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Hematology, Taussig Cancer Center, Cleveland, Ohio
| |
Collapse
|
32
|
Alsaidan AA, Al‐Kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Alsayed KA, Saad HM, Batiha GE. The potential role of SARS-CoV-2 infection in acute coronary syndrome and type 2 myocardial infarction (T2MI): Intertwining spread. Immun Inflamm Dis 2023; 11:e798. [PMID: 36988260 PMCID: PMC10022425 DOI: 10.1002/iid3.798] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/19/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a novel pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It has been shown that SARS-CoV-2 infection-induced inflammatory and oxidative stress and associated endothelial dysfunction may lead to the development of acute coronary syndrome (ACS). Therefore, this review aimed to ascertain the link between severe SARS-CoV-2 infection and ACS. ACS is a spectrum of acute myocardial ischemia due to a sudden decrease in coronary blood flow, ranging from unstable angina to myocardial infarction (MI). Primary or type 1 MI (T1MI) is mainly caused by coronary plaque rupture and/or erosion with subsequent occlusive thrombosis. Secondary or type 2 MI (T2MI) is due to cardiac and systemic disorders without acute coronary atherothrombotic disruption. Acute SARS-CoV-2 infection is linked with the development of nonobstructive coronary disorders such as coronary vasospasm, dilated cardiomyopathy, myocardial fibrosis, and myocarditis. Furthermore, SARS-CoV-2 infection is associated with systemic inflammation that might affect coronary atherosclerotic plaque stability through augmentation of cardiac preload and afterload. Nevertheless, major coronary vessels with atherosclerotic plaques develop minor inflammation during COVID-19 since coronary arteries are not initially and primarily targeted by SARS-CoV-2 due to low expression of angiotensin-converting enzyme 2 in coronary vessels. In conclusion, SARS-CoV-2 infection through hypercytokinemia, direct cardiomyocyte injury, and dysregulation of the renin-angiotensin system may aggravate underlying ACS or cause new-onset T2MI. As well, arrhythmias induced by anti-COVID-19 medications could worsen underlying ACS.
Collapse
Affiliation(s)
- Aseel Awad Alsaidan
- Department of Family and Community Medicine, College of MedicineJouf UniversitySakakaSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineALmustansiriyia UniversityBaghdadIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- Department of Research and DevelopmentAFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐Herdecke, Heusnerstrasse 40University of Witten‐HerdeckeWuppertalGermany
| | - Khalid Adel Alsayed
- Department of Family and Community MedicineSecurity Forces Hospital ProgramRiyadhSaudi Arabia
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityAlBeheiraEgypt
| |
Collapse
|
33
|
Matrix metallopeptidase-9 prognostic role in STEMI patients after percutaneous coronary intervention (PCI) in one-year follow-up period. COR ET VASA 2023. [DOI: 10.33678/cor.2022.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
34
|
Millar B, de Gaetano M. Posing the rationale for synthetic lipoxin mimetics as an adjuvant treatment to gold standard atherosclerosis therapies. Front Pharmacol 2023; 14:1125858. [PMID: 36865918 PMCID: PMC9971729 DOI: 10.3389/fphar.2023.1125858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Atherosclerosis is a progressive, multifactorial inflammatory, and dyslipidaemic disease, responsible for the majority of cardiovascular diseases globally. The chronic inflammation is the main driver of the initiation and progression of such disease, as a result of an imbalanced lipid metabolism and an ineffective immune response to attenuate the inflammatory component. The importance of inflammation resolution is being increasingly recognised in atherosclerosis and cardiovascular disease. It has a complex mechanism consisting of multiple stages, including restoring an effective removal of apoptotic bodies (efferocytosis) and their degradation (effero-metabolism), a macrophage phenotype switching towards resolving phenotypes, and the promotion of tissue healing and regeneration. The low-grade inflammation associated with atherosclerosis development is a driving force in disease exacerbation, and hence inflammation resolution is a key area of research. In this review, we explore the complex disease pathogenesis and its many contributing factors to gain a greater understanding of the disease and identify the current and potential therapeutic targets. First-line treatments and their efficacy will also be discussed in detail, to highlight the emerging field of resolution pharmacology. Despite the great efforts made by current gold-standard treatments, such as lipid-lowering and glucose-lowering drugs, they remain ineffective at tackling residual inflammatory risk and residual cholesterol risk. Resolution pharmacology represents a new era of atherosclerosis therapy, as endogenous ligands associated with inflammation resolution are exploited for their pharmacological benefits in a more potent and longer-acting manner. Novel FPR2-agonists, such as synthetic lipoxin analogues, provide an exciting new approach to enhance the pro-resolving response of the immune system and subsequently end the pro-inflammatory response to allow for an anti-inflammatory and pro-resolving environment for tissue healing, regeneration, and return to homeostasis.
Collapse
Affiliation(s)
| | - Monica de Gaetano
- Diabetes Complications Research Centre, Conway Institute & School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
35
|
Artha IMJR, Bakta IM, Manuaba IBP, Wita IW, Rohman MS, Astawa INM, Bhargah A. The Effects of Percutaneous Coronary Intervention on Biomarkers and Quality of Life in Patients With Chronic Total Coronary Artery Obstruction. Cardiol Res 2023; 14:69-78. [PMID: 36896223 PMCID: PMC9990544 DOI: 10.14740/cr1455] [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: 11/24/2022] [Accepted: 02/02/2023] [Indexed: 02/27/2023] Open
Abstract
Background Chronic total occlusion (CTO) is an angiographic picture of total occlusion without blood flow which is estimated to have lasted at least 3 months. This study attempted to provide an overview of the levels of matrix metalloproteinase-9 (MMP-9), soluble suppression tumorigenicity 2 (sST2), and N-terminal pro-B-type natriuretic peptide (NT-pro-BNP) as remodeling, inflammatory, and atherosclerotic markers, as well as changes in the angina severity in patients with CTO who underwent percutaneous coronary intervention (PCI) compared to those without PCI. Methods This study is a preliminary report with quasi-experimental design study with a pre-test and post-test approach to compare PCI's effect in CTO patients towards changes in MMP-9, sST2, NT-pro-BNP levels, and changes in the angina severity. Twenty subjects underwent PCI and 20 subjects with optimal medical therapy, who were then assessed at baseline and 8 weeks after intervention. Results The results of this preliminary report showed that decreased MMP-9 (pre-test: 12.07 ± 1.27 ng/mL vs. post-test: 9.91 ± 5.19 ng/mL, P = 0.049), sST2 (pre-test: 37.65 ± 20.00 ng/mL vs. post-test: 29.74 ± 15.17 ng/mL, P = 0.026) and NT-pro-BNP (pre-test: 0.63 ± 0.23 ng/mL vs. post-test: 0.24 ± 0.10 ng/mL, P < 0.001) levels were found after 8 weeks of PCI compared to those without such intervention. The levels of NT-pro-BNP were lower in the PCI group (0.24 ± 0.10 ng/mL) than in the non-PCI group (0.56 ± 0.23 ng/mL; P < 0.001). Moreover, there was an improvement of angina severity in PCI group than without PCI (P < 0.039). Conclusions Although this preliminary report found a significant decrease in MMP-9, NT-pro-BNP, and sST2 levels in CTO patients who had undergone PCI compared to those without PCI, as well as improved angina severity in these patients, this study still has limitations. The number of samples was so small that similar studies with larger sample sizes or multicenter investigations are required to deliver more trustworthy and useful results. Nevertheless, we encourage this study as a preliminary baseline for further studies in the future.
Collapse
Affiliation(s)
- I Made Junior Rina Artha
- Cardiology and Vascular Medicine Department, Faculty of Medicine, Universitas Udayana-Prof. I.G.N.G. Ngoerah General Hospital, Denpasar, Bali, Indonesia
| | - I Made Bakta
- Internal Medicine Department, Faculty of Medicine Universitas Udayana-Prof. I.G.N.G. Ngoerah General Hospital, Denpasar, Bali, Indonesia
| | | | - I Wayan Wita
- Cardiology and Vascular Medicine Department, Faculty of Medicine, Universitas Udayana-Prof. I.G.N.G. Ngoerah General Hospital, Denpasar, Bali, Indonesia
| | - Mohammad Saifur Rohman
- Department of Cardiology, Universitas Brawijaya-Saiful Anwar Hospital, Malang, Indonesia
| | - I Nyoman Mantik Astawa
- Virology Laboratory, Department of Animal Disease, Faculty of Veterinary Medicine, Universitas Udayana, Bali, Indonesia
| | - Agha Bhargah
- Cardiology and Vascular Medicine Department, Faculty of Medicine, Universitas Udayana-Prof. I.G.N.G. Ngoerah General Hospital, Denpasar, Bali, Indonesia
| |
Collapse
|
36
|
The Role of Neutrophils in Lower Limb Peripheral Artery Disease: State of the Art and Future Perspectives. Int J Mol Sci 2023; 24:ijms24021169. [PMID: 36674682 PMCID: PMC9866688 DOI: 10.3390/ijms24021169] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
In recent years, increasing attention has been paid to the role of neutrophils in cardiovascular (CV) disease (CVD) with evidence supporting their role in the initiation, progression, and rupture of atherosclerotic plaque. Although these cells have long been considered as terminally differentiated cells with a relatively limited spectrum of action, recent research has revealed intriguing novel cellular functions, including neutrophil extracellular trap (NET) generation and inflammasome activation, which have been linked to several human diseases, including CVD. While most research to date has focused on the role of neutrophils in coronary artery and cerebrovascular diseases, much less information is available on lower limb peripheral artery disease (PAD). PAD is a widespread condition associated with great morbidity and mortality, though physician and patient awareness of the disease remains low. To date, several studies have produced some evidence on the role of certain biomarkers of neutrophil activation in this clinical setting. However, the etiopathogenetic role of neutrophils, and in particular of some of the newly discovered mechanisms, has yet to be fully elucidated. In the future, complementary assessment of neutrophil activity should improve CV risk stratification and provide personalized treatments to patients with PAD. This review aims to summarize the basic principles and recent advances in the understanding of neutrophil biology, current knowledge about the role of neutrophils in atherosclerosis, as well as available evidence on their role of PAD.
Collapse
|
37
|
Bao H, Li J, Zhang B, Huang J, Su D, Liu L. Integrated bioinformatics and machine-learning screening for immune-related genes in diagnosing non-alcoholic fatty liver disease with ischemic stroke and RRS1 pan-cancer analysis. Front Immunol 2023; 14:1113634. [PMID: 37090698 PMCID: PMC10115222 DOI: 10.3389/fimmu.2023.1113634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Background The occurrence of ischemic stroke (IS) is associated with nonalcoholic fatty liver disease (NAFLD). The cancer burden of NAFLD complicated by IS also warrants attention. This study aimed to identify candidate immune biomarkers linked to NAFLD and IS and analyze their association with cancer. Methods Two of each of the NAFLD and IS datasets were downloaded, differentially expressed genes (DEGs) were identified, and module genes were screened via weighted gene coexpression network analysis (WGCNA). Subsequently, utilizing machine learning (least absolute shrinkage and selection operator regression, random forest and support vector machine-recursive feature elimination) and immune cell infiltration analysis, immune-related candidate biomarkers for NAFLD with IS were determined. Simultaneously, a nomogram was established, the diagnostic efficacy was assessed, and the role of candidate biomarkers in cancer was ascertained through pan-cancer analyses. Results In this study, 117 and 98 DEGs were identified from the combined NAFLD and IS datasets, respectively, and 279 genes were obtained from the most significant modules of NAFLD. NAFLD module genes and IS DEGs were intersected to obtain nine genes, which were enriched in the inflammatory response and immune regulation. After overlapping the results of the three machine learning algorithms, six candidate genes were obtained, based on which a nomogram was constructed. The calibration curve demonstrated good accuracy, and the candidate genes had high diagnostic values. The genes were found to be related to the immune dysregulation of stroke, and RRS1 was strongly associated with the prognosis, immune cell infiltration, microsatellite instability (MSI), and tumor mutation burden (TMB). Conclusion Six common candidate immune-related genes (PTGS2, FCGR1A, MMP9, VNN3, S100A12, and RRS1) of NAFLD and IS were identified, and a nomogram for diagnosing NAFLD with IS was established. RRS1 may serve as a candidate gene for predicting the prognosis of patients with cancer who have NAFLD complicated by IS, which could aid in their diagnosis and treatment.
Collapse
Affiliation(s)
- Huayan Bao
- Department of Medical Imaging Center; Guangxi Key Clinical Specialty (Medical Imaging Department); Dominant Cultivation Discipline of Guangxi Medical University Cancer Hospital (Medical Imaging Department), Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jianwen Li
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Boyang Zhang
- Department of Medical Imaging Center; Guangxi Key Clinical Specialty (Medical Imaging Department); Dominant Cultivation Discipline of Guangxi Medical University Cancer Hospital (Medical Imaging Department), Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ju Huang
- Department of Medical Imaging Center; Guangxi Key Clinical Specialty (Medical Imaging Department); Dominant Cultivation Discipline of Guangxi Medical University Cancer Hospital (Medical Imaging Department), Guangxi Medical University Cancer Hospital, Nanning, China
| | - Danke Su
- Department of Medical Imaging Center; Guangxi Key Clinical Specialty (Medical Imaging Department); Dominant Cultivation Discipline of Guangxi Medical University Cancer Hospital (Medical Imaging Department), Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Danke Su, ; Lidong Liu,
| | - Lidong Liu
- Department of Medical Imaging Center; Guangxi Key Clinical Specialty (Medical Imaging Department); Dominant Cultivation Discipline of Guangxi Medical University Cancer Hospital (Medical Imaging Department), Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Danke Su, ; Lidong Liu,
| |
Collapse
|
38
|
Seim BE, Holt MF, Ratajska A, Michelsen A, Ringseth MM, Halvorsen BE, Skjelland M, Kvitting JPE, Lundblad R, Krohg-Sørensen K, Osnes LTN, Aukrust P, Paus B, Ueland T. Markers of extracellular matrix remodeling and systemic inflammation in patients with heritable thoracic aortic diseases. Front Cardiovasc Med 2022; 9:1073069. [PMID: 36606286 PMCID: PMC9808784 DOI: 10.3389/fcvm.2022.1073069] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/29/2022] [Indexed: 01/07/2023] Open
Abstract
Background In approximately 20% of patients with thoracic aortic aneurysms or dissections a heritable thoracic aortic disease (HTAD) is suspected. Several monogenic connective tissue diseases imply high risk of aortic disease, including both non-syndromic and syndromic forms. There are some studies assessing inflammation and extracellular matrix remodeling in patients with non-hereditary aortic disease, but such studies in patients with hereditary diseases are scarce. Aims To quantify markers of extracellular matrix (ECM) and inflammation in patients with vascular connective tissue diseases versus healthy controls. Methods Patients with Loeys-Dietz syndrome (LDS, n = 12), Marfan syndrome (MFS, n = 11), and familial thoracic aortic aneurysm 6 (FTAA6, n = 9), i.e., actin alpha 2 (ACTA2) pathogenic variants, were recruited. Exome or genome sequencing was performed for genetic diagnosis. Several markers of inflammation and ECM remodeling were measured in plasma by enzyme immunoassays. Flow cytometry of T-cell subpopulations was performed on a subgroup of patients. For comparison, blood samples were drawn from 14 healthy controls. Results (i) All groups of HTAD patients had increased levels matrix metalloproteinase-9 (MMP-9) as compared with healthy controls, also in adjusted analyses, reflecting altered ECM remodeling. (ii) LDS patients had increased levels of pentraxin 3 (PTX3), reflecting systemic inflammation. (iii) LDS patients have increased levels of soluble CD25, a marker of T-cell activation. Conclusion Our data suggest that upregulated MMP-9, a matrix degrading enzyme, is a common feature of several subgroups of HTAD. In addition, LDS patients have increased levels of PTX3 reflecting systemic and in particular vascular inflammation.
Collapse
Affiliation(s)
- Bjørn Edvard Seim
- Department of Cardiothoracic Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Margrethe Flesvig Holt
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway,Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway,Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,*Correspondence: Margrethe Flesvig Holt,
| | | | - Annika Michelsen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway,Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Bente Evy Halvorsen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway,Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Mona Skjelland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway,Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - John-Peder Escobar Kvitting
- Department of Cardiothoracic Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Runar Lundblad
- Department of Cardiothoracic Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kirsten Krohg-Sørensen
- Department of Cardiothoracic Surgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Liv T. N. Osnes
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway,Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway,Faculty of Health Sciences, K. G. Jebsen Thrombosis Research Center, University of Tromsø – The Arctic University of Norway, Tromsø, Norway
| | - Benedicte Paus
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway,Faculty of Health Sciences, K. G. Jebsen Thrombosis Research Center, University of Tromsø – The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
39
|
NATH D, SHIVASEKAR M, VINODHINI V. Smoking Induces the Circulating Levels of Matrix Metalloproteinase-9 and Its Association with Cardiovascular Risk in Young Smokers. Medeni Med J 2022; 37:306-312. [PMID: 36578139 PMCID: PMC9808855 DOI: 10.4274/mmj.galenos.2022.45057] [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] [Indexed: 11/07/2022] Open
Abstract
Objective Smoking causes cardiovascular risk, which may alter the stability between the production and degradation of the extracellular matrix. Matrix metalloproteinase-9 (MMP-9) is a zinc-containing endopeptidase that degrades the extracellular matrix and is involved in tissue remodelling and several physiological processes. As a result, smoking-induced elevated serum MMP-9 levels, particularly at a younger age, raise the risk of coronary heart disease (CHD). Thus, this study aimed to determine the possible relationship between smoking-induced circulating MMP-9 and the risk of cardiovascular disease in young smokers. Methods In this cross-sectional study, the patients were divided into three groups. Each group contains 120 study participants. Group one consisted of 120 healthy individuals with no physical and mental illness, group two consisted of 120 active smokers with a heart disease, and group three consisted of 120 active smokers with a heart disease and diabetes, who attended Sri Ramaswamy Memorial Hospital for cardiology checkup at the age of 20-55 years. The serum MMP-9, high-sensitivity C-reactive protein (hs-CRP), and apolipoprotein-E (APO-E) levels were analyzed using the ELISA method, and the lipid levels were measured enzymatically using AU480 automatic analyzer (Beckman Coulter). Results Compared with non-smokers, the study shows that the mean serum MMP-9, hs-CRP, and APO-E levels were significantly higher in smokers (p<0.001). A strong relationship was also found between MMP-9 and hs-CRP, APO-E, smoking load, and smoking intensity. Conclusions A significant association was found between cigarette smoking with MMP-9, and relative exposure to circulating inflammation markers plays a potential role in the pathogenesis of CHD.
Collapse
Affiliation(s)
- Dinesh NATH
- SRM Medical College Hospital and Research Centre, SRMIST, Department of Biochemistry, Kattankulthur, Tamil Nadu, India
| | - Meera SHIVASEKAR
- SRM Medical College Hospital and Research Centre, SRMIST, Department of Biochemistry, Kattankulthur, Tamil Nadu, India,* Address for Correspondence: SRM Medical College Hospital and Research Centre, SRMIST, Department of Biochemistry, Kattankulthur, Tamil Nadu, India E-mail:
| | - V.M. VINODHINI
- SRM Medical College Hospital and Research Centre, SRMIST, Department of Biochemistry, Kattankulthur, Tamil Nadu, India
| |
Collapse
|
40
|
Chiorescu RM, Mocan M, Inceu AI, Buda AP, Blendea D, Vlaicu SI. Vulnerable Atherosclerotic Plaque: Is There a Molecular Signature? Int J Mol Sci 2022; 23:13638. [PMID: 36362423 PMCID: PMC9656166 DOI: 10.3390/ijms232113638] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2023] Open
Abstract
Atherosclerosis and its clinical manifestations, coronary and cerebral artery diseases, are the most common cause of death worldwide. The main pathophysiological mechanism for these complications is the rupture of vulnerable atherosclerotic plaques and subsequent thrombosis. Pathological studies of the vulnerable lesions showed that more frequently, plaques rich in lipids and with a high level of inflammation, responsible for mild or moderate stenosis, are more prone to rupture, leading to acute events. Identifying the vulnerable plaques helps to stratify patients at risk of developing acute vascular events. Traditional imaging methods based on plaque appearance and size are not reliable in prediction the risk of rupture. Intravascular imaging is a novel technique able to identify vulnerable lesions, but it is invasive and an operator-dependent technique. This review aims to summarize the current data from literature regarding the main biomarkers involved in the attempt to diagnose vulnerable atherosclerotic lesions. These biomarkers could be the base for risk stratification and development of the new therapeutic drugs in the treatment of patients with vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Roxana Mihaela Chiorescu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Mihaela Mocan
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Andreea Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine, 400349 Cluj-Napoca, Romania
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Andreea Paula Buda
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Dan Blendea
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
- Department of Cardiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400437 Cluj-Napoca, Romania
| | - Sonia Irina Vlaicu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| |
Collapse
|
41
|
Liu T, Ji H, Jian X, Wang W, Fan Z. Novel nomogram for predicting coronary vulnerable plaque risk in patients with coronary artery disease. Biomark Med 2022; 16:1139-1149. [PMID: 36625283 DOI: 10.2217/bmm-2022-0855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Objective: To develop and validate a nomogram for predicting coronary vulnerable plaques (VPs) in coronary artery disease (CAD) patients. Methods: One hundred seventy-seven CAD patients were enrolled in the training group. Another 60 patients were included for validation. Based on the identified independent risk factors, a nomogram model was developed and then validated. Results: Type 2 diabetes, hypertension, neutrophil-to-lymphocyte ratio, low-density lipoprotein cholesterol, MCP-1 and MMP-9 were found to be independent risk factors for coronary VPs. Both internal and external validation showed this nomogram had satisfactory discrimination via receiver operating characteristic curves, calibration via calibration plots and clinical application values via decision curve analysis. Conclusion: The authors established a nomogram model predicting coronary VP risk in CAD patients with promising clinical application value.
Collapse
Affiliation(s)
- Tao Liu
- Department of Cardiovascular Diseases, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, Beijing, China
| | - Hanhua Ji
- Department of Cardiovascular Diseases, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, Beijing, China
| | - Xinwen Jian
- Department of Cardiovascular Diseases, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, Beijing, China
| | - Weiyi Wang
- Department of Cardiovascular Diseases, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, Beijing, China
| | - Zeyuan Fan
- Department of Cardiovascular Diseases, Civil Aviation General Hospital, Civil Aviation Clinical Medical College of Peking University, Beijing, China
| |
Collapse
|
42
|
Liu D, Ghani D, Wain J, Szeto WY, Laudanski K. Concomitant elevated serum levels of tenascin, MMP-9 and YKL-40, suggest ongoing remodeling of the heart up to 3 months after cardiac surgery after normalization of the revascularization markers. Eur J Med Res 2022; 27:208. [PMID: 36271425 PMCID: PMC9585873 DOI: 10.1186/s40001-022-00831-8] [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: 08/31/2022] [Accepted: 09/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The recovery from cardiac surgery involves resolving inflammation and remodeling with significant connective tissue turnover. Dynamics of smoldering inflammation and injury (white blood cells, platelets, CRP, IL-8, IL-6), vascular inflammation (IL-15, VEGF, RANTES), connective tissue remodeling (tenascin, MMP-9), cardiac injury and remodeling (YKL-40), and vascular remodeling (epiregulin, MCP-1, VEGF) were assessed up to 3 months after cardiac surgery. We hypothesize that at 3 months, studied markers will return to pre-surgical levels. METHODS Patients (n = 139) scheduled for non-emergent heart surgery were included, except for patients with pre-existing immunological aberrancies. Blood was collected before surgery(tbaseline), 24 h later(t24h) after the first sample, 7 days(t7d), and 3 months(t3m) after tbaseline. Serum markers were measured via multiplex or ELISA. Electronic medical records (EMR) were used to extract demographical, pre-existing conditions and clinical data. Disposition (discharge home, discharge to facility, death, re-admission) was determined at 28 days and 3 months from admission. RESULTS Not all inflammatory markers returned to baseline (CRP↑↑, leukocytosis, thrombocytosis, IL-8↓, IL-6↓). Tenascin and YKL-40 levels remained elevated even at t3m. YKL-40 serum levels were significantly elevated at t24h and t7d while normalized at t3m. VEGF returned to the baseline, yet MCP-1 remained elevated at 3 months. CCL28 increased at 3 months, while RANTES and IL-15 declined at the same time. Disposition at discharge was determined by serum MMP-9, while YKL-40 correlated with duration of surgery and APACHE II24h. CONCLUSIONS The data demonstrated an ongoing extracellular matrix turnover at 3 months, while acute inflammation and vascular remodeling resolved only partially.
Collapse
Affiliation(s)
- Da Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Danyal Ghani
- College of Art and Sciences, Drexel University, Philadelphia, PA, USA
| | - Justin Wain
- Campbell University School of Osteopathic Medicine, Buies Creek, NC, USA
| | - Wilson Y Szeto
- Department of Cardiac Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA. .,Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA. .,Leonard Davis Institute for Health Economics, University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA.
| |
Collapse
|
43
|
Caimi G, Urso C, Brucculeri S, Amato C, Lo Presti R, Carlisi M. Leukocyte subtypes, gelatinases, and their tissue inhibitors in a group of subjects with asymptomatic carotid atherosclerosis. Clin Hemorheol Microcirc 2022; 82:37-51. [PMID: 35599473 PMCID: PMC9741743 DOI: 10.3233/ch-221380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In a cohort of subjects with asymptomatic carotid atherosclerosis (ACA), we have evaluated the neutrophil and lymphocyte count and their ratio (NLR), the gelatinases (MMP-2 and MMP-9) and their tissue inhibitors (TIMP-1 and TIMP-2). At baseline, no difference was observed between ACA subjects and subject control group regarding neutrophil and lymphocyte count while was evident in ACA subjects a significant increase in MMP-2, MMP-9 and TIMP-2 associated to a significant decrease in TIMP-1. Dividing the ACA according to the number of cardiovascular risk factors (CRFs) we have observed an increase in lymphocyte count in the subgroup with 3-5 CRFs. Evaluating the leukocyte subtypes according to all the surrogate markers of insulin resistance has been noted, in the subgroups that exceed the medians of these markers, a significant increase in neutrophil and lymphocyte count without any variation of the NLR. Effecting the same evaluation for the MMP/TIMP pattern we observed, instead, that the same subgroups tend to show a decrease in MMP-2 and an increase in MMP-9. No difference instead for TIMP-1 and TIMP-2. The abnormality of the MMP/TIMP pattern, bearing in mind the cardiometabolic clustering present in this cohort of ACA subjects, would induce to use drugs able not only to cure the cardiometabolic risk factors but also to influence the MMP/TIMP profile.
Collapse
Affiliation(s)
- Gregorio Caimi
- Department of Health Promotion and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy,Corresponding author: Gregorio Caimi, Via Leonardo Da Vinci, 52, 90145, Palermo, Italy. E-mails: ;
| | - C. Urso
- Fondazione Istituto “G. Giglio” Cefalù, Palermo, Italy
| | - S. Brucculeri
- Fondazione Istituto “G. Giglio” Cefalù, Palermo, Italy
| | - C. Amato
- Department of Health Promotion and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - R. Lo Presti
- Department of Psychology, Educational Science and Human Movement, Università degli Studi di Palermo, Palermo, Italy
| | - M. Carlisi
- Department of Health Promotion and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| |
Collapse
|
44
|
Nordeng J, Schandiz H, Solheim S, Åkra S, Hoffman P, Roald B, Bendz B, Arnesen H, Helseth R, Seljeflot I. TIMP-1 expression in coronary thrombi associate with myocardial injury in ST-elevation myocardial infarction patients. Coron Artery Dis 2022; 33:446-455. [PMID: 35102064 DOI: 10.1097/mca.0000000000001128] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) are considered important both in atherosclerosis and remodeling after acute myocardial infarction (AMI). We aimed to study genetic expression and presence of MMP-2, MMP-9, TIMP-1, TIMP-2 and the extracellular MMP-inducer (EMMPRIN) in coronary thrombi. Circulating levels and genetic expression in circulating leukocytes were also assessed, and relations to degree of myocardial injury measured by troponin T and time from symptom to PCI were explored. Expression of cell markers were also analyzed, indicating relations to cell types. METHODS Intracoronary thrombi were aspirated from 33 patients with ST-elevation myocardial infarction (STEMI). Blood samples with Pax-gene tubes were drawn at end of PCI and the next day. RNA was isolated from thrombi and leukocytes, and genes were relatively quantified by RT-PCR. Each thrombus was preserved for histology and immunohistochemistry analyzes. RESULTS Genes coding for the five markers were present in 84-100% of thrombi and immunohistochemically stained in 96-100%. Expression of TIMP-1 in thrombi and in leukocytes correlated significantly to peak troponin T ( r = 0.393 P = 0.026, r = 0.469 P = 0.006, respectively). No significant correlations between genes expressed in thrombi and time from symptom to PCI were observed. TIMP-1 was connected mainly to monocytes/macrophages in the thrombi. CONCLUSION MMP-2, MMP-9, TIMP-1, TIMP-2 and EMMPRIN were highly expressed in human coronary thrombi. The correlation between troponin T and the expression of TIMP-1 both in thrombi and in leukocytes at time of PCI indicates that TIMP-1 plays a role in myocardial damage early post-MI.
Collapse
Affiliation(s)
- Jostein Nordeng
- Center for Clinical Heart Research Oslo University Hospital Ullevål
- Department of Cardiology, Oslo University Hospital Ullevål
- Faculty of Medicine, University of Oslo
| | | | - Svein Solheim
- Center for Clinical Heart Research Oslo University Hospital Ullevål
- Department of Cardiology, Oslo University Hospital Ullevål
| | - Sissel Åkra
- Center for Clinical Heart Research Oslo University Hospital Ullevål
| | - Pavel Hoffman
- Section for Interventional Cardiology, Department of Cardiology, Oslo University Hospital Ullevål
| | - Borghild Roald
- Faculty of Medicine, University of Oslo
- Department of Pathology, Oslo University Hospital Ullevål
| | - Bjørn Bendz
- Faculty of Medicine, University of Oslo
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Harald Arnesen
- Center for Clinical Heart Research Oslo University Hospital Ullevål
- Faculty of Medicine, University of Oslo
| | - Ragnhild Helseth
- Center for Clinical Heart Research Oslo University Hospital Ullevål
- Department of Cardiology, Oslo University Hospital Ullevål
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research Oslo University Hospital Ullevål
- Department of Cardiology, Oslo University Hospital Ullevål
- Faculty of Medicine, University of Oslo
| |
Collapse
|
45
|
Electronic Cigarette and Atherosclerosis: A Comprehensive Literature Review of Latest Evidences. Int J Vasc Med 2022; 2022:4136811. [PMID: 36093338 PMCID: PMC9453087 DOI: 10.1155/2022/4136811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/29/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022] Open
Abstract
Coronary artery diseases (CAD), also known as coronary heart disease (CHD), are the world’s leading cause of death. The basis of coronary artery disease is the narrowing of the heart coronary artery lumen due to atherosclerosis. The use of electronic cigarettes has increased significantly over the years. However, harmful effects of electronic cigarettes are still not firm. The aim of this article is to review the impact of electronic cigarette and its role in the pathogenesis of atherosclerosis from recent studies. The results showed that several chemical compounds, such as nicotine, propylene glycol, particulate matters, heavy metals, and flavorings, in electronic cigarette induce atherosclerosis with each molecular mechanism that lead to atherosclerosis progression by formation of ROS, endothelial dysfunction, and inflammation. Further research is still needed to determine the exact mechanism and provide more clinical evidence.
Collapse
|
46
|
Wang Y, Chuang CY, Hawkins CL, Davies MJ. Activation and Inhibition of Human Matrix Metalloproteinase-9 (MMP9) by HOCl, Myeloperoxidase and Chloramines. Antioxidants (Basel) 2022; 11:antiox11081616. [PMID: 36009335 PMCID: PMC9405048 DOI: 10.3390/antiox11081616] [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: 07/25/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022] Open
Abstract
Matrix metalloproteinase-9 (MMP9, gelatinase B) plays a key role in the degradation of extracellular-matrix (ECM) proteins in both normal physiology and multiple pathologies, including those linked with inflammation. MMP9 is excreted as an inactive proform (proMMP9) by multiple cells, and particularly neutrophils. The proenzyme undergoes subsequent processing to active forms, either enzymatically (e.g., via plasmin and stromelysin-1/MMP3), or via the oxidation of a cysteine residue in the prodomain (the “cysteine-switch”). Activated leukocytes, including neutrophils, generate O2− and H2O2 and release myeloperoxidase (MPO), which catalyzes hypochlorous acid (HOCl) formation. Here, we examine the reactivity of HOCl and a range of low-molecular-mass and protein chloramines with the pro- and activated forms of MMP9. HOCl and an enzymatic MPO/H2O2/Cl− system were able to generate active MMP9, as determined by fluorescence-activity assays and gel zymography. The inactivation of active MMP9 also occurred at high HOCl concentrations. Low (nM—low μM) concentrations of chloramines formed by the reaction of HOCl with amino acids (taurine, lysine, histidine), serum albumin, ECM proteins (laminin and fibronectin) and basement membrane extracts (but not HEPES chloramines) also activate proMMP9. This activation is diminished by the competitive HOCl-reactive species, methionine. These data indicate that HOCl-mediated oxidation and MMP-mediated ECM degradation are synergistic and interdependent. As previous studies have shown that modified ECM proteins can also stimulate the cellular expression of MMP proteins, these processes may contribute to a vicious cycle of increasing ECM degradation during disease development.
Collapse
Affiliation(s)
- Yihe Wang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christine Y Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
47
|
Gkaliagkousi E, Lazaridis A, Dogan S, Fraenkel E, Tuna BG, Mozos I, Vukicevic M, Yalcin O, Gopcevic K. Theories and Molecular Basis of Vascular Aging: A Review of the Literature from VascAgeNet Group on Pathophysiological Mechanisms of Vascular Aging. Int J Mol Sci 2022; 23:ijms23158672. [PMID: 35955804 PMCID: PMC9368987 DOI: 10.3390/ijms23158672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Vascular aging, characterized by structural and functional alterations of the vascular wall, is a hallmark of aging and is tightly related to the development of cardiovascular mortality and age-associated vascular pathologies. Over the last years, extensive and ongoing research has highlighted several sophisticated molecular mechanisms that are involved in the pathophysiology of vascular aging. A more thorough understanding of these mechanisms could help to provide a new insight into the complex biology of this non-reversible vascular process and direct future interventions to improve longevity. In this review, we discuss the role of the most important molecular pathways involved in vascular ageing including oxidative stress, vascular inflammation, extracellular matrix metalloproteinases activity, epigenetic regulation, telomere shortening, senescence and autophagy.
Collapse
Affiliation(s)
- Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece
- Correspondence: (E.G.); (K.G.)
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Faculty of Medicine, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, 34755 Istanbul, Turkey
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital, Pavol Jozef Šafárik University of Košice, Trieda SNP 1, 04066 Košice, Slovakia
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, 34755 Istanbul, Turkey
| | - Ioana Mozos
- Department of Functional Sciences-Pathophysiology, Center for Translational Research and Systems Medicine, “Victor Babes” University of Medicine and Pharmacy, 300173 Timisoara, Romania
| | - Milica Vukicevic
- Cardiac Surgery Clinic, Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Ozlem Yalcin
- Department of Physiology, School of Medicine, Koc University, 34450 Istanbul, Turkey
| | - Kristina Gopcevic
- Laboratory for Analytics of Biomolecules, Department of Chemistry in Medicine, Faculty of Medicine, 11000 Belgrade, Serbia
- Correspondence: (E.G.); (K.G.)
| |
Collapse
|
48
|
Saleh S, George J, Kott KA, Meikle PJ, Figtree GA. The Translation and Commercialisation of Biomarkers for Cardiovascular Disease—A Review. Front Cardiovasc Med 2022; 9:897106. [PMID: 35722087 PMCID: PMC9201254 DOI: 10.3389/fcvm.2022.897106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
As a leading cause of mortality and morbidity worldwide, cardiovascular disease and its diagnosis, quantification, and stratification remain significant health issues. Increasingly, patients present with cardiovascular disease in the absence of known risk factors, suggesting the presence of yet unrecognized pathological processes and disease predispositions. Fortunately, a host of emerging cardiovascular biomarkers characterizing and quantifying ischaemic heart disease have shown great promise in both laboratory settings and clinical trials. These have demonstrated improved predictive value additional to widely accepted biomarkers as well as providing insight into molecular phenotypes beneath the broad umbrella of cardiovascular disease that may allow for further personalized treatment regimens. However, the process of translation into clinical practice – particularly navigating the legal and commercial landscape – poses a number of challenges. Practical and legal barriers to the biomarker translational pipeline must be further considered to develop strategies to bring novel biomarkers into the clinical sphere and apply these advances at the patient bedside. Here we review the progress of emerging biomarkers in the cardiovascular space, with particular focus on those relevant to the unmet needs in ischaemic heart disease.
Collapse
Affiliation(s)
- Soloman Saleh
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jacob George
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Katharine A. Kott
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Gemma A. Figtree
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Gemma A. Figtree
| |
Collapse
|
49
|
The Effect of Chronic Immunosuppressive Regimens Treatment on Aortal Media Morphology and the Balance between Matrix Metalloproteinases (mmp-2 and mmp-9) and Their Inhibitors in the Abdominal Aorta of Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19116399. [PMID: 35681984 PMCID: PMC9180580 DOI: 10.3390/ijerph19116399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023]
Abstract
Immunosuppressive drugs are widely and chronically used to avoid graft rejection in transplant recipients. However, they are also known to have organotoxic effects and can exert numerous side effects. The aim of this study was to assess whether the chronic treatment of rats with the most commonly used clinical immunosuppressive regimens in organ recipients had an effect on the morphology and function of the aorta. The rats were divided into five groups and each group was chronically treated with different sets of three immunosuppressive drugs (TRG, CRG, MRG, CMG, TMG) for 6 months. The changes were most profound in calcineurin inhibitor-based protocols. TMG protocol treatment was characterized by the most numerous alterations such as morphological changes, changes in the thickness of the tunic media, wider distances between elastic lamellae, an increase in the number of vSMCs and changes in collagen deposition. We concluded that the morphological changes were connected with MMP-2 and MMP-9/TIMP-2 and TIMP-1 imbalances, which was also determined and noticed.
Collapse
|
50
|
Construction and Analysis of lncRNA-Associated ceRNA Network in Atherosclerotic Plaque Formation. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4895611. [PMID: 35463977 PMCID: PMC9033352 DOI: 10.1155/2022/4895611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/05/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022]
Abstract
Atherosclerosis (AS) is a vascular disease with plaque formation. Unstable plaques can be expected to result in cardiovascular disease, such as myocardial infarction and stroke. Studies have verified that long noncoding RNAs (lncRNAs) play a critical role in atherosclerotic plaque formation (APF), including MALAT1, GAS5, and H19. A ceRNA network is a combination of these two interacting processes, which regulate the occurrence and progression of many diseases. However, lncRNA-associated ceRNA network in terms of APF is limited. This study sought to discover novel potential biomarkers and ceRNA network for APF. We designed a triple network based on the lncRNA-miRNA and mRNA-miRNA pairs obtained from lncRNASNP and starBase. Differentially expressed genes (DEGs) and lncRNAs in human vascular tissues derived from the Gene Expression Omnibus database (GSE43292, GSE97210) were systematically selected and analyzed. A ceRNA network was constructed by hypergeometric test, including 8 lncRNAs, 243 miRNAs, and 8 mRNAs. APF-related ceRNA structure was discovered for the first time by combining network analysis and statistical validation. Topological analysis determined the key lncRNAs with the highest centroid. GO and KEGG enrichment analysis indicated that the ceRNA network was primarily enriched in “regulation of platelet-derived growth factor receptor signaling pathway,” “negative regulation of leukocyte chemotaxis,” and “axonal fasciculation.” A functional lncRNA, HAND2-AS1, was identified in the ceRNA network, and the main miRNA (miRNA-570-3p) regulated by HAND2-AS1 was further screened. This present study elucidated the important function of lncRNA in the origination and progression of APF and indicated the potential use of these hub nodes as diagnostic biomarkers and therapeutic targets.
Collapse
|